Morbidity and Mortality Weekly Report
Recommendations and Reports / Vol. 70 / No. 4 July 23, 2021
Sexually Transmitted Infections Treatment
Guidelines, 2021
U.S. Department of Health and Human Services
Centers for Disease Control and Prevention
The MMWR series of publications is published by the Center for Surveillance, Epidemiology, and Laboratory Services, Centers for Disease Control and Prevention (CDC),
U.S. Department of Health and Human Services, Atlanta, GA 30329-4027.
Suggested citation: [Author names; first three, then et al., if more than six.] [Title]. MMWR Recomm Rep 2021;70(No. RR-#):[inclusive page numbers].
Centers for Disease Control and Prevention
Rochelle P. Walensky, MD, MPH, Director
Debra Houry, MD, MPH, Acting Principal Deputy Director
Daniel B. Jernigan, MD, MPH, Acting Deputy Director for Public Health Science and Surveillance
Rebecca Bunnell, PhD, MEd, Director, Office of Science
Jennifer Layden, MD, PhD, Deputy Director, Office of Science
Michael F. Iademarco, MD, MPH, Director, Center for Surveillance, Epidemiology, and Laboratory Services
MMWR Editorial and Production Staff (Serials)
Charlotte K. Kent, PhD, MPH, Editor in Chief
Christine G. Casey, MD, Editor
Mary Dott, MD, MPH, Online Editor
Terisa F. Rutledge, Managing Editor
David C. Johnson, Lead Technical Writer-Editor
Marella Meadows, Project Editor
Martha F. Boyd, Lead Visual Information Specialist
Alexander J. Gottardy, Maureen A. Leahy,
Julia C. Martinroe, Stephen R. Spriggs, Tong Yang,
Visual Information Specialists
Quang M. Doan, MBA, Phyllis H. King,
Terraye M. Starr, Moua Yang,
Information Technology Specialists
MMWR Editorial Board
Timothy F. Jones, MD, Chairman
Matthew L. Boulton, MD, MPH
Carolyn Brooks, ScD, MA
Jay C. Butler, MD
Virginia A. Caine, MD
Jonathan E. Fielding, MD, MPH, MBA
David W. Fleming, MD
William E. Halperin, MD, DrPH, MPH
Jewel Mullen, MD, MPH, MPA
Jeff Niederdeppe, PhD
Celeste Philip, MD, MPH
Patricia Quinlisk, MD, MPH
Patrick L. Remington, MD, MPH
Carlos Roig, MS, MA
William Schaffner, MD
Nathaniel Smith, MD, MPH
Morgan Bobb Swanson, BS
Abbigail Tumpey, MPH
Ian Branam, MA, Acting Lead
Health Communication Specialist
Shelton Bartley, MPH,
Lowery Johnson, Amanda Ray,
Jacqueline N. Sanchez, MS,
Health Communication Specialists
Will Yang, MA,
Visual Information Specialist
Recommendations and Reports
CONTENTS
Introduction ............................................................................................................1
Methods ....................................................................................................................1
Clinical Prevention Guidance ............................................................................ 2
STI Detection Among Special Populations ............................................... 11
HIV Infection ......................................................................................................... 24
Diseases Characterized by Genital, Anal, or Perianal Ulcers ............... 27
Syphilis ................................................................................................................... 39
Management of Persons Who Have a History of Penicillin Allergy .. 56
Diseases Characterized by Urethritis and Cervicitis ............................... 60
Chlamydial Infections ....................................................................................... 65
Gonococcal Infections ...................................................................................... 71
Mycoplasma genitalium .................................................................................... 80
Diseases Characterized by Vulvovaginal Itching, Burning, Irritation,
Odor, or Discharge ........................................................................................... 82
Pelvic Inflammatory Disease .......................................................................... 94
Epididymitis .......................................................................................................... 98
Human Papillomavirus Infections ..............................................................100
Viral Hepatitis ..................................................................................................... 113
Proctitis, Proctocolitis, and Enteritis ..........................................................124
Ectoparasitic Infections .................................................................................. 126
Sexual Assault and Abuse and STIs ............................................................128
References ...........................................................................................................135
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 1
US Department of Health and Human Services/Centers for Disease Control and Prevention
Sexually Transmitted Infections Treatment Guidelines, 2021
Kimberly A. Workowski, MD
1,2
; Laura H. Bachmann, MD
1
; Philip A. Chan, MD
1,3
; Christine M. Johnston, MD
1,4
; Christina A. Muzny, MD
1,5
;
Ina Park, MD
1,6
; Hilary Reno, MD
1,7
; Jonathan M. Zenilman, MD
1,8
; Gail A. Bolan, MD
1
1
Division of STD Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, CDC, Atlanta, Georgia;
2
Emory University, Atlanta, Georgia;
3
Brown University, Providence, Rhode Island;
4
University of Washington, Seattle, Washington;
5
University of Alabama at Birmingham, Birmingham, Alabama;
6
University of California San Francisco, San Francisco, California;
7
Washington University, St. Louis, Missouri;
8
Johns Hopkins University, Baltimore, Maryland
Summary
These guidelines for the treatment of persons who have or are at risk for sexually transmitted infections (STIs) were updated by
CDC after consultation with professionals knowledgeable in the field of STIs who met in Atlanta, Georgia, June 11–14, 2019.
The information in this report updates the 2015 guidelines. These guidelines discuss 1) updated recommendations for treatment of
Neisseria gonorrhoeae, Chlamydia trachomatis, and Trichomonas vaginalis; 2) addition of metronidazole to the recommended
treatment regimen for pelvic inflammatory disease; 3) alternative treatment options for bacterial vaginosis; 4) management of
Mycoplasma genitalium; 5) human papillomavirus vaccine recommendations and counseling messages; 6) expanded risk factors
for syphilis testing among pregnant women; 7) one-time testing for hepatitis C infection; 8) evaluation of men who have sex with
men after sexual assault; and 9) two-step testing for serologic diagnosis of genital herpes simplex virus. Physicians and other health
care providers can use these guidelines to assist in prevention and treatment of STIs.
Corresponding preparer: Kimberly A. Workowski, MD, Division of
STD Prevention, National Center for HIV/AIDS, Viral Hepatitis,
STD, and TB Prevention, CDC. Telephone: 404-639-1898; Email:
Introduction
The term “sexually transmitted infection” (STI) refers to
a pathogen that causes infection through sexual contact,
whereas the term “sexually transmitted disease” (STD) refers
to a recognizable disease state that has developed from an
infection. Physicians and other health care providers have a
crucial role in preventing and treating STIs. These guidelines
are intended to assist with that effort. Although the guidelines
emphasize treatment, prevention strategies and diagnostic
recommendations also are discussed.
This report updates Sexually Transmitted Diseases Treatment
Guidelines, 2015 (1) and should be regarded as a source of
clinical guidance rather than prescriptive standards. Health care
providers should always consider the clinical circumstances of
each person in the context of local disease prevalence. These
guidelines are applicable to any patient care setting that serves
persons at risk for STIs, including family planning clinics,
HIV care clinics, correctional health care settings, private
physicians’ offices, Federally Qualified Health Centers, clinics
for adolescent care, and other primary care facilities. These
guidelines are focused on treatment and counseling and do
not address other community services and interventions that
are essential to STI and HIV prevention efforts.
These STI treatment guidelines complement Recommendations
for Providing Quality Sexually Transmitted Diseases Clinical
Services, 2020 (2) regarding quality clinical services for STIs
in primary care and STD specialty care settings. This guidance
specifies operational determinants of quality services in various
clinical settings, describes on-site treatment and partner
services, and indicates when STI-related conditions should be
managed through consultation with or referral to a specialist.
Methods
These guidelines were developed by CDC staff who worked
with subject matter experts with expertise in STI clinical
management from other federal agencies, nongovernmental
academic and research institutions, and professional medical
organizations. CDC staff identified governmental and
nongovernmental subject matter experts on the basis of their
expertise and assisted them in developing questions to guide
individual literature reviews. CDC staff informed the subject
matter experts that they were being consulted to exchange
information and observations and to obtain their individual
input. All subject matter experts disclosed potential conflicts
of interest. STI Treatment Guidelines, 2021, Work Group
members are listed at the end of this report.
In 2018, CDC staff identified key questions about treatment
and clinical management to guide an update of the 2015
STD treatment guidelines (1). To answer these questions
and synthesize new information available since publication
of the 2015 guidelines, subject matter experts and CDC staff
Recommendations and Reports
2 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
collaborated to conduct systematic literature reviews by using
an extensive MEDLINE database evidence-based approach for
each section of the 2015 guidelines (e.g., using English-language
published abstracts and peer reviewed journal articles). These
systematic reviews were focused on four principal outcomes
of STI therapy for each disease or infection: 1) treatment
of infection on the basis of microbiologic eradication;
2) alleviation of signs and symptoms; 3) prevention of sequelae;
and 4) prevention of transmission, including advantages (e.g.,
cost-effectiveness, single-dose formulations, and directly
observed therapy) and disadvantages (e.g., adverse effects)
of specific regimens. The outcome of the literature reviews
guided development of background materials, including tables
of evidence from peer-reviewed publications summarizing
the type of study (e.g., randomized controlled trial or case
series), study population and setting, treatments or other
interventions, outcome measures assessed, reported findings,
and weaknesses and biases in study design and analysis.
In June 2019, the subject matter experts presented their
assessments of the literature reviews at an in-person meeting
of governmental and nongovernmental participants. Each
key question was discussed and pertinent publications were
reviewed in terms of strengths, weaknesses, and relevance.
Participants evaluated the quality of evidence, provided their
input, and discussed findings in the context of the modified
rating system used by the U.S. Preventive Services Task Force
(USPSTF). The discussions were informal and not structured
to reach consensus. CDC staff also reviewed the publications
from other professional organizations, including the American
College of Obstetricians and Gynecologists (ACOG), USPSTF,
the American Cancer Society (ACS), the American Society
for Colposcopy and Cervical Pathology (ASCCP), and the
Advisory Committee on Immunization Practices (ACIP).
The discussion culminated in a list of participants’ opinions
on all the key STI topic areas for consideration by CDC.
(More detailed descriptions of the key questions, search terms,
systematic search, evidence tables, and review process are
available at https://www.cdc.gov/std/treatment-guidelines/
default.htm).
CDC staff then independently reviewed the tables of evidence
prepared by the subject matter experts, individual comments
from the participants and professional organizations, and existing
guidelines from other organizations to determine whether
revisions to the 2015 STD treatment guidelines were warranted.
CDC staff ranked evidence as high, medium, and low on the
basis of each studys strengths and weaknesses according to the
USPSTF ratings (https://www.uspreventiveservicestaskforce.
org/uspstf/us-preventive-services-task-force-ratings). CDC staff
then developed draft recommendations that were peer reviewed
by public health and clinical experts as defined by the Office of
Management and Budget for influential scientific information.
A public webinar was held to provide an overview of the draft
recommendations and invite questions and comments on the
draft recommendations. The peer review comments, webinar,
questions, and responses were considered by CDC staff in
developing the final recommendations for the updated STI
treatment guidelines. Recommendations for HIV, hepatitis C,
cervical cancer screening, STI screening in pregnancy, human
papillomavirus (HPV) testing, and hepatitis A virus (HAV) and
hepatitis B virus (HBV) vaccination were developed after CDC
staff reviewed existing published recommendations. The English-
language literature was searched periodically by CDC staff to
identify subsequently published articles warranting consideration.
Throughout this report, the evidence used as the basis for
specific recommendations is discussed briefly. Publication
of comprehensive, annotated discussions of such evidence
is planned in a supplemental issue of the journal Clinical
Infectious Diseases after publication of the treatment guidelines.
When more than one therapeutic regimen is recommended
and the listed regimens have similar efficacy and similar
rates of intolerance or toxicity, the recommendations are
listed alphabetically. If differences are specified, regimens are
prioritized on the basis of these differences. Recommended
regimens should be used primarily; alternative regimens can be
considered in instances of notable drug allergy or other medical
contraindications to the recommended regimens. Alternative
regimens are considered inferior to recommended regimens on
the basis of available evidence regarding the principal outcomes
and disadvantages of the regimens.
Clinical Prevention Guidance
Prevention and control of STIs are based on the following
five major strategies (3):
1. Accurate risk assessment and education and counseling
of persons at risk regarding ways to avoid STIs through
changes in sexual behaviors and use of recommended
prevention services
2. Pre-exposure vaccination for vaccine-preventable STIs
3. Identification of persons with an asymptomatic
infection and persons with symptoms associated with
an STI
4. Effective diagnosis, treatment, counseling, and follow-
up of persons who are infected with an STI
5. Evaluation, treatment, and counseling of sex partners
of persons who are infected with an STI
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 3
US Department of Health and Human Services/Centers for Disease Control and Prevention
STI and HIV Infection Risk Assessment
Primary prevention of STIs includes assessment of behavioral
risk (i.e., assessing the sexual behaviors that can place persons
at risk for infection) and biologic risk (i.e., testing for risk
markers for STI and HIV acquisition or transmission). As part
of the clinical encounter, health care providers should routinely
obtain sexual histories from their patients and address risk
reduction as indicated in this report. Guidance for obtaining
a sexual history is available at the Division of STD Prevention
resource page (https://www.cdc.gov/std/treatment/resources.
htm) and in the curriculum provided by the National Network
of STD Clinical Prevention Training Centers (https://www.
nnptc.org). Effective interviewing and counseling skills,
characterized by respect, compassion, and a nonjudgmental
attitude toward all patients, are essential to obtaining a
thorough sexual history and delivering effective prevention
messages. Effective techniques for facilitating rapport with
patients include using open-ended questions (e.g., “Tell me
about any new sex partners youve had since your last visit” and
“What has your experience with using condoms been like?”);
understandable, nonjudgmental language (e.g., “What gender
are your sex partners?”and “Have you ever had a sore or scab
on your penis?”); and normalizing language (e.g., “Some of my
patients have difficulty using a condom with every sex act. How
is it for you?”). The “Five P’s” approach to obtaining a sexual
history is one strategy for eliciting information about the key
areas of interest (Box 1). In addition, health care professionals
can consider assessing sexual history by asking patients such
questions as, “Do you have any questions or concerns about
your sexual health?” Additional information about gaining
cultural competency when working with certain populations
(e.g., gay, bisexual, or other men who have sex with men
[MSM]; women who have sex with women [WSW] or with
women and men [WSWM]; or transgender men and women
or adolescents) is available in sections of these guidelines related
to these populations.
In addition to obtaining a behavioral risk assessment, a
comprehensive STI and HIV risk assessment should include
STI screening as recommended in these guidelines because
STIs are biologic markers of risk, particularly for HIV
acquisition and transmission among certain MSM. In most
clinical settings, STI screening is an essential and underused
component of an STI and HIV risk assessment. Persons
seeking treatment or evaluation for a particular STI should be
screened for HIV and other STIs as indicated by community
prevalence and individual risk factors (see Chlamydial
Infections; Gonococcal Infections; Syphilis). Persons should
be informed about all the tests for STIs they are receiving and
notified about tests for common STIs (e.g., genital herpes,
BOX 1. The Five P’s approach for health care providers obtaining sexual histories: partners, practices, protection from sexually
transmitted infections, past history of sexually transmitted infections, and pregnancy intention
1. Partners
Are you currently having sex of any kind?”
What is the gender(s) of your partner(s)?”
2. Practices
To understand any risks for sexually transmitted
infections (STIs), I need to ask more specific questions
about the kind of sex you have had recently.
What kind of sexual contact do you have or have you had?”
ű
“Do you have vaginal sex, meaning ‘penis in vagina’ sex?”
ű
“Do you have anal sex, meaning ‘penis in rectum/anus
sex?”
ű
“Do you have oral sex, meaning ‘mouth on penis/vagina’?”
3. Protection from STIs
“Do you and your partner(s) discuss prevention of STIs
and human immunodeficiency virus (HIV)?”
“Do you and your partner(s) discuss getting tested?”
For condoms:
ű
“What protection methods do you use? In what
situations do you use condoms?”
4. Past history of STIs
“Have you ever been tested for STIs and HIV?”
“Have you ever been diagnosed with an STI in the past?”
“Have any of your partners had an STI?”
Additional questions for identifying HIV and viral
hepatitis risk:
“Have you or any of your partner(s) ever injected drugs?”
“Is there anything about your sexual health that you
have questions about?”
5. Pregnancy intention
“Do you think you would like to have (more) children
in the future?”
“How important is it to you to prevent pregnancy
(until then)?”
Are you or your partner using contraception or
practicing any form of birth control?”
Would you like to talk about ways to prevent
pregnancy?”
Recommendations and Reports
4 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
trichomoniasis, Mycoplasma genitalium, and HPV) that are
available but not being performed and reasons why they are
not always indicated. Persons should be informed of their test
results and recommendations for future testing. Efforts should
be made to ensure that all persons receive STI care regardless
of personal circumstances (e.g., ability to pay, citizenship
or immigration status, gender identity, language spoken, or
specific sex practices).
STI and HIV Infection
Prevention Counseling
After obtaining a sexual history from their patients, all
providers should encourage risk reduction by offering
prevention counseling. Prevention counseling is most effective
if provided in a nonjudgmental and empathetic manner
appropriate to the patients culture, language, sex and gender
identity, sexual orientation, age, and developmental level.
Prevention counseling for STIs and HIV should be offered
to all sexually active adolescents and to all adults who have
received an STI diagnosis, have had an STI during the
previous year, or have had multiple sex partners. USPSTF
recommends intensive behavioral counseling for all sexually
active adolescents and for adults at increased risk for STIs and
HIV (4). Such interactive counseling, which can be resource
intensive, is directed at a persons risk, the situations in which
risk occurs, and the use of personalized goal-setting strategies.
One such approach, known as client-centered STI and HIV
prevention counseling, involves tailoring a discussion of risk
reduction to the persons situation. Although one large study in
STI clinics (Project RESPECT) demonstrated that this approach
was associated with lower acquisition of curable STIs (e.g.,
trichomoniasis, chlamydia, gonorrhea, and syphilis) (5), another
study conducted 10 years later in the same settings but different
contexts (Project AWARE) did not replicate this result (6).
With the challenges that intensive behavioral counseling poses,
health care professionals might find brief prevention messages
and those delivered through video or in a group session to be
more accessible for the client. A review of 11 studies evaluated
brief prevention messages delivered by providers and health
counselors and reported them to be feasible and to decrease
subsequent STIs in STD clinic settings (7) and HIV care
settings (8). Other approaches use motivational interviewing
to move clients toward achievable risk-reduction goals. Client-
centered counseling and motivational interviewing can be used
effectively by clinicians and staff trained in these approaches.
CDC provides additional information on these and other
effective behavioral interventions at https://www.cdc.gov/
std/program/interventions.htm. Training in client-centered
counseling and motivational interviewing is available through
the STD National Network of Prevention Training Centers
(https://www.nnptc.org).
In addition to one-on-one STI and HIV prevention
counseling, videos and large group presentations can provide
explicit information concerning STIs and reducing disease
transmission (e.g., how to use condoms consistently and
correctly and the importance of routine screening). Group-
based strategies have been effective in reducing the occurrence
of STIs among persons at risk, including those attending
STD clinics (9). Brief, online, electronic-learning modules for
young MSM have been reported to be effective in reducing
incident STIs and offer a convenient client platform for
effective interventions (10). Because the incidence of certain
STIs, most notably syphilis, is higher among persons with
HIV infection, use of client-centered STI counseling for
persons with HIV continues to be encouraged by public health
agencies and other health organizations (https://www.cdc.gov/
std/statistics/2019/default.htm). A 2014 guideline from CDC,
the Health Resources and Services Administration, and the
National Institutes of Health recommends that clinical and
nonclinical providers assess a persons behavioral and biologic
risks for acquiring or transmitting STIs and HIV, including
having sex without condoms, having recent STIs, and having
partners recently treated for STIs (https://stacks.cdc.gov/
view/cdc/44064). That federal guideline is for clinical and
nonclinical providers to offer or make referral for regular
screening for multiple STIs, on-site STI treatment when
indicated, and risk-reduction interventions tailored to the
persons risks. Brief risk-reduction counseling delivered by
medical providers during HIV primary care visits, coupled
with routine STI screening, has been reported to reduce STI
incidence among persons with HIV infection (8). Other
specific methods have been designed for the HIV care setting
(https://www.cdc.gov/hiv/effective-interventions/index.html).
Primary Prevention Methods
Pre-Exposure Vaccination
Pre-exposure vaccination is one of the most effective methods
for preventing transmission of HPV, HAV, and HBV, all
of which can be sexually transmitted. HPV vaccination is
recommended routinely for males and females aged 11 or
12 years and can be administered beginning at age 9 years.
HPV vaccination is recommended through age 26 years for
those not previously vaccinated (11). Sharing clinical decision-
making about HPV vaccination is recommended for certain
adults aged 27–45 years who are not adequately vaccinated
in accordance with existing guidance (https://www.cdc.gov/
vaccines/hcp/acip-recs/vacc-specific/hpv.html).
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 5
US Department of Health and Human Services/Centers for Disease Control and Prevention
Hepatitis B vaccination is recommended for all unvaccinated,
uninfected persons who are sexually active with more than
one partner or are being evaluated or treated for an STI (12).
In addition, hepatitis A and B vaccines are recommended for
MSM, persons who inject drugs, persons with chronic liver
disease, and persons with HIV or hepatitis C infections who
have not had hepatitis A or hepatitis B (12). HAV vaccine is
also recommended for persons who are homeless (13). Details
regarding HAV and HBV vaccination, including routine
childhood vaccination, are available at https://www.cdc.gov/
hepatitis and at the ACIP website (https://www.cdc.gov/
vaccines/hcp/acip-recs/vacc-specific/index.html).
Condoms
External Condoms
When used consistently and correctly, external latex
condoms, also known as male condoms, are effective in
preventing the sexual transmission of HIV infection (http://
www.ashasexualhealth.org/pdfs/Male_and_Female_Condoms.
pdf). In heterosexual HIV mixed-status relationships (i.e., those
involving one infected and one uninfected partner) in which
condoms were used consistently, HIV-negative partners were
71%–80% less likely to become infected with HIV, compared
with persons in similar relationships in which condoms were
not used (14,15). Two analyses of MSM mixed-status couple
studies estimated the protective effect of condom use to be 70%
and 91%, respectively (16,17). Moreover, studies demonstrate
that consistent condom use reduces the risk for other STIs,
including chlamydia, gonorrhea, hepatitis B, and trichomoniasis
(1821). By limiting lower genital tract infections, condoms
also might reduce the risk for pelvic inflammatory disease
(PID) among women (22). In addition, consistent and correct
use of latex condoms reduces the risk for HPV infection
and HPV-associated diseases, genital herpes, syphilis, and
chancroid when the infected area or site of potential exposure
is covered (2327). Additional information is available at
https://www.cdc.gov/condomeffectiveness/index.html and
www.factsaboutcondoms.com/professional.php. Condoms
are regulated as medical devices and are subject to random
sampling and testing by the Food and Drug Administration
(FDA). Each latex condom manufactured in the United States
is tested electronically for holes before packaging. The rate of
condom breakage during sexual intercourse and withdrawal in
the United States is approximately two broken condoms per
100 condoms. Rates of breakage and slippage might be slightly
higher during anal intercourse (28,29). The failure of condoms
to protect against STIs or unintended pregnancy usually results
from inconsistent or incorrect use rather than condom breakage
(30). Users should check the expiration or manufacture date
on the box or individual package. Latex condoms should not
be used beyond their expiration date or >5 years after the
manufacturing date. Condoms made of materials other than
latex are available in the United States and can be classified
into two general categories: 1) polyurethane, polyisoprene, or
other synthetic condoms and 2) natural membrane condoms.
Polyurethane external condoms provide protection against
STIs and HIV and pregnancy comparable to that of latex
condoms (20,31). These can be substituted for latex condoms
by persons with latex sensitivity, are typically more resistant to
deterioration, and are compatible with use of both oil-based
and water-based lubricants. The effectiveness of other synthetic
external condoms to prevent STIs has not been extensively
studied, and FDA labeling restricts their recommended use
to persons who are sensitive to or allergic to latex. Natural
membrane condoms (frequently called natural skin condoms
or [incorrectly] lambskin condoms) are made from lamb cecum
and can have pores up to 1,500 nm in diameter. Although
these pores do not allow the passage of sperm, they are more
than 10 times the diameter of HIV and more than 25 times
that of HBV. Moreover, laboratory studies demonstrate that
sexual transmission of viruses, including HBV, herpes simplex
virus (HSV), and HIV, can occur with natural membrane
condoms (31). Therefore, natural membrane condoms are not
recommended for prevention of STIs and HIV.
Providers should advise that condoms must be used
consistently and correctly to be effective in preventing STIs and
HIV while noting that any condom use is better than no condom
use. Providing instructions about the correct use of condoms
can be useful. Communicating the following recommendations
can help ensure that patients use external condoms correctly:
Use a new condom with each sex act (i.e., oral, vaginal,
and anal).
Carefully handle the condom to avoid damaging it with
fingernails, teeth, or other sharp objects.
Put the condom on after the penis is erect and before any
genital, oral, or anal contact with the partner.
Use only water-based or silicone-based lubricants (e.g.,
K-Y Jelly, Astroglide, AquaLube, or glycerin) with latex
condoms. Oil-based lubricants (e.g., petroleum jelly,
shortening, mineral oil, massage oils, body lotions, or
cooking oil) can weaken latex and should not be used;
however, oil-based lubricants typically can be used with
polyurethane or other synthetic condoms.
Ensure adequate lubrication during vaginal and anal sex,
which might require using exogenous water-based
lubricants.
Hold the condom firmly against the base of the penis
during withdrawal, and withdraw while the penis is still
erect to prevent the condom from slipping off.
Recommendations and Reports
6 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Additional information about external condoms is available
at https://www.cdc.gov/condomeffectiveness.
Internal Condoms
Condoms for internal vaginal use, also known as female
condoms, are available worldwide (e.g., the FC2 Female
Condom, Reddy condom, Cupid female condom, and Womans
condom) (31,32). Use of internal condoms can provide
protection from acquisition and transmission of STIs, although
data are limited. Internal condoms are more costly compared
with external condoms; however, they offer the advantage of
being controlled by the receptive partner as an STI and HIV
prevention method, and the newer versions might be acceptable
to all persons. Although the internal condom also has been used
during receptive anal intercourse, efficacy associated with this
practice remains unknown (33). Additional information about
the internal condom is available at http://www.ashasexualhealth.
org/pdfs/Male_and_Female_Condoms.pdf.
Cervical Diaphragms
In observational studies, diaphragm use has been
demonstrated to protect against cervical gonorrhea, chlamydia,
and trichomoniasis (34). However, a trial examining the effect
of a diaphragm plus lubricant on HIV acquisition among
women in Africa reported no additional protective effect when
compared with the use of male condoms alone. Likewise, no
difference by study arm in the rate of acquisition of chlamydia,
gonorrhea, or herpes occurred (35,36). Diaphragms should
not be relied on as the sole source of protection against HIV
and other STIs.
Multipurpose Prevention Technologies
Methods that combine STI and HIV prevention
with pregnancy prevention are known as multipurpose
prevention technologies (MPTs) (37) (https://www.who.int/
reproductivehealth/topics/linkages/mpts/en). Internal and
external condoms are both examples of MPTs because they are
effective prevention measures when used correctly for STI and
HIV transmission or pregnancy prevention. The multicenter
Evidence for Contraception Options and HIV Outcomes
(ECHO) trial observed no statistically significant differences in
HIV incidence rates among women randomly assigned to one
of three contraceptive methods (depot medroxyprogesterone
acetate [DMPA], levonorgestrel implant, and copper-
containing intrauterine device [IUD]); however, rates of HIV
infection were high in all groups, indicating a need for MPTs
(38). Development of MPTs is complex and ongoing; products
under study include microbicides with contraceptive devices
(e.g., tenofovir with a vaginal ring contraceptive delivery
package) and other innovative methods (39).
Topical Microbicides and Spermicides
Nonspecific topical microbicides are ineffective for
preventing HIV infection (4045). Tenofovir gel has been
studied for prevention of herpes simplex virus 2 (HSV-2)
and HIV infections (46,47). Adherence can be low (48), and
prevention of HIV infection, especially among women, has not
been demonstrated (47,49).Vaginal rings containing dapivirine
have provided some reduction in HIV infection (50,51). For
men and transgender women who have anal intercourse,
tenofovir gel appears safe when applied before and after anal
sex (52). Spermicides containing nonoxynol-9 (N-9) might
disrupt genital or rectal epithelium and have been associated
with an increased risk for HIV infection. Condoms with N-9
are no more effective than condoms without N-9; therefore,
N-9 alone or in a condom is not recommended for STI and
HIV prevention (40). N-9 use also has been associated with
an increased risk for bacterial urinary tract infections among
women (53,54).
Nonbarrier Contraception, Female Surgical
Sterilization, and Hysterectomy
Contraceptive methods that are not mechanical barriers
offer no protection against HIV or other STIs. The ECHO
study observed no differences in HIV incidence rates among
women randomly assigned to DMPA, levonorgestrel implant,
or copper-containing IUD contraceptive methods (38). A
systematic review of epidemiologic evidence reported that the
majority of studies demonstrated no association between use
of oral contraceptives and HIV acquisition among women
(55). Whether hormonal contraception alters a womans risk
for other STIs is uncertain (56,57).
Sexually active women who use contraceptive methods
other than condoms should be counseled about STI and HIV
infection prevention measures. These include pre-exposure
prophylaxis (PrEP) and postexposure prophylaxis (PEP),
limiting the number of sex partners, and correct and consistent
use of condoms.
Emergency Contraception
Unprotected intercourse exposes women to risks for STIs
and unplanned pregnancy. Providers should offer counseling
about the option of emergency contraception if pregnancy
is not desired. Options for emergency contraception in the
United States include copper-containing IUDs and emergency
contraceptive pills (ECPs) (58,59). More information is available
at https://www.acog.org/clinical/clinical-guidance/practice-
bulletin/articles/2015/09/emergency-contraception?utm_
source=redirect&utm_medium=web&utm_campaign=otn.
ECPs are available in the following formulations: ulipristal
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 7
US Department of Health and Human Services/Centers for Disease Control and Prevention
acetate in a single dose (30 mg) available by prescription,
levonorgestrel in a single dose (1.5 mg) available over the
counter or by prescription, or a combined estrogen and
progestin pill regimen. Insertion of a copper-containing
IUD ≤5 days after unprotected sex can reduce pregnancy risk
from a sex act by approximately 99% (60). ECPs are most
efficacious when initiated as soon as possible after unprotected
sex. Ulipristal acetate is effective ≤5 days after unprotected sex,
and levonorgestrel is most effective ≤3 days after unprotected
sex but has some efficacy at ≤5 days. ECPs are ineffective
(but not harmful) if the woman is already pregnant (61). A
2019 Cochrane review summarized the efficacy, safety, and
convenience of different emergency contraception methods (61).
More information about emergency contraception is
available in Contraceptive Technology, 21st Edition (31), in the
2016 U.S. Selected Practice Recommendations (U.S. SPR)
for Contraceptive Use (emergency contraception) available
at https://www.cdc.gov/reproductivehealth/contraception/
mmwr/spr/emergency.html, and in the 2016 U.S. Medical
Eligibility Criteria (U.S. MEC) for Contraceptive Use (copper
IUDs for emergency contraception) available at https://
www.cdc.gov/reproductivehealth/contraception/mmwr/mec/
appendixj.html.
Providers should educate males and females about emergency
contraception, especially if other methods of contraception
were used incorrectly or not at all and pregnancy is not desired
(62). An advance supply of ECPs can be provided or prescribed
so that ECPs will be available when needed (59).
Male Circumcision
Male circumcision reduces the risk for HIV infection and
certain STIs among heterosexual men. Three randomized,
controlled trials performed in regions of sub-Saharan Africa,
where generalized HIV epidemics involving predominantly
heterosexual transmission were occurring, demonstrated that
male circumcision reduces the risk for HIV acquisition among
men by 50%–60% (6365). In those trials, circumcision
also was protective against other STIs, including high-risk
genital HPV infection and genital herpes (6668). Follow-up
studies have demonstrated sustained benefit of circumcision
for HIV prevention (69) and that the effect is not mediated
solely through a reduction in HSV-2 infection or genital ulcer
disease (GUD) (70).
The World Health Organization (WHO) and the Joint
United Nations Programme on HIV/AIDS (UNAIDS)
recommend that male circumcision efforts be scaled up as an
effective intervention for preventing heterosexually acquired HIV
infection (71) in countries with hyperendemic and generalized
HIV epidemics within the context of ensuring universal access
to comprehensive HIV prevention, treatment, care, and support
(https://www.afro.who.int/publications/voluntary-medical-male-
circumcision-hiv-prevention). In the United States, the American
Academy of Pediatrics (AAP) recommends that newborn male
circumcision be available to families that desire it because the
benefits of the procedure, including prevention of penile cancers,
urinary tract infections, GUD, and HIV infection, outweigh the
risks. ACOG has also endorsed AAP’s policy statement. In light
of these benefits, the American Urological Association states
that male circumcision should be considered an option for risk
reduction, among other strategies (72). Additional information
for providers counseling male patients and parents regarding
male circumcision for preventing HIV, STIs, and other adverse
health outcomes is available at https://www.cdc.gov/hiv/risk/
male-circumcision.html.
No definitive data exist to determine whether male
circumcision reduces HIV acquisition among MSM, although
one meta-analysis of 62 observational studies reported that
circumcision was protective against HIV acquisition in low- to
middle-income countries but not in high-income countries
(73). Further studies are needed to confirm any potential
benefit of male circumcision for this population.
Pre-Exposure Prophylaxis for HIV
Daily oral antiretroviral PrEP with a fixed-dose combination
of emtricitabine (FTC) and either tenofovir disoproxil fumarate
(TDF) or tenofovir alafenamide (TAF) have demonstrated
safety (74) and a substantial reduction in the rate of HIV
acquisition for MSM (75). TDF/FTC has demonstrated
safety and efficacy for mixed-status heterosexual couples (76)
and heterosexual men and women recruited individually
(77); however, no evidence is yet available regarding TAF/
FTC among heterosexually active women. In addition, one
clinical trial involving persons who inject drugs (78) and one
involving heterosexual mixed-status couples (76) demonstrated
substantial efficacy and safety of daily oral PrEP with TDF
alone. High adherence to oral PrEP was strongly associated
with protection from HIV infection. Studies conducted with
MSM have demonstrated that taking PrEP at specific times
before and after sexual intercourse was effective in preventing
HIV; however, less experience exits with this regimen, it is
not FDA cleared, and it has not been studied among other
populations (79).
Comprehensive clinical practice guidelines are available
for providers in prescribing PrEP to reduce the risk for HIV
infection (80). Among HIV-negative sexually active men
and women, bacterial STIs are key indicators of risk for
HIV acquisition. Studies have documented the risk for HIV
acquisition among MSM within 1 year after infection with
rectal gonorrhea or chlamydia (one in 15 men), primary
or secondary syphilis (one in 18), and among men with no
Recommendations and Reports
8 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
rectal STI or syphilis infection (one in 53) (8183). Sexually
active adults and adolescents should be screened for STIs
(e.g., chlamydia, gonorrhea, and syphilis) in accordance with
recommendations, and persons with infection should be
offered PrEP. The USPSTF recommends that persons at risk
for HIV acquisition be offered PrEP (84). Persons at risk for
HIV acquisition include HIV-negative persons whose sexual
partner or partners have HIV infection (especially if viral load
is detectable or unknown), persons who have had gonorrhea
or syphilis during the previous 6 months, and injecting drug
users who share injection equipment (84). Clinical practice
guidelines recommend STI screening for persons taking PrEP
(80) because increased rates of STI acquisition have been
described (8587).
Pre-Exposure Prophylaxis for STIs
Providing HSV treatment to persons with HIV and HSV
infection has not demonstrated benefit in reducing HIV
acquisition among uninfected partners. A large randomized
controlled trial evaluated mixed-status heterosexual couples
among the partners with HIV infection who also were
seropositive for HSV-2 (88). Use of acyclovir had no effect
on HIV transmission. These findings are consistent with a
previous trial that reported no benefit of acyclovir in preventing
HIV acquisition among persons seropositive for HSV-2 (89).
Doxycycline prophylaxis has been examined for preventing
bacterial STIs. In a pilot study, 30 MSM living with HIV with
previous syphilis (two or more episodes since HIV diagnosis)
were randomly assigned to doxycycline 100 mg for 48 weeks
versus a financial incentive–based behavioral intervention (90).
That study demonstrated a 73% reduction in any bacterial STI
at any site, without substantial differences in sexual behavior.
Additional studies examining doxycycline prophylaxis are
under way or in development (91).
Postexposure Prophylaxis for HIV and STIs
Guidelines for using PEP aimed at preventing HIV and other
STIs as a result of sexual exposure are available at https://www.
cdc.gov/hiv/pdf/programresources/cdc-hiv-npep-guidelines.
pdf. Sexually active persons seeking HIV PEP should be
evaluated for PrEP after completing their PEP course and
testing negative for HIV. HIV PEP is also discussed elsewhere
in this report (see Sexual Assault and Abuse and STIs). Genital
hygiene methods (e.g., vaginal washing and douching) after
sexual exposure are ineffective in protecting against HIV and
STIs and might increase the risk for bacterial vaginosis (BV),
certain STIs, and HIV infection (92).
STI PEP in the form of doxycycline 200 mg taken after
unprotected anal sex has been studied among MSM and
transgender women; results demonstrated reduction in incident
chlamydia and syphilis by 70% and 73%, respectively, but no
effect on gonorrhea (93). Other studies are under way or in
development regarding doxycycline prophylaxis for bacterial
STIs (91). No long-term data are available regarding the impact
of STI PEP on antimicrobial resistance and the microbiome.
Further studies are needed to determine whether STI PEP is
an effective and beneficial strategy for STI prevention.
HIV Treatment as Prevention: Antiretroviral
Treatment of Persons with HIV to Prevent HIV
Among Partners
In 2011, the randomized controlled trial HPTN 052
demonstrated that, among HIV mixed-status heterosexual
couples, HIV antiretroviral therapy (ART) for the infected
partner decreased the risk for transmission to the uninfected
partner by 96% (94). Therefore, ART not only is beneficial to
the health of persons with HIV infection, it also reduces the
risk for transmission. Additional studies of HIV mixed-status
couples, heterosexual and MSM couples (PARTNER study),
and MSM couples (Opposites Attract and PARTNERS2
studies) reported that patients with HIV taking ART who
maintain an undetectable viral load demonstrate no risk for
transmitting HIV to their HIV-negative sex partners (9597).
For those reasons, ART should be offered to all persons with
HIV infection to obtain viral suppression. Detailed guidance
regarding ART regimens is available in the U.S. Department of
Health and Human Services’ HIV treatment guidelines (98).
HIV Seroadaptive Strategies
Seroadaptive strategies for HIV prevention have largely
originated within communities of MSM. They are predicated
on knowledge of self and partner HIV status. One specific
seroadaptive practice is serosorting, which includes limiting
anal sex without a condom to partners with the same HIV
status as their own or choosing to selectively use condoms
with HIV mixed-status partners. Another practice among
mixed-status couples is seropositioning, in which the
person with HIV infection is the receptive partner for anal
intercourse. Observational studies have consistently reported
that serosorting confers greater risk for HIV infection than
consistent condom use but has lower risk compared with anal
intercourse without a condom and without serosorting (99
101). Serosorting practices have been associated with increased
risk for STIs, including chlamydia and gonorrhea (102,103).
Serosorting is not recommended for the following reasons:
many MSM who have HIV infection do not know they
have HIV because they have not been tested recently, mens
assumptions about the HIV status of their partners might be
wrong, and some men with HIV infection might not disclose or
might misrepresent their HIV status. All of these factors increase
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 9
US Department of Health and Human Services/Centers for Disease Control and Prevention
the risk that serosorting can lead to HIV infection. Serosorting
has not been studied among heterosexually active persons.
Abstinence and Reduction of Number of
Sex Partners
Abstinence from oral, vaginal, and anal sex and participating
in a long-term, mutually monogamous relationship with a
partner known to be uninfected are prevention approaches to
avoid transmission of STIs. For persons who are being treated
for an STI (or whose partners are undergoing treatment),
counseling that encourages abstinence from sexual intercourse
until completion of the entire course of medication is vital
for preventing reinfection. A trial conducted among women
regarding the effectiveness of counseling messages when
patients have cervicitis or vaginal discharge demonstrated
that women whose sex partners have used condoms might
benefit from a hierarchical message that includes condoms
but women without such experience might benefit more from
an abstinence-only message (104). A more comprehensive
discussion of abstinence and other sexual practices that can help
persons reduce their risk for STIs is available in Contraceptive
Technology, 21st Edition (31).
Partner Services
The term “partner services” refers to a continuum of clinical
evaluation, counseling, diagnostic testing, and treatment
designed to increase the number of infected persons brought to
treatment and to reduce transmission among sexual networks.
This continuum includes efforts of health departments,
medical providers, and patients themselves. The term “public
health partner services” refers to efforts by public health
departments to identify the sex and needle-sharing partners
of infected persons to ensure their medical evaluation and
treatment. Health departments are increasingly incorporating
referral to additional services, as indicated, into the partner services
continuum. Aside from the general benefit to patients and partners,
service referrals and linkage can mitigate the circumstances that
increase risk for future STI and HIV acquisition.
The types and comprehensiveness of public health partner
services and the specific STIs for which they are offered vary
by public health agency, their resources, and the geographic
prevalence of STIs. In most areas of the United States, health
departments routinely attempt to provide partner services to
all persons with infectious syphilis (primary or secondary)
and persons with a new diagnosis of HIV infection. Health
departments should provide partner services for persons who
might have cephalosporin-resistant gonorrhea. In contrast,
relatively few U.S. health departments routinely provide
STI partner services to persons with gonorrhea, chlamydia,
trichomoniasis, or other STIs (105). Because STI diagnoses
often can serve as risk markers for HIV acquisition (83), public
health services might include follow-up of MSM with an STI
to offer HIV PrEP. Public health services can also include HIV
and STI prevention interventions including HIV and STI
testing, linkage and relinkage of persons with HIV infection
to HIV care clinics, and referral of partners of persons with
STIs or HIV infection to HIV PrEP, as indicated (106109).
Clinicians should familiarize themselves with public health
practices in their area; however, in most instances, providers
should understand that responsibility for discussing the
treatment of partners of persons with STIs rests with the
diagnosing provider and the patient. State laws require a good
faith effort by the provider to inform partners, and providers
should familiarize themselves with public health laws.
Clinicians who do not notify partners of patients directly
can still provide partner services by counseling infected persons
and providing them with written information and medication
to give to their partners (if recommended and allowable by
state law), directly evaluating and treating sex partners, and
cooperating with state and local health departments. Clinicians
efforts to ensure treatment of patients’ sex partners can reduce
the risk for reinfection and potentially diminish transmission
of STIs (110). Therefore, clinicians should encourage all
persons with STIs to notify their sex partners and urge them
to seek medical evaluation and treatment. Exceptions to this
practice include circumstances posing a risk for intimate
partner violence (111). Available data are limited regarding
the rate of intimate partner violence directly attributable
to partner notification (112,113); however, because of the
reported prevalence of intimate partner violence in the general
population (114), providers should consider the potential
risk before notifying partners of persons or encouraging
partner notification. Time spent counseling patients about the
importance of notifying partners is associated with improved
notification outcomes (115). When possible, clinicians should
advise persons to bring their primary sex partner with them
when returning for treatment and should concurrently treat
both persons. Although this approach can be effective for a
main partner (116,117), it might not be a feasible approach
for additional sex partners. Evidence indicates that providing
patients with written information to share with sex partners
can increase rates of partner treatment (110).
Certain health departments now use technology (e.g.,
email, texting, mobile applications, and social media outlets)
to facilitate partner services for locating and notifying the
sex partners of persons with STIs, including HIV (118,119).
Patients now have the option to use Internet sites to send
anonymous email or text messages advising partners of their
exposure to an STI (120); anonymous notification via the
Recommendations and Reports
10 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Internet is considered better than no notification at all.
However, because the extent to which these sites affect partner
notification and treatment is uncertain, patients should be
encouraged to notify their partners in person or by telephone,
email, or text message; alternatively, patients can authorize a
medical provider or public health professional to notify their
sex partners.
Expedited Partner Therapy
Expedited partner therapy (EPT) is a harm-reduction
strategy and the clinical practice of treating the sex partners
of persons with diagnosed chlamydia or gonorrhea, who are
unable or unlikely to seek timely treatment, by providing
medications or prescriptions to the patient as allowable by law.
Patients then provide partners with these therapies without
the health care provider having examined the partner (https://
www.cdc.gov/std/ept). Unless prohibited by law or other
regulations, medical providers should routinely offer EPT
to patients with chlamydia when the provider cannot ensure
that all of a patient’s sex partners from the previous 60 days
will seek timely treatment. If the patient has not had sex
during the 60 days before diagnosis, providers should offer
EPT for the patient’s most recent sex partner. Because EPT
must be an oral regimen and current gonorrhea treatment
involves an injection, EPT for gonorrhea should be offered
to partners unlikely to access timely evaluation after linkage
is explored. EPT is legal in the majority of states but varies
by chlamydial or gonococcal infection. Providers should visit
https://www.cdc.gov/std/ept to obtain updated information for
their state. Providing patients with packaged oral medication
is the preferred approach because the efficacy of EPT using
prescriptions has not been evaluated, obstacles to EPT can exist
at the pharmacy level (121,122), and many persons (especially
adolescents) do not fill the prescriptions provided to them by
a sex partner (123,124). Medication or prescriptions provided
for EPT should be accompanied by educational materials for
the partner, including treatment instructions, warnings about
taking medications (e.g., if the partner is pregnant or has an
allergy to the medication), general health counseling, and a
statement advising that partners seek medical evaluation as
soon as possible for HIV infection and any symptoms of STIs,
particularly PID.
Evidence supporting EPT is based on three U.S. clinical
trials involving heterosexual men and women with chlamydia
or gonorrhea (125127). All three trials reported that more
partners were treated when patients were offered EPT. Two
reported statistically significant decreases in the rate of
reinfection, and one observed a lower risk for persistent or
recurrent infection that was statistically nonsignificant. A
fourth trial in the United Kingdom did not demonstrate a
difference in the risk for reinfection or in the numbers of
partners treated between persons offered EPT and those
advised to notify their sex partners (128). U.S. trials and a
meta-analysis of EPT revealed that the magnitude of reduction
in reinfection of index patients, compared with patient referral,
differed according to the STI and the sex of the index patient
(110,125127). However, across trials, reductions in chlamydia
prevalence at follow-up were approximately 20%, and
reductions in gonorrhea were approximately 50% at follow-up.
Existing data indicate that EPT also might have a role in
partner management for trichomoniasis; however, no partner
management intervention has been reported to be more
effective than any other in reducing trichomoniasis reinfection
rates (129,130). No data support use of EPT in the routine
management of patients with syphilis.
Data are limited regarding use of EPT for gonococcal
or chlamydial infections among MSM, compared with
heterosexuals (131,132). Published studies, including recent
data regarding extragenital testing, indicated that male partners
of MSM with diagnosed gonorrhea or chlamydia might have
other bacterial STIs (gonorrhea or syphilis) or HIV (133135).
Studies have reported that 5% of MSM have a new diagnosis
of HIV when evaluated as partners of men with gonococcal
or chlamydial infections (133,134); however, more recent data
indicate that, in certain settings, the frequency of HIV infection
is much lower (135). Considering limited data and potential
for other bacterial STIs among MSM partners, shared clinical
decision-making regarding EPT is recommended. All persons
who receive bacterial STI diagnoses and their sex partners,
particularly MSM, should be tested for HIV, and those at risk
for HIV infection should be offered HIV PrEP (https://www.
cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-guidelines-2017.pdf).
Reporting and Confidentiality
Accurate and timely reporting of STIs is integral to public
health efforts in assessing morbidity trends, allocating limited
resources, and assisting local health authorities with partner
notification and treatment. STI and HIV/AIDS cases should
be reported in accordance with state and local statutory
requirements. Syphilis (including congenital syphilis),
gonorrhea, chlamydia, chancroid, and HIV are reportable
diseases in every state. Because the requirements for reporting
other STIs differ by state, clinicians should be familiar with the
reporting requirements applicable within their jurisdictions.
Reporting can be provider based, laboratory based, or
both. Clinicians who are unsure of state and local reporting
requirements should seek advice from state or local health
department STI programs. STI and HIV reports are kept
confidential. In most jurisdictions, such reports are protected
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 11
US Department of Health and Human Services/Centers for Disease Control and Prevention
by statute or regulation. Before conducting a follow-up
of a person with a positive STI test result, public health
professionals should consult the patients health care provider,
if possible, to inform them of the purpose of the public health
visit, verify the diagnosis, determine the treatments received,
and ascertain the best approaches to patient follow-up.
Retesting After Treatment to Detect
Repeat Infections
Retesting 3 months after diagnosis of chlamydia, gonorrhea,
or trichomoniasis can detect repeat infection and potentially
can be used to enhance population-based prevention (136,137).
Any person who has a positive test for chlamydia or gonorrhea,
along with women who have a positive test for trichomonas,
should be rescreened 3 months after treatment. Any person
who receives a syphilis diagnosis should undergo follow-up
serologic syphilis testing per current recommendations
and follow-up testing for HIV (see Syphilis). Additional
information regarding retesting is available elsewhere in this
report (see Chlamydial Infections; Gonococcal Infections;
Syphilis; Trichomoniasis).
STI Detection Among Special
Populations
Pregnant Women
Intrauterine or perinatally transmitted STIs can have
debilitating effects on pregnant women, their fetuses, and their
partners. All pregnant women and their sex partners should be
asked about STIs, counseled about the possibility of perinatal
infections, and provided access to recommended screening and
treatment, if needed.
Recommendations for screening pregnant women for
STIs to detect asymptomatic infections are based on disease
severity and sequelae, prevalence among the population, costs,
medicolegal considerations (e.g., state laws), and other factors.
The following screening recommendations for pregnant
women summarize clinical guidelines from federal agencies
and medical professional organizations.
Screening Recommendations
HIV Infection
All pregnant women in the United States should be tested
for HIV at the first prenatal visit, even if they have been
previously tested (138). Testing pregnant women for HIV
and prompt linkage to care of women with HIV infection are
vital for womens health and reducing perinatal transmission of
HIV through ART and obstetrical interventions. HIV testing
should be offered as part of the routine panel of prenatal tests
(i.e., opt-out testing). For women who decline HIV testing,
providers should address their concerns and, when appropriate,
continue to encourage testing. Partners of pregnant patients
should be offered HIV testing if their status is unknown (139).
Retesting in the third trimester (preferably before 36 weeks
gestation) is recommended for women at high risk for
acquiring HIV infection. Examples of women at high risk
include those who inject drugs, have STIs during pregnancy,
have multiple sex partners during pregnancy, have a new
sex partner during pregnancy, or have partners with HIV
infection; those who are receiving care in health care facilities
in settings with HIV incidence ≥1 per 1,000 women per year;
those who are incarcerated; those who live in areas with high
rates of HIV infection; or those who have signs or symptoms
of acute HIV infection (e.g., fever, lymphadenopathy, skin
rash, myalgia, arthralgia, headache, oral ulcers, leukopenia,
thrombocytopenia, or transaminase elevation) (140).
Rapid HIV testing should be performed for any woman in labor
who has not been tested for HIV during pregnancy or whose HIV
status is unknown, unless she declines. If a rapid HIV test result
is positive, ART should be administered without waiting for the
results of confirmatory testing (https://clinicalinfo.hiv.gov/sites/
default/files/inline-files/PerinatalGL.pdf).
Syphilis
During 2012–2019, congenital syphilis rates in the United
States increased from 8.4 to 48.5 cases per 100,000 births,
a 477.4% increase (141). At least 45 states have a prenatal
syphilis testing requirement, with high variability among those
requirements (142). In the United States, all pregnant women
should be screened for syphilis at the first prenatal visit, even
if they have been tested previously (143). Prenatal screening
for syphilis has been reported to be suboptimal in the United
States (144,145). Testing in the third trimester and at delivery
can prevent congenital syphilis cases (146,147). Partners of
pregnant women with syphilis should be evaluated, tested,
and treated.
When access to prenatal care is not optimal, a stat rapid
plasma reagin (RPR) card test and treatment, if that test is
reactive, should be administered at the time that a pregnancy
is confirmed or when the pregnancy test is performed, if
follow-up is uncertain. Pregnant women should be retested
for syphilis at 28 weeks’ gestation and at delivery if the mother
lives in a community with high syphilis rates or is at risk for
syphilis acquisition during pregnancy (e.g., misuses drugs or
has an STI during pregnancy, having multiple sex partners,
having a new sex partner, or having a sex partner with an STI).
Neonates should not be discharged from the hospital unless
Recommendations and Reports
12 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
the syphilis serologic status of the mother has been determined
at least once during pregnancy. Any woman who delivers a
stillborn infant should be tested for syphilis.
Hepatitis B
All pregnant women should be routinely tested for hepatitis
B surface antigen (HBsAg) at the first prenatal visit even if they
have been previously vaccinated or tested (148). Women who
are HBsAg positive should be provided with, or referred for,
counseling and medical management. Women who are HBsAg
negative but at risk for HBV infection should be vaccinated.
Women who were not screened prenatally, those who engage
in behaviors that put them at high risk for infection (e.g.,
having had more than one sex partner during the previous
6 months, having been evaluated or treated for an STI,
having had recent or current injection drug use, or having an
HBsAg-positive sex partner), and those with clinical hepatitis
should be tested at the time of admission to the hospital
for delivery. To avoid misinterpreting a transient positive
HBsAg result during the 21 days after vaccination, HBsAg
testing should be performed before vaccine administration.
All laboratories that conduct HBsAg tests should test initially
reactive specimens with a licensed neutralizing confirmatory
test. When pregnant women are tested for HBsAg at the time
of admission for delivery, shortened testing protocols can be
used, and initially reactive results should prompt expedited
administration of immunoprophylaxis to neonates (148).
Pregnant women who are HBsAg positive should be reported
to the local or state health department to ensure that they
are entered into a case-management system and that timely
and age-appropriate prophylaxis is provided to their infants.
Information concerning the pregnant womans HBsAg status
should be provided to the hospital where delivery is planned
and to the health care provider who will care for the newborn.
In addition, household and sexual contacts of women who are
HBsAg positive should be vaccinated.
Chlamydia
All pregnant women aged <25 years as well as older women
at increased risk for chlamydia (e.g., those aged ≥25 years who
have a new sex partner, more than one sex partner, a sex partner
with concurrent partners, or a sex partner who has an STI)
should be routinely screened for Chlamydia trachomatis at the first
prenatal visit (149). Pregnant women who remain at increased
risk for chlamydial infection also should be retested during the
third trimester to prevent maternal postnatal complications and
chlamydial infection in the neonate. Pregnant women identified
as having chlamydia should be treated immediately and have a
test of cure to document chlamydial eradication by a nucleic acid
amplification test (NAAT) 4 weeks after treatment. All persons
diagnosed with a chlamydial infection should be rescreened
3 months after treatment.
Gonorrhea
All pregnant women aged <25 years as well as women aged
≥25 years at increased risk for gonorrhea (e.g., those with other
STIs during pregnancy or those with a new sex partner, more
than one sex partner, a sex partner with concurrent partners,
or a sex partner who has an STI or is exchanging sex for money
or drugs) should be screened for Neisseria gonorrhoeae at the
first prenatal visit (149). Pregnant women who remain at high
risk for gonococcal infection also should be retested during the
third trimester to prevent maternal postnatal complications and
gonococcal infection in the neonate. Clinicians should consider
the communities they serve and might choose to consult local
public health authorities for guidance on identifying groups
that are more vulnerable to gonorrhea acquisition on the basis
of local disease prevalence. Gonococcal infection, in particular,
is concentrated among specific geographic locations and
communities (https://www.cdc.gov/std/statistics/2019/default.
htm). Pregnant women identified as having gonorrhea should
be treated immediately. All persons diagnosed with gonorrhea
should be rescreened 3 months after treatment.
Hepatitis C Virus
The rate of hepatitis C virus (HCV) infection has increased
among pregnant women in recent years (150153). HCV
screening should be performed for all pregnant women
during each pregnancy, except in settings where the HCV
infection (HCV positivity) rate is <0.1% (154156). The most
important risk factor for HCV infection is past or current
injecting drug use (157). Additional risk factors include having
had a blood transfusion or organ transplantation before July
1992, having received clotting factor concentrates produced
before 1987, having received an unregulated tattoo, having
been on long-term hemodialysis, having other percutaneous
exposures, or having HIV infection. All women with HCV
infection should receive counseling, supportive care, and
linkage to care (https://www.hcvguidelines.org). No vaccine
is available for preventing HCV transmission.
Cervical Cancer
Pregnant women should undergo cervical cancer screening
and at the same frequency as nonpregnant women; however,
management differs slightly during pregnancy (158).
Colposcopy is recommended for the same indications during
pregnancy as for nonpregnant women. However, biopsies
may be deferred, and endocervical sampling should not
be performed. Treatment should not be performed during
pregnancy unless cancer is detected.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 13
US Department of Health and Human Services/Centers for Disease Control and Prevention
Bacterial Vaginosis, Trichomoniasis, and
Genital Herpes
Evidence does not support routine screening for BV among
asymptomatic pregnant women at high risk for preterm
delivery (159). Symptomatic women should be evaluated
and treated (see Bacterial Vaginosis). Evidence does not
support routine screening for Trichomonas vaginalis among
asymptomatic pregnant women. Women who report symptoms
should be evaluated and treated (see Trichomoniasis). In
addition, evidence does not support routine HSV-2 serologic
screening among asymptomatic pregnant women. However,
type-specific serologic tests might be useful for identifying
pregnant women at risk for HSV-2 infection and for guiding
counseling regarding the risk for acquiring genital herpes
during pregnancy. Routine serial cultures for HSV are not
indicated for women in the third trimester who have a history
of recurrent genital herpes.
For more detailed discussions of STI screening and treatment
among pregnant women, refer to the following references:
Screening for HIV Infection: U.S. Preventive Services Task Force
Recommendation Statement (138); Recommendations for the Use
of Antiretroviral Drugs in Pregnant Women with HIV Infection
and Interventions to Reduce Perinatal HIV Transmission in
the United States (https://clinicalinfo.hiv.gov/sites/default/
files/inline-files/PerinatalGL.pdf); Guidelines for Perinatal
Care (160); Prevention of Hepatitis B Virus Infection in the
United States: Recommendations of the Advisory Committee
on Immunization Practices (12); Screening for Chlamydia and
Gonorrhea: U.S. Preventive Services Task Force Recommendation
Statement (149); Screening for Bacterial Vaginosis in Pregnant
Persons to Prevent Preterm Delivery: U.S. Preventive Services
Task Force Recommendation Statement (159); Screening for
Syphilis Infection in Pregnant Women: U.S. Preventive Services
Task Force Recommendation Statement (161); Serologic Screening
for Genital Herpes Infection: U.S. Preventive Services Task Force
Recommendation Statement (162); Screening for HIV Infection
in Pregnant Women: A Systematic Review for the U.S. Preventive
Services Task Force (163); Screening for Hepatitis B in Pregnant
Women: Updated Evidence Report and Systematic Review
for the U.S. Preventive Services Task Force (164); and CDC
Recommendations for Hepatitis C Screening Among Adults —
United States, 2020 (156).
Adolescents
In the United States, prevalence rates of certain STIs are
highest among adolescents and young adults (141). For
example, reported rates of chlamydia and gonorrhea are highest
among females during their adolescent and young adult years,
and many persons acquire HPV infection during that time.
Persons who initiate sex early in adolescence are at higher risk
for STIs, as are adolescents living in detention facilities; those
receiving services at STD clinics; those who are involved in
commercial sex exploitation or survival sex and are exchanging
sex for drugs, money, food, or housing; young males who
have sex with males (YMSM); transgender youths; and youths
with disabilities, substance misuse, or mental health disorders.
Factors contributing to increased vulnerability to STIs during
adolescence include having multiple sex partners, having
sequential sex partnerships of limited duration or concurrent
partnerships, failing to use barrier protection consistently
and correctly, having lower socioeconomic status, and facing
multiple obstacles to accessing health care (141,165).
All 50 states and the District of Columbia explicitly allow
minors to consent for their own STI services. No state requires
parental consent for STI care, although the age at which a
minor can provide consent for specified health care services
(i.e., HPV vaccination and HIV testing and treatment) varies
among states. In 2019, a total of 18 states allowed but did not
require physicians to notify parents of a minor’s receipt of STI
services, including states where minors can legally provide their
own consent to the service (https://www.cdc.gov/hiv/policies/
law/states/minors.html).
Protecting confidentiality for STI care, particularly for
adolescents enrolled in private health insurance plans, presents
multiple problems. After a claim has been submitted, many
states mandate that health plans provide a written statement
to the beneficiary indicating the service performed, the charges
covered, what the insurer allows, and the amount for which the
patient is responsible (i.e., explanation of benefits [EOB]) (166
169). In addition, federal laws obligate notices to beneficiaries
when claims are denied, including alerting beneficiaries who
need to pay for care until the allowable deductible is reached.
For STI testing and treatment-related care, an EOB or
medical bill that is received by a parent might disclose services
provided and list STI laboratory tests performed or treatment
administered. Some states have instituted mechanisms for
protecting adolescents’ confidentiality and limiting EOBs.
Additional risks to confidentiality breaches can inadvertently
occur through electronic health records, although technology
continues to evolve to assist with ensuring confidential care.
AAP and the Society for Adolescent Health and Medicine
(SAHM) have published guidance on strategies to address
emerging risks for confidentiality breaches associated with
health information technology (169).
AAP and the SAHM recommend that providers have
time alone with their adolescent patients that includes
assessment for sexual behavior. The AAP recommendations
are available at https://services.aap.org/en/news-room/
campaigns-and-toolkits/adolescent-health-care and the SAHM
Recommendations and Reports
14 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
recommendations are available at https://www.adolescenthealth.
org/My-SAHM/Login-or-Create-an-Account.aspx?returnurl=
%2fResources%2fClinical-Care-Resources%2fConfidentiality.
aspx. Discussions concerning sexual behavior should be tailored
for the patient’s developmental level and be aimed at identifying
risk behaviors (e.g., multiple partners; oral, anal, or vaginal
sex; or drug misuse behaviors). Careful, nonjudgmental, and
thorough counseling is particularly vital for adolescents who
might not feel comfortable acknowledging their engagement in
behaviors that make them more vulnerable to acquiring STIs.
Screening Recommendations
Recommendations for screening adolescents for STIs to
detect asymptomatic infections are based on disease severity
and sequelae, prevalence among the population, costs,
medicolegal considerations (e.g., state laws), and other factors.
Routine laboratory screening for common STIs is indicated
for all sexually active adolescents. The following screening
recommendations summarize published clinical prevention
guidelines for sexually active adolescents from federal agencies
and medical professional organizations.
Chlamydia
Routine screening for C. trachomatis infection on an annual
basis is recommended for all sexually active females aged
<25 years (149). Rectal chlamydial testing can be considered
for females on the basis of reported sexual behaviors and
exposure, through shared clinical decision-making between the
patient and the provider (170,171). Evidence is insufficient
to recommend routine screening for C. trachomatis among
sexually active young males, on the basis of efficacy and cost-
effectiveness. However, screening of sexually active young males
should be considered in clinical settings serving populations
of young men with a high prevalence of chlamydial infections
(e.g., adolescent service clinics, correctional facilities, and STD
clinics). Chlamydia screening, including pharyngeal or rectal
testing, should be offered to all YMSM at least annually on
the basis of sexual behavior and anatomic site of exposure (see
Men Who Have Sex with Men).
Gonorrhea
Routine screening for N. gonorrhoeae on an annual basis is
recommended for all sexually active females aged <25 years
(149). Extragenital gonorrhea screening (pharyngeal or rectal)
can be considered for females on the basis of reported sexual
behaviors and exposure, through shared clinical-decision
between the patient and the provider (170,171). Gonococcal
infection is more prevalent among certain geographic
locations and communities (141). Clinicians should consider
the communities they serve and consult local public health
authorities for guidance regarding identifying groups that are
more vulnerable to gonorrhea acquisition on the basis of local
disease prevalence. Evidence is insufficient to recommend
routine screening, on the basis of efficacy and cost-effectiveness,
for N. gonorrhoeae among asymptomatic sexually active young
males who have sex with females only. Screening for gonorrhea,
including pharyngeal or rectal testing, should be offered to
YMSM at least annually (see Men Who Have Sex with Men).
Providers might consider opt-out chlamydia and gonorrhea
screening (i.e., the patient is notified that testing will be
performed unless the patient declines, regardless of reported
sexual activity) for adolescent and young adult females during
clinical encounters. Cost-effectiveness analyses indicate that
opt-out chlamydia screening among adolescent and young
adult females might substantially increase screening, be cost-
saving (172), and identify infections among patients who do
not disclose sexual behavior (173).
HIV Infection
HIV screening should be discussed and offered to all
adolescents. Frequency of repeat screenings should be based on
the patient’s sexual behaviors and the local disease prevelance
(138). Persons with HIV infection should receive prevention
counseling and linkage to care before leaving the testing site.
Cervical Cancer
Guidelines from USPSTF and ACOG recommend that
cervical cancer screening begin at age 21 years (174,175). This
recommendation is based on the low incidence of cervical
cancer and limited usefulness of screening for cervical cancer
among adolescents (176). In contrast, the 2020 ACS guidelines
recommend that cervical cancer screening begin at age 25 years
with HPV testing. This change is recommended because the
incidence of invasive cervical cancer in women aged <25 years
is decreasing because of vaccination (177). Adolescents
with HIV infection who have initiated sexual intercourse
should have cervical screening cytology in accordance with
HIV/AIDS guidelines (https://clinicalinfo.hiv.gov/en/
guidelines/adult-and-adolescent-opportunistic-infection/
human-papillomavirus-disease?view=full).
Other Sexually Transmitted Infections
YMSM and pregnant females should be routinely screened
for syphilis (see Pregnant Women; Men Who Have Sex with
Men). Local disease prevalence can help guide decision-
making regarding screening for T. vaginalis, especially among
adolescent females in certain areas. Routine screening of
adolescents and young adults who are asymptomatic for certain
STIs (e.g., syphilis, trichomoniasis, BV, HSV, HAV, and HBV)
is not typically recommended.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 15
US Department of Health and Human Services/Centers for Disease Control and Prevention
Primary Prevention Recommendations
Primary prevention and anticipatory guidance for recognizing
symptoms and behaviors associated with STIs are strategies that
should be incorporated into all types of health care visits for
adolescents and young adults. The following recommendations
for primary prevention of STIs (i.e., vaccination and
counseling) are based on published clinical guidelines for
sexually active adolescents and young adults from federal
agencies and medical professional organizations.
HPV vaccination is recommended through age 26 years
for those not vaccinated previously at the routine age of
11 or 12 years (https://www.cdc.gov/vaccines/hcp/acip-
recs/vacc-specific/hpv.html).
The HBV vaccination series is recommended for all adolescents
and young adults who have not previously received the universal
HBV vaccine series during childhood (12).
The HAV vaccination series should be offered to
adolescents and young adults as well as those who have
not previously received the universal HAV vaccine series
during childhood (https://www.cdc.gov/vaccines/
schedules/hcp/imz/child-indications.html#note-hepa).
Information regarding HIV transmission, prevention, testing,
and implications of infection should be regarded as an essential
component of the anticipatory guidance provided to all
adolescents and young adults as part of routine health care.
CDC and USPSTF recommend offering HIV PrEP to
adolescents weighing ≥35 kg and adults who are HIV
negative and at substantial risk for HIV infection (80,178).
YMSM should be offered PrEP in youth-friendly settings
with tailored adherence support (e.g., text messaging and
visits per existing guidelines). Indications for PrEP, initial
and follow-up prescribing guidance, and laboratory testing
recommendations are the same for adolescents and adults
(https://www.cdc.gov/hiv/risk/prep).
Medical providers who care for adolescents and young
adults should integrate sexuality education into clinical
practice. Health care providers should counsel adolescents
about the sexual behaviors that are associated with risk for
acquiring STIs and should educate patients regarding
evidence-based prevention strategies, which includes a
discussion about abstinence and other risk-reduction
behaviors (e.g., consistent and correct condom use and
reduction in the number of sex partners including
concurrent partners). Interactive counseling approaches
(e.g., patient-centered counseling and motivational
interviewing) are effective STI and HIV prevention
strategies and are recommended by USPSTF. Educational
materials (e.g., handouts, pamphlets, and videos) can
reinforce office-based educational efforts.
Children
Management of children who have STIs requires close
cooperation among clinicians, laboratorians, and child-
protection authorities. Official investigations, when indicated,
should be initiated promptly. Certain diseases (e.g., gonorrhea,
syphilis, HIV, chlamydia, and trichomoniasis), if acquired after
the neonatal period, strongly indicate sexual contact. For other
diseases (e.g., HSV, HPV and anogenital warts, and vaginitis),
the association with sexual contact is not as clear (see Sexual
Assault and Abuse and STIs).
Men Who Have Sex with Men
MSM comprise a diverse group in terms of behaviors,
identities, and health care needs (179). The term “MSM often
is used clinically to refer to sexual behavior alone, regardless of
sexual orientation (e.g., a person might identify as heterosexual
but still be classified as MSM). Sexual orientation is
independent of gender identity. Classification of MSM can vary
in the inclusion of transgender men and women on the basis
of whether men are defined by sex at birth (i.e., transgender
women included) or current gender identity (i.e., transgender
men included). Therefore, sexual orientation as well as gender
identity of individual persons and their sex partners should be
obtained during health care visits. MSM might be at increased
risk for HIV and other STIs because of their sexual network or
behavioral or biologic factors, including number of concurrent
partners, condomless sex, anal sex, or substance use (180182).
These factors, along with sexual network or higher community
disease prevalence, can increase the risk for STIs among MSM
compared with other groups (183,184).
Performing a detailed and comprehensive sexual history is
the first step in identifying vulnerability and providing tailored
counseling and care (3). Factors associated with increased
vulnerability to STI acquisition among MSM include having
multiple partners, anonymous partners, and concurrent
partners (185,186). Repeat syphilis infections are common and
might be associated with HIV infection, substance use (e.g.,
methamphetamines), Black race, and multiple sex partners
(187). Similarly, gonorrhea incidence has increased among MSM
and might be more likely to display antimicrobial resistance
compared with other groups (188,189). Gonococcal infection
among MSM has been associated with similar risk factors to
syphilis, including having multiple anonymous partners and
substance use, especially methamphetamines (190). Disparities
in gonococcal infection are also more pronounced among certain
racial and ethnic groups of MSM (141).
Recommendations and Reports
16 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
HIV Risk Among Men Who Have Sex with Men
MSM are disproportionately at risk for HIV infection. In
the United States, the estimated lifetime risk for HIV infection
among MSM is one in six, compared with heterosexual men at
one in 524 and heterosexual women at one in 253 (191). These
disparities are further exacerbated by race and ethnicity, with
African American/Black and Hispanic/Latino MSM having a
one in two and a one in four lifetime risk for HIV infection,
respectively. For HIV, transmission occurs much more readily
through receptive anal sex, compared with penile-vaginal sex
(192). Similar to other STIs, multiple partners, anonymous
partners, condomless sex, and substance use are all associated
with HIV infection (193196). Importantly, other STIs
also might significantly increase the risk for HIV infection
(197199). An estimated 10% of new HIV infections were
attributable to chlamydial or gonococcal infection (81). A
substantial number of MSM remain unaware of their HIV
diagnosis (200). Clinical care involving MSM, including
those who have HIV infection, should involve asking about
STI-related risk factors and routine STI testing. Clinicians
should routinely ask MSM about their sexual behaviors and
symptoms consistent with common STIs, including urethral
discharge, dysuria, ulcers, rash, lymphadenopathy, and
anorectal symptoms that might be consistent with proctitis
(e.g., discharge, rectal bleeding, pain on defecation, or pain
during anal sex). However, certain STIs are asymptomatic,
especially at rectal and pharyngeal sites, and routine testing
is recommended. In addition, clinicians should provide
education and counseling regarding evidence-based safer-sex
approaches that have demonstrated effectiveness in reducing
STI incidence (see HIV Infection, Detection, Counseling,
and Referral).
Pre-Exposure Prophylaxis for HIV Prevention
PrEP is the use of medications for preventing an infection
before exposure. Studies have demonstrated that a daily oral
medication TDF/FTC is effective in preventing HIV acquisition,
and specifically among MSM (74,75,201). PrEP guidelines
provide information regarding sexually active persons who are at
substantial risk for acquiring HIV infection (having had anal or
vaginal sex during the previous 6 months with either a partner
with HIV infection, a bacterial STI in the past 6 months, or
inconsistent or no condom use with a sex partner) or persons
who inject drugs (injecting partner with HIV infection or sharing
injection equipment) (80). Those guidelines provide information
regarding daily PrEP use for either TDF/FTC (men or women)
or tenofovir alafenamide and emtricitabine for MSM. Screening
for bacterial STIs should occur at least every 6 months for all
sexually active patients and every 3 months among MSM or
among patients with ongoing risk behaviors. MSM taking PrEP
might compensate for decreased HIV acquisition risk by using
condoms less frequently or modifying their behavior in other ways
(202,203), although data regarding this behavior are inconsistent.
Studies have reported that MSM taking PrEP have high rates of
STIs, and frequent screening is warranted (204206).
Importance of Rectal and Pharyngeal Testing
Rectal and pharyngeal testing by NAAT for gonorrhea
and chlamydia is recognized as an important sexual health
consideration for MSM. Rectal gonorrhea and chlamydia are
associated with HIV infection (82,207), and men with repeat
rectal infections can be at substantially higher risk for HIV
acquisition (208). Pharyngeal infections with gonorrhea or
chlamydia might be a principal source of urethral infections
(209211). Studies have demonstrated that among MSM,
prevalence of rectal gonorrhea and chlamydia ranges from
0.2% to 24% and 2.1% to 23%, respectively, and prevalence
of pharyngeal gonorrhea and chlamydia ranges from 0.5% to
16.5% and 0% to 3.6%, respectively (171). Approximately
70% of gonococcal and chlamydial infections might be
missed if urogenital-only testing is performed among MSM
(212216) because most pharyngeal and rectal infections are
asymptomatic. Self-collected swabs have been reported to be
an acceptable means of collection for pharyngeal and rectal
specimens (217219), which can enhance patient comfort
and reduce clinical workloads.
A detailed sexual history should be taken for all MSM to
identify anatomic locations exposed to infection for screening.
Clinics that provide services for MSM at high risk should
consider implementing routine extragenital screening for
N. gonorrhoeae and C. trachomatis infections, and screening is
likely to be cost-effective (220).
Screening Recommendations
STI screening among MSM has been reported to be
suboptimal. In a cross-sectional sample of MSM in the United
States, approximately one third reported not having had an
STI test during the previous 3 years, and MSM with multiple
sex partners reported less frequent screening (221). MSM
living with HIV infection and engaged in care also experience
suboptimal rates of STI testing (222,223). Limited data exist
regarding the optimal frequency of screening for gonorrhea,
chlamydia, and syphilis among MSM, with the majority of
evidence derived from mathematical modeling. Models from
Australia have demonstrated that increasing syphilis screening
frequency from two times a year to four times a year resulted
in a relative decrease of 84% from peak prevalence (224). In
a compartmental model applied to different populations in
Canada, quarterly syphilis screening averted more than twice
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 17
US Department of Health and Human Services/Centers for Disease Control and Prevention
the number of syphilis cases, compared with semiannual
screening (225). Furthermore, MSM screening coverage
needed for eliminating syphilis among a population is
substantially reduced from 62% with annual screening to 23%
with quarterly screening (226,227). In an MSM transmission
model that explored the impact of HIV PrEP use on STI
prevalence, quarterly chlamydia and gonorrhea screening was
associated with an 83% reduction in incidence (205). The only
empiric data available that examined the impact of screening
frequency come from an observational cohort of MSM using
HIV PrEP in which quarterly screening identified more
bacterial STIs, and semiannual screening would have resulted
in delayed treatment of 35% of total identified STI infections
(206). In addition, quarterly screening was reported to have
prevented STI exposure in a median of three sex partners
per STI infection (206). On the basis of available evidence,
quarterly screening for gonorrhea, chlamydia, and syphilis for
certain sexually active MSM can improve case finding, which
can reduce the duration of infection at the population level,
reduce ongoing transmission and, ultimately, prevalence among
this population (228).
Preventive screening for common STIs is indicated for all
MSM. The following screening recommendations summarize
published federal agency and USPSTF clinical prevention
guidelines for MSM and should be performed at least annually.
HIV Infection
HIV serologic testing is indicated if HIV status is unknown
or if HIV negative and the patient or their sex partner has had
more than one sex partner since the most recent HIV test.
Syphilis
Syphilis serologic testing is indicated to establish whether
persons with reactive tests have untreated syphilis, have partially
treated syphilis, or are manifesting a slow or inadequate
serologic response to recommended previous therapy.
Gonorrhea and Chlamydia
The following testing is recommended for MSM:
A test for urethral infection* with N. gonorrhoeae and
C. trachomatis among men who have had insertive
intercourse during the preceding year (urine NAAT
is preferred).
A test for rectal infection* with N. gonorrhoeae and
C. trachomatis among men who have had receptive anal
intercourse during the preceding year (rectal NAAT
is preferred).
* Regardless of condom use during exposure.
A test for pharyngeal infection* with N. gonorrhoeae among
men who have had receptive oral intercourse during the
preceding year (pharyngeal NAAT is preferred).
Testing for C. trachomatis pharyngeal infection is not
recommended.
Basing screening practices solely on history might be
suboptimal because providers might feel uncomfortable
taking a detailed sexual history (229), men might also feel
uncomfortable sharing personal sexual information with
their provider, and rectal and pharyngeal infections can be
identified even in the absence of reported risk behaviors (171).
Furthermore, the role of saliva, kissing, and rimming (i.e.,
oral-rectal contact) in the transmission of N. gonorrhoeae and
C. trachomatis has not been well studied (230232).
Rectal and pharyngeal testing (provider-collected or self-
collected specimens) should be performed for all MSM who
report exposure at these sites. Testing can be offered to MSM
who do not report exposure at these sites after a detailed
explanation, due to known underreporting of risk behaviors.
All MSM with HIV infection entering care should be screened
for gonorrhea and chlamydia at appropriate anatomic sites of
exposure as well as for syphilis.
More frequent STI screening (i.e., for syphilis, gonorrhea,
and chlamydia) at 3- to 6-month intervals is indicated for
MSM, including those taking PrEP and those with HIV
infection, if risk behaviors persist or if they or their sex partners
have multiple partners. In addition, providers can consider
the benefits of offering more frequent HIV screening (e.g.,
every 3–6 months) to MSM at increased risk for acquiring
HIV infection.
Hepatitis B Virus
All MSM should be screened with HBsAg, HBV core
antibody, and HBV surface antibody testing to detect HBV
infection (233). Vaccination against both HAV and HBV
is recommended for all MSM for whom previous infection
or vaccination cannot be documented. Serologic testing can
be considered before vaccinating if the patient’s vaccination
history is unknown; however, vaccination should not be
delayed. Vaccinating persons who have had previous infection
or vaccination does not increase the risk for vaccine-related
adverse events (see Hepatitis A Virus; Hepatitis B Virus).
Hepatitis C Virus
CDC recommends HCV screening at least once for all
adults aged ≥18 years, except in settings where the prevalence
of HCV infection (HCV RNA positivity) is <0.1% (156).
The American Association for the Study of Liver Diseases/
Infectious Diseases Society of America guidelines recommend
all MSM with HIV infection be screened for HCV during the
Recommendations and Reports
18 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
initial HIV evaluation and at least annually thereafter (https://
www.hcvguidelines.org). More frequent screening depends
on ongoing risk behaviors, high-risk sexual behavior, and
concomitant ulcerative STIs or STI-related proctitis. Sexual
transmission of HCV can occur and is most common among
MSM with HIV infection (234237). Screening for HCV in
this setting is cost-effective (238,239). Screening should be
performed by using HCV antibody assays followed by HCV
RNA testing for those with a positive antibody test. Suspicion
for acute HCV infection (e.g., clinical evidence of hepatitis and
risk behaviors) should prompt consideration for HCV RNA
testing, despite a negative antibody test.
Human Papillomavirus
HPV infection and associated conditions (e.g., anogenital
warts and anal squamous intraepithelial lesions) are highly
prevalent among MSM. The HPV vaccination is recommended
for all men, including MSM and transgender persons or
immunocompromised males, including those with HIV
infection, through age 26 years (11). More information is
available at https://www.cdc.gov/hpv/downloads/9vhpv-
guidance.pdf.
A digital anorectal examination (DARE) should be
performed to detect early anal cancer among persons with HIV
and MSM without HIV but who have a history of receptive
anal intercourse. Data are insufficient to recommend routine
anal cancer screening with anal cytology in populations at risk
for anal cancer (see Anal Cancer). Health centers that initiate a
cytology-based screening program should only do so if referrals
to high-resolution anoscopy (HRA) and biopsy are available.
Herpes Simplex Virus-2
Evaluation for HSV-2 infection with type-specific serologic
tests also can be considered if infection status is unknown
among persons with previously undiagnosed genital tract
infection (see Genital Herpes).
Postexposure Prophylaxis and Pre-Exposure
Prophylaxis for STI Prevention
Studies have reported that a benefit might be derived
from STI PEP and PrEP for STI prevention. One study
demonstrated that monthly oral administration of a 1-g dose
of azithromycin reduced infection with N. gonorrhoeae and
C. trachomatis but did not decrease the incidence of HIV
transmission (240). Among MSM, doxycycline taken as PEP in
a single oral dose ≤24 hours after sex decreased infection with
Treponema pallidum and C. trachomatis; however, no substantial
effect was observed for infection with N. gonorrhoeae (93).
Doxycycline taken as STI PrEP as 100 mg orally once daily
also demonstrated a substantial reduction in gonorrhea,
chlamydia, and syphilis among MSM (90). However, these
studies had limitations because of small sample size, short
duration of therapy, and concerns about antibiotic resistance,
specifically regarding N. gonorrhoeae (241). Further study is
needed to determine the effectiveness of using antimicrobials
for STI PrEP or PEP.
Counseling and Education Approaches
Different counseling and STI prevention strategies are
needed to effectively engage different groups of MSM.
Outreach efforts should be guided by local surveillance efforts
and community input. Engaging MSM at risk through social
media, specifically online hookup sites, is an important
outreach effort to consider. Hookup sites are Internet sites
and mobile telephone applications that men might use for
meeting other men for sex. Internet use might facilitate sexual
encounters and STI transmission among MSM, and many
men report using hookup sites to meet partners (242245).
The ease and accessibility of meeting partners online might
reduce stigma and barriers of meeting partners through
other settings. Moreover, these sites offer an opportunity for
effective STI prevention messaging (246), although the cost
might be limiting (247). Different groups of MSM might use
different hookup sites, and efforts should be guided by local
community input. Studies have demonstrated the acceptability
and feasibility of reaching MSM through these hookup sites
to promote STI prevention efforts (248,249).
Enteric Infections Among Men Who Have Sex
with Men
The importance of sexual transmission of enteric pathogens
among MSM has been recognized since the 1970s, after the
first report of MSM-associated shigellosis was reported in
San Francisco (250,251). Global increases in the incidence
of shigellosis among adult MSM have been more recently
observed (252256). Sporadic outbreaks of Shigella sonnei
and Shigella flexneri have been reported among MSM
(257262). Transmission occurs through oral-anal contact
or sexual contact, and transmission efficiency is enhanced by
both biologic or host and behavioral factors. HIV without
viral suppression can be an independent risk factor that
can contribute to transmission by increasing shedding of
the enteric pathogen, increasing susceptibility of the host,
or both (255,263). Surveillance data in England during
2004–2015 demonstrated that 21% of nontravel-associated
Shigella diagnoses among MSM were among persons with
HIV infection (255).
Other enteric organisms might also cause disease among
MSM through sexual activities leading to oral-anal contact,
including bacteria such as Escherichia coli (264) and
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 19
US Department of Health and Human Services/Centers for Disease Control and Prevention
Campylobacter jejuni or Campylobacter coli (265,266); viruses
such as HAV (267); and parasites such as Giardia lamblia or
Entamoeba histolytica (268,269). Behavioral characteristics
associated with the sexual transmission of enteric infections
are broadly similar to those associated with other STIs (e.g.,
gonorrhea, syphilis, and lymphogranuloma venereum [LGV]).
This includes multiple sex partners and online hookup sites
that increase opportunities for sexual mixing, which might
create dense sexual networks that facilitate STI transmission
among MSM (270). Specific behaviors associated with sexually
transmitted enteric infections among MSM involve attendance
at sex parties and recreational drug use including chem sex
(i.e., using crystal methamphetamine, gamma-butyrolactone,
or mephedrone before or during sex), which might facilitate
condomless sex, group sex, fisting, use of sex toys, and scat play
(253,271). The growing number of sexually transmitted enteric
infections might be attributable in part to the emergence of
antimicrobial resistance. This is well reported regarding Shigella
species, for which rapid intercontinental dissemination of a
S. flexneri 3a lineage with high-level resistance to azithromycin
through sexual transmission among MSM (272) and clusters
of multidrug resistant shigella cases among MSM have recently
been reported (273). Multidrug-resistant Campylobacter species
have also been documented (266,274). For MSM patients with
diarrhea, clinicians should request laboratory examinations,
including stool culture; provide counseling about the risk for
infection with enteric pathogens during sexual activity (oral-
anal, oral-genital, anal-genital, and digital-anal contact) that
could expose them to enteric pathogens; and choose treatment,
when needed, according to antimicrobial drug susceptibility.
Women Who Have Sex with Women and
Women Who Have Sex with
Women and Men
WSW and WSWM comprise diverse groups with variations
in sexual identity, practices, and risk behaviors. Studies
indicate that certain WSW, particularly adolescents, young
women, and WSWM, might be at increased risk for STIs
and HIV on the basis of reported risk behaviors (275280).
Studies have highlighted the diversity of sexual practices and
examined use of protective or risk-reduction strategies among
WSW populations (281283). Use of barrier protection with
female partners (e.g., gloves during digital-genital sex, external
condoms with sex toys, and latex or plastic barriers [also known
as dental dams for oral-genital sex]) was infrequent in all
studies. Although health organizations have online materials
directed to patients, few comprehensive and reliable resources
of sexual health information for WSW are available (284).
Recent studies regarding STI rates among WSW and
WSWM indicate that WSWM experience higher rates of STIs
than WSW, with rates comparable with women who have sex
with men (WSM) in all studies reviewed (279,285,286). These
studies indicate that WSW might experience STIs at lower
rates than WSWM and WSM, although still at significant
rates (287). One study reported higher sexual-risk behaviors
among adolescent WSWM and WSW than among adolescent
WSM (280). WSW report reduced knowledge of STI risks
(288), and both WSW and WSWM experience barriers to care,
especially Black WSW and WSWM (289,290). In addition, a
continuum of sexual behaviors reported by WSW and WSWM
indicates the need for providers to not assume lower risk for
WSW, highlighting the importance of an open discussion
about sexual health.
Few data are available regarding the risk for STIs conferred
by sex between women; however, transmission risk probably
varies by the specific STI and sexual practice (e.g., oral-genital
sex; vaginal or anal sex using hands, fingers, or penetrative
sex items; and oral-anal sex) (291,292). Practices involving
digital-vaginal or digital-anal contact, particularly with shared
penetrative sex items, present a possible means for transmission
of infected cervicovaginal or anal secretions. This possibility
is most directly supported by reports of shared trichomonas
infections (293,294) and by concordant drug-resistance
genotype testing and phylogenetic linkage analysis identifying
HIV transmitted sexually between women (295,296). The
majority of WSW (53%–97%) have had sex with men in the
past and continue to do so, with 5%–28% of WSW reporting
male partners during the previous year (292,297300).
HPV can be transmitted through skin-to-skin contact,
and sexual transmission of HPV likely occurs between WSW
(301303). HPV DNA has been detected through polymerase
chain reaction (PCR)–based methods from the cervix, vagina,
and vulva among 13%–30% of WSW (301,302) and can
persist on fomites, including sex toys (304). Among WSW
who report no lifetime history of sex with men, 26% had
antibodies to HPV-16, and 42% had antibodies to HPV-6
(301). High-grade squamous intraepithelial lesions (HSIL) and
low-grade squamous intraepithelial lesions (LSIL) have been
detected on Papanicolaou smears (Pap tests) among WSW
who reported no previous sex with men (301,302). WSWM
are at risk for acquiring HPV from both their female partners
and male partners and thus are at risk for cervical cancer.
Therefore, routine cervical cancer screening should be offered
to all women, regardless of sexual orientation or practices,
and young adult WSW and WSWM should be offered HPV
vaccination in accordance with recommendations (11) (https://
www.cdc.gov/vaccines/hcp/acip-recs/vacc-specific/hpv.html).
Recommendations and Reports
20 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Genital transmission of HSV-2 between female sex partners
is inefficient but can occur. A U.S. population-based survey
among women aged 18–59 years demonstrated an HSV-2
seroprevalence of 30% among women reporting same-sex
partners during the previous year, 36% among women
reporting same-sex partners in their lifetime, and 24% among
women reporting no lifetime same-sex behavior (299). HSV-2
seroprevalence among women self-identifying as homosexual
or lesbian was 8%, similar to a previous clinic-based study of
WSW (299,305) but was 26% among Black WSW in one
study (287). The relatively frequent practice of orogenital sex
among WSW and WSWM might place them at higher risk
for genital infection with HSV-1, a hypothesis supported by
the recognized association between HSV-1 seropositivity and
previous number of female partners. Thus, sexual transmission
of HSV-1 and HSV-2 can occur between female sex partners.
This information should be communicated to women as part
of sexual health counseling.
Trichomonas is a relatively common infection among WSW
and WSWM, with prevalence rates higher than for chlamydia
or gonorrhea (306,307), and direct transmission of trichomonas
between female partners has been demonstrated (293,294).
Limited information is available regarding transmission
of bacterial STIs between female partners. Transmission of
syphilis between female sex partners, probably through oral
sex, has been reported. Although the rate of transmission of
C. trachomatis or N. gonorrhoeae between women is unknown,
infection also might be acquired from past or current male
partners. Data indicate that C. trachomatis infection among
WSW can occur (275,286,308,309). Data are limited
regarding gonorrhea rates among WSW and WSWM (170).
Reports of same-sex behavior among women should not deter
providers from offering and providing screening for STIs,
including chlamydia, according to guidelines.
BV is common among women, and even more so among
women with female partners (310312). Epidemiologic data
strongly demonstrate that BV is sexually transmitted among
women with female partners. Evidence continues to support
the association of such sexual behaviors as having a new partner,
having a partner with BV, having receptive oral sex, and having
digital-vaginal and digital-anal sex with incident BV (313,314).
A study including monogamous couples demonstrated
that female sex partners frequently share identical genital
Lactobacillus strains (315). Within a community-based
cohort of WSW, extravaginal (i.e., oral and rectal) reservoirs
of BV-associated bacteria were a risk factor for incident BV
(316). Studies have examined the impact of specific sexual
practices on the vaginal microflora (306,317319) and on
recurrent (320) or incident (321,322) BV among WSW. A
BV pathogenesis study in WSW reported that Prevotella bivia,
Gardnerella vaginalis, and Atopobium vaginae might have
substantial roles in development of incident BV (323). These
studies have continued to support, although have not proven,
the hypothesis that sexual behaviors, specific BV-associated
bacteria, and possibly exchange of vaginal or extravaginal
microbiota (e.g., oral bacterial communities) between partners
might be involved in the pathogenesis of BV among WSW.
Although BV is common among WSW, routine screening
for asymptomatic BV is not recommended. Results of one
randomized trial used a behavioral intervention to reduce
persistent BV among WSW through reduced sharing of vaginal
fluid on hands or sex toys. Women randomly assigned to the
intervention were 50% less likely to report receptive digital-
vaginal contact without gloves than control subjects, and they
reported sharing sex toys infrequently. However, these women
had no reduction in persistent BV at 1 month posttreatment
and no reduction in incident episodes of recurrent BV (324).
Trials have not been reported examining the benefits of treating
female partners of women with BV. Recurrent BV among
WSW is associated with having a same-sex partner and a lack of
condom use (325). Increasing awareness of signs and symptoms
of BV among women and encouraging healthy sexual practices
(e.g., avoiding shared sex toys, cleaning shared sex toys, and
using barriers) might benefit women and their partners.
Sexually active women are at risk for acquiring bacterial,
viral, and protozoal STIs from current and previous partners,
both male and female. WSW should not be presumed to be at
low or no risk for STIs on the basis of their sexual orientation.
Report of same-sex behavior among women should not deter
providers from considering and performing screening for STIs
and cervical cancer according to guidelines. Effective screening
requires that care providers and their female patients engage in
a comprehensive and open discussion of sexual and behavioral
risks that extends beyond sexual identity.
Transgender and Gender Diverse Persons
Transgender persons often experience high rates of stigma and
socioeconomic and structural barriers to care that negatively
affect health care usage and increase susceptibility to HIV and
STIs (326332). Persons who are transgender have a gender
identity that differs from the sex that they were assigned at birth
(333,334). Transgender women (also known as trans women,
transfeminine persons, or women of transgender experience)
are women who were assigned male sex at birth (born with
male anatomy). Transgender men (also known as trans men,
transmasculine persons, or men of transgender experience)
are men who were assigned female sex at birth (i.e., born with
female anatomy). In addition, certain persons might identify
outside the gender binary of male or female or move back and
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 21
US Department of Health and Human Services/Centers for Disease Control and Prevention
forth between different gender identities and use such terms
as “gender nonbinary,” “genderqueer,” or “gender fluid” to
describe themselves. Persons who use terms such as “agender”
or “null gender” do not identify with having any gender. The
term “cisgender” is used to describe persons who identify with
their assigned sex at birth. Prevalence studies of transgender
persons among the overall population have been limited and
often are based on small convenience samples.
Gender identity is independent of sexual orientation. Sexual
orientation identities among transgender persons are diverse.
Persons who are transgender or gender diverse might have sex
with cisgender men, cisgender women, or other transgender
or gender nonbinary persons.
Clinical Environment Assessment
Providers should create welcoming environments that
facilitate disclosure of gender identity and sexual orientation.
Clinics should document gender identity and sex assigned
at birth for all patients to improve sexual health care for
transgender and gender nonbinary persons. Assessment of
gender identity and sex assigned at birth has been validated
among diverse populations, has been reported to be acceptable
(335,336), and might result in increased patients identifying
as transgender (337).
Lack of medical provider knowledge and other barriers to care
(e.g., discrimination in health care settings or denial of services)
often result in transgender and gender nonbinary persons
avoiding or delaying preventive care services (338340) and
incurring missed opportunities for HIV and STI prevention
services. Gender-inclusive and trauma-guided health care
might increase the number of transgender patients who seek
sexual health services, including STI testing (341), because
transgender persons are at high risk for sexual violence (342).
Primary care providers should take a comprehensive sexual
history, including a discussion of STI screening, HIV PrEP
and PEP, behavioral health, and social determinants of sexual
health. Clinicians can improve the experience of sexual health
screening and counseling for transgender persons by asking for
their choice of terminology or modifying language (e.g., asking
patients their gender pronouns) to be used during clinic visits
and history taking and examination (343). Options for fertility
preservation, pregnancy potential, and contraception options
should also be discussed, if indicated. For transgender persons
who retain a uterus and ovaries, ovulation might continue in
the presence of testosterone therapy, and pregnancy potential
exists (https://transcare.ucsf.edu).
Transgender Women
A systematic review and meta-analysis of HIV infection
among transgender women estimated that HIV prevalence in
the United States is 14% among transgender women, with the
highest prevalence among Black (44%) and Hispanic (26%)
transgender women (344). Data also demonstrate high rates of
HIV infection among transgender women worldwide (345).
Bacterial STI prevalence varies among transgender women and
is based largely on convenience samples. Despite limited data,
international and U.S. studies have indicated elevated incidence
and prevalence of gonorrhea and chlamydia among transgender
women similar to rates among cisgender MSM (346348). A
recent study using data from the STD Surveillance Network
revealed that the proportions of transgender women with
extragenital chlamydial or gonococcal infections were similar
to those of cisgender MSM (349).
Providers caring for transgender women should have
knowledge of their patients’ current anatomy and patterns
of sexual behavior before counseling them about STI and
HIV prevention. The majority of transgender women have
not undergone genital-affirmation surgery and therefore
might retain a functional penis; in these instances, they
might engage in insertive oral, vaginal, or anal sex as well as
receptive oral or anal sex. In the U.S. Transgender Survey, 12%
of transgender women had undergone vaginoplasty surgery,
and approximately 50% more were considering surgical
intervention (350). Providers should have knowledge about
the type of tissue used to construct the neovagina, which
can affect future STI and HIV preventive care and screening
recommendations. The majority of vaginoplasty surgeries
conducted in the United States use penile and scrotal tissue
to create the neovagina (351). Other surgical techniques
use intestinal tissue (e.g., sigmoid colon graft) or split-skin
grafts (352). Although these surgeries involve penectomy and
orchiectomy, the prostate remains intact. Transgender women
who have had a vaginoplasty might engage in receptive vaginal,
oral, or anal sex.
Neovaginal STIs have infrequently been reported in
the literature and include HSV and HPV/genital warts in
penile-inversion vaginoplasty, C. trachomatis in procedures
that involved penile skin and grafts with urethra mucosa or
abdominal peritoneal lining (353), and N. gonorrhoeae in
both penile-inversion and colovaginoplasty (354359). If
the vaginoplasty used an intestinal graft, a risk also exists for
bowel-related disease (e.g., adenocarcinoma, inflammatory
bowel disease, diversion colitis, and polyps) (360362).
Transgender Men
The few studies of HIV prevalence among transgender men
indicated that they have a lower prevalence of HIV infection
than transgender women. A recent estimate of HIV prevalence
among transgender men was 2% (344). However, transgender
men who have sex with cisgender men might be at elevated
Recommendations and Reports
22 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
risk for HIV infection (332,363,364). Data are limited
regarding STI prevalence among transgender men, and the
majority of studies have used clinic-based data or convenience
sampling. Recent data from the STD Surveillance Network
demonstrated higher prevalence of gonorrhea and chlamydia
among transgender men, similar to rates reported among
cisgender MSM (365).
The U.S. Transgender Survey indicated that the proportion
of transgender men and gender diverse persons assigned
female sex at birth who have undergone gender-affirmation
genital surgery is low. Providers should consider the anatomic
diversity among transgender men because a person can undergo
a metoidioplasty (a procedure to increase the length of the
clitoris), with or without urethral lengthening, and might not
have a hysterectomy and oophorectomy and therefore be at risk
for bacterial STIs, HPV, HSV, HIV, and cervical cancer (366).
For transgender men using gender-affirming hormone therapy,
the decrease in estradiol levels caused by exogenous testosterone
can lead to vaginal atrophy (367,368) and is associated with a
high prevalence of unsatisfactory sample acquisition (369). The
impact of these hormonal changes on mucosal susceptibility
to HIV and STIs is unknown.
Transgender men who have not chosen to undergo
hysterectomy with removal of the cervix remain at risk for
cervical cancer. These persons often avoid cervical cancer
screening because of multiple factors, including discomfort
with medical examinations and fear of discrimination
(338,370). Providers should be aware that conducting a
speculum examination can be technically difficult after
metoidioplasty surgery because of narrowing of the introitus.
In these situations, high-risk HPV testing using a swab can
be considered; self-collected swabs for high-risk HPV testing
has been reported to be an acceptable option for transgender
men (371).
Screening Recommendations
The following are screening recommendations for
transgender and gender diverse persons:
Because of the diversity of transgender persons regarding
surgical gender-affirming procedures, hormone use, and
their patterns of sexual behavior, providers should remain
aware of symptoms consistent with common STIs and
screen for asymptomatic infections on the basis of the
patient’s sexual practices and anatomy.
Gender-based screening recommendations should be
adapted on the basis of anatomy (e.g., routine screening
for C. trachomatis and N. gonorrhoeae) as recommended
for all sexually active females aged <25 years on an annual
basis and should be extended to transgender men and
nonbinary persons with a cervix among this age group.
HIV screening should be discussed and offered to all
transgender persons. Frequency of repeat screenings should
be based on level of risk.
For transgender persons with HIV infection who have sex
with cisgender men and transgender women, STI
screening should be conducted at least annually, including
syphilis serology, HCV testing, and urogenital and
extragenital NAAT for gonorrhea and chlamydia.
Transgender women who have had vaginoplasty surgery
should undergo routine STI screening for all exposed sites
(e.g., oral, anal, or vaginal). No data are available regarding
the optimal screening method (urine or vaginal swab) for
bacterial STIs of the neovagina. The usual techniques for
creating a neovagina do not result in a cervix; therefore,
no rationale exists for cervical cancer screening (368).
If transgender men have undergone metoidioplasty surgery
with urethral lengthening and have not had a vaginectomy,
assessment of genital bacterial STIs should include a
cervical swab because a urine specimen will be inadequate
for detecting cervical infections.
Cervical cancer screening for transgender men and
nonbinary persons with a cervix should follow current
screening guidelines (see Human Papillomavirus Infections).
Persons in Correctional Facilities
Multiple studies have demonstrated that persons entering
correctional facilities have a high prevalence of STIs, HIV, and
viral hepatitis, especially those aged ≤35 years (141,372,373).
Risk behaviors for acquiring STIs (e.g., having condomless
sex, having multiple sex partners, substance misuse, and
engaging in commercial, survival, or coerced sex) are common
among incarcerated populations. Before their incarceration,
many persons have had limited access to medical care. Other
social determinants of health (e.g., insufficient social and
economic support or living in communities with high local
STI prevalence) are common. Addressing STIs in correctional
settings is vital for addressing the overall STI impact among
affected populations.
Growing evidence demonstrates the usefulness of expanded
STI screening and treatment services in correctional settings,
including short-term facilities (jails), long-term institutions
(prisons), and juvenile detention centers. For example, in
jurisdictions with comprehensive, targeted jail screening, more
chlamydial infections among females (and males if screened)
are detected and subsequently treated in the correctional setting
than in any other single reporting source (141,374) and might
represent the majority of reported cases in certain jurisdictions
(375). Screening in the jail setting has the potential to reach
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 23
US Department of Health and Human Services/Centers for Disease Control and Prevention
substantially more persons at risk than screening among the
prison population alone.
Both males and females aged ≤35 years in juvenile and adult
detention facilities have been reported to have higher rates of
chlamydia and gonorrhea than nonincarcerated persons in
the community (141,374,376). Syphilis seroprevalence rates,
which can indicate previously treated or current infection, are
considerably higher among incarcerated adult men and women
than among adolescents, which is consistent with the overall
national syphilis trends (141,374). Detection and treatment
of early syphilis in correctional facilities might affect rates
of transmission among adults and prevention of congenital
syphilis (377).
In jails, approximately half of entrants are released back
into the community within 48 hours. As a result, treatment
completion rates for those screened for STIs and who receive
STI diagnoses in short-term facilities might not be optimal.
However, because of the mobility of incarcerated populations
in and out of the community, the impact of screening in
correctional facilities on the prevalence of infections among
detainees and subsequent transmission in the community
after release might be considerable (378). Moreover, treatment
completion rates of ≥95% in short-term facilities can be
achieved by offering screening at or shortly after intake,
thus facilitating earlier receipt of test results and, if needed,
follow-up of untreated persons can be conducted through
public health outreach.
Universal, opt-out screening for chlamydia and gonorrhea
among females aged ≤35 years entering juvenile and adult
correctional facilities is recommended (379). Males aged
<30 years entering juvenile and adult correctional facilities
should also be screened for chlamydia and gonorrhea (380).
Opt-out screening has the potential to substantially increase
the number tested and the number of chlamydia and gonorrhea
infections detected (381385). Point-of-care (POC) NAAT
might also be considered if the tests have demonstrated
sufficient sensitivity and specificity. Studies have demonstrated
high prevalence of trichomoniasis among incarcerated
females (386392).
Screening Recommendations
Chlamydia and Gonorrhea
Females aged ≤35 years and males aged <30 years housed
in correctional facilities should be screened for chlamydia and
gonorrhea. This screening should be conducted at intake and
offered as opt-out screening.
Trichomonas
Females aged ≤35 years housed in correctional facilities
should be screened for trichomonas. This screening should be
conducted at intake and offered as opt-out screening.
Syphilis
Opt-out screening for incarcerated persons should be
conducted on the basis of the local area and institutional
prevalence of early (primary, secondary, or early latent)
infectious syphilis. Correctional facilities should stay apprised
of local syphilis prevalence. In short-term facilities, screening
at entry might be indicated.
Viral Hepatitis
All persons housed in juvenile and adult correctional facilities
should be screened at entry for viral hepatitis, including HAV,
HBV, and HCV, depending on local prevalence and the
persons vaccination status. Vaccination for HAV and HBV
should be offered if the person is susceptible.
Cervical Cancer
Women and transgender men who are housed in correctional
facilities should be screened for cervical cancer as for women
who are not incarcerated (393,394) (see Cervical Cancer).
HIV Infection
All persons being housed in juvenile and adult correctional
facilities should be screened at entry for HIV infection;
screening should be offered as opt-out screening. For those
identified as being at risk for HIV infection (e.g., with
diagnosed gonorrhea or syphilis or persons who inject drugs)
and being released into the community, starting HIV PrEP
(or providing linkage to a community clinic for HIV PrEP)
for HIV prevention should be considered (395,396). Persons
are likely to engage in high-risk activities immediately after
release from incarceration (397). For those identified with
HIV infection, treatment should be initiated. Those persons
receiving PrEP or HIV treatment should have linkage to
care established before release. Correctional settings should
consider implementing other STI prevention approaches,
both during incarceration and upon release, which might
include educational and behavioral counseling interventions
(398401), vaccination (e.g., for HPV) (402,403), condom
distribution (404,405), EPT (125), and PrEP to prevent HIV
infection (see Primary Prevention Methods).
Recommendations and Reports
24 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
HIV Infection
Detection, Counseling, and Referral
Infection with HIV causes an acute but brief and nonspecific
influenza-like retroviral syndrome that can include fever,
malaise, lymphadenopathy, pharyngitis, arthritis, or skin
rash. Most persons experience at least one symptom; however,
some might be asymptomatic or have no recognition of illness
(406409). Acute infection transitions to a multiyear, chronic
illness that progressively depletes CD4
+
T lymphocytes crucial
for maintenance of effective immune function. Ultimately,
persons with untreated HIV infection experience symptomatic,
life-threatening immunodeficiency (i.e., AIDS).
Effective ART that suppresses HIV replication to undetectable
levels reduces morbidity, provides a near-normal lifespan, and
prevents sexual transmission of HIV to others (9597,410
412). Early diagnosis of HIV and rapid linkage to care are
essential for achieving these goals. Guidelines from both the
U.S. Department of Health and Human Services and the
International AIDS Society–USA Panel recommend that all
persons with HIV infection be offered effective ART as soon
as possible, both to reduce morbidity and mortality and to
prevent HIV transmission (413).
STD specialty or sexual health clinics are a vital partner in
reducing HIV infections in the United States. These clinics
provide safety net services to vulnerable populations in need
of HIV prevention services who are not served by the health
care system and HIV partner service organizations. Diagnosis
of an STI is a biomarker for HIV acquisition, especially among
persons with primary or secondary syphilis or, among MSM,
rectal gonorrhea or chlamydia (197). STD clinics perform only
approximately 20% of all federally funded HIV tests nationally
but identify approximately 30% of all new infections (414).
Among testing venues, STD clinics are high performing in
terms of linkage to HIV care within 90 days of diagnosis;
during 2013–2017, the percentage of persons with a new
diagnosis in an STD clinic and linked to care within 90 days
increased from 55% to >90% (415,415).
Screening Recommendations
The following recommendations apply to testing for HIV:
HIV testing is recommended for all persons seeking STI
evaluation who are not already known to have HIV infection.
Testing should be routine at the time of the STI evaluation,
regardless of whether the patient reports any specific behavioral
risks for HIV. Testing for HIV should be performed at the time
of STI diagnosis and treatment if not performed at the initial
STI evaluation and screening (82,195,416).
CDC and USPSTF recommend HIV screening at least
once for all persons aged 15–65 years (417).
Persons at higher risk for HIV acquisition, including
sexually active gay, bisexual, and other MSM, should be
screened for HIV at least annually. Providers can consider
the benefits of offering more frequent screening (e.g., every
3–6 months) among MSM at increased risk for acquiring
HIV (418,419).
All pregnant women should be tested for HIV during the
first prenatal visit. A second test during the third trimester,
preferably at <36 weeks’ gestation, should be considered
and is recommended for women who are at high risk for
acquiring HIV infection, women who receive health care
in jurisdictions with high rates of HIV, and women
examined in clinical settings in which HIV incidence is
≥1 per 1,000 women screened per year (138,140).
HIV screening should be voluntary and free from coercion.
Patients should not be tested without their knowledge.
Opt-out HIV screening (notifying the patient that an HIV
test will be performed, unless the patient declines) is
recommended in all health care settings. CDC also
recommends that consent for HIV screening be
incorporated into the general informed consent for
medical care in the same manner as other screening or
diagnostic tests.
Requirement of specific signed consent for HIV testing is
not recommended. General informed consent for medical
care is considered sufficient to encompass informed
consent for HIV testing.
Providers should use a laboratory-based antigen/antibody
(Ag/Ab) combination assay as the first test for HIV, unless
persons are unlikely to follow up with a provider to receive
their HIV test results; in those cases screening with a rapid
POC test can be useful.
Preliminary positive screening tests for HIV should be
followed by supplemental testing to establish the diagnosis.
Providing prevention counseling as part of HIV screening
programs or in conjunction with HIV diagnostic testing is
not required (6). However, persons might be more likely to
think about HIV and consider their risk-related behavior
when undergoing an HIV test. HIV testing gives providers
an opportunity to conduct STI and HIV prevention
counseling and communicate risk-reduction messages.
Acute HIV infection can occur among persons who report
recent sexual or needle-sharing behavior or who have had
an STI diagnosis.
Providers should test for HIV RNA if initial testing according
to the HIV testing algorithm recommended by CDC is
negative or indeterminate when concerned about acute HIV
infection (https://stacks.cdc.gov/view/cdc/50872).
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 25
US Department of Health and Human Services/Centers for Disease Control and Prevention
Providers should not assume that a laboratory report of a
negative HIV Ag/Ab or antibody test indicates that the requisite
HIV RNA testing for acute HIV infection has been conducted.
They should consider explicitly requesting HIV RNA testing
when concerned about early acute HIV infection.
Providers should assess eligibility of all persons seeking
STI services for HIV PrEP and PEP. For persons with
substantial risk whose results are HIV negative, providers
should offer or provide referral for PrEP services, unless
the last potential HIV exposure occurred <72 hours, in
which case PEP might be indicated.
Diagnostic Considerations
HIV infection can be diagnosed by HIV 1/2 Ag/Ab
combination immunoassays. All FDA-cleared HIV tests are
highly sensitive and specific. Available serologic tests can
detect all known subtypes of HIV-1. The majority also detect
HIV-2 and uncommon variants of HIV-1 (e.g., group O and
group N).
According to an algorithm for HIV diagnosis, CDC
recommends that HIV testing begin with a laboratory-
based HIV-1/HIV-2 Ag/Ab combination assay, which, if
repeatedly reactive, is followed by a laboratory-based assay
with a supplemental HIV-1/HIV-2 antibody differentiation
assay (https://stacks.cdc.gov/view/cdc/50872). This algorithm
confers an additional advantage because it can detect HIV-2
antibodies after the initial immunoassay. Although HIV-2 is
uncommon in the United States, accurate identification is
vital because monitoring and therapy for HIV-2 differs from
that for HIV-1 (420). RNA testing should be performed
on all specimens with reactive immunoassay but negative
supplemental antibody test results to determine whether the
discordance represents acute HIV infection.
Rapid POC HIV tests can enable clinicians to make a
preliminary diagnosis of HIV infection in <20 minutes. The
majority of rapid antibody assays become reactive later in the
course of HIV infection than conventional laboratory-based
assays and thus can produce negative results among persons
recently infected (e.g., acutely infected persons). Furthermore,
HIV home-test kits only detect HIV antibodies and therefore
will not detect acute HIV infection. If early or acute infection
is suspected and a rapid HIV antibody assay is negative,
confirmatory testing with combined laboratory-based assays or
RNA testing should be performed. CDC recommends that all
persons with reactive rapid tests be assessed with a laboratory-
based Ag/Ab assay. Additional details about interpretation of
results by using the HIV testing algorithm recommended by
CDC are available at https://stacks.cdc.gov/view/cdc/48472.
Acute HIV Infection
Providers serving persons at risk for STIs are in a position
to diagnose HIV infection during its acute phase. Diagnosing
HIV infection during the acute phase is particularly important
because persons with acute HIV have highly infectious
disease due to the concentration of virus in plasma and
genital secretions, which is extremely elevated during that
stage of infection (421,422) (https://clinicalinfo.hiv.gov/en/
guidelines/adult-and-adolescent-arv/acute-and-recent-early-
hiv-infection?view=full). ART during acute HIV infection
is recommended because it substantially reduces infection
transmission to others, improves laboratory markers of
disease, might decrease severity of acute disease, lowers viral
setpoint, reduces the size of the viral reservoir, decreases
the rate of viral mutation by suppressing replication, and
preserves immune function (https://clinicalinfo.hiv.gov/en/
guidelines/adult-and-adolescent-arv/acute-and-recent-early-
hiv-infection?view=full). Persons who receive an acute HIV
diagnosis should be referred immediately to an HIV clinical
care provider, provided prevention counseling (e.g., advised to
reduce the number of partners and to use condoms correctly
and consistently), and screened for STIs. Information should be
provided regarding availability of PEP for sexual and injecting
drug use partners not known to have HIV infection if the
most recent contact was <72 hours preceding HIV diagnosis.
When providers test by using the CDC algorithm, specimens
collected during acute infection might give indeterminate or
negative results because insufficient anti-HIV antibodies and
potentially insufficient antigen are present to be reactive on
Ag/Ab combination assays and supplemental HIV-1/HIV-2
antibody differentiation assays. Whenever acute HIV infection
is suspected (e.g., initial testing according to the CDC algorithm
is negative or indeterminate after a possible sexual exposure to
HIV within the previous few days to weeks, especially if the
person has symptoms or has primary or secondary syphilis,
gonorrhea, or chlamydia), additional testing for HIV RNA
is recommended. If this additional testing for HIV RNA is
also negative, repeat testing in a few weeks is recommended
to rule out very early acute infection when HIV RNA might
not be detectable. A more detailed discussion of testing in
the context of acute HIV infection is available at https://
clinicalinfo.hiv.gov/en/guidelines/adult-and-adolescent-arv/
initiation-antiretroviral-therapy?view=full.
Treatment
ART should be initiated as soon as possible for all persons with
HIV infection regardless of CD4
+
T-cell count, both for individual
health and to prevent HIV transmission (https://clinicalinfo.hiv.
gov/sites/default/files/inline-files/AdultandAdolescentGL.pdf).
Recommendations and Reports
26 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Persons with HIV infection who achieve and maintain a viral load
suppressed to <200 copies/mL with ART have effectively no risk
for sexually transmitting HIV (9597,421). Early HIV diagnosis
and treatment is thus not only vital for individual health but also as
a public health intervention to prevent new infections. Knowledge
of the prevention benefit of treatment can help reduce stigma and
increase the persons commitment to start and remain adherent
to ART (423). The importance of adherence should be stressed
as well as the fact that ART does not protect against other STIs
that can be prevented by using condoms. Interventions to assist
persons to remain adherent to their prescribed HIV treatment, to
otherwise reduce the possibility of transmission to others, and to
protect themselves against STIs, have been developed for diverse
populations at risk (424) (https://clinicalinfo.hiv.gov/sites/default/
files/inline-files/AdultandAdolescentGL.pdf).
Comprehensive HIV treatment and care services might not
be available in facilities focused primarily on STI treatment.
Providers in such settings should be knowledgeable about HIV
treatment and care options available in their communities
and promptly link persons who have newly diagnosed HIV
infection and any persons with HIV infection who are not
engaged in ongoing effective care to a health care provider
or facility experienced in caring for persons living with HIV
(https://clinicalinfo.hiv.gov/sites/default/files/inline-files/
AdultandAdolescentGL.pdf).
Other HIV Management Considerations
Behavioral and psychosocial services are integral to caring for
persons with HIV infection. Providers should expect persons
to be distressed when first informed that they have HIV. They
face multiple adaptive challenges, including coping with the
reactions of others to a stigmatizing illness, developing and
adopting strategies to maintain physical and emotional health,
initiating changes in behavior to prevent HIV transmission to
others, and reducing the risk for acquiring additional STIs.
Many persons will require assistance gaining access to health
care and other support services and coping with changes in
personal relationships.
Persons with HIV infection might have additional needs
(e.g., referral for substance use or mental health disorders).
Others require assistance to secure and maintain employment
and housing. Persons capable of reproduction might require
family planning counseling, information about reproductive
health choices, and referral for reproductive health care.
The following recommendations apply to managing persons
with diagnosed HIV infection:
Link persons with HIV infection to care and start them
on ART as soon as possible.
Report cases (in accordance with local requirements) to
public health and initiate partner services.
Provide prevention counseling to persons with diagnosed
HIV infection.
Ensure all persons with HIV infection are informed that
if they achieve and maintain a suppressed viral load, they
have effectively no risk for transmitting HIV. Stress that
a suppressed viral load is not a substitute for condoms and
behavioral modifications because ART does not protect
persons with HIV against other STIs.
Provide additional counseling, either on-site or through
referral, about the psychosocial and medical implications
of having HIV infection.
Assess the need for immediate medical care and
psychosocial support.
Link persons with diagnosed HIV infection to services
provided by health care personnel experienced in managing
HIV infection. Additional services that might be needed
include substance misuse counseling and treatment,
treatment for mental health disorders or emotional distress,
reproductive counseling, risk-reduction counseling, and
case management. Providers should follow up to ensure
that patients have received services for any identified needs.
Persons with HIV infection should be educated about the
importance of ongoing medical care and what to expect
from these services.
STI Screening of Persons with HIV Infection in HIV
Care Settings
At the initial HIV care visit, providers should screen all
sexually active persons for syphilis, gonorrhea, and chlamydia,
and perform screening for these infections at least annually
during the course of HIV care (425). Specific testing
includes syphilis serology and NAAT for N. gonorrhoeae and
C. trachomatis at the anatomic site of exposure. Women should
also be screened for trichomoniasis at the initial visit and
annually thereafter. Women should be screened for cervical
cancer precursor lesions per existing guidelines (98).
More frequent screening for syphilis, gonorrhea, and
chlamydia (e.g., every 3 or 6 months) should be tailored
to individual risk and the local prevalence of specific STIs.
Certain STIs can be asymptomatic; their diagnosis might
prompt referral for partner services, might identify sexual and
needle-sharing partners who can benefit from early diagnosis
and treatment of HIV, and might prompt reengagement in
care or HIV prevention services (e.g., PEP or PrEP) (8). More
detailed information on screening, testing, and treatment is
provided in pathogen-specific sections of this report.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 27
US Department of Health and Human Services/Centers for Disease Control and Prevention
Partner Services and Reporting
Partner notification is a key component in the evaluation
of persons with HIV infection. Early diagnosis and treatment
of HIV among all potentially exposed sexual and injecting
drug sharing partners can improve their health and reduce
new infections. For those partners without HIV infection,
partner services also provide an opportunity for offering HIV
prevention services, including PrEP or PEP (if exposure was
<72 hours previous) and STI testing and treatment.
Health care providers should inform persons with diagnosed
HIV infection about any legal obligations of providers to report
cases of HIV to public health; the local confidential processes
for managing partner services, including that a public health
department still might be in contact to follow up in their care
and partner services; and the benefits and risks of partner
notification and services. Health care providers should also
encourage persons with a new HIV diagnosis to notify their
partners and provide them with referral information for their
partners about HIV testing. Partner notification for exposure
to HIV should be confidential. Health care providers can assist
in the partner notification process, either directly or by referral
to health department partner notification programs. Health
department staff are trained to use public health investigation
strategies for confidentially locating persons who can benefit
from HIV treatment, care, or prevention services. Guidance
regarding spousal notification varies by jurisdiction. Detailed
recommendations for notification, evaluation, and treatment of
exposed partners are available in Recommendations for Partner
Services Programs for HIV Infection, Syphilis, Gonorrhea, and
Chlamydial Infections (111).
Special Considerations
Pregnancy
All pregnant women should be tested for HIV during the
first prenatal visit. A second test during the third trimester,
preferably at <36 weeks’ gestation, should be considered and
is recommended for women who are at high risk for acquiring
HIV, women who receive health care in jurisdictions with high
rates of HIV infection, and women served in clinical settings
in which prenatal screening identifies ≥1 pregnant woman
with HIV per 1,000 women screened (138). Diagnostic
algorithms for HIV for pregnant women do not differ from
those for nonpregnant women (see STI Detection Among
Special Populations). Pregnant women should be informed
that HIV testing will be performed as part of the routine panel
of prenatal tests (138); for women who decline HIV testing,
providers should address concerns that pose obstacles, discuss
the benefits of testing (e.g., early HIV detection, treatment, and
care for improving health of the mother and reducing perinatal
transmission of HIV), and encourage testing at subsequent
prenatal visits. Women who decline testing because they have
had a previous negative HIV test result should be informed
about the importance of retesting during each pregnancy.
Women with no prenatal care should be tested for HIV at the
time of delivery.
Testing pregnant women is crucial because knowledge of
infection status can help maintain the womans health, and
it enables receipt of interventions (i.e., ART or specialized
obstetrical care) that can substantially reduce the risk for
perinatal transmission of HIV. Pregnant women with
diagnosed HIV infection should be educated about the benefits
of ART for their own health and for reducing the risk for HIV
transmission to their infant. In the absence of ART, a mother’s
risk for transmitting HIV to her neonate is approximately
30%; however, risk can be reduced to <2% through ART,
obstetrical interventions (i.e., elective cesarean delivery at
38 weeks’ pregnancy), and breastfeeding avoidance (https://
clinicalinfo.hiv.gov/sites/default/files/inline-files/PerinatalGL.
pdf). Pregnant women with HIV infection should be linked
to an HIV care provider experienced in managing HIV in
pregnancy and provided antenatal and postpartum treatment
and advice. Detailed and regularly updated recommendations
for managing pregnant patients with HIV infection are
available at https://clinicalinfo.hiv.gov/sites/default/files/inline-
files/PerinatalGL.pdf.
HIV Infection Among Neonates, Infants, and Children
Diagnosis of HIV infection in a pregnant woman indicates
the need for evaluating and managing the HIV-exposed
neonate and considering whether the womans other children,
if any, might be infected. Detailed recommendations regarding
diagnosis and management of HIV infection among neonates
and children of mothers with HIV are beyond the scope of
these guidelines but are available at https://clinicalinfo.hiv.
gov/en/guidelines. Exposed neonates and children with HIV
infection should be referred to physicians with expertise in
neonatal and pediatric HIV management.
Diseases Characterized by Genital,
Anal, or Perianal Ulcers
In the United States, the majority of young, sexually active
patients who have genital, anal, or perianal ulcers have either
genital herpes or syphilis. The frequency of each condition
differs by geographic area and population; however, genital
herpes is the most prevalent of these diseases. More than one
etiologic agent (e.g., herpes and syphilis) can be present in
any genital, anal, or perianal ulcer. Less common infectious
Recommendations and Reports
28 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
causes of genital, anal, or perianal ulcers include chancroid,
LGV, and granuloma inguinale (donovanosis). GUDs (e.g.,
syphilis, herpes, and LGV) might also present as oral ulcers.
Genital herpes, syphilis, chlamydia, gonorrhea, and chancroid
have been associated with an increased risk for HIV acquisition
and transmission. Genital, anal, or perianal lesions can also be
associated with infectious and noninfectious conditions that
are not sexually transmitted (e.g., yeast, trauma, carcinoma,
aphthae or Behcets disease, fixed drug eruption, or psoriasis).
A diagnosis based only on medical history and physical
examination frequently can be inaccurate. Therefore, all
persons who have genital, anal, or perianal ulcers should be
evaluated. Specific evaluation of genital, anal, or perianal ulcers
includes syphilis serology tests and darkfield examination
from lesion exudate or tissue, or NAAT if available; NAAT
or culture for genital herpes type 1 or 2; and serologic testing
for type-specific HSV antibody. In settings where chancroid is
prevalent, a NAAT or culture for Haemophilus ducreyi should
be performed.
No FDA-cleared NAAT for diagnosing syphilis is available
in the United States; however, multiple FDA-cleared NAATs
are available for diagnosing HSV-1 and HSV-2 in genital
specimens. Certain clinical laboratories have developed their
own syphilis and HSV NAATs and have conducted Clinical
Laboratory Improvement Amendment (CLIA) verification
studies with genital specimens. Type-specific serology for
HSV-2 might aid in identifying persons with genital herpes
(see Genital Herpes). In addition, biopsy of ulcers with
immunohistochemistry can help identify the cause of ulcers
that are unusual or that do not respond to initial therapy. HIV
testing should be performed on all persons not known to have
HIV infection who present with genital, anal, or perianal ulcers
(see Diagnostic Considerations in disease-specific sections).
NAAT testing at extragenital sites should be considered for
cases in which GUDs are suspected (e.g., oral manifestations
of syphilis, herpes, or LGV). Commercially available NAATs
have not been cleared by FDA for these indications; however,
they can be used by laboratories that have met regulatory
requirements for an off-label procedure.
Because early syphilis treatment decreases transmission
possibility, public health standards require health care
providers to presumptively treat any patient with a suspected
case of infectious syphilis at the initial visit, even before test
results are available. Presumptive treatment of a patient with a
suspected first episode of genital herpes also is recommended
because HSV treatment benefits depend on prompt therapy
initiation. The clinician should choose the presumptive
treatment on the basis of the clinical presentation (i.e., HSV
lesions begin as vesicles and primary syphilis as a papule) and
epidemiologic circumstances (e.g., high incidence of disease
among populations and communities and travel history). For
example, syphilis is so common among MSM that any male
who has sex with men presenting with a genital ulcer should be
presumptively treated for syphilis at the initial visit after syphilis
and HSV tests are performed. After a complete diagnostic
evaluation, >25% of patients who have genital ulcers might
not have a laboratory-confirmed diagnosis (426).
Chancroid
Chancroid prevalence has declined in the United States
(141). When infection does occur, it is usually associated
with sporadic outbreaks. Worldwide, chancroid appears to
have decreased as well, although infection might still occur in
certain Africa regions and the Caribbean. Chancroid is a risk
factor in HIV transmission and acquisition (197).
Diagnostic Considerations
A definitive diagnosis of chancroid requires identifying
H. ducreyi on special culture media that is not widely
available from commercial sources; even when these media
are used, sensitivity is <80% (427). No FDA-cleared NAAT
for H. ducreyi is available in the United States; however,
such testing can be performed by clinical laboratories that
have developed their own NAAT and have conducted CLIA
verification studies on genital specimens.
The combination of one or more deep and painful
genital ulcers and tender suppurative inguinal adenopathy
indicates the chancroid diagnosis; inguinal lymphadenitis
typically occurs in <50% of cases (428). For both clinical and
surveillance purposes, a probable diagnosis of chancroid can
be made if all of the following four criteria are met: 1) the
patient has one or more painful genital ulcers; 2) the clinical
presentation, appearance of genital ulcers and, if present,
regional lymphadenopathy are typical for chancroid; 3) the
patient has no evidence of T. pallidum infection by darkfield
examination or NAAT (i.e., ulcer exudate or serous fluid) or
by serologic tests for syphilis performed at least 7–14 days
after onset of ulcers; and 4) HSV-1 or HSV-2 NAAT or HSV
culture performed on the ulcer exudate or fluid are negative.
Treatment
Successful antimicrobial treatment for chancroid cures
the infection, resolves the clinical symptoms, and prevents
transmission to others. In advanced cases, genital scarring and
rectal or urogenital fistulas from suppurative buboes can result
despite successful therapy.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 29
US Department of Health and Human Services/Centers for Disease Control and Prevention
Recommended Regimens for Chancroid
Azithromycin 1 g orally in a single dose
or
Ceftriaxone 250 mg IM in a single dose
or
Ciprofloxacin 500 mg orally 2 times/day for 3 days
or
Erythromycin base 500 mg orally 3 times/day for 7 days
Azithromycin and ceftriaxone offer the advantage of
single-dose therapy (429). Worldwide, several isolates with
intermediate resistance to either ciprofloxacin or erythromycin
have been reported. However, because cultures are not routinely
performed, and chancroid is uncommon, data are limited
regarding prevalence of H. ducreyi antimicrobial resistance.
Other Management Considerations
Men who are uncircumcised and persons with HIV infection
do not respond as well to treatment as persons who are
circumcised or are HIV negative (430). Patients should be
tested for HIV at the time chancroid is diagnosed. If the initial
HIV test results were negative, the provider can consider the
benefits of offering more frequent testing and HIV PrEP to
persons at increased risk for HIV infection.
Follow-Up
Patients should be reexamined 3–7 days after therapy
initiation. If treatment is successful, ulcers usually improve
symptomatically within 3 days and objectively within 7 days
after therapy. If no clinical improvement is evident, the clinician
should consider whether the diagnosis is correct, another STI
is present, the patient has HIV infection, the treatment was
not used as instructed, or the H. ducreyi strain causing the
infection is resistant to the prescribed antimicrobial. The time
required for complete healing depends on the size of the ulcer;
large ulcers might require >2 weeks. In addition, healing can
be slower for uncircumcised men who have ulcers under the
foreskin. Clinical resolution of fluctuant lymphadenopathy is
slower than that of ulcers and might require needle aspiration
or incision and drainage, despite otherwise successful therapy.
Although needle aspiration of buboes is a simpler procedure,
incision and drainage might be preferred because of reduced
need for subsequent drainage procedures.
Management of Sex Partners
Regardless of whether disease symptoms are present, sex
partners of patients with chancroid should be examined and
treated if they had sexual contact with the patient during the
10 days preceding the patients symptom onset.
Special Considerations
Pregnancy
Data indicate ciprofloxacin presents a low risk to the
fetus during pregnancy but has potential for toxicity during
breastfeeding (431). Alternative drugs should be used if the
patient is pregnant or lactating. No adverse effects of chancroid
on pregnancy outcome have been reported.
HIV Infection
Persons with HIV infection who have chancroid infection
should be monitored closely because they are more likely to
experience chancroid treatment failure and to have ulcers
that heal slowly (430,432). Persons with HIV might require
repeated or longer courses of therapy, and treatment failures
can occur with any regimen. Data are limited concerning
the therapeutic efficacy of the recommended single-dose
azithromycin and ceftriaxone regimens among persons with
HIV infection.
Children
Because sexual contact is the major primary transmission route
among U.S. patients, diagnosis of chancroid ulcers among infants
and children, especially in the genital or perineal region, is highly
suspicious of sexual abuse. However, H. ducreyi is recognized as a
major cause of nonsexually transmitted cutaneous ulcers among
children in tropical regions and, specifically, countries where
yaws is endemic (433435). Acquisition of a lower-extremity
ulcer attributable to H. ducreyi in a child without genital ulcers
and reported travel to a region where yaws is endemic should
not be considered evidence of sexual abuse.
Genital Herpes
Genital herpes is a chronic, lifelong viral infection. Two types
of HSV can cause genital herpes: HSV-1 and HSV-2. Most
cases of recurrent genital herpes are caused by HSV-2, and
11.9% of persons aged 14–49 years are estimated to be infected
in the United States (436). However, an increasing proportion
of anogenital herpetic infections have been attributed to
HSV-1, which is especially prominent among young women
and MSM (186,437,438).
The majority of persons infected with HSV-2 have not
had the condition diagnosed, many of whom have mild or
unrecognized infections but shed virus intermittently in the
anogenital area. Consequently, most genital herpes infections
are transmitted by persons unaware that they have the
infection or who are asymptomatic when transmission occurs.
Management of genital HSV should address the chronic nature
of the infection rather than focusing solely on treating acute
episodes of genital lesions.
Recommendations and Reports
30 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Diagnostic Considerations
Clinical diagnosis of genital herpes can be difficult because the
self-limited, recurrent, painful, and vesicular or ulcerative lesions
classically associated with HSV are absent in many infected persons
at the time of clinical evaluation. If genital lesions are present,
clinical diagnosis of genital herpes should be confirmed by type-
specific virologic testing from the lesion by NAAT or culture (186).
Recurrences and subclinical shedding are much more frequent for
HSV-2 genital herpes infection than for HSV-1 genital herpes
(439,440). Therefore, prognosis and counseling depend on which
HSV type is present. Type-specific serologic tests can be used to
aid in the diagnosis of HSV infection in the absence of genital
lesions. Both type-specific virologic and type-specific serologic tests
for HSV should be available in clinical settings that provide care
to persons with or at risk for STIs. HSV-2 genital herpes infection
increases the risk for acquiring HIV twofold to threefold; therefore,
all persons with genital herpes should be tested for HIV (441).
Virologic Tests
HSV NAAT assays are the most sensitive tests because
they detect HSV from genital ulcers or other mucocutaneous
lesions; these tests are increasingly available (442444).
Although multiple FDA-cleared assays exist for HSV detection,
these tests vary in sensitivity from 90.9% to 100%; however,
they are considered highly specific (445447). PCR is also
the test of choice for diagnosing HSV infections affecting the
central nervous system (CNS) and systemic infections (e.g.,
meningitis, encephalitis, and neonatal herpes). HSV PCR
of the blood should not be performed to diagnose genital
herpes infection, except in cases in which concern exists for
disseminated infection (e.g., hepatitis). In certain settings, viral
culture is the only available virologic test. The sensitivity of viral
culture is low, especially for recurrent lesions, and decreases
rapidly as lesions begin to heal (443,448). Viral culture
isolates and PCR amplicons should be typed to determine
whether HSV-1 or HSV-2 is causing the infection. Failure to
detect HSV by NAAT or culture, especially in the presence
of older lesions or the absence of active lesions, does not
indicate an absence of HSV infection because viral shedding
is intermittent. Similarly, random or blind genital swabs in
the absence of lesions should not be used to diagnose genital
HSV infection because sensitivity is low, and a negative result
does not exclude the presence of HSV infection.
Cytologic detection of cellular changes associated with HSV
infection is an insensitive and nonspecific method of diagnosing
genital lesions (i.e., Tzanck preparation) and therefore should
not be relied on. Although a direct immunofluorescence
assay using fluorescein-labeled monoclonal antibodies is also
available for detecting HSV antigen from genital specimens,
this assay lacks sensitivity and is not recommended (449).
Type-Specific Serologic Tests
Both type-specific and type-common antibodies to HSV
develop during the first weeks after infection and persist
indefinitely. The majority of available, accurate type-
specific HSV serologic assays are based on the HSV-specific
glycoprotein G2 (gG2) (HSV-2) and glycoprotein G1 (gG1)
(HSV-1). Type-common antibody tests do not distinguish
between HSV-1 and HSV-2 infection; therefore, type-specific
serologic assays should be requested (450452).
Both laboratory-based assays and POC tests that provide
results for HSV-2 antibodies from capillary blood or serum
during a clinic visit are available. The sensitivity of glycoprotein
G type-specific tests for detecting HSV-2 antibody varies from
80% to 98%; false-negative results might be more frequent
at early stages of infection (451,453,454). Therefore, in
cases of recent suspected HSV-2 acquisition, repeat type-
specific antibody testing 12 weeks after the presumed time
of acquisition is indicated. The most commonly used test,
HerpeSelect HSV-2 enzyme immunoassay (EIA), often is
falsely positive at low index values (1.1–3.0) (457457). One
study reported an overall specificity of 57.4%, with a specificity
of 39.8% for index values of 1.1–2.9 (458). Because of the
poor specificity of commercially available type-specific EIAs,
particularly with low index values (<3.0), a confirmatory test
(Biokit or Western blot) with a second method should be
performed before test interpretation. Use of confirmatory
testing with the Biokit or the Western blot assays have been
reported to improve accuracy of HSV-2 serologic testing (459).
The HerpeSelect HSV-2 immunoblot should not be used for
confirmation because it uses the same antigen as the HSV-2
EIA. If confirmatory tests are unavailable, patients should be
counseled about the limitations of available testing before
obtaining serologic tests, and health care providers should
be aware that false-positive results occur. Immunoglobulin
M (IgM) testing for HSV-1 or HSV-2 is not useful because
IgM tests are not type specific and might be positive during
recurrent genital or oral episodes of herpes (460). Therefore,
HSV IgM testing is not recommended.
Because approximately all HSV-2 infections are sexually
acquired, presence of type-specific HSV-2 antibody implies
anogenital infection. In this instance, education and counseling
for persons with genital HSV infections should be provided.
The presence of HSV-1 antibody alone is more difficult
to interpret. HSV-1 serologic testing does not distinguish
between oral and genital infection and typically should not
be performed for diagnosing genital HSV-1 infection. Persons
with HSV-1 antibodies often have oral HSV infection acquired
during childhood, which might be asymptomatic. Lack of
symptoms in a person who is HSV-1 seropositive does not
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 31
US Department of Health and Human Services/Centers for Disease Control and Prevention
distinguish anogenital from orolabial or cutaneous infection,
and, regardless of site of infection, these persons remain at risk
for acquiring HSV-2. In addition, HSV-1 serologic testing
has low sensitivity for detection of HSV-1 antibody (458).
However, acquisition of HSV-1 genital herpes is increasing,
and HSV-1 genital herpes also can be asymptomatic (437
439,461,462). Diagnosis of HSV-1 infection is confirmed by
virologic tests from genital lesions.
Type-specific HSV-2 serologic assays for diagnosing
HSV-2 are useful in the following scenarios: recurrent or
atypical genital symptoms or lesions with a negative HSV
PCR or culture result, clinical diagnosis of genital herpes
without laboratory confirmation, and a patient’s partner has
genital herpes. HSV-2 serologic screening among the general
population is not recommended. Patients who are at higher
risk for infection (e.g., those presenting for an STI evaluation,
especially for persons with ≥10 lifetime sex partners, and
persons with HIV infection) might need to be assessed for a
history of genital herpes symptoms, followed by type-specific
HSV serologic assays to diagnose genital herpes for those with
genital symptoms.
Genital Herpes Management
Antiviral medication offers clinical benefits to symptomatic
patients and is the mainstay of management. The goals for
use of antiviral medications to treat genital herpes infection
are to treat or prevent symptomatic genital herpes recurrences
and improve quality of life and suppress the virus to prevent
transmission to sexual partners. Counseling regarding the
natural history of genital herpes, risks for sexual and perinatal
transmission, and methods for reducing transmission is also
integral to clinical management.
Systemic antiviral drugs can partially control the signs and
symptoms of genital herpes when used to treat first clinical and
recurrent episodes or when used as daily suppressive therapy.
However, these drugs neither eradicate latent virus nor affect
the risk, frequency, or severity of recurrences after the drug
is discontinued. Randomized trials have indicated that three
FDA-approved antiviral medications provide clinical benefit
for genital herpes: acyclovir, valacyclovir, and famciclovir
(463471). Valacyclovir is the valine ester of acyclovir and has
enhanced absorption after oral administration, allowing for
less frequent dosing than acyclovir. Famciclovir also has high
oral bioavailability. Topical therapy with antiviral drugs offers
minimal clinical benefit and is discouraged.
First Clinical Episode of Genital Herpes
Newly acquired genital herpes can cause a prolonged
clinical illness with severe genital ulcerations and neurologic
involvement. Even persons with first-episode herpes who have
mild clinical manifestations initially can experience severe or
prolonged symptoms during recurrent infection. Therefore,
all patients with first episodes of genital herpes should receive
antiviral therapy.
Recommended Regimens for First Clinical Episode of Genital
Herpes*
Acyclovir
400 mg orally 3 times/day for 7–10 days
or
Famciclovir 250 mg orally 3 times/day for 7–10 days
or
Valacyclovir 1 g orally 2 times/day for 7–10 days
* Treatment can be extended if healing is incomplete after 10 days of therapy.
Acyclovir 200 mg orally 5 times/day is also effective but is not
recommended because of the frequency of dosing.
Recurrent HSV-2 Genital Herpes
Almost all persons with symptomatic first-episode HSV-2
genital herpes subsequently experience recurrent episodes of
genital lesions. Intermittent asymptomatic shedding occurs
among persons with HSV-2 genital herpes infection, even
those with longstanding clinically silent infection. Antiviral
therapy for recurrent genital herpes can be administered either
as suppressive therapy to reduce the frequency of recurrences
or episodically to ameliorate or shorten the duration of lesions.
Certain persons, including those with mild or infrequent
recurrent outbreaks, benefit from antiviral therapy; therefore,
options for treatment should be discussed. Many persons prefer
suppressive therapy, which has the additional advantage of
decreasing the risk for transmitting HSV-2 genital herpes to
susceptible partners (472,473).
Suppressive Therapy for Recurrent HSV-2
Genital Herpes
Suppressive therapy reduces frequency of genital herpes
recurrences by 70%–80% among patients who have frequent
recurrences (469472). Persons receiving such therapy
often report having experienced no symptomatic outbreaks.
Suppressive therapy also is effective for patients with less
frequent recurrences. Long-term safety and efficacy have
been documented among patients receiving daily acyclovir,
valacyclovir, and famciclovir (474). Quality of life is improved
for many patients with frequent recurrences who receive
suppressive therapy rather than episodic treatment (475).
Providers should discuss with patients on an annual basis
whether they want to continue suppressive therapy because
frequency of genital HSV-2 recurrence diminishes over time
for many persons. However, neither treatment discontinuation
nor laboratory monitoring is necessary because adverse events
and development of HSV antiviral resistance related to long-
term antiviral use are uncommon.
Recommendations and Reports
32 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Treatment with valacyclovir 500 mg daily decreases the rate
of HSV-2 transmission for discordant heterosexual couples
in which a partner has a history of genital HSV-2 infection
(473). Such couples should be encouraged to consider
suppressive antiviral therapy as part of a strategy for preventing
transmission, in addition to consistent condom use and
avoidance of sexual activity during recurrences. Suppressive
antiviral therapy for persons with a history of symptomatic
genital herpes also is likely to reduce transmission when used
by those who have multiple partners. HSV-2 seropositive
persons without a history of symptomatic genital herpes
have a 50% decreased risk for genital shedding, compared
with those with symptomatic genital herpes (476). No data
are available regarding efficacy of suppressive therapy for
preventing HSV-2 transmission among discordant couples in
which a partner has a history of asymptomatic HSV-2 infection
identified by a positive HSV-2 serologic test. Among HSV-2
seropositive persons without HIV infection, oral TDF/FTC
and intravaginal tenofovir are ineffective at reducing the risk
for HSV-2 shedding or recurrences (477).
Recommended Regimens for Suppression of Recurrent HSV-2
Genital Herpes
Acyclovir 400 mg orally 2 times/day
or
Valacyclovir 500 mg orally once a day*
or
Valacyclovir 1 g orally once a day
or
Famciclovir 250 mg orally 2 times/day
* Valacyclovir 500 mg once a day might be less effective than other
valacyclovir or acyclovir dosing regimens for persons who have frequent
recurrences (i.e., ≥10 episodes/year).
Famciclovir appears somewhat less effective for suppression of
viral shedding (478). Ease of administration and cost also are
key considerations for prolonged treatment.
Recurrent HSV-1 Genital Herpes
Recurrences are less frequent after the first episode of HSV-1
genital herpes, compared with genital HSV-2 genital herpes,
and genital shedding rapidly decreases during the first year of
infection (479). No data are available regarding the efficacy
of suppressive therapy for preventing transmission among
persons with HSV-1 genital herpes infection. Because of
the decreased risk for recurrences and shedding, suppressive
therapy for HSV-1 genital herpes should be reserved for those
with frequent recurrences through shared clinical decision-
making between the patient and the provider.
Episodic Therapy for Recurrent HSV-2 Genital Herpes
Episodic treatment of recurrent herpes is most effective if
therapy is initiated within 1 day of lesion onset or during the
prodrome that precedes some outbreaks. The patient should
be provided with a supply of drug or a prescription for the
medication with instructions to initiate treatment immediately
when symptoms begin. Acyclovir, famciclovir, and valacyclovir
appear equally effective for episodic treatment of genital herpes
(466470).
Recommended Regimens for Episodic Therapy for Recurrent
HSV-2 Genital Herpes*
Acyclovir 800 mg orally 2 times/day for 5 days
or
Acyclovir 800 mg orally 3 times/day for 2 days
or
Famciclovir 1 g orally 2 times/day for 1 day
or
Famciclovir 500 mg orally once, followed by 250 mg 2 times/day for
2 days
or
Famciclovir 125 mg orally 2 times/day for 5 days
or
Valacyclovir 500 mg orally 2 times/day for 3 days
or
Valacyclovir 1 g orally once daily for 5 days
* Acyclovir 400 mg orally 3 times/day for 5 days is also effective but is not
recommended because of frequency of dosing.
Severe Disease
Intravenous (IV) acyclovir therapy (5–10 mg/kg body weight
IV every 8 hours) should be provided for patients who have severe
HSV disease or complications that necessitate hospitalization
(e.g., disseminated infection, pneumonitis, or hepatitis) or
CNS complications (e.g., meningitis or encephalitis). HSV-2
meningitis is a rare complication of HSV-2 genital herpes
infection that affects women more than men (480). IV therapy
should be considered until clinical improvement followed by
oral antiviral therapy to complete >10 days of total therapy.
Longer duration is recommended for CNS complications.
HSV-2 meningitis is characterized clinically by signs of
headache, photophobia, fever, meningismus, and cerebrospinal
fluid (CSF) lymphocytic pleocytosis, accompanied by mildly
elevated protein and normal glucose (481). Optimal therapies
for HSV-2 meningitis have not been well studied (482);
however, acyclovir 5–10 mg/kg body weight IV every 8 hours
until clinical improvement is observed, followed by high-dose
oral antiviral therapy (valacyclovir 1 g 3 times/day) to complete
a 10- to 14-day course of total therapy, is recommended.
For patients with previous episodes of documented HSV-2
meningitis, oral valacyclovir may be used for the entire
course during episodes of recurrent HSV-2 meningitis. A
randomized clinical trial indicated that suppressive therapy
(valacyclovir 500 mg 2 times/day) did not prevent recurrent
HSV-2 meningitis episodes; however, the dose might not
have been sufficient for CNS penetration (483). Valacyclovir
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 33
US Department of Health and Human Services/Centers for Disease Control and Prevention
500 mg 2 times/day is not recommended for suppression of
HSV-2 meningitis; higher doses have not been studied in
clinical trials. HSV meningitis should be distinguished from
encephalitis, which requires a longer course (14–21 days) of
IV therapy. Impaired renal function warrants an adjustment
in acyclovir dosage.
Hepatitis
Hepatitis is a rare manifestation of disseminated HSV
infection, often reported among pregnant women who
acquire HSV during pregnancy (484). Pregnant women in
any trimester can present with fever and hepatitis (markedly
elevated transaminases) but might not have any genital or skin
lesions. HSV hepatitis is associated with fulminant liver failure
and high mortality (25%). Therefore, a high index of suspicion
for HSV is necessary, with a confirmatory diagnosis by HSV
PCR from blood (485). Among pregnant women with fever
and unexplained severe hepatitis, disseminated HSV infection
should be considered, and empiric IV acyclovir should be
initiated pending confirmation (484).
Prevention
Consistent and correct condom use has been reported
in multiple studies to decrease, but not eliminate, the risk
for HSV-2 transmission from men to women (486488).
Condoms are less effective for preventing transmission from
women to men (489). Two randomized clinical trials of
medical male circumcision (MMC) demonstrated a decreased
risk for HSV-2 acquisition among men in Uganda and
South Africa (66,68). Results from a third trial conducted
in Kenya did not demonstrate a substantial difference in
HSV-2 acquisition among men who received MMC (490).
A systematic review indicated high consistency for decreased
risk for HSV-2 acquisition among women with a male partner
who underwent MMC (491). These data indicate that MMC
can be associated with decreased risk for HSV-2 acquisition
among adult heterosexual men and with decreased risk for
HSV-2 transmission from male to female partners.
Randomized clinical trials have demonstrated that PrEP with
daily oral TDF/FTC decreases the risk for HSV-2 acquisition
by 30% in heterosexual partnerships (492). Pericoital
intravaginal tenofovir 1% gel also decreases the risk for HSV-2
acquisition among heterosexual women (493). Among MSM
and transgender women, daily oral TDF/FTC decreases the
risk for severe ulcers with symptomatic genital HSV-2 infection
but not for HSV-2 acquisition (494). Insufficient evidence
exists that TDF/FTC use among those who are not at risk for
HIV acquisition will prevent HSV-2 infection, and it should
not be used for that sole purpose. Oral TDF does not prevent
HSV-2 acquisition among persons with HIV infection who
are taking TDF as part of their ART regimen (495). No data
indicate that antivirals (acyclovir, valacyclovir, or famciclovir)
can be taken as PrEP by persons without HSV-2 to prevent
its acquisition.
Counseling
Counseling of persons with genital herpes and their sex
partners is crucial for management. The goals of counseling
include helping patients cope with the infection and preventing
sexual and perinatal transmission. Although initial counseling
can be provided at the first visit, patients often benefit from
learning about the chronic aspects of the disease after the acute
illness subsides. Multiple resources, including Internet sites and
printed materials, are available to assist patients, their partners,
and clinicians who provide counseling (496,497) (https://www.
ashasexualhealth.org and https://www.cdc.gov/std/herpes).
Although the psychological effect of a serologic diagnosis of
HSV-2 infection in a person with asymptomatic or unrecognized
genital herpes appears minimal and transient (498,499), certain
persons with HSV infection might express anxiety concerning
genital herpes that does not reflect the actual clinical severity
of their disease; the psychological effect of HSV infection can
be substantial. Common concerns about genital herpes include
the severity of initial clinical manifestations, recurrent episodes,
sexual relationships and transmission to sex partners, and ability
to bear healthy children.
Symptomatic HSV-2 Genital Herpes
When counseling persons with symptomatic HSV-2 genital
herpes infection, the provider should discuss the following:
The natural history of the disease, with emphasis on the
potential for recurrent episodes, asymptomatic viral
shedding, and the attendant risks for sexual transmission
of HSV to occur during asymptomatic periods
(asymptomatic viral shedding is most frequent during the
first 12 months after acquiring HSV-2).
The effectiveness of daily suppressive antiviral therapy for
preventing symptomatic recurrent episodes of genital
herpes for persons experiencing a first episode or recurrent
genital herpes.
The effectiveness of daily use of valacyclovir in reducing
risk for transmission of HSV-2 among persons without
HIV (473) and use of episodic therapy to shorten the
duration of recurrent episodes.
The importance of informing current sex partners about
genital herpes and informing future partners before
initiating a sexual relationship.
The importance of abstaining from sexual activity with
uninfected partners when lesions or prodromal symptoms
are present.
Recommendations and Reports
34 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
The effectiveness of male latex condoms, which when used
consistently and correctly can reduce, but not eliminate,
the risk for genital herpes transmission (486488).
The type-specific serologic testing of partners of persons
with symptomatic HSV-2 genital herpes to determine
whether such partners are already HSV seropositive or
whether risk for acquiring HSV exists.
The low risk for neonatal HSV except when genital herpes
is acquired late in pregnancy or if prodrome or lesions are
present at delivery.
The increased risk for HIV acquisition among HSV-2
seropositive persons who are exposed to HIV (76,471).
The lack of effectiveness of episodic or suppressive therapy
among persons with HIV infection to reduce risk for
transmission to partners who might be at risk for
HSV-2 acquisition.
Asymptomatic HSV-2 Genital Herpes
When counseling persons with asymptomatic HSV-2 genital
herpes infection, the provider should consider the following:
Asymptomatic persons who receive a diagnosis of HSV-2
by type-specific serologic testing (with confirmatory testing,
if needed) should receive education about the symptoms of
genital herpes infection (see Diagnostic Considerations).
Episodic and suppressive antiviral therapies are used
predominantly to treat recurrences, prevent recurrences,
and prevent transmission to sex partners of persons with
symptomatic HSV-2 infection.
For patients with serological evidence of HSV-2 (with
combination testing if needed) without symptomatic
recurrences, neither episodic nor suppressive therapy is
indicated for prevention of recurrences (see Diagnostic
Considerations).
Among persons with asymptomatic infection, the efficacy
of suppressive therapy to prevent HSV-2 transmission to
sex partners has not been studied.
Because of the decreased risk for shedding among those
with asymptomatic HSV-2 genital herpes, the benefit of
suppressive therapy for preventing transmission is
unknown among this population.
HSV-1 Genital Herpes
When counseling persons with HSV-1 genital herpes
infection, the provider should consider the following:
Persons with virologic laboratory-documented
symptomatic HSV-1 genital herpes infection should be
educated that the risk for recurrent genital herpes and
genital shedding is lower with HSV-1 infection, compared
with HSV-2 infection.
Because of the decreased risk for recurrences and shedding,
suppressive therapy for HSV-1 genital herpes should be
reserved for those with frequent recurrences.
For patients with frequently recurring HSV-1 genital
herpes, suppressive therapy might be considered.
Suppressive therapy to prevent HSV-1 transmission to sex
partners has not been studied.
For persons with symptomatic HSV-1 genital herpes or
asymptomatic HSV-2 genital herpes, suppressive therapy can
be considered for those who have substantial psychosocial
distress caused by the diagnosis of genital herpes. For women
who have genital herpes, the providers who care for them
during pregnancy and those who will care for their newborn
infant should be informed of their infection (see Genital
Herpes During Pregnancy).
Management of Sex Partners
The sex partners of persons who have symptomatic
genital herpes can benefit from evaluation and counseling.
Symptomatic sex partners should be evaluated and treated in
the same manner as patients who have symptomatic genital
herpes. Asymptomatic sex partners of patients who have
symptomatic genital herpes should be asked about a history
of genital symptoms and offered type-specific serologic testing
for HSV-2. For partners without genital herpes, no data are
available on which to base a recommendation for PEP or
PrEP with antiviral medications or that they would prevent
acquisition, and this should not be offered to patients as a
prevention strategy.
Special Considerations
Drug Allergy, Intolerance, or Adverse Reactions
Allergic and other adverse reactions to oral acyclovir,
valacyclovir, and famciclovir are rare. Desensitization to
acyclovir has been described (500).
HIV Infection
Immunocompromised patients can have prolonged or severe
episodes of genital, perianal, or oral herpes. Lesions caused by
HSV are common among persons with HIV infection and
might be severe, painful, and atypical (501). HSV shedding is
increased among persons with HIV infection (502). Whereas
ART reduces the severity and frequency of symptomatic genital
herpes, frequent subclinical shedding still occurs (503,504).
Clinical manifestations of genital herpes might worsen during
immune reconstitution early after initiation of ART. HSV-2
type-specific serologic testing can be considered for persons
with HIV infection during their initial evaluation, particularly
among those with a history of genital symptoms indicative of
HSV infection.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 35
US Department of Health and Human Services/Centers for Disease Control and Prevention
Recommended therapy for first-episode genital herpes is
the same as for persons without HIV infection, although
treatment courses might need to be extended for lesion
resolution. Suppressive or episodic therapy with oral antiviral
agents is effective in decreasing the clinical manifestations of
HSV infection among persons with HIV (503,504). The risk
for GUD increases during the first 6 months after starting
ART, especially among persons who have a CD4
+
T-cell count
<200 cell/mm
3
. Suppressive antiviral therapy reduces the risk
for GUD among this population and can be continued for
6 months after ART initiation (504) when the risk for GUD
returns to baseline levels. Suppressive antiviral therapy among
persons with HIV and HSV infection does not reduce the
risk for either HIV transmission or HSV-2 transmission to
susceptible sex partners (88,505). Suppressive antiviral therapy
does not delay HIV disease progression and is not associated
with decreased risk for HIV-related inflammation among
persons taking ART (506). For severe HSV disease, initiating
therapy with acyclovir 5–10 mg/kg IV every 8 hours might
be necessary.
Recommended Regimens for Daily Suppression of Genital
Herpes Among Persons with HIV Infection
Acyclovir 400–800 mg orally 2–3 times/day
or
Famciclovir 500 mg orally 2 times/day
or
Valacyclovir 500 mg orally 2 times/day
Recommended Regimens for Episodic Genital Herpes Infection
Among Persons with HIV Infection
Acyclovir 400 mg orally 3 times/day for 5–10 days
or
Famciclovir 500 mg orally 2 times/day for 5–10 days
or
Valacyclovir 1 g orally 2 times/day for 5–10 days
Antiviral-Resistant HSV Infection
If lesions persist or recur in a patient receiving antiviral
treatment, acyclovir resistance should be suspected and a viral
culture obtained for phenotypic sensitivity testing (507).
Molecular testing for acyclovir resistance is not available.
Such persons should be managed in consultation with an
infectious disease specialist, and alternative therapy should be
administered. All acyclovir-resistant strains are also resistant
to valacyclovir, and the majority are resistant to famciclovir.
Foscarnet (40–80 mg/kg body weight IV every 8 hours until
clinical resolution is attained) is the treatment of choice for
acyclovir-resistant genital herpes (508,509). Intravenous
cidofovir 5 mg/kg body weight once weekly might also be
effective. Foscarnet and cidofovir are nephrotoxic medications
that require intensive laboratory monitoring and infectious
disease specialist consultation. Imiquimod 5% applied to the lesion
for 8 hours 3 times/week until clinical resolution is an alternative
that has been reported to be effective (510,511). Topical cidofovir
gel 1% can be applied to lesions 2–4 times daily; however, cidofovir
must be compounded at a pharmacy (512).
Prevention of antiviral resistance remains challenging among
persons with HIV infection. Experience with another group
of immunocompromised persons (e.g., hematopoietic stem-
cell recipients) demonstrated that persons receiving daily
suppressive antiviral therapy were less likely to experience
acyclovir-resistant HSV infection compared with those who
received episodic therapy for outbreaks (513).
Genital Herpes During Pregnancy
Prevention of neonatal herpes depends both on preventing
acquisition of genital herpes during late pregnancy and
avoiding exposure of the neonate to herpetic lesions and viral
shedding during delivery. Mothers of newborns who acquire
neonatal herpes often lack histories of clinically evident genital
herpes (514,515). The risk for transmission to the neonate from
an infected mother is high (30%–50%) among women who
acquire genital herpes near the time of delivery and low (<1%)
among women with prenatal histories of recurrent herpes or
who acquire genital herpes during the first half of pregnancy
(516,517). Women who acquire HSV in the second half of
pregnancy should be managed in consultation with maternal-
fetal medicine and infectious disease specialists.
All pregnant women should be asked whether they have
a history of genital herpes or genital symptoms concerning
for HSV infection. At the onset of labor, all women should
be questioned thoroughly about symptoms of genital herpes,
including prodromal symptoms (e.g., pain or burning at site
before appearance of lesion), and all women should be examined
thoroughly for herpetic lesions. Women without symptoms or
signs of genital herpes or its prodrome can deliver vaginally.
Although cesarean delivery does not eliminate the risk for HSV
transmission to the neonate (517), women with recurrent genital
herpetic lesions at the onset of labor should have a cesarean
delivery to reduce the risk for neonatal HSV infection.
Routine HSV-2 serologic screening of pregnant women is not
recommended. Women without known genital herpes should
be counseled to abstain from vaginal intercourse during the
third trimester with partners known to have or suspected of
having genital herpes. In addition, to prevent HSV-1 genital
herpes, pregnant women without known orolabial herpes
should be advised to abstain from receptive oral sex during the
third trimester with partners known to have or suspected to
have orolabial herpes. Type-specific serologic tests can be useful
for identifying pregnant women at risk for HSV infection and
for guiding counseling regarding the risk for acquiring genital
Recommendations and Reports
36 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
herpes during pregnancy. For example, such testing might be
offered to a woman with no history of genital herpes whose
sex partner has HSV infection. Many fetuses are exposed to
acyclovir each year, and the medication is believed to be safe
for use during all trimesters of pregnancy. A case-control study
reported an increased risk for the rare neonatal outcome of
gastroschisis among women who used antiviral medications
between the month before conception and the third month of
pregnancy (518). Acyclovir is also believed to be safe during
breastfeeding (431,519). Although data regarding prenatal
exposure to valacyclovir and famciclovir are limited, data from
animal trials indicate that these drugs also pose a low risk
among pregnant women (520). Acyclovir can be administered
orally to pregnant women with first-episode genital herpes or
recurrent herpes and should be administered IV to pregnant
women with severe HSV (see Genital Herpes, Hepatitis).
Suppressive acyclovir treatment starting at 36 weeks’ gestation
reduces the frequency of cesarean delivery among women who
have recurrent genital herpes by diminishing the frequency of
recurrences at term (521523). However, such treatment might
not protect against transmission to neonates in all cases (524).
No data support use of antiviral therapy among asymptomatic
HSV-seropositive women without a history of genital herpes.
In addition, the effectiveness of antiviral therapy among sex
partners with a history of genital herpes to decrease the risk
for HSV transmission to a pregnant woman has not been
studied. Additional information on the clinical management
of genital herpes in pregnancy is available through existing
guidelines (525).
Recommended Regimen for Suppression of Recurrent Genital
Herpes Among Pregnant Women*
Acyclovir 400 mg orally 3 times/day
or
Valacyclovir 500 mg orally 2 times/day
* Treatment recommended starting at 36 weeks’ gestation.
Neonatal Herpes
Newborn infants exposed to HSV during birth, as
documented by virologic testing of maternal lesions at delivery
or presumed by observation of maternal lesions, should be
followed clinically in consultation with a pediatric infectious
disease specialist. Detailed guidance is available regarding
management of neonates who are delivered vaginally in the
presence of maternal genital herpes lesions and is beyond the
scope of these guidelines; more information is available from
the AAP (https://redbook.solutions.aap.org). Surveillance
cultures or PCR of mucosal surfaces of the neonate to detect
HSV infection might be considered before the development of
clinical signs of neonatal herpes to guide treatment initiation.
In addition, administration of acyclovir might be considered
for neonates born to women who acquired HSV near term
because the risk for neonatal herpes is high for these newborn
infants. All newborn infants who have neonatal herpes should
be promptly evaluated and treated with systemic acyclovir.
The recommended regimen for infants treated for known or
suspected neonatal herpes is acyclovir 20 mg/kg body weight
IV every 8 hours for 14 days if disease is limited to the skin and
mucous membranes, or for 21 days for disseminated disease
and disease involving the CNS.
Granuloma Inguinale (Donovanosis)
Granuloma inguinale (donovanosis) is a genital ulcerative
disease caused by the intracellular gram-negative bacterium
Klebsiella granulomatis (formerly known as Calymmatobacterium
granulomatis). The disease occurs rarely in the United States;
however, sporadic cases have been described in India, South
Africa, and South America (526535). Although granuloma
inguinale was previously endemic in Australia, it is now
extremely rare (536,537). Clinically, the disease is characterized
as painless, slowly progressive ulcerative lesions on the genitals
or perineum without regional lymphadenopathy; subcutaneous
granulomas (pseudobuboes) also might occur. The lesions
are highly vascular (i.e., beefy red appearance) and can bleed.
Extragenital infection can occur with infection extension to
the pelvis, or it can disseminate to intra-abdominal organs,
bones, or the mouth. The lesions also can develop secondary
bacterial infection and can coexist with other sexually
transmitted pathogens.
Diagnostic Considerations
The causative organism of granuloma inguinale is difficult
to culture, and diagnosis requires visualization of dark-staining
Donovan bodies on tissue crush preparation or biopsy.
Although no FDA-cleared molecular tests for the detection of
K. granulomatis DNA exist, molecular assays might be useful
for identifying the causative agent.
Treatment
Multiple antimicrobial regimens have been effective;
however, only a limited number of controlled trials have
been published (538). Treatment has been reported to halt
progression of lesions, and healing typically proceeds inward
from the ulcer margins. Prolonged therapy is usually required to
permit granulation and reepithelialization of the ulcers. Relapse
can occur 6–18 months after apparently effective therapy.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 37
US Department of Health and Human Services/Centers for Disease Control and Prevention
Recommended Regimen for Granuloma Inguinale (Donovanosis)
Azithromycin 1 g orally once/week or 500 mg daily for >3 weeks and
until all lesions have completely healed
Alternative Regimens
Doxycycline 100 mg orally 2 times/day for at least 3 weeks and until all
lesions have completely healed
or
Erythromycin base 500 mg orally 4 times/day for >3 weeks and until all
lesions have completely healed
or
Trimethoprim-sulfamethoxazole one double-strength (160 mg/800
mg) tablet orally 2 time/day for >3 weeks and until all lesions have
completely healed
The addition of another antibiotic to these regimens can be
considered if improvement is not evident within the first few
days of therapy.
Other Management Considerations
Patients should be followed clinically until signs and
symptoms have resolved. All persons who receive a diagnosis
of granuloma inguinale should be tested for HIV.
Follow-Up
Patients should be followed clinically until signs and
symptoms resolve.
Management of Sex Partners
Persons who have had sexual contact with a patient who has
granuloma inguinale within the 60 days before onset of the
patient’s symptoms should be examined and offered therapy.
However, the value of empiric therapy in the absence of clinical
signs and symptoms has not been established.
Special Considerations
Pregnancy
Use of doxycycline in pregnancy might be associated
with discoloration of teeth; however, the risk is not well
defined. Doxycycline is compatible with breastfeeding (431).
Sulfonamides can be associated with neonatal kernicterus
among those with glucose-6-phospate dehydrogenase
deficiency and should be avoided during the third trimester
and while breastfeeding (431). For these reasons, pregnant and
lactating women with granuloma inguinale should be treated
with a macrolide regimen (erythromycin or azithromycin).
HIV Infection
Persons with granuloma inguinale and HIV infection should
receive the same regimens as those who do not have HIV.
Lymphogranuloma Venereum
LGV is caused by C. trachomatis serovars L1, L2, or L3
(539,540). LGV can cause severe inflammation and invasive
infection, in contrast with C. trachomatis serovars A–K that
cause mild or asymptomatic infection. Clinical manifestations
of LGV can include GUD, lymphadenopathy, or proctocolitis.
Rectal exposure among MSM or women can result in
proctocolitis, which is the most common presentation of
LGV infection (541), and can mimic inflammatory bowel
disease with clinical findings of mucoid or hemorrhagic
rectal discharge, anal pain, constipation, fever, or tenesmus
(542,543). Outbreaks of LGV proctocolitis have been
reported among MSM with high rates of HIV infection
(544547). LGV proctocolitis can be an invasive, systemic
infection and, if it is not treated early, can lead to chronic
colorectal fistulas and strictures; reactive arthropathy has also
been reported. However, reports indicate that rectal LGV can
also be asymptomatic (548). A common clinical manifestation
of LGV among heterosexuals is tender inguinal or femoral
lymphadenopathy that is typically unilateral. A self-limited
genital ulcer or papule sometimes occurs at the site of inoculation.
However, by the time persons seek care, the lesions have often
disappeared. LGV-associated lymphadenopathy can be severe,
with bubo formation from fluctuant or suppurative inguinal or
femoral lymphadenopathy. Oral ulceration can occur and might
be associated with cervical adenopathy (549551). Persons with
genital or colorectal LGV lesions can also experience secondary
bacterial infection or can be infected with other sexually and
nonsexually transmitted pathogens.
Diagnostic Considerations
A definitive LGV diagnosis can be made only with LGV-
specific molecular testing (e.g., PCR-based genotyping). These
tests can differentiate LGV from non–LGV C. trachomatis in
rectal specimens. However, these tests are not widely available,
and results are not typically available in a time frame that
would influence clinical management. Therefore, diagnosis is
based on clinical suspicion, epidemiologic information, and a
C. trachomatis NAAT at the symptomatic anatomic site, along
with exclusion of other etiologies for proctocolitis, inguinal
lymphadenopathy, or genital, oral, or rectal ulcers (551,552).
Genital or oral lesions, rectal specimens, and lymph node
specimens (i.e., lesion swab or bubo aspirate) can be tested
for C. trachomatis by NAAT or culture. NAAT is the preferred
approach for testing because it can detect both LGV strains and
non–LGV C. trachomatis strains (553). Therefore, all persons
presenting with proctocolitis should be tested for chlamydia
with a NAAT performed on rectal specimens. Severe symptoms
of proctocolitis (e.g., bloody discharge, tenesmus, and rectal
Recommendations and Reports
38 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
ulcers) indicate LGV. A rectal Gram stain with >10 white
blood cells (WBCs) has also been associated with rectal LGV
(545,554,555).
Chlamydia serology (complement fixation or
microimmunofluorescence) should not be used routinely as a
diagnostic tool for LGV because the utility of these serologic
methods has not been established, interpretation has not been
standardized, and validation for clinical proctitis presentation
has not been done. It might support an LGV diagnosis in
cases of isolated inguinal or femoral lymphadenopathy for
which diagnostic material for C. trachomatis NAAT cannot
be obtained.
Treatment
At the time of the initial visit (before diagnostic NAATs
for chlamydia are available), persons with a clinical syndrome
consistent with LGV should be presumptively treated.
Presumptive treatment for LGV is indicated among patients
with symptoms or signs of proctocolitis (e.g., bloody
discharge, tenesmus, or ulceration); in cases of severe inguinal
lymphadenopathy with bubo formation, particularly if the
patient has a recent history of a genital ulcer; or in the presence
of a genital ulcer if other etiologies have been ruled out. The
goal of treatment is to cure infection and prevent ongoing tissue
damage, although tissue reaction to the infection can result in
scarring. Buboes might require aspiration through intact skin
or incision and drainage to prevent formation of inguinal or
femoral ulcerations.
Recommended Regimen for Lymphogranuloma Venereum
Doxycycline 100 mg orally 2 times/day for 21 days
Alternative Regimens
Azithromycin 1 g orally once weekly for 3 weeks*
or
Erythromycin base 500 mg orally 4 times/day for 21 days
* Because this regimen has not been validated, a test of cure with C. trachomatis
NAAT 4 weeks after completion of treatment can be considered.
The optimal treatment duration for symptomatic LGV has
not been studied in clinical trials. The recommended 21-day
course of doxycycline is based on long-standing clinical practice
and is highly effective, with an estimated cure rate of >98.5%
(555,556). Shorter courses of doxycycline might be effective
on the basis of a small retrospective study of MSM with rectal
LGV, 50% of whom were symptomatic, who received a 7- to
14-day course of doxycycline and had a 97% cure rate (558).
Randomized prospective studies of shorter-course doxycycline
for treating LGV are needed. Longer courses of therapy might
be required in the setting of fistulas, buboes, and other forms
of severe disease (559).
A small nonrandomized study from Spain involving patients
with rectal LGV demonstrated cure rates of 97% with a
regimen of azithromycin 1 g once weekly for 3 weeks (560).
Pharmacokinetic data support this dosing strategy (561);
however, this regimen has not been validated. Fluoroquinolone-
based treatments also might be effective; however, the optimal
duration of treatment has not been evaluated. The clinical
significance of asymptomatic LGV is unknown, and it is
effectively treated with a 7-day course of doxycycline (562).
Other Management Considerations
Patients should be followed clinically until signs and
symptoms have resolved. Persons who receive an LGV diagnosis
should be tested for other STIs, especially HIV, gonorrhea, and
syphilis. Those whose HIV test results are negative should be
offered HIV PrEP.
Follow-Up
All persons who have been treated for LGV should be retested
for chlamydia approximately 3 months after treatment. If
retesting at 3 months is not possible, providers should retest
at the patient’s next visit for medical care within the 12-month
period after initial treatment.
Management of Sex Partners
Persons who have had sexual contact with a patient who
has LGV within the 60 days before onset of the patients
symptoms should be evaluated, examined, and tested for
chlamydial infection, depending on anatomic site of exposure.
Asymptomatic partners should be presumptively treated with
a chlamydia regimen (doxycycline 100 mg orally 2 times/day
for 7 days).
Special Considerations
Pregnancy
Use of doxycycline in pregnancy might be associated with
discoloration of teeth; however, the risk is not well defined
(563). Doxycycline is compatible with breastfeeding (431).
Azithromycin might prove useful for LGV treatment during
pregnancy, at a presumptive dose of 1 g weekly for 3 weeks;
no published data are available regarding an effective dose and
duration of treatment. Pregnant and lactating women with
LGV can be treated with erythromycin, although this regimen
is associated with frequent gastrointestinal side effects. Pregnant
women treated for LGV should have a test of cure performed
4 weeks after the initial C. trachomatis NAAT-positive test.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 39
US Department of Health and Human Services/Centers for Disease Control and Prevention
HIV Infection
Persons with LGV and HIV infection should receive the
same regimens as those who do not have HIV. Prolonged
therapy might be required because a delay in resolution of
symptoms might occur.
Syphilis
Syphilis is a systemic disease caused by T. pallidum. The
disease has been divided into stages on the basis of clinical
findings, which guide treatment and follow-up. Persons who
have syphilis might seek treatment for signs or symptoms.
Primary syphilis classically presents as a single painless ulcer
or chancre at the site of infection but can also present with
multiple, atypical, or painful lesions (564). Secondary syphilis
manifestations can include skin rash, mucocutaneous lesions,
and lymphadenopathy. Tertiary syphilis can present with
cardiac involvement, gummatous lesions, tabes dorsalis, and
general paresis.
Latent infections (i.e., those lacking clinical manifestations)
are detected by serologic testing. Latent syphilis acquired
within the preceding year is referred to as early latent syphilis;
all other cases of latent syphilis are classified as late latent
syphilis or latent syphilis of unknown duration.
T. pallidum can infect the CNS, which can occur at any stage
of syphilis and result in neurosyphilis. Early neurologic clinical
manifestations or syphilitic meningitis (e.g., cranial nerve
dysfunction, meningitis, meningovascular syphilis, stroke,
and acute altered mental status) are usually present within
the first few months or years of infection. Late neurologic
manifestations (e.g., tabes dorsalis and general paresis) occur
10 to >30 years after infection.
Infection of the visual system (ocular syphilis) or auditory
system (otosyphilis) can occur at any stage of syphilis but is
commonly identified during the early stages and can present
with or without additional CNS involvement. Ocular syphilis
often presents as panuveitis but can involve structures in
both the anterior and posterior segment of the eye, including
conjunctivitis, anterior uveitis, posterior interstitial keratitis,
optic neuropathy, and retinal vasculitis. Ocular syphilis can
result in permanent vision loss. Otosyphilis typically presents
with cochleo-vestibular symptoms, including tinnitus, vertigo,
and sensorineural hearing loss. Hearing loss can be unilateral or
bilateral, have a sudden onset, and progress rapidly. Otosyphilis
can result in permanent hearing loss.
Diagnostic Considerations
Darkfield examinations and molecular tests for detecting
T. pallidum directly from lesion exudate or tissue are
the definitive methods for diagnosing early syphilis and
congenital syphilis (565). Although no T. pallidum direct-
detection molecular NAATs are commercially available,
certain laboratories provide locally developed and validated
PCR tests for detecting T. pallidum DNA. A presumptive
diagnosis of syphilis requires use of two laboratory serologic
tests: a nontreponemal test (i.e., Venereal Disease Research
Laboratory [VDRL] or rapid plasma reagin [RPR] test)
and a treponemal test (i.e., the T. pallidum passive particle
agglutination [TP-PA] assay, various EIAs, chemiluminescence
immunoassays [CIAs] and immunoblots, or rapid treponemal
assays) (566568). At least 18 treponemal-specific tests are
cleared for use in the United States. Use of only one type of
serologic test (nontreponemal or treponemal) is insufficient
for diagnosis and can result in false-negative results among
persons tested during primary syphilis and false-positive results
among persons without syphilis or previously treated syphilis.
Nontreponemal Tests and Traditional Algorithm
False-positive nontreponemal test results can be associated
with multiple medical conditions and factors unrelated to
syphilis, including other infections (e.g., HIV), autoimmune
conditions, vaccinations, injecting drug use, pregnancy,
and older age (566,569). Therefore, persons with a reactive
nontreponemal test should always receive a treponemal test
to confirm the syphilis diagnosis (i.e., traditional algorithm).
Nontreponemal test antibody titers might correlate with disease
activity and are used for monitoring treatment response. Serum
should be diluted to identify the highest titer, and results should
be reported quantitatively. A fourfold change in titer, equivalent
to a change of two dilutions (e.g., from 1:16 to 1:4 or from 1:8
to 1:32), is considered necessary for demonstrating a clinically
significant difference between two nontreponemal test results
obtained by using the same serologic test, preferably from the
same manufacturer to avoid variation in results. Sequential
serologic tests for a patient should be performed using the
same testing method (VDRL or RPR), preferably by the same
laboratory. VDRL and RPR are equally valid assays; however,
quantitative results from the two tests cannot be compared
directly with each other because the methods are different,
and RPR titers frequently are slightly higher than VDRL titers.
Nontreponemal test titers usually decrease after treatment
and might become nonreactive with time. However, for certain
persons, nontreponemal antibodies might decrease less than
fourfold after treatment (i.e., inadequate serologic response)
or might decline appropriately but fail to serorevert and
Recommendations and Reports
40 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
persist for a long period. Atypical nontreponemal serologic
test results (e.g., unusually high, unusually low, or fluctuating
titers) might occur regardless of HIV status. When serologic
tests do not correspond with clinical findings indicative of
primary, secondary, or latent syphilis, presumptive treatment is
recommended for persons with risk factors for syphilis, and use
of other tests (e.g., biopsy for histology and immunostaining
and PCR of lesion) should be considered. For the majority
of persons with HIV infection, serologic tests are accurate
and reliable for diagnosing syphilis and evaluating response
to treatment.
Treponemal Tests and Reverse
Sequence Algorithm
The majority of patients who have reactive treponemal tests
will have reactive tests for the remainder of their lives, regardless
of adequate treatment or disease activity. However, 15%–25%
of patients treated during the primary stage revert to being
serologically nonreactive after 2–3 years (570). Treponemal
antibody titers do not predict treatment response and therefore
should not be used for this purpose.
Clinical laboratories sometimes screen syphilis serologic
samples by using automated treponemal immunoassays,
typically by EIA or CIA (571573). This reverse sequence
algorithm for syphilis testing can identify persons previously
treated for syphilis, those with untreated or incompletely
treated syphilis, and those with false-positive results that can
occur with a low likelihood of infection (574). Persons with
a positive treponemal screening test should have a standard
quantitative nontreponemal test with titer performed
reflexively by the laboratory to guide patient management
decisions. If the nontreponemal test is negative, the laboratory
should perform a treponemal test different from the one used
for initial testing, preferably TP-PA or treponemal assay based
on different antigens than the original test, to adjudicate the
results of the initial test.
If a second treponemal test is positive (e.g., EIA reactive,
RPR nonreactive, or TP-PA reactive), persons with a history
of previous treatment will require no further management
unless sexual history indicates a reexposure. In this instance, a
repeat nontreponemal test 2–4 weeks after a confirmed medical
history and physical examination is recommended to evaluate
for early infection. Those without a history of treatment for
syphilis should be offered treatment. Unless a medical history
or results of a physical examination indicate a recent infection,
previously untreated persons should be treated for syphilis of
unknown duration or late latent syphilis.
If the second treponemal test is negative (e.g., EIA reactive,
RPR nonreactive, TP-PA nonreactive) and the epidemiologic
risk and clinical probability for syphilis are low, further
evaluation or treatment is not indicated.
Multiple studies demonstrate that high quantitative index
values or high signal-to-cutoff ratio from treponemal EIA
or CIA tests correlate with TP-PA positivity, which might
eliminate the need for additional confirmatory testing;
however, the range of index values varies among different
treponemal immunoassays, and the values that correspond to
high levels of reactivity with confirmatory testing might differ
by immunoassay (567,575582).
Cerebrospinal Fluid Evaluation
Further testing with CSF evaluation is warranted for
persons with clinical signs of neurosyphilis (e.g., cranial nerve
dysfunction, meningitis, stroke, acute or chronic altered mental
status, or loss of vibration sense). All patients with ocular
symptoms and reactive syphilis serology need a full ocular
examination, including cranial nerve evaluation. If cranial nerve
dysfunction is present, a CSF evaluation is needed. Among
persons with isolated ocular symptoms (i.e., no cranial nerve
dysfunction or other neurologic abnormalities), confirmed
ocular abnormalities on examination, and reactive syphilis
serology, a CSF examination is unnecessary before treatment.
CSF analysis can be helpful in evaluating persons with ocular
symptoms and reactive syphilis serology who do not have ocular
findings or cranial nerve dysfunction on examination. Among
patients with isolated auditory abnormalities and reactive
syphilis serology, CSF evaluation is likely to be normal and is
unnecessary before treatment (583,584).
Laboratory testing is helpful in supporting the diagnosis of
neurosyphilis; however, no single test can be used to diagnose
neurosyphilis in all instances. Diagnosis of neurosyphilis
depends on a combination of CSF tests (e.g., CSF cell count,
protein, or reactive CSF-VDRL) in the presence of reactive
serologic test (nontreponemal and treponemal) results and
neurologic signs and symptoms. CSF laboratory abnormalities
are common for persons with early syphilis and are of
unknown medical significance in the absence of neurologic
signs or symptoms (585). CSF-VDRL is highly specific but
insensitive. For a person with neurologic signs or symptoms, a
reactive CSF-VDRL (in the absence of blood contamination)
is considered diagnostic of neurosyphilis.
When CSF-VDRL is negative despite clinical signs of
neurosyphilis, reactive serologic tests results, lymphocytic
pleocytosis, or protein, neurosyphilis should be considered.
In that instance, additional evaluation by using fluorescent
treponemal-antibody absorption (FTA-ABS) or TP-PA testing
on CSF might be warranted. The CSF FTA-ABS test is less
specific for neurosyphilis than the CSF-VDRL but is highly
sensitive. Fewer data are available regarding CSF TP-PA;
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 41
US Department of Health and Human Services/Centers for Disease Control and Prevention
however, the sensitivity and specificity appear similar to the
CSF FTA-ABS (586). Neurosyphilis is highly unlikely with
a negative CSF FTA-ABS or TP-PA test, especially among
persons with nonspecific neurologic signs and symptoms (587).
Among persons with HIV infection, CSF leukocyte count
can be elevated (>5 WBCs/mm
3
); the association with CSF
leukocyte count and plasma HIV viral suppression has not been
well characterized. Using a higher cutoff (>20 WBCs/mm
3
)
might improve the specificity of neurosyphilis diagnosis among
this population (588).
Treatment
Penicillin G, administered parenterally, is the preferred drug
for treating patients in all stages of syphilis. The preparation
used (i.e., benzathine, aqueous procaine, or aqueous
crystalline), dosage, and length of treatment depend on the
stage and clinical manifestations of the disease. Treatment for
late latent syphilis (>1 years’ duration) and tertiary syphilis
requires a longer duration of therapy because organisms
theoretically might be dividing more slowly (the validity of this
rationale has not been assessed). Longer treatment duration is
required for persons with latent syphilis of unknown duration
to ensure that those who did not acquire syphilis within the
preceding year are adequately treated.
Selection of the appropriate penicillin preparation is
important because T. pallidum can reside in sequestered sites
(e.g., the CNS and aqueous humor) that are poorly accessed
by certain forms of penicillin. Combinations of benzathine
penicillin, procaine penicillin, and oral penicillin preparations
are not considered appropriate for syphilis treatment. Reports
have indicated that practitioners have inadvertently prescribed
combination long- and short-acting benzathine-procaine
penicillin (Bicillin C-R) instead of the standard benzathine
penicillin product (Bicillin L-A) recommended in the United
States for treating primary, secondary, and latent syphilis.
Practitioners, pharmacists, and purchasing agents should be
aware of the similar names of these two products to avoid
using the incorrect combination therapy agent for treating
syphilis (589).
Penicillins effectiveness for treating syphilis was well
established through clinical experience even before the value of
randomized controlled clinical trials was recognized. Therefore,
approximately all recommendations for treating syphilis are
based not only on clinical trials and observational studies, but
on many decades of clinical experience.
Special Considerations
Pregnancy
Parenteral penicillin G is the only therapy with documented
efficacy for syphilis during pregnancy. Pregnant women with
syphilis at any stage who report penicillin allergy should be
desensitized and treated with penicillin (see Management of
Persons Who Have a History of Penicillin Allergy).
Jarisch-Herxheimer Reaction
The Jarisch-Herxheimer reaction is an acute febrile reaction
frequently accompanied by headache, myalgia, and fever that
can occur within the first 24 hours after the initiation of any
syphilis therapy; it is a reaction to treatment and not an allergic
reaction to penicillin. Patients should be informed about this
possible adverse reaction and how to manage it if it occurs. The
Jarisch-Herxheimer reaction occurs most frequently among
persons who have early syphilis, presumably because bacterial
loads are higher during these stages. Antipyretics can be used
to manage symptoms; however, they have not been proven to
prevent this reaction. The Jarisch-Herxheimer reaction might
induce early labor or cause fetal distress in pregnant women;
however, this should not prevent or delay therapy (590) (see
Syphilis During Pregnancy).
Management of Sex Partners
Sexual transmission of T. pallidum is thought to occur only
when mucocutaneous syphilitic lesions are present. Such
manifestations are uncommon after the first year of infection.
Persons exposed through sexual contact with a person who
has primary, secondary, or early latent syphilis should be
evaluated clinically and serologically and treated according to
the following recommendations:
Persons who have had sexual contact with a person who
receives a diagnosis of primary, secondary, or early latent
syphilis <90 days before the diagnosis should be treated
presumptively for early syphilis, even if serologic test results
are negative.
Persons who have had sexual contact with a person who
receives a diagnosis of primary, secondary, or early latent
syphilis >90 days before the diagnosis should be treated
presumptively for early syphilis if serologic test results are
not immediately available and the opportunity for
follow-up is uncertain. If serologic tests are negative, no
treatment is needed. If serologic tests are positive,
treatment should be based on clinical and serologic
evaluation and syphilis stage.
In certain areas or among populations with high syphilis
infection rates, health departments recommend notification
and presumptive treatment of sex partners of persons with
Recommendations and Reports
42 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
syphilis of unknown duration who have high nontreponemal
serologic test titers (i.e., >1:32) because high titers might
be indicative of early syphilis. These partners should be
managed as if the index patient had early syphilis.
Long-term sex partners of persons who have late latent
syphilis should be evaluated clinically and serologically for
syphilis and treated on the basis of the evaluations findings.
The following sex partners of persons with syphilis are
considered at risk for infection and should be confidentially
notified of the exposure and need for evaluation: partners
who have had sexual contact within 3 months plus the
duration of symptoms for persons who receive a diagnosis
of primary syphilis, within 6 months plus duration of
symptoms for those with secondary syphilis, and within
1 year for persons with early latent syphilis.
Primary and Secondary Syphilis
Treatment
Parenteral penicillin G has been used effectively for achieving
clinical resolution (i.e., the healing of lesions and prevention of
sexual transmission) and for preventing late sequelae. However,
no comparative trials have been conducted to guide selection
of an optimal penicillin regimen. Substantially fewer data are
available for nonpenicillin regimens.
Recommended Regimen for Primary and Secondary Syphilis*
Among Adults
Benzathine penicillin G 2.4 million units IM in a single dose
* Recommendations for treating syphilis among persons with HIV infection
and pregnant women are discussed elsewhere in this report (see Syphilis
Among Persons with HIV Infection; Syphilis During Pregnancy).
Available data demonstrate that use of additional doses of
benzathine penicillin G, amoxicillin, or other antibiotics do
not enhance efficacy of this recommended regimen when used
to treat primary and secondary syphilis, regardless of HIV
status (591593).
Recommended Regimen for Syphilis Among Infants and
Children
Benzathine penicillin G 50,000 units/kg body weight IM, up to the adult
dose of 2.4 million units in a single dose
Infants and children aged ≥1 month who receive a syphilis
diagnosis should have birth and maternal medical records
reviewed to assess whether they have congenital or acquired
syphilis (see Congenital Syphilis). Infants and children aged
≥1 month with primary and secondary syphilis should be
managed by a pediatric infectious disease specialist and
evaluated for sexual abuse (e.g., through consultation with child
protective services) (see Sexual Assault or Abuse of Children).
Other Management Considerations
All persons who have primary and secondary syphilis should
be tested for HIV at the time of diagnosis and treatment. Those
persons whose HIV test results are negative should be offered
HIV PrEP. In geographic areas in which HIV prevalence is
high, persons who have primary or secondary syphilis should
be offered PrEP and retested for HIV in 3 months if the initial
HIV test result was negative.
Persons who have syphilis and symptoms or signs indicating
neurologic disease (e.g., cranial nerve dysfunction, meningitis,
stroke, or altered mental state) should have an evaluation
that includes CSF analysis. Persons with syphilis who have
symptoms or signs of ocular syphilis (e.g., uveitis, iritis,
neuroretinitis, or optic neuritis) should have a thorough cranial
nerve examination and ocular slit-lamp and ophthalmologic
examinations. CSF evaluation is not always needed for persons
with ocular syphilis if no evidence of cranial nerves 2, 3, 4,
5, and 6 dysfunction or other evidence of neurologic disease
exists. If symptoms and signs of otic syphilis are present then
an otologic examination is needed; CSF evaluation in persons
with otic syphilis does not aid in the clinical management
and therefore is not recommended (see Cerebrospinal Fluid
Evaluation). Treatment should be guided by the results of these
evaluations. Invasion of CSF by T. pallidum accompanied
by CSF laboratory abnormalities is common among adults
who have primary or secondary syphilis but has unknown
medical significance (585). In the absence of clinical
neurologic findings, no evidence supports variation from the
recommended treatment regimen for primary or secondary
syphilis. Symptomatic neurosyphilis after treatment with the
penicillin regimens recommended for primary and secondary
syphilis is rare. Therefore, unless clinical signs or symptoms of
neurologic or ophthalmic involvement are present, routine CSF
analysis is not recommended for persons who have primary or
secondary syphilis.
Follow-Up
Clinical and serologic evaluation should be performed at
6 and 12 months after treatment; more frequent evaluation
might be prudent if opportunity for follow-up is uncertain
or if repeat infection is a clinical concern. Serologic response
(i.e., titer) should be compared with the titer at the time of
treatment. However, assessing serologic response to treatment
can be difficult, and definitive criteria for cure or failure by
serologic criteria have not been well established. In addition,
nontreponemal test titers might decrease more slowly for
persons previously treated for syphilis (594,595).
Persons who have signs or symptoms that persist or recur
and those with at least a fourfold increase in nontreponemal
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 43
US Department of Health and Human Services/Centers for Disease Control and Prevention
test titer persisting for >2 weeks likely were reinfected or
experienced treatment failure. Among persons who have
neurologic findings or persons with no neurologic findings
without any reported sexual exposure during the previous
3–6 months indicating that treatment failure might be possible,
a CSF examination is recommended with treatment guided
by CSF findings. These persons should also be reevaluated for
HIV infection.
Among persons with no neurologic findings after a thorough
neurologic examination and who are sexually active, reinfection
is likely and repeat treatment for early syphilis is recommended.
These persons should also be reevaluated for HIV infection.
Failure of nontreponemal test titers to decrease fourfold
within 12 months after therapy for primary or secondary
syphilis (inadequate serologic response) might be indicative
of treatment failure. However, clinical trial data have
demonstrated that 10%–20% of persons with primary and
secondary syphilis treated with the recommended therapy
will not achieve the fourfold decrease in nontreponemal titer
within 12 months after treatment (591,596,597). Serologic
response to treatment appears to be associated with multiple
factors, including the persons syphilis stage (earlier stages are
more likely to decrease fourfold and become nonreactive),
initial nontreponemal antibody titers (titers <1:8 are less likely
to decline fourfold than higher titers), and age (titers among
older patients might be less likely to decrease fourfold than
those of younger patients) (596598). Optimal management
of persons who have an inadequate serologic response after
syphilis treatment is unclear. At a minimum, these persons
should receive additional neurologic examinations, clinical
and serologic follow-up annually, and reevaluation for HIV
infection. If neurologic symptoms or signs are identified,
a CSF evaluation is recommended, with findings guiding
management. If additional follow-up cannot be ensured,
retreatment is recommended. Because treatment failure might
be the result of unrecognized CNS infection, CSF examination
can be considered in situations in which follow-up is uncertain.
For retreatment, weekly injections of benzathine penicillin
G 2.4 million units intramuscularly (IM) for 3 weeks is
recommended, unless CSF examination indicates that
neurosyphilis is present (see Neurosyphilis, Ocular Syphilis,
and Otosyphilis). Serologic titers might not decrease, despite
a negative CSF examination and a repeated 3-week therapy
course (599). In these circumstances, the benefit of additional
therapy or repeated CSF examinations is unclear, and it is not
typically recommended. Serologic and clinical monitoring at
least annually should continue to monitor for any sustained
increases in nontreponemal titer.
Management of Sex Partners
See Syphilis, Management of Sex Partners.
Special Considerations
Penicillin Allergy
Data to support use of alternatives to penicillin in treating
primary and secondary syphilis are limited. However, multiple
therapies might be effective for nonpregnant persons with
penicillin allergy who have primary or secondary syphilis.
Doxycycline (100 mg orally 2 times/day for 14 days) (600,601)
and tetracycline (500 mg orally 4 times/day for 14 days) have
been used for years and can be effective. Compliance is likely to
be better with doxycycline than tetracycline because tetracycline
can cause more gastrointestinal side effects and requires more
frequent dosing. Limited clinical studies, along with biologic
and pharmacologic evidence, indicate that ceftriaxone (1 g daily
either IM or IV for 10 days) is effective for treating primary
and secondary syphilis; however, the optimal dose and duration
of ceftriaxone therapy have not been defined (602,603).
Azithromycin as a single 2-g oral dose has been effective
for treating primary and secondary syphilis among certain
populations (602,604,605). However, because of T. pallidum
chromosomal mutations associated with azithromycin and
other macrolide resistance and documented treatment failures
in multiple U.S. geographic areas, azithromycin should not be
used as treatment for syphilis (606608). Thorough clinical
and serologic follow-up of persons receiving any alternative
therapy is essential.
Persons with a penicillin allergy whose compliance with
therapy or follow-up cannot be ensured should be desensitized
and treated with benzathine penicillin G. Skin testing for
penicillin allergy might be useful in circumstances in which
the reagents and expertise are available for performing the test
adequately (see Management of Persons Who Have a History
of Penicillin Allergy).
Pregnancy
Pregnant women with primary or secondary syphilis who
are allergic to penicillin should be desensitized and treated
with penicillin G. Skin testing or oral graded penicillin dose
challenge might be helpful in identifying women at risk for
acute allergic reactions (see Management of Persons Who Have
a History of Penicillin Allergy; Syphilis During Pregnancy).
HIV Infection
Persons with HIV infection who have primary or secondary
syphilis should be treated similarly to those without HIV (see
Syphilis Among Persons with HIV Infection).
Recommendations and Reports
44 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Latent Syphilis
Latent syphilis is defined as syphilis characterized by
seroreactivity without other evidence of primary, secondary,
or tertiary disease. Persons who have latent syphilis and who
acquired syphilis during the preceding year are classified as
having early latent syphilis (early nonprimary, nonsecondary).
Persons can receive a diagnosis of early latent syphilis if, during
the year preceding the diagnosis, they had a documented
seroconversion or a sustained (>2 weeks) fourfold or greater
increase in nontreponemal test titers in a previously treated
person; unequivocal symptoms of primary or secondary
syphilis; or a sex partner documented to have primary,
secondary, or early latent syphilis. In addition, for persons
with reactive nontreponemal and treponemal tests whose only
possible exposure occurred during the previous 12 months,
early latent syphilis can be assumed.
In the absence of these conditions associated with latent
syphilis, an asymptomatic person should be considered to
have latent syphilis of unknown duration or late latent syphilis
(>1 years duration). Nontreponemal serologic titers usually are
higher early in the course of syphilis infection. However, early
latent syphilis cannot be reliably diagnosed solely on the basis
of nontreponemal titers. All persons with latent syphilis should
have careful examination of all accessible mucosal surfaces to
evaluate for mucosal lesions (primary or secondary syphilis)
before making a latent syphilis diagnosis. Physical examination
should include the oral cavity, perianal area, perineum, rectum,
and genitals (vagina and cervix for women; scrotum, penis, and
underneath the foreskin for uncircumcised men).
Treatment
Because latent syphilis is not transmitted sexually, the
objective of treating persons in this disease stage is to prevent
medical complications of syphilis. Latent syphilis can also be
vertically transmitted to a fetus; therefore, the goal of treating
a pregnant woman is to prevent congenital syphilis. Although
clinical experience supports the effectiveness of penicillin in
achieving this goal, limited evidence is available for guiding
choice of specific regimens or duration. Available data
demonstrate that additional doses of benzathine penicillin G,
amoxicillin, or other antibiotics in early latent syphilis do not
enhance efficacy, regardless of HIV status (592,593,609).
Recommended Regimens for Latent Syphilis* Among Adults
Early latent syphilis: Benzathine penicillin G 2.4 million units IM in a
single dose
Late latent syphilis: Benzathine penicillin G 7.2 million units total,
administered as 3 doses of 2.4 million units IM each at 1-week intervals
* Recommendations for treating syphilis in persons with HIV and pregnant
women are discussed elsewhere in this report (see Syphilis Among
Persons with HIV Infection; Syphilis During Pregnancy).
Infants and children aged ≥1 month with diagnosed latent
syphilis should be managed by a pediatric infectious disease
specialist and receive a CSF examination. In addition, birth
and maternal medical records should be reviewed to assess
whether these infants and children have congenital or acquired
syphilis. For those with congenital syphilis, treatment should
be undertaken as described (see Congenital Syphilis). Those
with acquired syphilis should be evaluated for sexual abuse
(e.g., through consultation with child protection services) (see
Sexual Assault or Abuse of Children). These regimens are for
children who are not allergic to penicillin who have acquired
syphilis and who have normal CSF examinations.
Other Management Considerations
All persons who have latent syphilis should be tested for HIV
at the time of diagnosis or treatment. Those persons whose
HIV test results are negative should be offered HIV PrEP. In
geographic areas in which the prevalence of HIV infection is
high or among populations vulnerable to HIV acquisition,
persons who have early latent or late latent syphilis should be
offered PrEP and retested for HIV in 3 months if the first HIV
test result was negative.
Persons who receive a diagnosis of latent syphilis and have
neurologic or ocular signs and symptoms (e.g., cognitive
dysfunction, motor or sensory deficits, ophthalmic or auditory
symptoms, cranial nerve palsies, or symptoms or signs of
meningitis or stroke) should be evaluated for neurosyphilis, ocular
syphilis, or otosyphilis according to their clinical presentation
(see Neurosyphilis, Ocular Syphilis, and Otosyphilis).
If a person receives a delayed dose of penicillin in a course of
weekly therapy for late latent syphilis or syphilis of unknown
duration, the course of action that should be recommended
is unclear. Clinical experience indicates that an interval of
10–14 days between doses of benzathine penicillin for latent
syphilis might be acceptable before restarting the sequence of
injections (i.e., if dose 1 is administered on day 0, dose 2 is
administered on days 10–14). Pharmacologic considerations
indicate that an interval of 7–9 days between doses, if feasible,
might be preferred (610612). Delayed doses are not optimal
for pregnant women receiving therapy for latent syphilis (613).
Pregnant women who have delays in any therapy dose >9 days
between doses should repeat the full course of therapy.
Follow-Up
Quantitative nontreponemal serologic tests should be
repeated at 6, 12, and 24 months. These serologic titers should
be compared with the titer at the time of treatment. Persons
with at least a fourfold sustained increase in nontreponemal
test titer persisting for >2 weeks or who experienced signs
or symptoms attributable to primary or secondary syphilis
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 45
US Department of Health and Human Services/Centers for Disease Control and Prevention
were likely reinfected or experienced treatment failure. These
persons should be retreated and reevaluated for HIV infection.
Among persons who have neurologic findings after a thorough
neurologic examination or among persons with no neurologic
findings and no sexual exposure during the previous year, a CSF
examination is recommended. Treatment should be guided by
CSF findings. Among persons with no neurologic findings after
neurologic examination and who are sexually active, treatment
with weekly injections of benzathine penicillin G 2.4 million
units IM for 3 weeks is recommended.
Optimal management of persons who have less than a
fourfold decrease in titers 24 months after treatment (i.e., an
inadequate serologic response) is unclear, especially if the initial
titer was <1:8. At a minimum, these persons should receive
additional clinical and serologic follow-up and be evaluated for
HIV infection. If neurologic symptoms or signs are identified,
a CSF evaluation is recommended, with the findings guiding
management. If additional follow-up cannot be ensured or if
an initially high titer (>1:32) does not decrease at least fourfold
24 months after treatment, retreatment with weekly injections
of benzathine penicillin G 2.4 million units IM for 3 weeks is
recommended. Because treatment failure might be the result
of unrecognized CNS infection, CSF examination can be
considered in such situations where follow-up is uncertain or
initial high titers do not decrease after 24 months.
If the CSF examination is negative, repeat treatment for
latent syphilis is recommended. Serologic titers might not
decrease despite a negative CSF examination and a repeated
course of therapy, especially if the initial nontreponemal titer
is low (<1:8); in these circumstances, the need for additional
therapy or repeated CSF examinations is unclear but is usually
not recommended. Serologic and clinical monitoring at
least annually should continue to monitor for any sustained
increases in nontreponemal titer.
Management of Sex Partners
See Syphilis, Management of Sex Partners.
Special Considerations
Penicillin Allergy
The effectiveness of alternatives to penicillin in treating
latent syphilis has not been well documented. Nonpregnant
patients allergic to penicillin who have clearly defined early
latent syphilis should respond to antibiotics recommended as
alternatives to penicillin for treating primary and secondary
syphilis (see Primary and Secondary Syphilis). The only
acceptable alternatives for treating late latent syphilis or
syphilis of unknown duration are doxycycline (100 mg orally
2 times/day) or tetracycline (500 mg orally 4 times/day),
each for 28 days. The efficacy of these alternative regimens
among persons with HIV infection has not been well studied.
These therapies should be used only in conjunction with close
serologic and clinical follow-up, especially among persons
with HIV infection. On the basis of biologic plausibility and
pharmacologic properties, ceftriaxone might be effective for
treating latent syphilis. However, the optimal dose and duration
of ceftriaxone therapy have not been defined; treatment
decisions should be discussed in consultation with a specialist.
Persons with a penicillin allergy whose compliance with
therapy or follow-up cannot be ensured should be desensitized
and treated with benzathine penicillin G. Skin testing for
penicillin allergy might be useful in circumstances in which
the reagents and expertise are available for performing the test
adequately (see Management of Persons Who Have a History
of Penicillin Allergy).
Pregnancy
Pregnant women who are allergic to penicillin should be
desensitized and treated with penicillin G. Skin testing for
penicillin allergy might be useful in circumstances in which
the reagents and expertise are available for performing the test
adequately (see Management of Persons Who Have a History
of Penicillin Allergy; Syphilis During Pregnancy).
HIV Infection
Persons with HIV infection who have latent syphilis should
be treated similarly to persons who do not have HIV (see
Syphilis Among Persons with HIV Infection).
Tertiary Syphilis
Tertiary syphilis refers to gummas, cardiovascular syphilis,
psychiatric manifestations (e.g., memory loss or personality
changes), or late neurosyphilis. Guidelines for all forms of
neurosyphilis (e.g., early or late neurosyphilis) are discussed
elsewhere in these recommendations (see Neurosyphilis,
Ocular Syphilis, and Otosyphilis). Persons with gummas
and cardiovascular syphilis who are not allergic to penicillin
and have no evidence of neurosyphilis by clinical and CSF
examination should be treated with the following regimen.
Recommended Regimen for Tertiary Syphilis Among Adults
Tertiary syphilis with normal CSF examination: Benzathine penicillin
G 7.2 million units total, administered as 3 doses of 2.4 million units IM
each at 1-week intervals
Other Management Considerations
All persons who have tertiary syphilis should receive a CSF
examination before therapy is initiated and have an HIV test.
Recommendations and Reports
46 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Those persons whose HIV test results are negative should be
offered HIV PrEP. Persons with CSF abnormalities should be
treated with a neurosyphilis regimen. Certain providers treat all
persons who have cardiovascular syphilis with a neurosyphilis
regimen. These persons should be managed in consultation
with an infectious disease specialist. Limited information is
available concerning clinical response and follow-up of persons
who have tertiary syphilis.
Management of Sex Partners
See Syphilis, Management of Sex Partners.
Special Considerations
Penicillin Allergy
Any person allergic to penicillin should be treated in
consultation with an infectious disease specialist.
Pregnancy
Pregnant women who are allergic to penicillin should be
desensitized and treated with penicillin G. Skin testing or oral
graded penicillin dose challenge might be helpful in identifying
women at risk for acute allergic reactions (see Management
of Persons Who Have a History of Penicillin Allergy; Syphilis
During Pregnancy).
HIV Infection
Persons with HIV infection who have tertiary syphilis should
be treated as described for persons without HIV (see Syphilis
Among Persons with HIV Infection).
Neurosyphilis, Ocular Syphilis, and
Otosyphilis
Treatment
CNS involvement can occur during any stage of syphilis,
and CSF laboratory abnormalities are common among
persons with early syphilis, even in the absence of clinical
neurologic findings. No evidence exists to support variation
from recommended diagnosis and treatment for syphilis at
any stage for persons without clinical neurologic findings,
except tertiary syphilis. If clinical evidence of neurologic
involvement is observed (e.g., cognitive dysfunction, motor or
sensory deficits, cranial nerve palsies, or symptoms or signs of
meningitis or stroke), a CSF examination should be performed
before treatment.
Syphilitic uveitis or other ocular syphilis manifestations
(e.g., neuroretinitis and optic neuritis) can occur at any stage
of syphilis and can be isolated abnormalities or associated with
neurosyphilis. All persons with ocular symptoms and reactive
syphilis serology need a full ocular examination, including
cranial nerve evaluation. If cranial nerve dysfunction is present,
a CSF evaluation is needed. Among persons with isolated ocular
symptoms (no cranial nerve dysfunction or other neurologic
abnormalities), reactive syphilis serology, and confirmed ocular
abnormalities on examination, CSF examination is unnecessary
before treatment. CSF analysis might be helpful in evaluating
persons with ocular symptoms and reactive syphilis serology
who do not have ocular findings on examination. If ocular
syphilis is suspected, immediate referral to and management
in collaboration with an ophthalmologist is crucial. Ocular
syphilis should be treated similarly to neurosyphilis, even if a
CSF examination is normal.
Hearing loss and other otologic symptoms can occur at any
stage of syphilis and can be isolated abnormalities or associated
with neurosyphilis, especially of cranial nerve 8. However,
among persons with isolated auditory symptoms, normal
neurologic examination, and reactive syphilis serology, CSF
examination is likely to be normal and is not recommended
before treatment. Otosyphilis should be managed in
collaboration with an otolaryngologist and treated by using
the same regimen as for neurosyphilis.
Recommended Regimen for Neurosyphilis, Ocular Syphilis, or
Otosyphilis Among Adults
Aqueous crystalline penicillin G 18–24 million units per day,
administered as 3–4 million units IV every 4 hours or continuous
infusion for 10–14 days
If compliance with therapy can be ensured, the following
alternative regimen might be considered.
Alternative Regimen
Procaine penicillin G 2.4 million units IM once daily
plus
Probenecid 500 mg orally 4 times/day, both for 10–14 days
The durations of the recommended and alternative regimens
for neurosyphilis are shorter than the duration of the regimen
used for latent syphilis. Therefore, benzathine penicillin,
2.4 million units IM once per week for 1–3 weeks, can be
considered after completion of these neurosyphilis treatment
regimens to provide a comparable total duration of therapy.
Other Management Considerations
The following are other considerations in the management
of persons who have neurosyphilis:
All persons who have neurosyphilis, ocular syphilis, or
otosyphilis should be tested for HIV at the time of
diagnosis. Those whose HIV test results are negative
should be offered HIV PrEP.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 47
US Department of Health and Human Services/Centers for Disease Control and Prevention
Although systemic steroids are used frequently as
adjunctive therapy for otosyphilis and for ocular syphilis,
such drugs have not been proven to be beneficial.
Follow-Up
Data from two studies indicate that, among immunocompetent
persons and persons with HIV infection who are on effective
ART, normalization of the serum RPR titer predicts
normalization of abnormal CSF parameters after neurosyphilis
treatment (614,615). Therefore, repeated CSF examinations
are unnecessary for persons without HIV infection or persons
with HIV infection who are on ART and who exhibit serologic
and clinical responses after treatment.
Management of Sex Partners
See Syphilis, Management of Sex Partners.
Special Considerations
Penicillin Allergy
Limited data indicate that ceftriaxone 1–2 g daily either IM
or IV for 10–14 days can be used as an alternative treatment
for persons with neurosyphilis (603,616,617). Cross-sensitivity
between ceftriaxone and penicillin can occur; however, the
risk for penicillin cross-reactivity between third-generation
cephalosporins is negligible (618621) (see Management of
Persons Who Have a History of Penicillin Allergy). If concern
exists regarding ceftriaxone safety for a patient with neurosyphilis,
skin testing should be performed to confirm penicillin allergy
and, if necessary, penicillin desensitization in consultation with
a specialist is recommended. Other regimens have not been
adequately evaluated for treatment of neurosyphilis.
Pregnancy
Pregnant women who are allergic to penicillin should be
desensitized and treated with penicillin G. Skin testing or oral
graded penicillin dose challenge might be helpful in identifying
women at risk for acute allergic reactions (see Management of
Persons Who Have a History of Penicillin Allergy).
HIV Infection
Persons with HIV infection who have neurosyphilis should
be treated as described for persons without HIV (see Syphilis
Among Persons with HIV Infection).
Syphilis Among Persons with HIV Infection
Diagnostic Considerations
Interpretation of treponemal and nontreponemal serologic
tests for persons with HIV infection is the same as for persons
without HIV. Although rare, unusual serologic responses have
been observed among persons with HIV infection who have
syphilis. The majority of reports have involved posttreatment
serologic titers that were higher than expected (i.e., high serofast)
or fluctuated, and false-negative serologic test results and delayed
appearance of seroreactivity have also been reported (622).
When clinical findings are indicative of syphilis, but serologic
tests are nonreactive or their interpretation is unclear, alternative
tests (e.g., biopsy of a lesion, darkfield examination, or PCR of
lesion material) might be useful for diagnosis. Neurosyphilis,
ocular syphilis, and otosyphilis should be considered in the
differential diagnosis of neurologic, ocular, and other signs
and symptoms among persons with HIV infection.
Treatment
Persons with HIV infection who have early syphilis might
be at increased risk for neurologic complications (623) and
might have higher rates of inadequate serologic response
with recommended regimens. The magnitude of these risks is
not defined precisely but is likely small. Although long-term
(>1 year) comparative data are lacking, no treatment regimens
for syphilis have been demonstrated to be more effective in
preventing neurosyphilis among persons with HIV infection
than the syphilis regimens recommended for persons without
HIV (609). Careful follow-up after therapy is essential. Using
ART per current HIV guidelines might improve clinical
outcomes among persons coinfected with HIV and syphilis;
concerns regarding adequate treatment of syphilis among
persons with HIV infection might not apply to those with
HIV virologic suppression (624,625).
Primary and Secondary Syphilis Among Persons
with HIV Infection
Recommended Regimen for Primary and Secondary Syphilis
Among Persons with HIV Infection
Benzathine penicillin G 2.4 million units IM in a single dose
Available data demonstrate that additional doses of
benzathine penicillin G, amoxicillin, or other antibiotics in
primary and secondary syphilis among persons with HIV
infection do not result in enhanced efficacy (592,593,609).
Other Management Considerations
The majority of persons with HIV infection respond
appropriately to the recommended benzathine penicillin G
treatment regimen for primary and secondary syphilis (626).
CSF abnormalities (e.g., mononuclear pleocytosis and elevated
protein levels) can be common among persons with HIV, even
those without syphilis. The clinical and prognostic significance
of such CSF laboratory abnormalities among persons with
primary and secondary syphilis who lack neurologic symptoms
Recommendations and Reports
48 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
is unknown. Certain studies have demonstrated that among
persons with HIV infection and syphilis, CSF abnormalities
are associated with a CD4
+
T-cell count of ≤350 cells/mL or
an RPR titer of ≥1:32 (614,627). However, CSF examination
followed by treatment for neurosyphilis on the basis of
laboratory abnormalities has not been associated with improved
clinical outcomes in the absence of neurologic signs and
symptoms. All persons with HIV infection and primary and
secondary syphilis should have a thorough neurologic, ocular,
and otic examination (614,622,625). CSF examination should
be reserved for those with an abnormal neurologic examination.
Follow-Up
Persons with HIV infection and primary or secondary
syphilis should be evaluated clinically and serologically for
possible treatment failure at 3, 6, 9, 12, and 24 months after
therapy; those who meet the criteria for treatment failure
(i.e., signs or symptoms that persist or recur or a sustained
[>2 weeks] fourfold or greater increase in titer) should be
managed in the same manner as persons without HIV infection
(i.e., depending on history of sexual activity and on findings of
neurologic examination, either repeat treatment with weekly
injections of benzathine penicillin G 2.4 million units IM for
3 weeks or CSF examination and repeat treatment guided by
CSF findings) (see Primary and Secondary Syphilis).
In addition, CSF examination and retreatment can be
considered for persons whose nontreponemal test titers do
not decrease fourfold within 24 months of therapy. If CSF
examination is normal, treatment with benzathine penicillin
G administered as 2.4 million units IM at weekly intervals for
3 weeks is recommended. Serologic titers might not decrease
despite a negative CSF examination and a repeated 3-week
course of therapy (599). Especially if the initial nontreponemal
titer is low (<1:8) in these circumstances, the benefit of
additional therapy or repeated CSF examinations is unclear but
is not usually recommended. Serologic and clinical monitoring
at least annually should continue to monitor for any sustained
increases in nontreponemal titer.
Management of Sex Partners
See Syphilis, Management of Sex Partners.
Special Considerations
Penicillin Allergy
Persons with HIV infection who are allergic to penicillin
and have primary or secondary syphilis should be managed
according to the recommendations for persons without HIV
who are allergic to penicillin (see Primary and Secondary
Syphilis). Persons with penicillin allergy whose compliance
with alternative therapy or follow-up cannot be ensured should
be desensitized and treated with penicillin G (see Management
of Persons Who Have a History of Penicillin Allergy). Using
penicillin alternatives has not been well studied among persons
with HIV infection; azithromycin is not recommended for
persons with HIV and primary or secondary syphilis infection.
Alternative therapies should be used only in conjunction with
close serologic and clinical follow-up. Persons with HIV and
latent syphilis should be treated similarly to persons who do
not have HIV (see Latent Syphilis).
Latent Syphilis Among Persons with HIV Infection
Recommended Regimen for Early Latent Syphilis Among
Persons with HIV Infection
Benzathine penicillin G 2.4 million units IM in a single dose
Recommended Regimen for Late Latent Syphilis or Latent
Syphilis of Unknown Duration Among Persons with HIV
Infection
Benzathine penicillin G 7.2 million units total, administered as 3 doses
of 2.4 million units IM at 1-week intervals
Other Management Considerations
All persons with HIV and latent syphilis infection should
undergo a thorough neurologic, ocular, and otic examination;
those with neurologic symptoms or signs should undergo
immediate CSF examination. In the absence of neurologic
symptoms or signs, CSF examination has not been associated
with improved clinical outcomes and therefore is not
recommended. Those with ocular or otic symptoms or
signs should be evaluated for ocular syphilis and otosyphilis
according to those clinical presentations (see Neurosyphilis,
Ocular Syphilis, and Otosyphilis).
Follow-Up
Patients with HIV and latent syphilis infection should
be evaluated clinically and serologically at 6, 12, 18, and
24 months after therapy. Those persons who meet the criteria
for treatment failure (i.e., signs or symptoms that persist or
recur or a sustained [>2 weeks] fourfold or greater increase
in titer) should be managed in the same manner as persons
without HIV (i.e., depending on history of sexual activity and
on findings of neurologic examination, either repeat treatment
with weekly injections of benzathine penicillin G 2.4 million
units IM for 3 weeks or CSF examination and repeat treatment
guided by CSF findings) (see Latent Syphilis).
In addition, CSF examination and retreatment can be
considered for persons whose nontreponemal test titers do
not decrease fourfold within 24 months of therapy. If CSF
examination is normal, treatment with benzathine penicillin
G administered as 2.4 million units IM at weekly intervals for
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 49
US Department of Health and Human Services/Centers for Disease Control and Prevention
3 weeks is recommended. Serologic titers might not decrease
despite a negative CSF examination and a repeated 3-week
course of therapy (599). Especially if the initial nontreponemal
titer is low (<1:8) in these circumstances, the benefit of
additional therapy or repeated CSF examinations is unclear but
is not usually recommended. Serologic and clinical monitoring
at least annually should continue to ensure nontreponemal
titers remain stable without any sustained titer increases.
Management of Sex Partners
See Syphilis, Management of Sex Partners.
Special Considerations
Penicillin Allergy
The efficacy of alternative nonpenicillin regimens for latent
syphilis for persons living with HIV infection has not been well
studied, and these therapies should be used only in conjunction
with close serologic and clinical follow-up. Patients with
penicillin allergy whose compliance with alternative therapy
or follow-up cannot be ensured should be desensitized and
treated with penicillin G (see Management of Persons Who
Have a History of Penicillin Allergy).
Neurosyphilis, Ocular Syphilis, and Otic Syphilis
Among Persons with HIV Infection
All persons with HIV and syphilis infection should receive a
careful neurologic ocular and otic examination. Persons with
HIV infection and neurosyphilis should be treated according
to the recommendations for persons with neurosyphilis and
without HIV infection (see Neurosyphilis, Ocular Syphilis,
and Otosyphilis).
Follow-Up
Persons with HIV and neurosyphilis infection should be
managed according to the recommendations for persons without
HIV infection. Serum RPR can be followed for necessary
treatment success rather than following CSF parameters (see
Neurosyphilis, Ocular Syphilis, and Otosyphilis). Limited data
indicate that changes in CSF parameters might occur more
slowly among persons with HIV infection, especially those
with more advanced immunosuppression (588,624).
Management of Sex Partners
See Syphilis, Management of Sex Partners.
Special Considerations
Penicillin Allergy
Persons with HIV who are allergic to penicillin and have
neurosyphilis infection should be managed according to the
recommendations for persons without HIV infection with
neurosyphilis who are allergic to penicillin (see Neurosyphilis,
Ocular Syphilis, and Otosyphilis). Small observational studies
conducted among persons with HIV and neurosyphilis report
that ceftriaxone 1–2 g IV daily for 10–14 days might be
effective as an alternative agent (628630). The possibility
of cross-sensitivity between ceftriaxone and penicillin exists;
however, the risk for penicillin cross-reactivity between
third-generation cephalosporins is negligible (619621,631)
(see Management of Persons Who Have a History of Penicillin
Allergy). If concern exists regarding the safety of ceftriaxone
for a person with HIV and neurosyphilis, skin testing should
be performed to confirm penicillin allergy and, if necessary,
penicillin desensitization in consultation with a specialist is
recommended. Other regimens have not been adequately
evaluated for treatment of neurosyphilis.
Syphilis During Pregnancy
All women should be screened serologically for syphilis
at the first prenatal care visit (174), which is mandated by
the majority of states (142). Among populations for whom
receipt of prenatal care is not optimal, serologic screening and
treatment (if serologic test is reactive) should be performed at
the time of pregnancy testing (632). Antepartum screening can
be performed by manual nontreponemal antibody testing (e.g.,
RPR) by using the traditional syphilis screening algorithm or
by treponemal antibody testing (e.g., immunoassays) using the
reverse sequence algorithm.
Pregnant women with positive treponemal screening tests
(e.g., EIA, CIA, or immunoblot) should have additional
quantitative nontreponemal testing because titers are essential
for monitoring treatment response. Serologic testing should
also be performed twice during the third trimester: at 28 weeks
gestation and at delivery for pregnant women who live in
communities with high rates of syphilis and for women who
have been at risk for syphilis acquisition during pregnancy.
Maternal risk factors for syphilis during pregnancy include
sex with multiple partners, sex in conjunction with drug
use or transactional sex, late entry to prenatal care (i.e., first
visit during the second trimester or later) or no prenatal
care, methamphetamine or heroine use, incarceration of the
woman or her partner, and unstable housing or homelessness
(174,633636). Moreover, as part of the management of
pregnant women who have syphilis, providers should obtain
information concerning ongoing risk behaviors and treatment
of sex partners to assess the risk for reinfection.
Any woman who has a fetal death after 20 weeks’ gestation
should be tested for syphilis. No mother or neonate should
leave the hospital without maternal serologic status having been
Recommendations and Reports
50 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
documented at least once during pregnancy. Any woman who at
the time of delivery has no prenatal care history or has been at
risk for syphilis acquisition during pregnancy (e.g., misuses drugs;
has had another STI during pregnancy; or has had multiple sex
partners, a new partner, or a partner with an STI) should have the
results of a syphilis serologic test documented before discharge.
Diagnostic Considerations
Pregnant women seropositive for syphilis should be
considered infected unless an adequate treatment history is
clearly documented in the medical records and sequential
serologic antibody titers have decreased as recommended for
the syphilis stage. The risk for antepartum fetal infection or
congenital syphilis at delivery is related to the syphilis stage
during pregnancy, with the highest risk occurring during
the primary and secondary stages. Quantitative maternal
nontreponemal titer, especially if >1:8, might be a marker of
early infection and bacteremia. However, risk for fetal infection
is still substantial among pregnant women with late latent
syphilis and low titers. Pregnant women with stable, serofast
low nontreponemal titers who have previously been treated
for syphilis might not require additional treatment; however,
increasing or high antibody titers in a pregnant woman
previously treated might indicate reinfection or treatment
failure, and treatment should be offered.
If an automated treponemal test (e.g., EIA or CIA) is used for
antepartum syphilis screening, all positive tests should be reflexed
to a quantitative nontreponemal test (e.g., RPR or VDRL). If
the nontreponemal test is negative, the results are considered
discrepant and a second treponemal test (TP-PA is preferred)
should be performed, preferably on the same specimen.
If the second treponemal test is positive (e.g., EIA positive,
RPR negative, or TP-PA positive), current or previous syphilis
infection can be confirmed. For women with a history of
adequately treated syphilis who do not have ongoing risk, no
further treatment is necessary. Women without a history of
treatment should have the syphilis stage determined and should
be treated accordingly with a recommended penicillin regimen.
If the second treponemal test is negative (e.g., EIA positive,
RPR negative, or TP-PA negative), the positive EIA or CIA is
more likely to represent a false-positive test result for women
who are living in communities with low rates of syphilis, have
a partner who is uninfected, and have no history of treated
syphilis (637,638). If the woman is at low risk for syphilis,
lacks signs or symptoms of primary syphilis, has a partner
with no clinical or serologic evidence of syphilis, and is likely
to follow up with clinical care, repeat serologic testing within 4
weeks can be considered to determine whether the EIA or CIA
remains positive or if the RPR, VDRL, or TP-PA result becomes
positive. If both the RPR and TP-PA remain negative, no further
treatment is necessary. If follow-up is not likely, women with an
isolated reactive treponemal test and without a history of treated
syphilis should be treated according to the syphilis stage.
Treatment
Penicillin G is the only known effective antimicrobial for
treating fetal infection and preventing congenital syphilis
(639). Evidence is insufficient to determine the optimal
penicillin regimen during pregnancy (640).
Recommended Regimen for Syphilis During Pregnancy
Pregnant women should be treated with the recommended penicillin
regimen for their stage of infection
Other Management Considerations
The following recommendations should be considered for
pregnant women with syphilis infection:
Certain evidence indicates that additional therapy is
beneficial for pregnant women to prevent congenital
syphilis. For women who have primary, secondary, or early
latent syphilis, a second dose of benzathine penicillin G
2.4 million units IM can be administered 1 week after the
initial dose (641643).
When syphilis is diagnosed during the second half of
pregnancy, management should include a sonographic fetal
evaluation for congenital syphilis. However, this evaluation
should not delay therapy. Sonographic signs of fetal or
placental syphilis (e.g., hepatomegaly, ascites, hydrops, fetal
anemia, or a thickened placenta) indicate a greater risk for
fetal treatment failure (644); cases accompanied by these
signs should be managed in consultation with obstetric
specialists. A second dose of benzathine penicillin G
2.4 million units IM after the initial dose might be beneficial
for fetal treatment in these situations.
Women treated for syphilis during the second half of
pregnancy are at risk for premature labor or fetal distress
if the treatment precipitates the Jarisch-Herxheimer
reaction (590). These women should be advised to seek
obstetric attention after treatment if they notice any fever,
contractions, or decrease in fetal movements. Stillbirth is
a rare complication of treatment; however, concern for
this complication should not delay necessary treatment.
No data are available to support that corticosteroid
treatment alters the risk for treatment-related complications
during pregnancy.
Missed doses >9 days between doses are not acceptable for
pregnant women receiving therapy for late latent syphilis
(613). An optimal interval between doses is 7 days for
pregnant women. If a pregnant woman does not return
for the next dose on day 7, every effort should be made
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 51
US Department of Health and Human Services/Centers for Disease Control and Prevention
to contact her and link her to immediate treatment within
2 days to avoid retreatment. Pregnant women who miss a
dose of therapy should repeat the full course of therapy.
All women who have syphilis should be offered testing for
HIV at the time of diagnosis.
Follow-Up
Coordinated prenatal care and treatment are vital because
providers should document that women are adequately
treated for the syphilis stage and ensure that the clinical and
antibody responses are appropriate for the patient’s disease
stage. If syphilis is diagnosed and treated at or before 24 weeks
gestation, serologic titers should not be repeated before 8 weeks
after treatment (e.g., at 32 weeks’ gestation) but should be
repeated again at delivery. Titers should be repeated sooner
if reinfection or treatment failure is suspected. For syphilis
diagnosed and treated after 24 weeks’ gestation, serologic titers
should be repeated at delivery.
A majority of women will not achieve a fourfold decrease
in titers before delivery, although this does not indicate
treatment failure (645). However, a fourfold increase in titer
after treatment (e.g., from 1:8 to 1:32) that is sustained for
>2 weeks is concerning for reinfection or treatment failure.
Nontreponemal titers can increase immediately after treatment,
presumably related to the treatment response. Therefore, unless
symptoms and signs exist of primary or secondary syphilis,
follow-up titer should not be repeated until approximately
8 weeks after treatment. Inadequate maternal treatment is
likely if delivery occurs within 30 days of therapy, clinical signs
of infection are present at delivery, or the maternal antibody
titer at delivery is fourfold higher than the pretreatment titer.
Management of Sex Partners
See Syphilis, Management of Sex Partners.
Special Considerations
Penicillin Allergy
No proven alternatives to penicillin are available for
treatment of syphilis during pregnancy. Pregnant women who
have a history of penicillin allergy should be desensitized and
treated with penicillin G. Skin testing or oral graded penicillin
dose challenge might be helpful in identifying women at risk
for acute allergic reactions (see Management of Persons Who
Have a History of Penicillin Allergy).
Tetracycline and doxycycline are to be avoided in the second
and third trimesters of pregnancy (431). Erythromycin and
azithromycin should not be used because neither reliably cures
maternal infection nor treats an infected fetus (640). Data are
insufficient to recommend ceftriaxone or other cephalosporins
for treatment of maternal infection and prevention of
congenital syphilis (646,647).
HIV Infection
Placental inflammation from congenital syphilis infection
might increase the risk for perinatal transmission of HIV. All
women with HIV infection should be evaluated for syphilis and
receive a penicillin regimen appropriate for the syphilis stage.
Data are insufficient to recommend any alternative regimens
for pregnant women with syphilis and HIV infection (see
Syphilis Among Persons with HIV).
Congenital Syphilis
The rate of reported congenital syphilis in the United States
has increased dramatically since 2012. During 2019, a total of
1,870 cases of congenital syphilis were reported, including 94
stillbirths and 34 infant deaths (141). The 2019 national rate
of 48.5 cases per 100,000 live births represents a 41% increase
relative to 2018 (34.3 cases per 100,000 live births) and a 477%
increase relative to 2012 (8.4 cases per 100,000 live births).
During 2015–2019, the rate of congenital syphilis increased
291.1% (12.4 to 48.5 per 100,000 live births), which mirrors
increases in the rate of primary and secondary syphilis among
females aged 15–44 years (a 171.9% increase, from 3.2 to 8.7
per 100,000 females).
Effective prevention and detection of congenital syphilis
depend on identifying syphilis among pregnant women and,
therefore, on the routine serologic screening of pregnant
women during the first prenatal visit and at 28 weeks’ gestation
and at delivery for women who live in communities with high
rates of syphilis, women with HIV infection, or those who
are at increased risk for syphilis acquisition. Certain states
have recommended screening three times during pregnancy
for all women; clinicians should screen according to their
states guidelines.
Maternal risk factors for syphilis during pregnancy include
sex with multiple partners, sex in conjunction with drug
use or transactional sex, late entry to prenatal care (i.e., first
visit during the second trimester or later) or no prenatal
care, methamphetamine or heroine use, incarceration of the
woman or her partner, and unstable housing or homelessness
(174,633636). Moreover, as part of the management of
pregnant women who have syphilis, providers should obtain
information concerning ongoing risk behaviors and treatment
of sex partners to assess the risk for reinfection.
Routine screening of neonatal sera or umbilical cord blood
is not recommended because diagnosis at that time does not
prevent congenital syphilis in certain newborns. No mother
or newborn infant should leave the hospital without maternal
Recommendations and Reports
52 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
serologic status having been documented at least once during
pregnancy. Any woman who had no prenatal care before
delivery or is considered at increased risk for syphilis acquisition
during pregnancy should have the results of a syphilis serologic
test documented before she or her neonate is discharged. A
quantitative RPR is needed at the time of delivery to compare
with the neonates nontreponemal test result. If a stat RPR
is unavailable and a rapid treponemal test is performed at
delivery, the results should be confirmed by using standard
syphilis serologic laboratory tests (e.g., RPR and treponemal
test) and algorithms.
Evaluation and Treatment of Neonates
Diagnosis of congenital syphilis can be difficult because
maternal nontreponemal and treponemal immunoglobulin G
(IgG) antibodies can be transferred through the placenta to the
fetus, complicating the interpretation of reactive serologic tests
for syphilis among neonates (infants aged <30 days). Therefore,
treatment decisions frequently must be made on the basis of
identification of syphilis in the mother; adequacy of maternal
treatment; presence of clinical, laboratory, or radiographic
evidence of syphilis in the neonate; and comparison of maternal
(at delivery) and neonatal nontreponemal serologic titers (e.g.,
RPR or VDRL) by using the same test, preferably conducted
by the same laboratory. Any neonate at risk for congenital
syphilis should receive a full evaluation and testing for HIV.
All neonates born to mothers who have reactive nontreponemal
and treponemal test results should be evaluated with a
quantitative nontreponemal serologic test (RPR or VDRL)
performed on the neonate’s serum because umbilical cord blood
can become contaminated with maternal blood and yield a
false-positive result, and Whartons jelly within the umbilical
cord can yield a false-negative result. The nontreponemal
test performed on the neonate should be the same type of
nontreponemal test performed on the mother.
Conducting a treponemal test (e.g., TP-PA, immunoassay-
EIA, CIA, or microbead immunoassay) on neonatal serum is
not recommended because it is difficult to interpret, as passively
transferred maternal antibodies can persist for >15 months.
Commercially available IgM tests are not recommended.
All neonates born to women who have reactive nontreponemal
serologic tests for syphilis at delivery should be examined
thoroughly for evidence of congenital syphilis (e.g., nonimmune
hydrops, conjugated or direct hyperbilirubinemia
or
cholestatic jaundice or cholestasis, hepatosplenomegaly,
rhinitis, skin rash, or pseudoparalysis of an extremity).
Pathologic examination of the placenta or umbilical cord using
specific staining (e.g., silver) or a T. pallidum PCR test using
Direct hyperbilirubinemia is direct bilirubin level >2 mg/dL (34 umol/L) or
20% of the total bilirubin level.
a CLIA-validated test should be considered; direct fluorescence
antibody (DFA-TP) reagents are unavailable (565). Darkfield
microscopic examination or PCR testing of suspicious lesions
or body fluids (e.g., bullous rash or nasal discharge) also should
be performed. In addition to these tests, for stillborn infants,
skeletal survey demonstrating typical osseous lesions might aid
in the diagnosis of congenital syphilis because these
abnormalities are not detected on fetal ultrasound.
The following scenarios describe the recommended
congenital syphilis evaluation and treatment of neonates born
to women who had reactive nontreponemal and treponemal
serologic tests for syphilis during pregnancy (e.g., RPR reactive,
TP-PA reactive or EIA reactive, RPR reactive) and have a
reactive nontreponemal test at delivery (e.g., RPR reactive).
Maternal history of infection with T. pallidum and treatment
for syphilis should be considered when evaluating and treating
the neonate for congenital syphilis in most scenarios, except
when congenital syphilis is proven or highly probable.
Scenario 1: Confirmed Proven or Highly Probable
Congenital Syphilis
Any neonate with
an abnormal physical examination that is consistent with
congenital syphilis;
a serum quantitative nontreponemal serologic titer that is
fourfold
§
(or greater) higher than the mothers titer at
delivery (e.g., maternal titer = 1:2, neonatal titer ≥1:8 or
maternal titer=1:8, neonatal titer ≥1:32)
; or
a positive darkfield test or PCR of placenta, cord, lesions, or
body fluids or a positive silver stain of the placenta or cord.
Recommended Evaluation
CSF analysis for VDRL, cell count, and protein**
§
One dilution is within the test performance of nontreponemal tests and is not
a significant change.
The absence of a fourfold or greater titer for a neonate does not exclude
congenital syphilis.
** Interpretation of CSF test results requires a nontraumatic lumbar puncture
(i.e., a CSF sample that is not contaminated with blood). CSF test results
obtained during the neonatal period can be difficult to interpret; normal values
differ by gestational age and are higher among preterm infants. Studies indicate
that 95% of healthy neonates have values of ≤16–19 WBCs/mm
3
or protein
levels of ≤115–118 mg/dL on CSF examination. During the second month
of life, 95% of healthy infants have ≤9–11 WBCs/mm
3
or protein levels of
≤89–91 mg/dL. Lower values (i.e., 5 WBCs/mm
3
and protein level of 40 mg/
dL) might be considered the upper limits of normal for older infants. Other
causes of elevated values should be considered when an infant is being evaluated
for congenital syphilis (Sources: Kestenbaum LA, Ebberson J, Zorc JJ,
Hodinka RL, Shah SS. Defining cerebrospinal fluid white blood cell count
reference values in neonates and young infants. Pediatrics 2010;125:257–64;
Shah SS, Ebberson J, Kestenbaum LA, Hodinka RL, Zorc JJ. Age-specific
reference values for cerebrospinal fluid protein concentration in neonates and
young infants. J Hosp Med 2011;6:22–7; Thomson J, Sucharew H, Cruz AT,
et al.; Pediatric Emergency Medicine Collaborative Research Committee [PEM
CRC] HSV Study Group. Cerebrospinal fluid reference values for young
infants undergoing lumbar puncture. Pediatrics 2018;141:e20173405.)
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 53
US Department of Health and Human Services/Centers for Disease Control and Prevention
Complete blood count (CBC) and differential and
platelet count
Long-bone radiographs
Other tests as clinically indicated (e.g., chest radiograph,
liver function tests, neuroimaging, ophthalmologic
examination, and auditory brain stem response)
Recommended Regimens, Confirmed or Highly Probable
Congenital Syphilis
Aqueous crystalline penicillin G 100,000–150,000 units/kg/body
weight/day, administered as 50,000 units/kg body weight/dose IV every
12 hours during the first 7 days of life and every 8 hours thereafter for a
total of 10 days
or
Procaine penicillin G 50,000 units/kg body weight/dose IM in a single
daily dose for 10 days
If >1 day of therapy is missed, the entire course should
be restarted. Data are insufficient regarding use of other
antimicrobial agents (e.g., ampicillin). When possible, a full
10-day course of penicillin is preferred, even if ampicillin was
initially provided for possible sepsis (648650). Using agents
other than penicillin requires close serologic follow-up for
assessing therapy adequacy.
Scenario 2: Possible Congenital Syphilis
Any neonate who has a normal physical examination and a
serum quantitative nontreponemal serologic titer equal to or
less than fourfold of the maternal titer at delivery (e.g., maternal
titer=1:8, neonatal titer ≤1:16) and one of the following:
The mother was not treated, was inadequately treated, or
has no documentation of having received treatment.
The mother was treated with erythromycin or a regimen
other than those recommended in these guidelines (i.e., a
nonpenicillin G regimen).
††
The mother received the recommended regimen but
treatment was initiated <30 days before delivery.
Recommended Evaluation
CSF analysis for VDRL, cell count, and protein**
CBC, differential, and platelet count
Long-bone radiographs
This evaluation is not necessary if a 10-day course of
parenteral therapy is administered, although such evaluations
might be useful. For instance, a lumbar puncture might
document CSF abnormalities that would prompt close
follow-up. Other tests (e.g., CBC, platelet count, and long-
bone radiographs) can be performed to further support a
diagnosis of congenital syphilis.
††
A women treated with a regimen other than recommended in these guidelines
should be considered untreated.
Recommended Regimens, Possible Congenital Syphilis
Aqueous crystalline penicillin G 100,000–150,000 units/kg body
weight/day, administered as 50,000 units/kg body weight/dose IV every
12 hours during the first 7 days of life and every 8 hours thereafter for a
total of 10 days
or
Procaine penicillin G 50,000 units/kg body weight/dose IM in a single
daily dose for 10 days
or
Benzathine penicillin G 50,000 units/kg body weight/dose IM in a
single dose
Before using the single-dose benzathine penicillin G regimen,
the recommended evaluation (i.e., CSF examination, long-
bone radiographs, and CBC with platelets) should be normal,
and follow-up should be certain. If any part of the neonates
evaluation is abnormal or not performed, if the CSF analysis
is uninterpretable because of contamination with blood, or
if follow-up is uncertain, a 10-day course of penicillin G
is required.
If the neonates nontreponemal test is nonreactive and
the provider determines that the mothers risk for untreated
syphilis is low, treatment of the neonate with a single IM
dose of benzathine penicillin G 50,000 units/kg body weight
for possible incubating syphilis can be considered without
an evaluation. Neonates born to mothers with untreated
early syphilis at the time of delivery are at increased risk for
congenital syphilis, and the 10-day course of penicillin G
should be considered even if the neonates nontreponemal
test is nonreactive, the complete evaluation is normal, and
follow-up is certain.
Scenario 3: Congenital Syphilis Less Likely
Any neonate who has a normal physical examination and a
serum quantitative nontreponemal serologic titer equal or less
than fourfold of the maternal titer at delivery (e.g., maternal
titer=1:8, neonatal titer ≤1:16) and both of the following
are true:
The mother was treated during pregnancy, treatment was
appropriate for the infection stage, and the treatment
regimen was initiated ≥30 days before delivery.
The mother has no evidence of reinfection or relapse.
Recommended Evaluation
No evaluation is recommended.
Recommended Regimen, Congenital Syphilis Less Likely
Benzathine penicillin G 50,000 units/kg body weight/dose IM in a
single dose*
* Another approach involves not treating the newborn if follow-up is
certain but providing close serologic follow-up every 2–3 months for 6
months for infants whose mothers nontreponemal titers decreased at
least fourfold after therapy for early syphilis or remained stable for low-
titer, latent syphilis (e.g., VDRL <1:2 or RPR <1:4).
Recommendations and Reports
54 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Scenario 4: Congenital Syphilis Unlikely
Any neonate who has a normal physical examination and a
serum quantitative nontreponemal serologic titer equal to or
less than fourfold of the maternal titer at delivery
§
and both
of the following are true:
The mother’s treatment was adequate before pregnancy.
The mother’s nontreponemal serologic titer remained low
and stable (i.e., serofast) before and during pregnancy and
at delivery (e.g., VDRL ≤1:2 or RPR ≤1:4).
Recommended Evaluation
No evaluation is recommended.
Recommended Regimen, Congenital Syphilis Unlikely
No treatment is required. However, any neonate with reactive
nontreponemal tests should be followed serologically to ensure the
nontreponemal test returns to negative (see Follow-Up). Benzathine
penicillin G 50,000 units/kg body weight as a single IM injection might
be considered, particularly if follow-up is uncertain and the neonate has
a reactive nontreponemal test.
The following situations describe management of neonates
born to women screened during pregnancy by using the reverse
sequence algorithm with reactive treponemal serologic tests
and a nonreactive nontreponemal serologic test.
Reactive maternal treponemal serologies with a
nonreactive nontreponemal serology (e.g., EIA reactive,
RPR nonreactive, or TP-PA reactive) during pregnancy.
Syphilis is highly unlikely for neonates born to mothers with
a nonreactive nontreponemal test after adequate treatment
for syphilis during pregnancy or documentation of adequate
treatment before pregnancy (with no evidence of reinfection
of relapse). If testing is performed again at delivery and 1) the
maternal nontreponemal test remains nonreactive and 2) the
neonate has a normal physical examination and nonreactive
nontreponemal test (e.g., RPR nonreactive), the provider
should consider managing similarly to Scenario 4 without
a laboratory evaluation and with no treatment required.
Benzathine penicillin G 50,000 units/kg body weight as a
single IM injection might be considered if syphilis exposure
is possible within 1 month of delivery and follow-up of the
mother and infant is uncertain.
Isolated reactive maternal treponemal serology (e.g.,
EIA reactive, RPR nonreactive, or TP-PA nonreactive)
during pregnancy. Syphilis is unlikely for neonates born to
mothers screened with the reverse sequence algorithm with
isolated reactive maternal treponemal serology. Among low-
prevalence populations, these are likely false-positive results and
might become nonreactive with repeat testing (638). If these
neonates have a normal physical examination and the risk for
syphilis is low in the mother, no evaluation and treatment are
recommended for the neonate. If syphilis exposure is possible
or unknown in the mother or the mother desires further
evaluation to definitively rule out syphilis, repeat serology
within 4 weeks is recommended to evaluate for early infection
(see Syphilis During Pregnancy).
Isolated reactive maternal treponemal serology (e.g., rapid
treponemal test) at delivery. For mothers with late or no
prenatal care with a reactive rapid treponemal test at delivery,
confirmatory laboratory-based testing should be performed;
however, results should not delay evaluation and treatment of
the neonate. These neonates should be evaluated and treated
with a 10-day course of penicillin as recommended in Scenario
1, and consultation with a specialist is recommended.
Follow-Up
All neonates with reactive nontreponemal tests should receive
thorough follow-up examinations and serologic testing (i.e., RPR
or VDRL) every 2–3 months until the test becomes nonreactive.
For a neonate who was not treated because congenital syphilis
was considered less likely or unlikely, nontreponemal antibody
titers should decrease by age 3 months and be nonreactive
by age 6 months, indicating that the reactive test result was
caused by passive transfer of maternal IgG antibody. At age
6 months, if the nontreponemal test is nonreactive, no further
evaluation or treatment is needed; if the nontreponemal test is
still reactive, the infant is likely infected and should be treated.
Treated neonates who exhibit persistent nontreponemal test
titers by age 6–12 months should be reevaluated through CSF
examination and managed in consultation with an expert.
Retreatment with a 10-day course of a penicillin G regimen
might be indicated.
Neonates with a negative nontreponemal test at birth and
whose mothers were seroreactive at delivery should be retested
at age 3 months to rule out serologically negative incubating
congenital syphilis at the time of birth. Treponemal tests should
not be used to evaluate treatment response because the results
are qualitative, and passive transfer of maternal IgG treponemal
antibody might persist for >15 months.
Neonates whose initial CSF evaluations are abnormal do
not need repeat lumbar puncture unless they exhibit persistent
nontreponemal serologic test titers at age 6–12 months.
Persistent nontreponemal titers and CSF abnormalities should
be managed in consultation with an expert.
Special Considerations
Penicillin Allergy
Neonates who require treatment for congenital syphilis
but who have a history of penicillin allergy or develop an
allergic reaction presumed secondary to penicillin should
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 55
US Department of Health and Human Services/Centers for Disease Control and Prevention
be desensitized and then treated with penicillin G (see
Management of Persons Who Have a History of Penicillin
Allergy). Skin testing remains unavailable for neonates because
the procedure has not been standardized for this age group.
Data are insufficient regarding use of other antimicrobial agents
(e.g., ceftriaxone) for congenital syphilis among neonates. If
a nonpenicillin G agent is used, close clinical and serologic
follow-up is required in consultation with an expert. Repeat
CSF examination should be performed if the initial CSF
examination was abnormal.
Penicillin Shortage
During periods when the availability of aqueous crystalline
penicillin G is compromised, the following is recommended
(https://www.cdc.gov/std/treatment/drug-notices.htm):
For neonates with clinical evidence of congenital syphilis
(see Scenario 1), check local sources for aqueous crystalline
penicillin G (potassium or sodium) and notify CDC and
FDA of limited supply. If IV penicillin G is limited,
substitute some or all daily doses with procaine penicillin G
(50,000 units/kg body weight/dose IM/day in a single
daily dose for 10 days).
If aqueous or procaine penicillin G is unavailable,
ceftriaxone (50–75 mg/kg body weight/day IV every
24 hours) can be considered with thorough clinical and
serologic follow-up and in consultation with an expert
because evidence is insufficient to support using ceftriaxone
for treating congenital syphilis. Ceftriaxone should be used
with caution in neonates with jaundice.
For neonates without any clinical evidence of congenital
syphilis (see Scenario 2 and Scenario 3), use
ű
procaine penicillin G 50,000 units/kg body weight/
dose/day IM in a single dose for 10 days, or
ű
benzathine penicillin G 50,000 units/kg body weight
IM as a single dose.
If any part of the evaluation for congenital syphilis is
abnormal or was not performed, CSF examination is not
interpretable, or follow-up is uncertain, procaine penicillin
G is recommended. A single dose of ceftriaxone is
inadequate therapy.
For premature neonates who have no clinical evidence of
congenital syphilis (see Scenario 2 and Scenario 3) and
might not tolerate IM injections because of decreased
muscle mass, IV ceftriaxone can be considered with
thorough clinical and serologic follow-up and in
consultation with an expert. Ceftriaxone dosing should
be adjusted according to birthweight.
HIV Infection
Evidence is insufficient to determine whether neonates who
have congenital syphilis and HIV infection or whose mothers
have HIV require different therapy or clinical management
than is recommended for all neonates. All neonates with
congenital syphilis should be managed similarly, regardless of
HIV status.
Evaluation and Treatment of Infants and Children
with Congenital Syphilis
Infants and children aged ≥1 month who are identified as
having reactive serologic tests for syphilis (e.g., RPR reactive,
TP-PA reactive or EIA reactive, RPR reactive) should be
examined thoroughly and have maternal serology and records
reviewed to assess whether they have congenital or acquired
syphilis (see Primary and Secondary Syphilis; Latent Syphilis;
Sexual Assault or Abuse of Children). In the case of extremely
early or incubating syphilis at the time of delivery, all maternal
serologic tests might have been negative; thus, infection might
be undetected until a diagnosis is made later in the infant or
child. Any infant or child at risk for congenital syphilis should
receive a full evaluation and testing for HIV infection.
International adoptee, immigrant, or refugee children from
countries where treponemal infections (e.g., yaws or pinta) are
endemic might have reactive nontreponemal and treponemal
serologic tests, which cannot distinguish between syphilis and
other subspecies of T. pallidum (651). These children might
also have syphilis (T. pallidum subspecies pallidum) and should
be evaluated for congenital syphilis.
Recommended Evaluation
The following evaluations should be performed:
CSF analysis for VDRL, cell count, and protein
CBC, differential, and platelet count
Other tests as clinically indicated (e.g., long-bone
radiographs, chest radiograph, liver function tests,
abdominal ultrasound, ophthalmologic examination,
neuroimaging, and auditory brain-stem response)
Recommended Regimen for Congenital Syphilis Among Infants
and Children
Aqueous crystalline penicillin G 200,000–300,000 units/kg body
weight/day IV, administered as 50,000 units/kg body weight every 4–6
hours for 10 days
If the infant or child has no clinical manifestations of
congenital syphilis and the evaluation (including the CSF
examination) is normal, treatment with <3 weekly doses of
benzathine penicillin G 50,000 units/kg body weight IM
can be considered. A single dose of benzathine penicillin
G 50,000 units/kg body weight IM up to the adult dose of
Recommendations and Reports
56 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
2.4 million units in a single dose can be considered after the
10-day course of IV aqueous penicillin G to provide more
comparable duration for treatment in those who have no
clinical manifestations and normal CSF. All of these treatment
regimens should also be adequate for children who might have
other treponemal infections.
Follow-Up
Thorough follow-up examinations and serologic testing (i.e.,
RPR or VDRL) of infants and children treated for congenital
syphilis after the neonatal period (aged >30 days) should be
performed every 3 months until the test becomes nonreactive
or the titer has decreased fourfold. The serologic response
after therapy might be slower for infants and children than
neonates. If these titers increase at any point >2 weeks or do
not decrease fourfold after 12–18 months, the infant or child
should be evaluated (e.g., CSF examination), treated with
a 10-day course of parenteral penicillin G, and managed in
consultation with an expert. Treponemal tests (e.g., EIA, CIA,
or TP-PA) should not be used to evaluate treatment response
because the results are qualitative and persist after treatment,
and passive transfer of maternal IgG treponemal antibody
might persist for >15 months after delivery. Infants or children
whose initial CSF evaluations are abnormal do not need repeat
lumbar puncture unless their serologic titers do not decrease
fourfold after 12–18 months. After 18 months of follow-up,
abnormal CSF indices that persist and cannot be attributed to
other ongoing illness indicate that retreatment is needed for
possible neurosyphilis and should be managed in consultation
with an expert.
Special Considerations
Penicillin Allergy
Infants and children who require treatment for congenital
syphilis but who have a history of penicillin allergy or develop
an allergic reaction presumed secondary to penicillin should be
desensitized and treated with penicillin G (see Management
of Persons Who Have a History of Penicillin Allergy). Skin
testing remains unavailable for infants and children because the
procedure has not been standardized for this age group. Data
are insufficient regarding use of other antimicrobial agents (e.g.,
ceftriaxone) for congenital syphilis among infants and children.
If a nonpenicillin G agent is used, close clinical, serologic, and
CSF follow-up is required in consultation with an expert.
Penicillin Shortage
During periods when availability of penicillin G is
compromised, management options are similar to options
for the neonate (see Evaluation and Treatment of Neonates).
For infants and children with clinical evidence of
congenital syphilis, if IV penicillin is limited after checking
local sources and notifying CDC and FDA about limited
supplies, procaine penicillin G (50,000 units/kg body
weight/dose IM up to the adult dose of 2.4 million units
a day in a single daily dose for 10 days) is recommended.
If procaine penicillin G is not available, ceftriaxone (in doses
for age and weight) can be considered with thorough clinical
and serologic follow-up. Infants and children receiving
ceftriaxone should be managed in consultation with an expert
because evidence is insufficient to support use of ceftriaxone
for treatment of congenital syphilis among infants or children.
For infants aged ≥30 days, use ceftriaxone 75 mg/kg body
weight/day IV or IM in a single daily dose for 10–14 days
(dose adjustment might be necessary on the basis of current
weight). For children, ceftriaxone 100 mg/kg body weight/
day in a single daily dose is recommended.
For infants and children without any clinical evidence of
infection (see Scenario 2 and Scenario 3), use
ű
procaine penicillin G 50,000 units/kg body weight/dose
IM up to the adult dose of 2.4 million units a day in a
single dose for 10 days, or
ű
benzathine penicillin G 50,000 units/kg body weight
IM up to the adult dose of 2.4 million units as a
single dose.
If any part of the evaluation for congenital syphilis is
abnormal or not performed, CSF examination is not
interpretable, or follow-up is uncertain, procaine penicillin
G is recommended. In these scenarios, a single dose of
ceftriaxone is inadequate therapy.
HIV Infection
Evidence is insufficient to determine whether infants and
children who have congenital syphilis and HIV infection or
whose mothers have HIV require different therapy or clinical
management than what is recommended for all infants and
children. All infants and children with congenital syphilis
should be managed similarly, regardless of HIV status.
Management of Persons Who Have a
History of Penicillin Allergy
Penicillin and other ß-lactam antibiotics have a crucial
role in treating STIs. Penicillin is recommended for all
clinical stages of syphilis, and no proven alternatives exist for
treating neurosyphilis, congenital syphilis, or syphilis during
pregnancy. Ceftriaxone, a third-generation cephalosporin, is
recommended for gonorrhea treatment. For extragenital site
infections, especially pharyngeal, failure rates of nonceftriaxone
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 57
US Department of Health and Human Services/Centers for Disease Control and Prevention
regimens can be substantial. In most clinical settings, patients
who report a penicillin allergy are not treated with ß-lactam
antimicrobials. For patients with a diagnosis of gonorrhea and
a concomitant reported allergy to penicillin, ceftriaxone is often
avoided, even though the cross-reactivity between penicillin
allergy and third-generation cephalosporins is low (652654).
Prevalence of reported allergy to penicillin is approximately
10% among the U.S. population and higher among hospital
inpatients and residents in health care–related facilities (655
658). One large study in an STI clinic revealed that 8.3% of
patients reported penicillin or another ß-lactam antibiotic
allergy (659). Penicillin allergy is often overreported, with the
majority of patients who report penicillin allergy able to tolerate
the medication (660). The prevalence of reported penicillin
allergy in low-income countries is unknown; however, limited
data indicate that penicillin is one of the most frequently
reported antibiotic allergies (661).
Patients often are incorrectly labeled as allergic to penicillin
and are therefore denied the benefit of a ß-lactam therapy.
The presence of a penicillin allergy label considerably reduces
prescribing options for affected patients. Moreover, penicillin
allergy labels lead to the use of more expensive and less effective
drugs and can result in adverse consequences, including longer
length of hospital stay and increased risk for infection. Multiple
studies have described that persons with reported penicillin or
another ß-lactam antibiotic allergy have higher rates of surgical-
site infections, methicillin-resistant Staphylococcus aureus
infections, and higher medical care usage (653,662664).
The overreported prevalence of penicillin allergy is
secondary to imprecise use of the term “allergy” by families
and clinicians and lack of clarity to differentiate between
immunoglobulin E (IgE)-mediated hypersensitivity reactions,
drug intolerances, and other idiosyncratic reactions that can
occur days after exposure. Approximately 80% of patients
with a true IgE-mediated allergic reaction to penicillin have
lost the sensitivity after 10 years (658). Thus, patients with
recent reactions are more likely to be allergic than patients
with remote reactions, and patients who had allergic reactions
in the distant past might no longer be reactive.
In a Baltimore, Maryland, STI clinic study, only 7.1% of
the patients who reported allergy to penicillin or to another
ß-lactam antibiotic had an objective positive test for penicillin
allergy (659). Moreover, in studies that have incorporated
penicillin skin testing and graded oral challenge among persons
with reported penicillin allergy, the true rates of allergy are low,
ranging from 1.5% to 6.1% (665667). Studies in preoperative
surgical patients with reported penicillin allergy, evaluated
for cardiovascular surgery (668) or orthopedics (669), have
rates of skin test positivity <8.5%. However, when patients
with high-risk penicillin allergy histories are excluded, 99%
of patients could receive ß-lactams. In hospitalized patients
and other populations with comorbidities, the typical rates of
validated penicillin allergy among patients who report a history
of penicillin allergy are 2.5%–9.0% (670673).
Cross-Reactivity with Cephalosporins
Penicillin and cephalosporins both contain a ß-lactam ring.
This structural similarity has led to considerable confusion
regarding cross-reactivity of these drugs and the risks for
allergic reactions from cephalosporins among penicillin-
allergic patients. In most clinical settings, patients with
reported penicillin allergy are precluded from treatment
with such cephalosporin antibiotics as ceftriaxone. Third-
generation cephalosporins (e.g., ceftriaxone and cefixime) have
lower cross-reactivity with IgE-mediated penicillin-allergic
patients (<1%) compared with first- and second-generation
cephalosporins (range: 1%–8%). Moreover, anaphylaxis
secondary to cephalosporins is extremely rare among persons
who report a penicillin allergy and is estimated to occur at a rate
of one per 52,000 persons (652). Data from the Kaiser health
care system reported that among 3,313 patients with self-
reported cephalosporin allergy who received a cephalosporin
(mostly first generation), no cases of anaphylaxis were reported
(652). Use of third- and fourth-generation cephalosporins
and carbapenems is safe for patients without a history of any
IgE-mediated symptoms (e.g., anaphylaxis or urticaria) from
penicillin during the preceding 10 years.
Validating Penicillin or Another ß-Lactam
Antibiotic Allergy
Evaluating a patient who reports a penicillin or another
ß-lactam antibiotic allergy involves three steps: 1) obtaining
a thorough medical history, including previous exposures to
penicillin or other ß-lactam antibiotics (658); 2) performing
a skin test evaluation by using the penicillin major and minor
determinants; and 3) among those who have a negative
penicillin skin test, performing an observed oral challenge with
250 mg amoxicillin before proceeding directly to treatment
with the indicated ß-lactam therapy (667,675).
For persons who have a positive skin test reactive to penicillin
(either to the major or minor determinants), treatment with a
ß-lactam antibiotic is not usually advised, and other effective
antimicrobials should be used (656,658). For persons among
whom the only therapy option is a penicillin antibiotic (e.g., a
patient with neurosyphilis or a pregnant woman with syphilis)
and among whom a penicillin skin test is positive, induction
of penicillin tolerance (also referred to as desensitization) is
required (675). Desensitization protocols to penicillin should
Recommendations and Reports
58 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
be performed by allergists, and they require a monitored
inpatient environment.
Penicillin Skin Testing
Penicillin skin testing with a major determinant
analog (penicilloyl-polylysine) and minor determinants
(benzylpenicilloate, benzylpenilloate, or benzylpenicillin
isomers of penicillin) are used for skin test evaluation for
IgE-dependent penicillin allergy and can reliably identify
persons at high risk for IgE-mediated reactions to penicillin
(658,660,676). Until recently, penicillin skin testing in the
United States only included the major determinant benzyl
penicillin poly-L-lysine (Pre-Pen) in addition to penicillin
G. This test identifies approximately 90%–99% of the IgE-
mediated penicillin-allergic patients. Because the remaining
1%–10% of penicillin-allergic patients who are not captured
by this penicillin skin test are due to minor determinants IgE
antibodies, the standard practice is to follow skin testing with
an observed oral challenge of amoxicillin 250 mg with 1 hour
of observation. If the skin test and oral challenge are both
negative, the risk for IgE-mediated anaphylaxis approaches zero
and is equivalent to that of a person who has never reported
an allergy to penicillin.
A revised version of the penicillin skin test kit, which includes
the major determinant reagent Pre-Pen, minor determinants,
and amoxicillin, is being evaluated by FDA. This penicillin
skin test kit has been evaluated among 455 patients (677)
with previous allergy history and has a negative predictive
value of 98%. If approved, this kit might eliminate the need
for oral challenge.
Penicillin skin testing has become a clinically significant
element in antibiotic stewardship programs, and the
procedure has been increasingly used by hospital-based
pharmacists, hospitalists, and infectious disease physicians
(670,672,673,678,679) as part of overall antibiotic stewardship
interventions. When integrated into stewardship, the rates of
ß-lactam antibiotic use increased substantially (670).
Recommendations
Persons with a history of severe adverse cutaneous
reaction (e.g., Stevens-Johnson syndrome or toxic epidermal
necrolysis) and other severe non–IgE-mediated reactions (e.g.,
interstitial nephritis or hemolytic anemia) are not candidates
for penicillin skin testing or challenge. Penicillin and any
other ß-lactam antibiotics should be avoided indefinitely
among these patients, who should be referred to an allergy
center for further evaluation. Similarly, patients who deny
penicillin allergy, but who report previous IgE-type reactions
to cephalosporins, should be referred to an allergist for specific
cephalosporin testing.
In a time of increasing antimicrobial resistance, following
recommended use of antibiotic treatments is crucial. STI
programs and clinicians should promote increased access to
penicillin allergy testing. Allergy testing is being provided by
clinicians in primary care and hospital settings. If appropriate,
STI programs and ambulatory settings should consider
developing expanded access to penicillin or ß-lactam allergy
assessment.
Persons with high-risk symptom histories (e.g., anaphylaxis
within the previous 10 years) should not be administered
penicillin or a ß-lactam antibiotic in an ambulatory setting.
Furthermore, these persons with high-risk symptoms should
not receive penicillin skin testing or amoxicillin oral challenge
in an ambulatory STI setting and should be referred to an
allergist for further evaluation.
High-risk symptom histories include development of
the following after penicillin or ß-lactam administration:
anaphylaxis within 6 hours or severe adverse cutaneous reaction
(e.g., eosinophilia and systemic symptoms, Stevens-Johnson
syndrome, toxic epidermal necrolysis, or acute generalized
exanthematous pustulosis) and other severe non–IgE-mediated
reactions (e.g., kidney or hepatic injury, hemolytic anemia, or
thrombocytopenia).
Direct Treatment Approach for Ceftriaxone
Among persons with confirmed IgE-mediated penicillin
allergy, the level of cross-reactivity with third-generation
cephalosporins is low (652,680,681). If a patient has a low-
risk history for an IgE-mediated penicillin allergy, ambulatory
settings often treat with third-generation cephalosporins
without further testing. Low-risk history includes one
nonspecific symptom (e.g., gastrointestinal intolerance,
headache, fatigue, or nonurticarial rash) (Box 2). In addition,
a family history of penicillin or ß-lactam allergy alone is not a
contraindication for treatment with ß-lactam antibiotics. This
practice is increasingly being used in ambulatory settings and
for preoperative prophylaxis (658,663,680,682684).
BOX 2. Low-risk history in patients who report penicillin allergy
Gastrointestinal symptoms
Headache
Pruritis without rash
Localized rash
Delayed onset rash (>24 hours)
Symptoms unknown
Family history of penicillin or another drug allergy
Patient denies allergy but it is on the medical record
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 59
US Department of Health and Human Services/Centers for Disease Control and Prevention
Patients at Low Risk for Oral Challenge
If the patient gives only a low-risk history of IgE-mediated
penicillin allergy that includes symptoms such gastrointestinal
intolerance, headache, fatigue, or nonspecific pruritus, or gives
a family history only, an oral challenge can be administered
to document the absence of allergy (Box 2). If the reaction
occurred in the distant past (>10 years), the likelihood is
reduced even further (653,658,663,682,683,685,686). The
risk for severe amoxicillin-mediated anaphylaxis has decreased
over time and is rare. In the United Kingdom during 1972–
2007, one fatal case of amoxicillin-medicated anaphylaxis was
reported (684).
Skin Testing for Penicillin Allergy
Skin testing for penicillin allergy should be performed if
any indication exists that the symptoms were secondary to an
IgE-mediated hypersensitivity. Testing is also indicated as a
potential diagnostic procedure to definitively rule out penicillin
allergy and document a negative allergy status in the medical
record (i.e., delabeling). Because penicillin allergy testing
does not test for multiple minor determinants, a person with
a negative skin test should follow up with an oral challenge to
confirm the negative status.
Persons with negative results of a penicillin skin test, followed by
an amoxicillin oral challenge, can receive conventional penicillin
therapy safely if needed. Persons with positive skin test results and
for whom no other clinical options exist (e.g., neurosyphilis and
syphilis in a pregnant woman) should be referred to an allergist
and desensitized before initiating treatment.
Testing Procedures
Penicillin skin testing includes use of skin test reagents for
identifying persons at risk for adverse reactions (Box 3), followed
by initial pinprick screening with penicillin major determinants
(Pre-Pen) and penicillin G, followed by intradermal testing if
pinprick results are negative. Penicillin testing procedures are
performed in accordance with the Pre-Pen test kit instructions
(https://penallergytest.com/wp-content/uploads/PRE-PEN-
Package-Insert.pdf). Saline negative controls and histamine
positive controls are an integral part of the procedure. Penicillin
skin testing should not be performed for patients who have
taken antihistamines within the past 7 days.
Skin testing can be safely performed by trained
nonallergists and has been implemented as an antimicrobial
stewardship intervention by internal medicine physicians,
pharmacists, hospitalists, and infectious disease physicians
(670,673,678,679). Patients tested should also receive
documentation of status, and the results should be entered in
the medical record.
Penicillin skin testing during pregnancy is considered safe.
For pregnant persons who report a penicillin or ß-lactam
allergy, penicillin allergy is an important consideration in
treating syphilis during pregnancy and the potential for group
B streptococcal infection and preoperative prophylaxis if a
cesarean delivery is required. However, oral challenges should
not be performed unless in a setting where additional support
services are available.
Managing Persons Being Tested
Patients who have a positive skin test should not receive
ß-lactam drugs in the ambulatory setting and should be
referred to an allergist or penicillin allergy expert for further
evaluation. The allergy testing results should be documented
in the medical record. Patients who test negative should be
informed that their risk for anaphylaxis is extremely low and is
equivalent to a person who does not report an allergy history. If
treatment with penicillin or ceftriaxone is indicated, it can be
administered safely. Documentation of testing results should
be provided to the patient.
Desensitization
Desensitization is required for persons who have a
documented penicillin allergy and for whom no therapeutic
alternatives exist (e.g., syphilis during pregnancy and persons
with neurosyphilis). Modified protocols might be considered
BOX 3. Skin test reagents for identifying persons at risk for
adverse reactions to penicillin
Major determinant
Benzylpenicilloyl polylysine injection (Pre-Pen)
(AllerQuest) (6 × 10
-5
M)
Minor determinant precursors
Benzylpenicillin G (10
-2
M, 3.3 mg/mL, 10,000 units/mL)
Benzylpenicilloate (10
-2
M, 3.3 mg/mL)
Benzylpenicilloate (or penicilloyl propylamine)
(10
-2
M, 3.3 mg/mL)
Aged penicillin is not an adequate source of minor
determinants. Penicillin G should either be freshly
prepared or come from a fresh-frozen source.
Positive control
Commercial histamine for scratch testing (1.0 mg/mL)
Negative control
Diluent (usually saline) or allergen diluent
Source: Adapted from Saxon A, Beall GN, Rohr AS, Adelman DC. Immediate
hypersensitivity reactions to beta-lactam antibiotics. Ann Intern Med
1987;107:204−15.
Recommendations and Reports
60 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
on the basis of the clinical syndrome, drug of choice, and route
of administration (687690). Patients might require referral
to a specialty center where desensitization can be performed.
Allergy Referral Resources
With increased access to skin testing kits and the need to
better target therapy for gonorrhea and syphilis, programs
should identify local allergy consultant resources.
Diseases Characterized by Urethritis
and Cervicitis
Urethritis
Urethritis, as characterized by urethral inflammation, can
result from either infectious or noninfectious conditions.
Symptoms, if present, include dysuria, urethral pruritis,
and mucoid, mucopurulent, or purulent discharge. Signs of
urethral discharge on examination can also be present among
persons without symptoms. Although N. gonorrhoeae and C.
trachomatis are well established as clinically important infectious
causes of urethritis, M. genitalium has been strongly associated
with urethritis and, less commonly, prostatitis (691697). If
POC diagnostic tools (e.g., Gram, methylene blue [MB], or
gentian violet [GV] stain microscopy) are unavailable, drug
regimens effective against both gonorrhea and chlamydia
should be administered. Further testing to determine the
specific etiology is recommended for preventing complications,
reinfection, and transmission because a specific diagnosis might
improve treatment compliance, delivery of risk-reduction
interventions, and partner services. Both chlamydia and
gonorrhea are reportable to health departments. NAATs are
preferred for detecting C. trachomatis and N. gonorrhoeae, and
urine is the preferred specimen for males (553). NAAT-based
tests for diagnosing T. vaginalis among men with urethritis
have not been cleared by FDA; however, laboratories have
performed the CLIA-compliant validation studies (698)
needed to provide such testing.
Etiology
Multiple organisms can cause infectious urethritis. The
presence of gram-negative intracellular diplococci (GNID)
or purple intracellular diplococci (MB or GV) on urethral
smear is indicative of presumed gonococcal infection,
which is frequently accompanied by chlamydial infection.
Nongonococcal urethritis (NGU), which is diagnosed when
microscopy of urethral secretions indicate inflammation
without GNID or MB or GV purple intracellular diplococci,
is caused by C. trachomatis in 15%–40% of cases; however,
prevalence varies by age group, with a lower proportion of
disease occurring among older men (699). Documentation
of chlamydial infection as NGU etiology is essential because
of the need for partner referral for evaluation and treatment
to prevent complications of chlamydia, especially for female
partners. Complications of C. trachomatis–associated NGU
among males include epididymitis, prostatitis, and reactive
arthritis.
M. genitalium is associated with symptoms of urethritis and
urethral inflammation and accounts for 15%–25% of NGU
cases in the United States (691693,696,697,700). Among
men with symptoms of urethritis, M. genitalium was detected
in 11% of those with urethritis in Australia (701), 12%–15%
in the United Kingdom (702704), 15% in South Africa (696),
19% in China (705), 21% in Korea, 22% in Japan (706), and
28.7% in the United States (range: 20.4%–38.8%) (697). Data
are inconsistent regarding other Mycoplasma and Ureaplasma
species as etiologic agents of urethritis (707). The majority
of men with Ureaplasma infections do not have overt disease
unless a high organism load is present.
T. vaginalis can cause urethritis among heterosexual
men; however, the prevalence varies substantially by U.S.
geographic region, age, and sexual behavior and within specific
populations. Studies among men with and without overt
urethritis in developed countries document relatively low
rates of T. vaginalis in the Netherlands (0.5%) (708), Japan
(1.3%) (706,709), the United States (2.4%) (710), and the
United Kingdom (3.6%) (703). Studies in other countries have
documented higher rates, such as in Croatia (8.2%) (711) and
Zimbabwe (8.4%) (712), particularly among symptomatic
patients.
Neisseria meningitidis can colonize mucosal surfaces and
cause urethritis (713). Urogenital N. meningitidis rates
and duration of carriage, prevalence of asymptomatic and
symptomatic infection, and modes of transmission have
not been systematically described; however, studies indicate
that N. meningitidis can be transmitted through oral-
penile contact (i.e., fellatio) (714716). N. meningitidis
has similar colony morphology appearance on culture and
cannot be distinguished from N. gonorrhoeae on Gram stain.
Identification of N. meningitidis as the etiologic agent with
presumed gonococcal urethritis on the basis of Gram stain
but negative NAAT for gonorrhea requires a confirmation
by culture. Meningococcal urethritis is treated with the same
antimicrobial regimens as gonococcal urethritis. Although
evidence is limited regarding the risk for sexual transmission or
recurrent infections with meningococcal urethritis, treatment
of sex partners of patients with meningococcal urethritis with
the same antimicrobial regimens as for exposure to gonococcal
infection can be considered. No indication exists for treating
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 61
US Department of Health and Human Services/Centers for Disease Control and Prevention
persons with N. meningitidis identified in their oropharynx
when not also associated with symptomatic urethritis.
In other instances, NGU can be caused by HSV, Epstein-
Barr virus, and adenovirus (699) acquired by fellatio (i.e., oral-
penile contact). In a retrospective review of 80 cases of HSV
urethritis in Australia (717), the majority of infections were
associated with HSV-1 with clinical findings of meatitis (62%),
genital ulceration (37%), and dysuria (20%). Adenovirus can
present with dysuria, meatal inflammation, and conjunctivitis
(718). Enteric bacteria have been identified as an uncommon
cause of NGU and might be associated with insertive anal
intercourse (699).
Other bacterial pathogens have been implicated as potential
causes of clinical urethritis, either in clustered case series or as
sporadic cases such as Haemophilus influenzae and Haemophilus
parainfluenzae (719723). Haemophilus was identified in 12.6%
of cases among 413 men (mostly MSM reporting insertive oral
sex) (724), and high rates of azithromycin resistance (39.5%)
were identified among Haemophilus urethritis patients (725).
Individual case reports have linked NGU to multiple bacterial
species, including Corynebacterium propinquum (726), Kurthia
gibsonii (727), Corynebacterium glucuronolyticum (728,729),
Corynebacterium striatrium (730), Aerococcus urinae (731),
and Neisseria elongata (732). Diagnostic testing and treatment
for less-common organisms are reserved for situations in
which these infections are suspected (e.g., sexual partner with
trichomoniasis, urethral lesions, or severe dysuria and meatitis)
or when NGU is not responsive to recommended therapy.
Even in settings that provide comprehensive diagnostic
testing, etiology can remain obscure in half of cases. Idiopathic
NGU was reported in 772 (59%) of 1,295 first presentations
of NGU among men seeking sexual health services in Australia
(701). In a case-control study of 211 men with NGU
symptoms in Denmark, no identifiable pathogen was identified
in 24% of acute cases and 33% of chronic cases (733). NGU’s
importance if not caused by a defined pathogen is uncertain;
neither complications (e.g., urethral stricture or epididymitis)
nor adverse outcomes among sex partners have been identified
in these cases.
Associations between NGU and insertive anal and oral
exposure have been reported (734), as have higher rates
of BV-associated Leptotrichia or Sneathia species among
heterosexual men with urethritis (735). These studies increase
concern for possible undetected infectious rectal or vaginal
pathogens, or alternatively, a transient reactive dysbiosis after
exposure to a new microbiome or even a noninfectious reactive
etiology (736).
Diagnostic Considerations
Clinicians should attempt to obtain objective evidence of
urethral inflammation. If POC diagnostic tests (e.g., Gram
stain or MB or GV microscopy) are unavailable, urethritis
can be documented on the basis of any of the following signs
or laboratory tests:
Mucoid, mucopurulent, or purulent discharge on examination.
Gram stain is a POC diagnostic test for evaluating
urethritis that is highly sensitive and specific for
documenting both urethritis and the presence or absence
of gonococcal infection; MB or GV stain of urethral
secretions is an alternative POC diagnostic test with
performance characteristics similar to Gram stain; thus,
the cutoff number for WBCs per oil immersion field
should be the same (737).
ű
Presumed gonococcal infection is established by
documenting the presence of WBCs containing GNID
in Gram stain or intracellular purple diplococci in MB
or GV smears; men should be tested for C. trachomatis
and N. gonorrhoeae by NAATs and presumptively treated
and managed accordingly for gonococcal infection (see
Gonococcal Infections).
ű
If no intracellular gram-negative or purple diplococci
are present, men should receive NAATs for C. trachomatis
and N. gonorrhoeae and can be managed for NGU as
recommended (see Nongonococcal Urethritis).
ű
Gram stain of urethral secretions exist that demonstrate
≥2 WBCs per oil immersion field (738). The microscopy
diagnostic cutoff might vary, depending on background
prevalence (≥2 WBCs/high power field [HPF] in high-
prevalence settings [STI clinics] or ≥5 WBCs/HPF in
lower-prevalence settings).
§§
§§
For urethral microscopy, the cutoff for diagnosing urethritis is ≥2 WBCs/HPF
(Sources: Rietmeijer CA, Mettenbrink CJ. Recalibrating the Gram stain
diagnosis of male urethritis in the era of nucleic acid amplification testing. Sex
Transm Dis 2012;39:18–20; Rietmeijer CA, Mettenbrink CJ. The diagnosis
of nongonococcal urethritis in men: can there be a universal standard? Sex
Transm Dis 2017;44:195–6). An additional evaluation supported this cutoff
by demonstrating NGU sensitivity of 92.6% for cutoff of ≥2 versus 55.6%
sensitivity for cutoff of ≥5 (Source: Sarier M, Sepin N, Duman I, et al.
Microscopy of Gram-stained urethral smear in the diagnosis of urethritis: which
threshold value should be selected? Andrologia 2018;50:e13143). Diagnostic
cutoffs for 369 symptomatic and asymptomatic heterosexual men seeking STI
care in Seattle revealed a maximal sensitivity and specificity achieved with a
cutoff of ≥5 WBCs/HPF. Using a lower cutoff of ≥2 WBCs/HPF would miss
13% of C. trachomatis and M. genitalium and overtreat 45% of persons who
have negative tests (Source: Leipertz G, Chambers L, Lowens S, et al. P796
Reassessing the Gram stain smear [GSS] polymorphonuclear leukocyte [PMN]
cutoff for diagnosing non-gonococcal urethritis [NGU]. Sex Transm Infect
2019;95[Suppl 1]:A339). Another study discussed that the WBC/HPF cutoff
value should discriminate on the basis of the prevalence of chlamydia,
mycoplasma, and gonorrhea among a clinic population (Source: Moi H,
Hartgill U, Skullerud KH, Reponen EJ, Syvertsen L, Moghaddam A.
Microscopy of stained urethral smear in male urethritis: which cutoff should
be used? Sex Transm Dis 2017;44:189–94).
Recommendations and Reports
62 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Positive leukocyte esterase test on first-void urine or
microscopic examination of sediment from a spun first-
void urine demonstrating ≥10 WBCs/HPF.
Men evaluated in settings in which Gram stain or MB or
GV smear is unavailable who meet at least one criterion for
urethritis (i.e., urethral discharge, positive leukocyte esterase
test on first void urine, or microscopic examination of first-
void urine sediment with ≥10 WBCs/HPF) should be tested
for C. trachomatis and N. gonorrhoeae by NAATs and treated
with regimens effective against gonorrhea and chlamydia.
If symptoms are present but no evidence of urethral
inflammation is present, NAATs for C. trachomatis and
N. gonorrhoeae might identify infections (739). Persons
with chlamydia or gonorrhea should receive recommended
treatment, and sex partners should be referred for evaluation
and treatment. If none of these clinical criteria are present,
empiric treatment of men with symptoms of urethritis is
recommended only for those at high risk for infection who are
unlikely to return for a follow-up evaluation or test results. Such
men should be treated with drug regimens effective against
gonorrhea and chlamydia.
Nongonococcal Urethritis
NGU is a nonspecific diagnosis that can have various
infectious etiologies. C. trachomatis has been well established
as an NGU etiology; however, prevalence varies across
populations and accounts for <50% of overall cases (712,740
742). M. genitalium is estimated to account for 10%–25% of
cases (696,697,701,703,704,706,733,743), and T. vaginalis
for 1%–8% of cases depending on population and location
(703,706,708,710,712). Other etiologies include different
bacteria, such as Haemophilus species (724,725), N. meningitidis
(713,716), HSV (706,717), and adenovirus (744). However,
even when extensive testing is performed, no pathogens are
identified in approximately half of cases (701,733).
Diagnostic Considerations
Clinical presentation can include urethral discharge,
irritation, dysuria, or meatal pruritus (697,743,745). NGU is
confirmed for symptomatic men when diagnostic evaluation of
urethral secretions indicates inflammation, without evidence
of diplococci by Gram, MB, or GV smear on microscopy
(712,746,747). Visible discharge or secretions can be collected
by a swab without inserting it into the urethra; if no visible
secretions, the swab can be inserted into the urethral meatus
and rotated, making contact with the urethral wall before
removal. If microscopy is unavailable, urine testing for
leukocyte esterase can be performed on first-void urine, and
microscopic examination of sediment from a spun first-void
urine demonstrating ≥10 WBCs/HPF has a high negative
predictive value.
All men who have suspected or confirmed NGU should
be tested for chlamydia and gonorrhea by using NAATs.
A specific diagnosis can potentially reduce complications,
reinfection, and transmission. M. genitalium testing should be
performed for men who have persistent or recurrent symptoms
after initial empiric treatment. Testing for T. vaginalis should
be considered in areas or among populations with high
prevalence, in cases where a partner is known to be infected,
or for men who have persistent or recurrent symptoms after
initial empiric treatment.
Treatment
Ideally, treatment should be pathogen based; however,
diagnostic information might not be immediately available.
Presumptive treatment should be initiated at NGU diagnosis.
Doxycycline is highly effective for chlamydial urethral
infections and is also effective for chlamydial infections of
the rectum; it also has some activity against M. genitalium.
In contrast, reports have increased of azithromycin treatment
failures for chlamydial infection (748,749), and the incidence
of macrolide resistance in M. genitalium also has been rapidly
rising (697,702,705,750,751). Pharmacokinetic data indicate
that changing azithromycin dosing from a single-dose strategy
to a multiday strategy might protect against inducing resistance
in M. genitalium infections (745,752) (see Mycoplasma
genitalium).
Recommended Regimen for Nongonococcal Urethritis
Doxycycline 100 mg orally 2 times/day for 7 days
Alternative Regimens
Azithromycin 1 g orally in a single dose
or
Azithromycin 500 mg orally in a single dose; then 250 mg orally daily
for 4 days
To maximize compliance with recommended therapies,
medications should be dispensed on-site at the clinic, and,
regardless of the number of doses involved in the regimen,
the first dose should be directly observed. Erythromycin is no
longer recommended for NGU because of its gastrointestinal
side effects and dosing frequency. Levofloxacin is no longer
recommended for NGU because of its inferior efficacy,
especially for M. genitalium.
Management Considerations
To minimize transmission and reinfections, men treated for
NGU should be instructed to abstain from sexual intercourse
until they and their partners have been treated (i.e., until
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 63
US Department of Health and Human Services/Centers for Disease Control and Prevention
completion of a 7-day regimen and symptoms have resolved or
for 7 days after single-dose therapy). Men with NGU should
be tested for HIV and syphilis.
Follow-Up
Men should be provided their testing results obtained as
part of the NGU evaluation. Those with a specific diagnosis
of chlamydia, gonorrhea, or trichomoniasis should be
offered partner services and instructed to return 3 months
after treatment for repeat testing because of high rates of
reinfection, regardless of whether their sex partners were treated
(136,137,753,754) (see Chlamydial Infections; Gonococcal
Infections; Trichomoniasis).
If symptoms persist or recur after therapy completion, men
should be instructed to return for reevaluation and should be
tested for M. genitalium and T. vaginalis. Symptoms alone,
without documentation of signs or laboratory evidence of
urethral inflammation, are insufficient basis for retreatment.
Providers should be alert to the possible diagnosis of chronic
prostatitis or chronic pelvic pain syndrome in men experiencing
persistent perineal, penile, or pelvic pain or discomfort;
voiding symptoms; pain during or after ejaculation; or new-
onset premature ejaculation lasting for >3 months. Men with
persistent pain should be referred to a urologist with expertise
in pelvic pain disorders.
Management of Sex Partners
All sex partners of men with NGU within the preceding
60 days should be referred for evaluation and testing and
presumptive treatment with a drug regimen effective against
chlamydia. All partners should be evaluated and treated
according to the management section for their respective
pathogen; EPT could be an alternate approach if a partner is
unable to access timely care. To avoid reinfection, sex partners
should abstain from sexual intercourse until they and their
partners are treated.
Persistent or Recurrent Nongonococcal Urethritis
The objective diagnosis of persistent or recurrent NGU
should be made before considering additional antimicrobial
therapy. Symptomatic recurrent or persistent urethritis might
be caused by treatment failure or reinfection after successful
treatment. Among men who have persistent symptoms after
treatment without objective signs of urethral inflammation, the
value of extending the duration of antimicrobials has not been
demonstrated. Treatment failure for chlamydial urethritis has
been estimated at 6%–12% (755). The most common cause
of persistent or recurrent NGU is M. genitalium, especially
after doxycycline therapy (756,757). Treatment failure for
M. genitalium is harder to determine because certain men
achieve clinical cure (i.e., resolution of symptoms) but can
still have detectable M. genitalium in urethral specimens (758).
The initial step in recurrent urethritis is assessing compliance
with treatment or potential reexposure to an untreated sex
partner (697,743). If the patient did not comply with the
treatment regimen or was reexposed to an untreated partner,
retreatment with the initial regimen can be considered. If
therapy was appropriately completed and no reexposure
occurred, therapy is dependent on the initial treatment
regimen. Ideally, diagnostic testing among men with recurrent
or persistent symptoms, including those with gonorrhea,
chlamydia, M. genitalium, and trichomoniasis, can be used to
guide further management decisions.
T. vaginalis is also known to cause urethritis among men who
have sex with women. In areas where T. vaginalis is prevalent,
men who have sex with women with persistent or recurrent
urethritis should be tested for T. vaginalis and presumptively
treated with metronidazole 2 g orally in a single dose or
tinidazole 2 g orally in a single dose; their partners should be
referred for evaluation and treatment, if needed.
If T. vaginalis is unlikely (MSM with NGU or negative
T. vaginalis NAAT), men with recurrent NGU should be tested
for M. genitalium by using an FDA-cleared NAAT. Treatment
for M. genitalium includes a two-stage approach, ideally using
resistance-guided therapy. If M. genitalium resistance testing is
available it should be performed, and the results should be used
to guide therapy (see Mycoplasma genitalium). If M. genitalium
resistance testing is not available, doxycycline 100 mg orally
2 times/day for 7 days followed by moxifloxacin 400 mg orally
once daily for 7 days should be used. The rationale for this
approach is that although not curative, doxycycline decreases
the M. genitalium bacterial load, thereby increasing likelihood
of moxifloxacin success (759). Higher doses of azithromycin
have not been effective for M. genitalium after azithromycin
treatment failures. Men with persistent or recurrent NGU after
treatment for M. genitalium or T. vaginalis should be referred
to an infectious disease or urology specialist.
Special Considerations
HIV Infection
NGU might facilitate HIV transmission (760). Persons with
NGU and HIV infection should receive the same treatment
regimen as those who do not have HIV.
Cervicitis
Two major diagnostic signs characterize cervicitis: 1) a
purulent or mucopurulent endocervical exudate visible in
the endocervical canal or on an endocervical swab specimen
(commonly referred to as mucopurulent cervicitis), and
Recommendations and Reports
64 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
2) sustained endocervical bleeding easily induced by gentle
passage of a cotton swab through the cervical os. Either or both
signs might be present. Cervicitis frequently is asymptomatic;
however, certain women might report an abnormal vaginal
discharge and intermenstrual vaginal bleeding (e.g., especially
after sexual intercourse). The criterion of using an increased
number of WBCs on endocervical Gram stain in the diagnosis
of cervicitis has not been standardized; it is not sensitive,
has a low positive predictive value for C. trachomatis and
N. gonorrhoeae infections, and is not available in most clinical
settings (297,761). Leukorrhea, defined as >10 WBCs/HPF on
microscopic examination of vaginal fluid, might be a sensitive
indicator of cervical inflammation with a high negative
predictive value (i.e., cervicitis is unlikely in the absence of
leukorrhea) (762,763). Finally, although the presence of gram-
negative intracellular diplococci on Gram stain of endocervical
exudate might be specific for diagnosing gonococcal cervical
infection when evaluated by an experienced laboratorian, it is
not a sensitive indicator of infection (764).
Etiology
C. trachomatis or N. gonorrhoeae is the most common etiology
of cervicitis defined by diagnostic testing. Trichomoniasis,
genital herpes (especially primary HSV-2 infection), or
M. genitalium (761,765768) also have been associated
with cervicitis. However, in many cases of cervicitis, no
organism is isolated, especially among women at relatively
low risk for recent acquisition of these STIs (e.g., women aged
>30 years) (769). Limited data indicate that BV and frequent
douching might cause cervicitis (770772). The majority
of persistent cases of cervicitis are not caused by reinfection
with C. trachomatis or N. gonorrhoeae; other factors might
be involved (e.g., persistent abnormality of vaginal flora,
M. genitalium, douching or exposure to other types of chemical
irritants, dysplasia, or idiopathic inflammation in the zone of
ectopy). Available data do not indicate an association between
group B streptococcus colonization and cervicitis (773,774).
No specific evidence exists for a role for Ureaplasma parvum or
Ureaplasma urealyticum in cervicitis (707,761,765,775,776).
Diagnostic Considerations
Because cervicitis might be a sign of upper genital tract
infection (e.g., endometritis), women should be assessed for
signs of PID and tested for C. trachomatis and N. gonorrhoeae
with NAAT on vaginal, cervical, or urine samples (553) (see
Chlamydial Infections; Gonococcal Infections). Women
with cervicitis also should be evaluated for concomitant BV
and trichomoniasis. Because sensitivity of microscopy for
detecting T. vaginalis is relatively low (approximately 50%),
symptomatic women with cervicitis and negative wet-mount
microscopy for trichomonads should receive further testing
(i.e., NAAT, culture, or other FDA-cleared diagnostic test)
(see Trichomoniasis). Testing for M. genitalium with the FDA-
cleared NAAT can be considered. Although HSV-2 infection
has been associated with cervicitis, the utility of specific testing
(i.e., PCR or culture) for HSV-2 is unknown. Testing for
U. parvum, U. urealyticum, Mycoplasma hominis, or genital
culture for group B streptococcus is not recommended.
Treatment
Multiple factors should affect the decision to provide
presumptive therapy for cervicitis. Presumptive treatment with
antimicrobials for C. trachomatis and N. gonorrhoeae should
be provided for women at increased risk (e.g., those aged
<25 years and women with a new sex partner, a sex partner
with concurrent partners, or a sex partner who has an STI), if
follow-up cannot be ensured, or if testing with NAAT is not
possible. Trichomoniasis and BV should be treated if detected
(see Bacterial Vaginosis; Trichomoniasis). For women at lower
risk for STIs, deferring treatment until results of diagnostic
tests are available is an option. If treatment is deferred and
C. trachomatis and N. gonorrhoeae NAATs are negative, a
follow-up visit to determine whether the cervicitis has resolved
can be considered.
Recommended Regimen for Cervicitis*
Doxycycline 100 mg orally 2 times/day for 7 days
* Consider concurrent treatment for gonococcal infection if the patient is
at risk for gonorrhea or lives in a community where the prevalence of
gonorrhea is high (see Gonococcal Infections).
Alternative Regimen
Azithromycin 1 g orally in a single dose
Other Management Considerations
To minimize transmission and reinfection, women treated
for cervicitis should be instructed to abstain from sexual
intercourse until they and their partners have been treated
(i.e., until completion of a 7-day regimen or for 7 days after
single-dose therapy) and symptoms have resolved. Women
who receive a cervicitis diagnosis should be tested for syphilis
and HIV in addition to other recommended diagnostic tests.
Follow-Up
Women receiving treatment should return to their provider
for a follow-up visit to determine whether cervicitis has
resolved. For women who are untreated, a follow-up visit gives
providers an opportunity to communicate test results obtained
as part of the cervicitis evaluation. Providers should treat on
the basis of any positive test results and determine whether
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 65
US Department of Health and Human Services/Centers for Disease Control and Prevention
cervicitis has resolved. Women with a specific diagnosis of
chlamydia, gonorrhea, or trichomoniasis should be offered
partner services and instructed to return in 3 months after
treatment for repeat testing because of high rates of reinfection,
regardless of whether their sex partners were treated (753). If
symptoms persist or recur, women should be instructed to
return for reevaluation.
Management of Sex Partners
Management of sex partners of women treated for cervicitis
should be tailored for the specific infection identified or
suspected. All sex partners during the previous 60 days should
be referred for evaluation, testing, and presumptive treatment
if chlamydia, gonorrhea, or trichomoniasis was identified. EPT
and other effective partner referral strategies are alternative
approaches for treating male partners of women who have
chlamydial or gonococcal infection (125127) (see Partner
Services). To avoid reinfection, sex partners should abstain
from sexual intercourse until they and their partners are treated.
Persistent or Recurrent Cervicitis
Women with persistent or recurrent cervicitis despite
antimicrobial therapy should be reevaluated for possible
reexposure or treatment failure. If relapse or reinfection with
a specific infection has been excluded, BV is not present, and
sex partners have been evaluated and treated, management
options for persistent cervicitis are undefined. In addition,
the usefulness of repeated or prolonged administration of
antimicrobial therapy for persistent symptomatic cervicitis
remains unknown. The etiology of persistent cervicitis,
including the potential role of M. genitalium (777), is unclear.
M. genitalium might be considered for cases of cervicitis that
persist after azithromycin or doxycycline therapy in which
reexposure to an infected partner or medical nonadherence is
unlikely. Among women with persistent cervicitis who were
previously treated with doxycycline or azithromycin, testing
for M. genitalium can be considered and treatment initiated on
the basis of results of diagnostic testing (318) (see Mycoplasma
genitalium). For women with persistent symptoms that are
clearly attributable to cervicitis, referral to a gynecologic
specialist can be considered for evaluation of noninfectious
causes (e.g., cervical dysplasia or polyps) (778).
Special Considerations
HIV Infection
Women with cervicitis and HIV infection should receive
the same treatment regimen as those who do not have HIV.
Cervicitis can increase cervical HIV shedding, and treatment
reduces HIV shedding from the cervix and thereby might
reduce HIV transmission to susceptible sex partners (779783).
Pregnancy
Diagnosis and treatment of cervicitis for pregnant women
does not differ from that for women who are not pregnant (see
Diagnostic Considerations; Treatment).
Contraceptive Management
According to U.S. Medical Eligibility Criteria for Contraceptive
Use, 2016, leaving an IUD in place during treatment for
cervicitis is advisable (58). However, current recommendations
specify that an IUD should not be placed if active cervicitis
is diagnosed (59).
Chlamydial Infections
Chlamydial Infection Among
Adolescents and Adults
Chlamydial infection is the most frequently reported
bacterial infectious disease in the United States, and prevalence
is highest among persons aged ≤24 years (141,784). Multiple
sequelae can result from C. trachomatis infection among
women, the most serious of which include PID, ectopic
pregnancy, and infertility. Certain women who receive a
diagnosis of uncomplicated cervical infection already have
subclinical upper genital tract infection.
Asymptomatic infection is common among both men and
women. To detect chlamydial infection, health care providers
frequently rely on screening tests. Annual screening of all
sexually active women aged <25 years is recommended, as is
screening of older women at increased risk for infection (e.g.,
women aged ≥25 years who have a new sex partner, more
than one sex partner, a sex partner with concurrent partners,
or a sex partner who has an STI) (149). In a community-
based cohort of female college students, incident chlamydial
infection was also associated with BV and high-risk HPV
infection (785). Although chlamydia incidence might be higher
among certain women aged ≥25 years in certain communities,
overall, the largest proportion of infection is among women
aged <25 years (141).
Chlamydia screening programs have been demonstrated
to reduce PID rates among women (786,787). Although
evidence is insufficient to recommend routine screening for
C. trachomatis among sexually active young men because of
certain factors (i.e., feasibility, efficacy, and cost-effectiveness),
screening of sexually active young men should be considered
in clinical settings with a high prevalence of chlamydia (e.g.,
adolescent clinics, correctional facilities, or STD specialty
Recommendations and Reports
66 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
clinics) or for populations with a high burden of infection
(e.g., MSM) (149,788). Among women, the primary focus of
chlamydia screening should be to detect and treat chlamydia,
prevent complications, and test and treat their partners,
whereas targeted chlamydia screening for men should be
considered only when resources permit, prevalence is high, and
such screening does not hinder chlamydia screening efforts for
women (789791). More frequent screening than annual for
certain women (e.g., adolescents) or certain men (e.g., MSM)
might be indicated on the basis of risk behaviors.
Diagnostic Considerations
For women, C. trachomatis urogenital infection can be
diagnosed by vaginal or cervical swabs or first-void urine. For
men, C. trachomatis urethral infection can be diagnosed by
testing first-void urine or a urethral swab. NAATs are the most
sensitive tests for these specimens and are the recommended
test for detecting C. trachomatis infection (553). NAATs that
are FDA cleared for use with vaginal swab specimens can be
collected by a clinician or patient in a clinical setting. Patient-
collected vaginal swab specimens are equivalent in sensitivity
and specificity to those collected by a clinician using NAATs
(792,793), and this screening strategy is highly acceptable
among women (794,795). Optimal urogenital specimen
types for chlamydia screening by using NAAT include first-
catch urine (for men) and vaginal swabs (for women) (553).
Recent studies have demonstrated that among men, NAAT
performance on self-collected meatal swabs is comparable
to patient-collected urine or provider-collected urethral
swabs (796798). Patient collection of a meatal swab for
C. trachomatis testing might be a reasonable approach for men
who are either unable to provide urine or prefer to collect their
own meatal swab over providing urine. Previous evidence
indicates that the liquid-based cytology specimens collected for
Pap smears might be acceptable specimens for NAAT, although
test sensitivity using these specimens might be lower than that
associated with use of cervical or vaginal swab specimens (799);
regardless, certain NAATs have been cleared by FDA for use
on liquid-based cytology specimens.
Rectal and oropharyngeal C. trachomatis infection among
persons engaging in receptive anal or oral intercourse can be
diagnosed by testing at the anatomic exposure site. NAATs have
been demonstrated to have improved sensitivity and specificity,
compared with culture, for detecting C. trachomatis at rectal
and oropharyngeal sites (553,800804), and certain NAAT
platforms have been cleared by FDA for these anatomic sites
(805). Data indicate that NAAT performance on self-collected
rectal swabs is comparable to clinician-collected rectal swabs,
and this specimen collection strategy for rectal C. trachomatis
screening is highly acceptable among men (217,806).
Self-collected rectal swabs are a reasonable alternative to
clinician-collected rectal swabs for C. trachomatis screening by
NAAT, especially when clinicians are not available or when self-
collection is preferred over clinician collection. Annual screening
for rectal C. trachomatis infection should be performed among
men who report sexual activity at the rectal site. Extragenital
chlamydial testing at the rectal site can be considered for
females on the basis of reported sexual behaviors and exposure
through shared clinical decision-making bythe patient and the
provider. The majority of persons with C. trachomatis detected
at oropharyngeal sites do not have oropharyngeal symptoms.
The clinical significance of oropharyngeal C. trachomatis
infection is unclear, and prevalence is low, even among
populations at high risk. However, when gonorrhea testing
is performed at the oropharyngeal site, chlamydia test results
might be reported because certain NAATs detect both bacteria
from a single specimen.
POC tests for C. trachomatis among asymptomatic persons
can expedite treatment of infected persons and their sex
partners. Among symptomatic patients, POC tests for
C. trachomatis can optimize treatment by limiting unnecessary
presumptive treatment at the time of clinical decision-making
and improve antimicrobial stewardship. Thus, using a POC
test will likely be a cost-effective diagnostic strategy for
C. trachomatis infection (807). Newer NAAT-based POC tests
have promising performance and are becoming commercially
available (807809).
Treatment
Treating persons with C. trachomatis prevents adverse
reproductive health complications and continued sexual
transmission. Furthermore, treating their sex partners
can prevent reinfection and infection of other partners.
Treating pregnant women usually prevents transmission of
C. trachomatis to neonates during birth. Treatment should be
provided promptly for all persons with chlamydial infection;
treatment delays have been associated with complications (e.g.,
PID) in a limited proportion of women (810).
Recommended Regimen for Chlamydial Infection Among
Adolescents and Adults
Doxycycline 100 mg orally 2 times/day for 7 days
Alternative Regimens
Azithromycin 1 g orally in a single dose
or
Levofloxacin 500 mg orally once daily for 7 days
A meta-analysis and a Cochrane systematic review evaluated
data from randomized clinical trials of azithromycin versus
doxycycline for treating urogenital chlamydial infection
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 67
US Department of Health and Human Services/Centers for Disease Control and Prevention
determined that microbiologic treatment failure among men
was higher for azithromycin than for doxycycline (748,749).
Observational studies have also demonstrated that doxycycline
is more efficacious for rectal C. trachomatis infection for men
and women than azithromycin (748,811). A randomized trial
for the treatment of rectal chlamydia infection among MSM
reported microbiologic cure was 100% with doxycycline and
74% with azithromycin (812). A published review reported
that C. trachomatis was detected at the anorectal site among
33%–83% of women who had urogenital C. trachomatis
infection, and its detection was not associated with report of
receptive anorectal sexual activity (813).
Although the clinical significance of oropharyngeal
C. trachomatis infection is unclear and routine oropharyngeal
screening is not recommended, oropharyngeal C. trachomatis
can be sexually transmitted to genital sites (211,814); therefore,
if C. trachomatis is identified from an oropharyngeal specimen
while screening for pharyngeal gonorrhea, it should be treated.
Evidence is limited regarding the efficacy of antimicrobial
regimens for oropharyngeal chlamydia; however, a recently
published observational study indicates doxycycline might
be more efficacious than azithromycin for oropharyngeal
chlamydia (815).
Available evidence supports that doxycycline is efficacious
for C. trachomatis infections of urogenital, rectal, and
oropharyngeal sites. Although azithromycin maintains high
efficacy for urogenital C. trachomatis infection among women,
concern exists regarding effectiveness of azithromycin for
concomitant rectal C. trachomatis infection, which can occur
commonly among women and cannot be predicted by reported
sexual activity. Inadequately treated rectal C. trachomatis
infection among women who have urogenital chlamydia
can increase the risk for transmission and place women at
risk for repeat urogenital C. trachomatis infection through
autoinoculation from the anorectal site (816). Doxycycline is
also available in a delayed-release 200-mg tablet formulation,
which requires once-daily dosing for 7 days and is as effective
as doxycycline 100 mg twice daily for 7 days for treating
urogenital C. trachomatis infection in men and women. It is
more costly but also has lower frequency of gastrointestinal
side effects (817). Levofloxacin is an effective treatment
alternative but is more expensive. Erythromycin is no longer
recommended because of the frequency of gastrointestinal side
effects, which can result in nonadherence. When nonadherence
to doxycycline regimen is a substantial concern, azithromycin
1 g regimen is an alternative treatment option but might
require posttreatment evaluation and testing because it has
demonstrated lower treatment efficacy among persons with
rectal infection.
Among persons receiving multidose regimens, medication
should be dispensed with all doses involved, on-site and in
the clinic, and the first dose should be directly observed. To
maximize adherence with recommended therapies, on-site,
directly observed single-dose therapy with azithromycin should
always be available for persons for whom adherence with
multiday dosing is a considerable concern.
Other Management Considerations
To minimize disease transmission to sex partners, persons
treated for chlamydia should be instructed to abstain from
sexual intercourse for 7 days after single-dose therapy or until
completion of a 7-day regimen and resolution of symptoms if
present. To minimize risk for reinfection, patients also should
be instructed to abstain from sexual intercourse until all of their
sex partners have been treated. Persons who receive a diagnosis
of chlamydia should be tested for HIV, gonorrhea, and syphilis.
MSM who are HIV negative with a rectal chlamydia diagnosis
should be offered HIV PrEP.
Follow-Up
Test of cure to detect therapeutic failure (i.e., repeat testing
4 weeks after completing therapy) is not advised for nonpregnant
persons treated with the recommended or alternative regimens,
unless therapeutic adherence is in question, symptoms persist,
or reinfection is suspected. Moreover, using chlamydial NAATs
at <4 weeks after completion of therapy is not recommended
because the continued presence of nonviable organisms
(553,818,819) can lead to false-positive results.
A high prevalence of C. trachomatis infection has been
observed among women and men who were treated
for chlamydial infection during the preceding months
(753,755,820822). The majority of posttreatment infections
do not result from treatment failure but rather from reinfection
caused by failure of sex partners to receive treatment or
initiation of sexual activity with a new infected partner (823),
indicating a need for improved education and treatment of
sex partners. Repeat infections confer an elevated risk for PID
and other complications among women. Men and women
who have been treated for chlamydia should be retested
approximately 3 months after treatment, regardless of whether
they believe their sex partners were treated; scheduling the
follow-up visit at the time of treatment is encouraged (753).
If retesting at 3 months is not possible, clinicians should retest
whenever persons next seek medical care <12 months after
initial treatment.
Management of Sex Partners
Sex partners should be referred for evaluation, testing, and
presumptive treatment if they had sexual contact with the
Recommendations and Reports
68 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
partner during the 60 days preceding the patient’s onset of
symptoms or chlamydia diagnosis. Although the exposure
intervals defining identification of sex partners at risk are
based on limited data, the most recent sex partner should be
evaluated and treated, even if the time of the last sexual contact
was >60 days before symptom onset or diagnosis.
If health department partner management strategies (e.g.,
disease intervention specialists) are impractical or unavailable
for persons with chlamydia, and if a provider is concerned
that sex partners are unable to promptly access evaluation and
treatment services, EPT should be considered as permitted by
law (see Partner Services). Compared with standard patient
referral of partners, this approach to therapy, which involves
delivering the medication itself or a prescription by the
patient or collaborating pharmacy, has been associated with
decreased rates of persistent or recurrent chlamydia among
women (125127). Providers should provide patients with
written educational materials to give to their partners about
chlamydia, which should include notification that partners
have been exposed and information about the importance
of treatment. These materials also should inform partners
about potential therapy-related allergies and adverse effects,
along with symptoms indicative of complications (e.g.,
testicular pain among men and pelvic or abdominal pain
among women). Educational materials for female partners
should include information about the importance of seeking
medical evaluation, especially if PID symptoms are present;
undertreatment of PID among female partners and missed
opportunities for diagnosing other STIs among women
are concerning. MSM with chlamydia have a high risk for
coexisting infections, especially undiagnosed HIV, among
their partners and might have partners without HIV who
could benefit from HIV PrEP. Data are also limited regarding
effectiveness of EPT in reducing persistent or recurrent
chlamydia among MSM (123,133,134); thus, shared clinical
decision-making regarding EPT for MSM is recommended.
Having partners accompany patients when they return for
treatment is another strategy that has been used successfully
for ensuring partner treatment (see Partner Services). To avoid
reinfection, sex partners should be instructed to abstain from
condomless sexual intercourse until they and their sex partners
have been treated (i.e., after completion of a 7-day regimen)
and any symptoms have resolved.
Special Considerations
Pregnancy
Clinical experience and published studies indicate that
azithromycin is safe and effective during pregnancy (824826).
Doxycycline is contraindicated during the second and third
trimesters of pregnancy because of risk for tooth discoloration.
Human data reveal that levofloxacin presents a low risk to the
fetus during pregnancy but has potential for toxicity during
breastfeeding; however, data from animal studies increase
concerns regarding cartilage damage to neonates (431).
Test of cure (i.e., repeat testing after completion of therapy)
to document chlamydial eradication, preferably by NAAT,
at approximately 4 weeks after therapy completion during
pregnancy is recommended because severe sequelae can occur
among mothers and neonates if the infection persists. In
addition, all pregnant women who have chlamydial infection
diagnosed should be retested 3 months after treatment.
Detection of C. trachomatis infection during the third semester
is not uncommon among adolescent and young adult women,
including those without C. trachomatis detected at the time of
initial prenatal screening (827). Women aged <25 years and
those at increased risk for chlamydia (i.e., those who have a
new sex partner, more than one sex partner, a sex partner with
concurrent partners, or a sex partner who has an STI) should
be screened at the first prenatal visit and rescreened during the
third trimester to prevent maternal postnatal complications
and chlamydial infection in the infant (149).
Recommended Regimen for Chlamydial Infection During
Pregnancy
Azithromycin 1 g orally in a single dose
Alternative Regimen
Amoxicillin 500 mg orally 3 times/day for 7 days
Because of concerns regarding chlamydia persistence
after exposure to penicillin-class antibiotics that has been
demonstrated in animal and in vitro studies, amoxicillin is
listed as an alternative therapy for C. trachomatis for pregnant
women (828,829). Erythromycin is no longer recommended
because of the frequency of gastrointestinal side effects that can
result in therapy nonadherence. In addition, systematic reviews
and meta-analyses have noted an association with macrolide
antimicrobials, especially erythromycin, during pregnancy and
adverse child outcomes, indicating cautious use in pregnancy
(830831).
HIV Infection
Persons who have chlamydia and HIV infection should
receive the same treatment regimen as those who do not
have HIV.
Chlamydial Infection Among Neonates
Prenatal screening and treatment of pregnant women is
the best method for preventing chlamydial infection among
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 69
US Department of Health and Human Services/Centers for Disease Control and Prevention
neonates. C. trachomatis infection of neonates results from
perinatal exposure to the mothers infected cervix. Initial
C. trachomatis neonatal infection involves the mucous
membranes of the eye, oropharynx, urogenital tract, and
rectum, although infection might be asymptomatic in these
locations. Instead, C. trachomatis infection among neonates
is most frequently recognized by conjunctivitis that develops
5–12 days after birth. C. trachomatis also can cause a subacute,
afebrile pneumonia with onset at ages 1–3 months. Although
C. trachomatis has been the most frequent identifiable
infectious cause of ophthalmia neonatorum, neonatal
chlamydial infections, including ophthalmia and pneumonia,
have occurred less frequently since institution of widespread
prenatal screening and treatment of pregnant women. Neonates
born to mothers at high risk for chlamydial infection, with
untreated chlamydia, or with no or unconfirmed prenatal care,
are at high risk for infection. However, presumptive treatment
of the neonate is not indicated because the efficacy of such
treatment is unknown. Infants should be monitored to ensure
prompt and age-appropriate treatment if symptoms develop.
Processes should be in place to ensure communication between
physicians and others caring for the mother and the newborn
to ensure thorough monitoring of the newborn after birth.
Ophthalmia Neonatorum Caused by
C. trachomatis
A chlamydial etiology should be considered for all infants aged
≤30 days who experience conjunctivitis, especially if the mother
has a history of chlamydial infection. These infants should receive
evaluation and age-appropriate care and treatment.
Preventing Ophthalmia Neonatorum Caused by
C. trachomatis
Neonatal ocular prophylaxis with erythromycin, the only
agent available in the United States for this purpose, is
ineffective against chlamydial ophthalmia neonatorum (or
pneumonia) (833). As an alternative, prevention efforts should
focus on prenatal screening for C. trachomatis, including
screening pregnant women at risk for C. trachomatis
infection at the first prenatal visit (e.g., women aged
<25 years and those aged ≥25 years who have a new sex
partner, more than one sex partner, a sex partner with
concurrent partners, or a sex partner who has an STI);
treating all pregnant women with C. trachomatis during
pregnancy and performing a test of cure 4 weeks after
treatment to verify chlamydial eradication; these women
should also be retested 3 months after treatment and again
in the third trimester or at time of delivery, and their
partners should also be tested and treated;
retesting pregnant women during the third trimester who
initially tested negative but remained at increased risk for
acquiring infection (e.g., women aged <25 years and those
aged ≥25 years who have a new sex partner, more than one
sex partner, a sex partner with concurrent partners, or a
sex partner who has an STI); and
screening at delivery those pregnant women who were not
screened for C. trachomatis during pregnancy if at risk or
who had no prenatal care; physicians and others caring
for the mother and the newborn should communicate to
ensure follow-up on the results of laboratory tests
performed at delivery, and if positive, prompt and age-
appropriate treatment for the newborn and the mother.
Neonates born to mothers for whom prenatal chlamydia
screening has been confirmed and the results are negative are
not at high risk for infection.
Diagnostic Considerations
Sensitive and specific methods for diagnosing chlamydial
ophthalmia in the neonate include both tissue culture and
nonculture tests (e.g., DFA tests and NAATs). DFA is the
only nonculture FDA-cleared test for detecting chlamydia
from conjunctival swabs. NAATs are not cleared by FDA for
detecting chlamydia from conjunctival swabs, and clinical
laboratories should verify the procedure according to CLIA
regulations. Specimens for culture isolation and nonculture
tests should be obtained from the everted eyelid by using a
Dacron (DuPont)-tipped swab or the swab specified by the
manufacturers test kit; for culture and DFA, specimens must
contain conjunctival cells, not exudate alone. Ocular specimens
from neonates being evaluated for chlamydial conjunctivitis
also should be tested for N. gonorrhoeae (see Ophthalmia
Neonatorum Caused by N. gonorrhoeae).
Treatment
Recommended Regimen for Chlamydial Infection Among
Neonates
Erythromycin base or ethyl succinate 50 mg/kg body weight/day
orally, divided into 4 doses daily for 14 days*
* An association between oral erythromycin and azithromycin and infantile
hypertrophic pyloric stenosis (IHPS) has been reported among infants
aged <6 weeks. Infants treated with either of these antimicrobials should
be followed for IHPS signs and symptoms.
Although data regarding use of azithromycin for treating
neonatal chlamydial infection are limited, available data
demonstrate that a short therapy course might be effective
(834). Topical antibiotic therapy alone is inadequate for
treating ophthalmia neonatorum caused by chlamydia and is
unnecessary when systemic treatment is administered.
Recommendations and Reports
70 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Follow-Up
Because the efficacy of erythromycin treatment for
ophthalmia neonatorum is approximately 80%, a second
course of therapy might be required (834,835). Data regarding
the efficacy of azithromycin for ophthalmia neonatorum are
limited. Therefore, follow-up of infants is recommended to
determine whether the initial treatment was effective. The
possibility of concomitant chlamydial pneumonia should be
considered (see Infant Pneumonia Caused by C. trachomatis).
Management of Mothers and Their Sex Partners
Mothers of infants who have ophthalmia caused by chlamydia
and the sex partners of these women should be evaluated and
presumptively treated for chlamydia (see Chlamydial Infection
Among Adolescents and Adults).
Infant Pneumonia Caused by C. trachomatis
Chlamydial pneumonia among infants typically occurs at age
1–3 months and is a subacute pneumonia. Characteristic signs
of chlamydial pneumonia among infants include a repetitive
staccato cough with tachypnea and hyperinflation and bilateral
diffuse infiltrates on a chest radiograph. In addition, peripheral
eosinophilia (≥400 cells/mm
3
) occurs frequently. Because
clinical presentations differ, all infants aged 1–3 months
suspected of having pneumonia, especially those whose
mothers have a history of, are at risk for (e.g., aged <25 years
and those aged ≥25 years who have a new sex partner, more
than one sex partner, a sex partner with concurrent partners,
or a sex partner who has an STI), or suspected of having a
chlamydial infection should be tested for C. trachomatis and
treated if infected.
Diagnostic Considerations
Specimens for chlamydial testing should be collected from
the nasopharynx. Tissue culture is the definitive standard
diagnostic test for chlamydial pneumonia. Nonculture
tests (e.g., DFA and NAAT) can be used. DFA is the only
nonculture FDA-cleared test for detecting C. trachomatis from
nasopharyngeal specimens; however, DFA of nasopharyngeal
specimens has a lower sensitivity and specificity than culture.
NAATs are not cleared by FDA for detecting chlamydia from
nasopharyngeal specimens, and clinical laboratories should
verify the procedure according to CLIA regulations (553).
Tracheal aspirates and lung biopsy specimens, if collected,
should be tested for C. trachomatis.
Treatment
Because test results for chlamydia often are unavailable at the
time initial treatment decisions are being made, treatment for
C. trachomatis pneumonia frequently is based on clinical and
radiologic findings, age of the infant (i.e., 1–3 months), and
risk for chlamydia in the mother (i.e., aged <25 years, history
of chlamydial infection, multiple sex partners, a sex partner
with a concurrent partner, or a sex partner with a history of
an STI). In the absence of laboratory results in a situation
with a high degree of suspicion of chlamydial infection and
the mother is unlikely to return with the infant for follow-up,
exposed infants can be presumptively treated with the shorter-
course regimen of azithromycin 20 mg/kg body weight/day
orally, 1 dose daily for 3 days.
Recommended Regimen for Chlamydial Pneumonia Among
Infants
Erythromycin base or ethyl succinate 50 mg/kg body weight/day orally
divided into 4 doses daily for 14 days
Alternative Regimen
Azithromycin suspension 20 mg/kg/day orally, 1 dose daily for 3 days
Follow-Up
Because erythromycin effectiveness in treating pneumonia
caused by C. trachomatis is approximately 80%, a second
course of therapy might be required (836). Data regarding
effectiveness of azithromycin in treating chlamydial pneumonia
are limited. Follow-up of infants is recommended to determine
whether the pneumonia has resolved, although certain infants
with chlamydial pneumonia continue to have abnormal
pulmonary function tests later during childhood.
Management of Mothers and Their Sex Partners
Mothers of infants who have chlamydial pneumonia and the
sex partners of these women should be evaluated, tested, and
presumptively treated for chlamydia (see Chlamydial Infection
Among Adolescents and Adults).
Chlamydial Infection Among Infants and
Children
Sexual abuse should be considered a cause of chlamydial
infection among infants and children. However, perinatally
transmitted C. trachomatis infection of the nasopharynx,
urogenital tract, and rectum can persist for 2–3 years (see
Sexual Assault or Abuse of Children).
Diagnostic Considerations
NAATs can be used to test vaginal and urine specimens
from girls and urine in boys (see Sexual Assault or Abuse
of Children). Data are lacking regarding use of NAATs for
specimens from extragenital sites (rectum and pharynx) among
boys and girls (553); other nonculture tests (e.g., DFA) are
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 71
US Department of Health and Human Services/Centers for Disease Control and Prevention
not recommended because of specificity concerns. Although
data regarding NAATs for specimens from extragenital sites for
children are more limited and performance is test dependent
(553), no evidence supports that NAAT performance
for detecting C. trachomatis for extragenital sites among
children would differ from that among adults. Because of
the implications of a diagnosis of C. trachomatis infection in
a child, only CLIA-validated, FDA-cleared NAAT should be
used for extragenital site specimens (837).
Recommended Regimens for Chlamydial Infection Among
Infants and Children
For infants and children weighing <45 kg: Erythromycin base or ethyl
succinate 50 mg/kg body weight/day orally divided into 4 doses daily
for 14 days
Data are limited regarding the effectiveness and optimal dose of
azithromycin for treating chlamydial infection among infants and
children weighing <45 kg.
For children weighing ≥45 kg but aged <8 years: Azithromycin 1 g
orally in a single dose
For children aged ≥8 years: Azithromycin 1 g orally in a single dose
or
Doxycycline 100 mg orally 2 times/day for 7 days
Other Management Considerations
See Sexual Assault or Abuse of Children.
Follow-Up
A test of cure to detect therapeutic failure ensures treatment
effectiveness and should be obtained at a follow-up visit
approximately 4 weeks after treatment is completed.
Gonococcal Infections
Gonococcal Infection Among
Adolescents and Adults
In the United States, an estimated 1,568,000 new
N. gonorrhoeae infections occur each year (141,838), and
gonorrhea is the second most commonly reported bacterial
communicable disease. Urethral infections caused by
N. gonorrhoeae can produce symptoms among men that cause
them to seek curative treatment soon enough to prevent
sequelae, but often not soon enough to prevent transmission
to others. Among women, gonococcal infections are commonly
asymptomatic or might not produce recognizable symptoms
until complications (e.g., PID) have occurred. PID can result in
tubal scarring that can lead to infertility or ectopic pregnancy.
Annual screening for N. gonorrhoeae infection is recommended
for all sexually active women aged <25 years and for older
women at increased risk for infection (e.g., those aged ≥25 years
who have a new sex partner, more than one sex partner, a sex
partner with concurrent partners, or a sex partner who has
an STI) (149). Additional risk factors for gonorrhea include
inconsistent condom use among persons who are not in
mutually monogamous relationships, previous or coexisting
STIs, and exchanging sex for money or drugs. Clinicians
should consider the communities they serve and consult local
public health authorities for guidance regarding identifying
groups at increased risk. Gonococcal infection, in particular, is
concentrated in specific geographic locations and communities.
MSM at high risk for gonococcal infection (e.g., those with
multiple anonymous partners or substance abuse) or those at
risk for HIV acquisition should be screened at all anatomic
sites of exposure every 3–6 months (see Men Who Have Sex
with Men). At least annual screening is recommended for all
MSM. Screening for gonorrhea among heterosexual men and
women aged >25 years who are at low risk for infection is
not recommended (149). A recent travel history with sexual
contacts outside the United States should be part of any
gonorrhea evaluation.
Diagnostic Considerations
Specific microbiologic diagnosis of N. gonorrhoeae infection
should be performed for all persons at risk for or suspected of
having gonorrhea; a specific diagnosis can potentially reduce
complications, reinfections, and transmission. Culture, NAAT,
and POC NAAT, such as GeneXpert (Cepheid), are available
for detecting genitourinary infection with N. gonorrhoeae
(149); culture requires endocervical (women) or urethral
(men) swab specimens. Culture is also available for detecting
rectal, oropharyngeal, and conjunctival gonococcal infection.
NAATs and POC NAATs) allow for the widest variety of
FDA-cleared specimen types, including endocervical and
vaginal swabs and urine for women, urethral swabs and urine
for men, and rectal swabs and pharyngeal swabs for men and
women (www.accessdata.fda.gov/cdrh_docs/reviews/K121710.
pdf). However, product inserts for each NAAT manufacturer
should be consulted carefully because collection methods
and specimen types vary. Certain NAATs that have been
demonstrated to detect commensal Neisseria species might
have comparable low specificity when testing oropharyngeal
specimens for N. gonorrhoeae (553). NAAT sensitivity for
detecting N. gonorrhoeae from urogenital and nongenital
anatomic sites is superior to culture but varies by NAAT type
(553,800803). For urogenital infections, optimal specimen
types for gonorrhea screening using NAATs include first-void
urine for men and vaginal swab specimens for women (553).
Patient-collected samples can be used in place of provider-
collected samples in clinical settings when testing by NAAT
Recommendations and Reports
72 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
for urine (men and women), vaginal swabs, rectal swabs, and
oropharyngeal swabs after patient instructions have been
provided (209,806,839842). Patient-collected specimens
are reasonable alternatives to provider-collected swabs for
gonorrhea screening by NAAT.
In cases of suspected or documented treatment failure,
clinicians should perform both culture and antimicrobial
susceptibility testing because NAATs cannot provide
antimicrobial susceptibility results. Because N. gonorrhoeae
has demanding nutritional and environmental growth
requirements, optimal recovery rates are achieved when
specimens are inoculated directly and when the growth
medium is promptly incubated in an increased carbon dioxide
(CO
2
) environment (553). Nonnutritive swab transport
systems are available that might maintain gonococcal viability
for <48 hours in ambient temperatures (843845).
Because of its high specificity (>99%) and sensitivity
(>95%), a Gram stain of urethral discharge or secretions that
demonstrate polymorphonuclear leukocytes with intracellular
gram-negative diplococci can be considered diagnostic for
infection with N. gonorrhoeae among symptomatic men.
However, because of lower sensitivity, a negative Gram stain
should not be considered sufficient for ruling out infection
among asymptomatic men. Infection detection by using
Gram stain of endocervical, pharyngeal, and rectal specimens
also is insensitive and is not recommended. MB or GV stain
of urethral secretions is an alternative POC diagnostic test
with performance characteristics similar to Gram stain.
Gonococcal infection is diagnosed among symptomatic men by
documenting the presence of a WBC-containing intracellular
purple diplococci in MB or GV smears.
Antimicrobial-Resistant N. gonorrhoeae
Gonorrhea treatment is complicated by the ability of
N. gonorrhoeae to develop resistance to antimicrobials
(846848). In 1986, the Gonococcal Isolate Surveillance
Project (GISP), a national sentinel surveillance system, was
established to monitor trends in antimicrobial susceptibilities
of urethral N. gonorrhoeae strains in the United States
(849). The epidemiology of antimicrobial resistance guides
decisions about gonococcal treatment recommendations
and has evolved because of shifts in antimicrobial resistance
patterns. During 2007, emergence of fluoroquinolone-
resistant N. gonorrhoeae in the United States prompted CDC
to cease recommending fluoroquinolones for gonorrhea
treatment, leaving cephalosporins as the only remaining
class of antimicrobials available for gonorrhea treatment in
the United States (850). Reflecting concern about emerging
gonococcal resistance, CDC’s 2010 STD treatment guidelines
recommended dual therapy for gonorrhea with a cephalosporin
plus either azithromycin or doxycycline, even if NAAT for
C. trachomatis was negative at the time of treatment (851).
However, during 2006–2011, the minimum concentrations of
cefixime needed to inhibit in vitro growth of the N. gonorrhoeae
strains circulating in the United States and other countries
increased, demonstrating that cefixime effectiveness might be
waning (851). In addition, treatment failures with cefixime or
other oral cephalosporins were reported in Asia (852855),
Europe (856860), South Africa (861), and Canada (862,863).
During that time, case reports of ceftriaxone treatment failures
for pharyngeal infections reported in Australia (864,865),
Japan (866), and Europe were concerning (856,867).
Consequently, CDC no longer recommends cefixime as a
first-line regimen for gonorrhea treatment in the United
States (868). Since 2013, the proportion of GISP isolates
that demonstrate reduced susceptibility (minimal inhibitory
concentration [MIC] ≥2.0 µg/mL) to azithromycin has
increased almost tenfold, to 5.1% in 2019 (141). Unlike the
appearance of ciprofloxacin resistance in the early 2000s, and
cefixime reduced-susceptibility isolates during 2010–2011,
emergence of azithromycin resistance is not concentrated
among certain populations (e.g., MSM in the western United
States). Azithromycin has unique pharmacokinetic properties
that might predispose to resistance due to its prolonged half-life
(869,870). With the exception of a small cluster of gonorrhea
strains with azithromycin resistance and reduced susceptibility
to cefixime and ceftriaxone among seven patients during 2016,
all gonorrhea strains identified by GISP are susceptible to
either or both azithromycin and ceftriaxone or cefixime. In
addition, since 2013, antimicrobial stewardship has become
an urgent public health concern in the United States as
described in Antimicrobial Resistant Threats in the United States
(871). Emergence of azithromycin resistance is not isolated to
N. gonorrhoeae; it has also been demonstrated in M. genitalium
and such enteric pathogens as Shigella and Campylobacter (see
Mycoplasma genitalium; Proctitis, Proctocolitis, and Enteritis).
Finally, concern exists regarding azithromycin treatment
efficacy for chlamydia (see Chlamydial Infections).
Dual therapy for gonococcal infection with ceftriaxone
and azithromycin recommended in previous guidance
might have mitigated emergence of reduced susceptibility to
ceftriaxone in N. gonorrhoeae; however, concerns regarding
potential harm to the microbiome and the effect on other
pathogens diminishes the benefits of maintaining dual therapy.
Consequently, only ceftriaxone is recommended for treating
gonorrhea in the United States (872). Clinicians remaining
vigilant for treatment failures is paramount, and CDC plans
to continue to monitor for changing ceftriaxone MICs
until additional antimicrobials or a vaccine is available. In
cases in which chlamydial infection has not been excluded,
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 73
US Department of Health and Human Services/Centers for Disease Control and Prevention
patients should also receive antichlamydial therapy. CDC
and state health departments participate in CDC-supported
gonorrhea surveillance activities (https://www.cdc.gov/std/
gisp) and can provide the most current information regarding
gonococcal susceptibility.
Criteria for resistance to cefixime and ceftriaxone have not
been defined by the Clinical and Laboratory Standards Institute
(CLSI). However, isolates with cefixime or ceftriaxone MICs
≥0.5 µg/mL are considered to have decreased susceptibility
(873). In the United States, the proportion of isolates in
GISP demonstrating decreased susceptibility to ceftriaxone
or cefixime has remained low; during 2019, <0.1% of isolates
with decreased susceptibility (MIC ≥0.5 µg/mL) to ceftriaxone
or cefixime were identified (141). Because increasing MICs
might predict resistance emergence, GISP established lower
cephalosporin MIC threshold values that are lower than the
susceptibility breakpoints set by CLSI to provide greater
sensitivity in detecting decreasing gonococcal susceptibility
for surveillance purposes. The percentage of isolates with
cefixime MICs ≥0.25 µg/mL increased from 0.1% during
2006 to 1.4% during 2011 (851,874) and declined to
0.3% during 2019 (141). The percentage of isolates with
ceftriaxone MICs ≥0.125 µg/mL increased from <0.1% in
2006 to 0.4% in 2011 and decreased to 0.1% in 2019 (141).
Isolates with high-level cefixime and ceftriaxone MICs (MICs
= 1.5–8.0 µg/mL and MICs = 1.5–4.0 µg/mL, respectively)
have been identified in Japan (866), France (867,875), Spain
(876,877), the United Kingdom, and Australia (878,879).
Decreased susceptibility of N. gonorrhoeae to cephalosporins
and other antimicrobials is expected to continue; state and
local surveillance for antimicrobial resistance is crucial for
guiding local therapy recommendations (846,847). Although
approximately 3% of all U.S. men who have gonococcal
infections are sampled through GISP, surveillance by clinicians
also is crucial. Clinicians who diagnose N. gonorrhoeae infection
in a person with suspected cephalosporin treatment failure
should perform culture and AST of relevant clinical specimens,
consult an infectious disease specialist or an STD clinical
expert (https://www.stdccn.org/render/Public) for guidance
in clinical management, and report the case to CDC through
state and local public health authorities within 24 hours.
Isolates should be saved and sent to CDC through local and
state public health laboratory mechanisms. Health departments
should prioritize notification and culture evaluation for sexual
partners of persons with N. gonorrhoeae infection thought
to be associated with cephalosporin treatment failure or
persons whose isolates demonstrate decreased susceptibility
to cephalosporin. Agar dilution is the reference standard
and preferred method of antimicrobial susceptibility testing
with N. gonorrhoeae. Antibiotic gradient strips, such as Etest
(bioMérieux), can be used and are considered an acceptable
alternative for quantitative antimicrobial susceptibility testing with
N. gonorrhoeae when manufacturer instructions are followed. Disc
diffusion only provides qualitative susceptibility results.
Uncomplicated Gonococcal Infection of the
Cervix, Urethra, or Rectum
Recommended Regimen for Uncomplicated Gonococcal
Infection of the Cervix, Urethra, or Rectum Among Adults and
Adolescents
Ceftriaxone 500 mg* IM in a single dose for persons weighing <150 kg
If chlamydial infection has not been excluded, treat for chlamydia with
doxycycline 100 mg orally 2 times/day for 7 days.
* For persons weighing ≥150 kg, 1 g ceftriaxone should be administered.
Although clinical data confirm that a single injection
of ceftriaxone 250 mg is >99% (95% confidence interval
[CI]:97.6%–99.7%) effective in curing anogenital gonorrhea
of circulating isolates (MIC = 0.03 µg/mL), a higher dose is
likely necessary for isolates with elevated MICs (880,881).
Effective treatment of uncomplicated urogenital gonorrhea with
ceftriaxone requires concentrations higher than the strain MIC
for approximately 24 hours; although individual variability
exists in the pharmacokinetics of ceftriaxone, a 500-mg dose
of ceftriaxone is expected to achieve in approximately 50 hours
MIC >0.03 µg/mL (880,881). The pharmacokinetics of
ceftriaxone might be different in the pharynx with longer times
higher than the strain MIC likely needed to prevent selection
of mutant strains in the pharynx (882).
Single-dose injectable cephalosporin regimens, other than
ceftriaxone, that are safe and have been effective against
uncomplicated urogenital and anorectal gonococcal infections
in the past include ceftizoxime (500 mg IM), cefoxitin (2 g IM
with probenecid 1 g orally), and cefotaxime (500 mg IM).
None of these injectable cephalosporins offer any advantage
over ceftriaxone 250 mg for urogenital infection, and efficacy
for pharyngeal infection is less certain (883,884). Because the
ceftriaxone dose has been increased and the pharmacokinetics
of other cephalosporins have not been evaluated, these dosing
regimens might be at a disadvantage over ceftriaxone 500 mg.
Alternative Regimens if Ceftriaxone Is Not Available
Gentamicin 240 mg IM in a single dose
plus
Azithromycin 2 g orally in a single dose
or
Cefixime* 800 mg orally in a single dose
* If chlamydial infection has not been excluded, providers should treat for
chlamydia with doxycycline 100 mg orally 2 times/day for 7 days.
Recommendations and Reports
74 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
In one clinical trial, dual treatment with single doses of
IM gentamicin 240 mg plus oral azithromycin 2 g cured
100% of cases (lower one-sided 95% CI bound:98.5%) and
can be considered an alternative to ceftriaxone for persons
with cephalosporin allergy (885). This trial was not powered
enough to provide reliable estimates of the efficacy of these
regimens for treatment of rectal or pharyngeal infection;
however, this regimen cured the few extragenital infections
among study participants. Notably, gastrointestinal adverse
events, primarily vomiting <1 hour after dosing, occurred
among 3%–4% of persons treated with gentamicin plus
azithromycin, necessitating retreatment with ceftriaxone
and azithromycin. A similar trial that studied gentamicin
240 mg plus azithromycin 1 g determined lower cure rates at
extragenital sites; 80% (95% CI:72%–88%) of pharyngeal
and 90% (95% CI:84%–95%) of rectal infections were cured
with this regimen (886). Gemifloxacin plus azithromycin has
been studied and is no longer recommended as an alternative
regimen because of limited availability, cost, and antimicrobial
stewardship concerns (885).
An 800-mg oral dose of cefixime should be considered only
as an alternative cephalosporin regimen because it does not
provide as high, nor as sustained, bactericidal blood levels as a
500-mg IM dose of ceftriaxone. Furthermore, it demonstrates
limited efficacy for treatment of pharyngeal gonorrhea (92.3%
cure; 95% CI:74.9%–99.1%); in older clinical studies,
cefixime cured 97.5% of uncomplicated urogenital and
anorectal gonococcal infections (95% CI:95.4%–99.8%)
(883,884). The increase in the prevalence of isolates obtained
through GISP with elevated cefixime MICs might indicate
early stages of development of clinically significant gonococcal
resistance to cephalosporins. Changes in cefixime MICs can
result in decreasing effectiveness of cefixime for treating
urogenital gonorrhea. Furthermore, as cefixime becomes
less effective, continued used of cefixime might hasten the
development of resistance to ceftriaxone, a safe, well-tolerated,
injectable cephalosporin and the last antimicrobial known to
be highly effective in a single dose for treatment of gonorrhea
at all anatomic infection sites. Other oral cephalosporins (e.g.,
cefpodoxime and cefuroxime) are not recommended because of
inferior efficacy and less favorable pharmacodynamics (883).
Monotherapy with azithromycin 2 g orally as a single
dose has been demonstrated to be 99.2% effective against
uncomplicated urogenital gonorrhea (95% CI:97.3%–99.9%)
(883). However, monotherapy is not recommended because
of concerns about the ease with which N. gonorrhoeae can
develop resistance to macrolides, the high proportion of isolates
with azithromycin decreased susceptibility, and documented
azithromycin treatment failures (859). Strains of N. gonorrhoeae
circulating in the United States are not adequately susceptible
to penicillin, tetracycline, and older macrolides (e.g.,
erythromycin), and thus use of these antimicrobials cannot
be recommended.
Spectinomycin is effective (98.2% in curing uncomplicated
urogenital and anorectal gonococcal infections) but has poor
efficacy for pharyngeal infections (883,887). It is unavailable
in the United States, and the gentamicin alternative regimen
has replaced the need for spectinomycin, if a cephalosporin
allergy exists, in the United States.
Uncomplicated Gonococcal Infection of
the Pharynx
The majority of gonococcal infections of the pharynx are
asymptomatic and can be relatively common among certain
populations (800,801,888890). Although these infections
rarely cause complications, they have been reported to be a
major source of community transmission and might be a driver
of antimicrobial resistance (891,892). Gonococcal infections
of the pharynx are more difficult to eradicate than infections
at urogenital and anorectal sites (862). Few antimicrobial
regimens reliably cure >90% of gonococcal pharyngeal
infections (883,884). Providers should ask their patients with
urogenital or rectal gonorrhea about oral sexual exposure; if
reported, pharyngeal testing should be performed.
Recommended Regimen for Uncomplicated Gonococcal
Infection of the Pharynx Among Adolescents and Adults
Ceftriaxone 500 mg* IM in a single dose for persons weighing <150 kg
* For persons weighing ≥150 kg, 1 g ceftriaxone should be administered.
If chlamydial infection is identified when pharyngeal
gonorrhea testing is performed, treat for chlamydia with
doxycycline 100 mg orally 2 times/day for 7 days. No reliable
alternative treatments are available for pharyngeal gonorrhea.
For persons with an anaphylactic or other severe reaction
(e.g., Stevens Johnson syndrome) to ceftriaxone, consult
an infectious disease specialist for an alternative treatment
recommendation.
Other Management Considerations
To maximize adherence with recommended therapies
and reduce complications and transmission, medication
for gonococcal infection should be provided on-site and
directly observed. If medications are unavailable when
treatment is indicated, linkage to an STI treatment facility
should be provided for same-day treatment. To minimize
disease transmission, persons treated for gonorrhea should
be instructed to abstain from sexual activity for 7 days after
treatment and until all sex partners are treated (7 days after
receiving treatment and resolution of symptoms, if present).
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 75
US Department of Health and Human Services/Centers for Disease Control and Prevention
All persons who receive a diagnosis of gonorrhea should be
tested for other STIs, including chlamydia, syphilis, and HIV.
Those persons whose HIV test results are negative should be
offered HIV PrEP.
Follow-Up
A test of cure (i.e., repeat testing after completion of
therapy) is unnecessary for persons who receive a diagnosis of
uncomplicated urogenital or rectal gonorrhea who are treated
with any of the recommended or alternative regimens. Any
person with pharyngeal gonorrhea should return 7–14 days
after initial treatment for a test of cure by using either culture or
NAAT; however, testing at 7 days might result in an increased
likelihood of false-positive tests. If the NAAT is positive, effort
should be made to perform a confirmatory culture before
retreatment, especially if a culture was not already collected. All
positive cultures for test of cure should undergo antimicrobial
susceptibility testing. Symptoms that persist after treatment
should be evaluated by culture for N. gonorrhoeae (with or
without simultaneous NAAT) and antimicrobial susceptibility.
Persistent urethritis, cervicitis, or proctitis also might be caused
by other organisms (see Urethritis; Cervicitis; Proctitis).
A high prevalence of N. gonorrhoeae infection has been
observed among men and women previously treated for
gonorrhea (137,753,754,893). The majority of these infections
result from reinfection caused by failure of sex partners to
receive treatment or the initiation of sexual activity with a
new infected partner, indicating a need for improved patient
education and treatment of sex partners. Men or women who
have been treated for gonorrhea should be retested 3 months
after treatment regardless of whether they believe their sex
partners were treated; scheduling the follow-up visit at the
time of treatment is encouraged. If retesting at 3 months is not
possible, clinicians should retest whenever persons next seek
medical care <12 months after initial treatment.
Management of Sex Partners
Recent sex partners (i.e., persons having sexual contact with
the infected patient <60 days preceding onset of symptoms or
gonorrhea diagnosis) should be referred for evaluation, testing,
and presumptive treatment. If the patient’s last potential sexual
exposure was >60 days before onset of symptoms or diagnosis,
the most recent sex partner should be treated.
If health department partner-management strategies (e.g.,
disease intervention specialists) are impractical or unavailable
for persons with gonorrhea and partners’ access to prompt
clinical evaluation and treatment is limited, EPT can be
delivered to the partner by the patient or a collaborating
pharmacy as permitted by law (see Partner Services). Treatment
of the sexual partner with cefixime 800 mg as a single dose
is recommended, provided that concurrent chlamydial
infection has been excluded. If a chlamydia test result has not
been documented, the partner may be treated with a single
dose of oral cefixime 800 mg plus oral doxycycline 100 mg
2 times/day for 7 days. If adherence with multiday dosing is
a considerable concern, azithromycin 1 g can be considered
but has lower treatment efficacy among persons with rectal
chlamydia (see Chlamydial Infections). Provision of medication
by EPT should be accompanied by written materials (125,127)
for educating partners about gonorrhea, their exposure to
gonorrhea, and the importance of therapy. These materials
should also educate partners about seeking clinical evaluation
for adverse reactions or complications and general follow-up
when able. Educational materials for female partners should
include information about the importance of seeking medical
evaluation for PID, especially if symptomatic; undertreatment
of PID among female partners and missed opportunities for
diagnosing other STIs among women are of concern. MSM
with gonorrhea have a high risk for coexisting infections
(especially undiagnosed HIV) among their partners, and they
might have partners without HIV who could benefit from
PrEP. Data are also limited regarding the effectiveness of EPT
in reducing persistent or recurrent gonorrhea among MSM
(133,135); thus, shared clinical decision-making regarding
EPT for MSM is recommended (see Partner Services). To avoid
reinfection, sex partners should be instructed to abstain from
condomless sexual intercourse for 7 days after they and their
sex partners have completed treatment and after resolution of
symptoms, if present.
Suspected Cephalosporin Treatment Failure
Cephalosporin treatment failure is the persistence of
N. gonorrhoeae infection despite recommended cephalosporin
treatment; such failure is indicative of infection with
cephalosporin-resistant gonorrhea among persons whose
partners were treated and whose risk for reinfection is
low. Suspected treatment failure has been reported among
persons receiving oral and injectable cephalosporins (852
855,857,859,861,863,864,867,875,894). Treatment failure
should be considered for persons whose symptoms do not
resolve within 3–5 days after recommended treatment and
report no sexual contact during the posttreatment follow-up
period and persons with a positive test of cure (i.e., positive
culture >72 hours or positive NAAT >7 days after receiving
recommended treatment) when no sexual contact is reported
during the posttreatment follow-up period (874). Treatment
failure should also be considered for persons who have a
positive culture on test of cure, if obtained, if evidence exists
of decreased susceptibility to cephalosporins on antimicrobial
Recommendations and Reports
76 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
susceptibility testing, regardless of whether sexual contact is
reported during the posttreatment follow-up period.
The majority of suspected treatment failures in the United
States are likely to be reinfections rather than actual treatment
failures (137,753,754,894). However, in cases in which
reinfection is unlikely and treatment failure is suspected,
before retreatment, relevant clinical specimens should be
obtained for culture (preferably with simultaneous NAAT)
and antimicrobial susceptibility testing if N. gonorrhoeae is
isolated. Phenotypic antimicrobial susceptibility testing should
be performed by using Etest or agar dilution. All isolates
of suspected treatment failures should be sent to CDC for
antimicrobial susceptibility testing by agar dilution; local
laboratories should store isolates for possible further testing if
needed. Testing or storage of specimens or isolates should be
facilitated by the state or local health department according to
local public health protocol. Instructions for shipping isolates
to CDC are available at https://www.cdc.gov/std/gonorrhea/
arg/specimen_shipping_instructions1-29-08.pdf.
For persons with suspected cephalosporin treatment failure,
the treating clinician should consult an infectious disease
specialist, the National Network of STD Clinical Prevention
Training Center clinical consultation line (https://www.stdccn.
org/render/Public), the local or state health department STI
program, or CDC (telephone: 800-232-4636) for advice about
obtaining cultures, antimicrobial susceptibility testing and
treatment. Suspected treatment failure should be reported to
CDC through the local or state health department <24 hours
after diagnosis.
Patients with suspected treatment failures should first be
retreated routinely with the initial regimen used (ceftriaxone
500 mg IM), with the addition of doxycycline if chlamydia
infection exists, because reinfections are more likely than
actual treatment failures. However, in situations with a higher
likelihood of treatment failure than reinfection, relevant
clinical specimens should be obtained for culture (preferably
with simultaneous NAAT) and antimicrobial susceptibility
testing before retreatment. Dual treatment with single doses
of IM gentamicin 240 mg plus oral azithromycin 2 g can be
considered, particularly when isolates are identified as having
elevated cephalosporin MICs (885,886,895). Persons with
suspected treatment failure after treatment with the alternative
regimen (cefixime or gentamicin) should be treated with
ceftriaxone 500 mg as a single IM dose or as a single dose with
or without an antichlamydial agent on the basis of chlamydia
infection status. A test of cure at relevant clinical sites should
be obtained 7–14 days after retreatment; culture is the
recommended test, preferably with simultaneous NAAT, and
antimicrobial susceptibility testing of N. gonorrhoeae if isolated.
Clinicians should ensure that the patients’ sex partners from
the preceding 60 days are evaluated promptly with culture
and presumptively treated by using the same regimen used
for the patients.
Special Considerations
Drug Allergy, Intolerance, and Adverse Reactions
The risk for penicillin cross-reactivity is highest with
first-generation cephalosporins but is rare (<1%) with
third-generation cephalosporins (e.g., ceftriaxone and
cefixime) (631,680,896). Clinicians should first thoroughly
assess a patient’s allergy history, including type of reaction,
associated medications, and previous prescription records.
If IgE-mediated penicillin allergy is strongly suspected, dual
treatment with single doses of IM gentamicin 240 mg plus oral
azithromycin 2 g can be administered (885,886). If a patient
is asymptomatic and the treating facility is able to perform
gyrase A (gyrA) testing to identify ciprofloxacin susceptibility
(wild type), oral ciprofloxacin 500 mg in a single dose can be
administered. Providers treating persons with IgE-mediated
cephalosporin or penicillin allergy should refer to the section
of these guidelines regarding evaluation (see Management of
Persons Who Have a History of Penicillin Allergy).
Pregnancy
Pregnant women infected with N. gonorrhoeae should be
treated with ceftriaxone 500 mg in a single IM dose plus
treatment for chlamydia if infection has not been excluded.
When cephalosporin allergy or other considerations preclude
treatment with this regimen, consultation with an infectious
disease specialist or an STD clinical expert is recommended.
Gentamicin use is cautioned during pregnancy because of risk
for neonatal birth defects, nephrotoxicity, or ototoxicity (897)
(https://www.stdccn.org/render/Public).
HIV Infection
Persons who have gonorrhea and HIV infection should
receive the same treatment regimen as those who do not
have HIV.
Gonococcal Conjunctivitis
In the only published study of the treatment regarding
gonococcal conjunctivitis among adults, all 12 study
participants responded to a single 1-g IM injection of
ceftriaxone (898). Because gonococcal conjunctivitis is
uncommon and data regarding treatment of gonococcal
conjunctivitis among adults are limited, consultation with an
infectious disease specialist should be considered.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 77
US Department of Health and Human Services/Centers for Disease Control and Prevention
Recommended Regimen for Gonococcal Conjunctivitis Among
Adolescents and Adults
Ceftriaxone 1 g IM in a single dose
Providers should consider one-time lavage of the infected eye with
saline solution.
Management of Sex Partners
Patients should be instructed to refer their sex partners
for evaluation and treatment (see Gonococcal Infections,
Management of Sex Partners).
Disseminated Gonococcal Infection
Infrequently, N. gonorrhoeae can cause disseminated
infection. Disseminated gonococcal infection (DGI) frequently
results in petechial or pustular acral skin lesions, asymmetric
polyarthralgia, tenosynovitis, or oligoarticular septic arthritis
(899901). Rarely, DGI is complicated by perihepatitis
associated with gonococcal PID, endocarditis, or meningitis.
Certain strains of N. gonorrhoeae that cause DGI can cause
minimal genital inflammation, and urogenital or anorectal
infections are often asymptomatic among DGI patients.
If DGI is suspected, NAATs or culture specimens from all
exposed urogenital and extragenital sites should be collected
and processed, in addition to disseminated sites of infection
(e.g., skin, synovial fluid, blood, or CSF). All N. gonorrhoeae
isolates should be tested for antimicrobial susceptibility.
Risk factors for dissemination have included female sex,
menstruation, pregnancy, and terminal complement deficiency
(899); however, reports are increasing among men (900,901).
Persons receiving eculizumab, a monoclonal antibody that
inhibits terminal complement activation, also might be at
higher risk for DGI (902).
Hospitalization and consultation with an infectious disease
specialist are recommended for initial therapy, especially
for persons who might not comply with treatment, have an
uncertain diagnosis, or have purulent synovial effusions or
other complications. Examination for clinical evidence of
endocarditis and meningitis should be performed.
Treatment of Arthritis and Arthritis-Dermatitis
Syndrome
Recommended Regimen for Gonococcal-Related Arthritis and
Arthritis-Dermatitis Syndrome
Ceftriaxone 1 g IM or IV every 24 hours
If chlamydial infection has not been excluded, providers should treat for
chlamydia with doxycycline 100 mg orally 2 times/day for 7 days.
Alternative Regimens
Cefotaxime 1 g IV every 8 hours
or
Ceftizoxime 1 g every 8 hours
If chlamydial infection has not been excluded, providers should treat for
chlamydia with doxycycline 100 mg orally 2 times/day for 7 days.
When treating for the arthritis-dermatitis syndrome, the
provider can switch to an oral agent guided by antimicrobial
susceptibility testing 24–48 hours after substantial clinical
improvement, for a total treatment course of >7 days.
Treatment of Gonococcal Meningitis and Endocarditis
Recommended Regimen for Gonococcal Meningitis and
Endocarditis
Ceftriaxone 1–2 g IV every 24 hours
If chlamydial infection has not been excluded, providers should treat for
chlamydia with doxycycline 100 mg orally 2 times/day for 7 days.
No recent studies have been published regarding treatment
of DGI involving the CNS or cardiovascular system. The
duration of treatment for DGI in these situations has not been
systematically studied and should be determined in consultation
with an infectious disease specialist. Treatment for DGI should
be guided by the results of antimicrobial susceptibility testing.
Length of treatment should be determined based on clinical
presentation. Therapy for meningitis should be continued with
recommended parenteral therapy for 10–14 days. Parenteral
antimicrobial therapy for endocarditis should be administered
for >4 weeks. Treatment of gonococcal perihepatitis should be
managed in accordance with the recommendations for PID
in these guidelines.
Management of Sex Partners
Gonococcal infection frequently is asymptomatic among sex
partners of persons who have DGI. Providers should instruct
patients to refer partners with whom they have had sexual
contact during the previous 60 days for evaluation, testing,
and presumptive treatment (see Gonococcal Infections,
Management of Sex Partners).
Gonococcal Infection Among Neonates
Prenatal screening and treatment of pregnant women for
gonorrhea is the best method for preventing N. gonorrhoeae
infection among neonates. Gonococcal infection among
neonates results from perinatal exposure to the mothers infected
cervix. It is usually an acute illness that manifests 2–5 days after
birth. Prevalence of infection among neonates depends on the
prevalence of infection among pregnant women and whether
pregnant women are screened and treated for gonorrhea during
Recommendations and Reports
78 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
pregnancy. The most severe manifestations of N. gonorrhoeae
infection among neonates are ophthalmia neonatorum and
sepsis, which can include arthritis and meningitis. Less severe
manifestations include rhinitis, vaginitis, urethritis, and scalp
infection at sites of previous fetal monitoring.
Preventing Ophthalmia Neonatorum Caused by
N. gonorrhoeae
Ocular prophylaxis and preventive gonorrhea screening
and treatment of infected pregnant women are especially
important because ophthalmia neonatorum can result in
perforation of the globe of the eye and blindness (903).
Ocular prophylaxis for gonococcal ophthalmia neonatorum
has a long history of preventing sight-threatening gonococcal
ocular infections. Cases in the United States are uncommon,
which is likely attributable to gonorrhea screening programs
for women, including pregnant women, that have contributed
substantially to reduction in ophthalmia neonatorum (904).
Neonatal ocular prophylaxis with erythromycin, the only agent
available in the United States, is required by law in most states
and is recommended because of safety, low cost, and ease of
administration. It can contribute to preventing gonococcal
blindness because not all pregnant women are screened for
gonorrhea. The USPSTF recommends ocular prophylaxis
with erythromycin ointment for all newborns <24 hours
after birth (903). In addition to continuing routine ocular
prophylaxis, prevention should focus on prenatal screening
for N. gonorrhoeae, including
screening pregnant women at risk (e.g., women aged
<25 years and those aged ≥25 years who have a new sex
partner, more than one sex partner, a sex partner with
concurrent partners, a sex partner who has an STI, or live
in a community with high rates of gonorrhea) for
N. gonorrhoeae infection at the first prenatal visit;
treating all pregnant women with N. gonorrhoeae infection
during pregnancy and retesting in 3 months, in the third
trimester or at time of delivery (sex partners should be
tested and treated);
retesting pregnant women in the third trimester who
initially tested negative but remained at increased risk for
acquiring infection (e.g., women aged <25 years and those
aged ≥25 years who have a new sex partner, more than one
sex partner, a sex partner with concurrent partners, a sex
partner who has an STI, or live in a community with high
rates of gonorrhea); and
screening for gonorrhea at delivery for women not tested
during pregnancy and at risk for infection (e.g., women
aged <25 years and those aged ≥25 years who have a new
sex partner, more than one sex partner, a sex partner with
concurrent partners, a sex partner who has an STI, or live
in a community with high rates of gonorrhea) or received
no prenatal care; providers caring for the mother and the
newborn should communicate to ensure follow-up on the
results of laboratory tests performed at delivery, and if
positive, prompt appropriate treatment of the newborn
and mother.
Erythromycin is the only ophthalmic ointment recommended
for use among neonates. Silver nitrate and tetracycline
ophthalmic ointments are no longer manufactured in the
United States, bacitracin is ineffective, and povidone iodine
has not been studied adequately (905,906). Gentamicin
ophthalmic ointment has been associated with severe ocular
reactions (907,908). If erythromycin ointment is unavailable,
infants at risk for exposure to N. gonorrhoeae, especially those
born to a mother at risk for gonococcal infection or with no
prenatal care, can be administered ceftriaxone 25–50 mg/kg
body weight IV or IM, not to exceed 250 mg in a single dose.
Recommended Regimen to Prevent Ophthalmia Neonatorum
Caused by N. gonorrhoeae
Erythromycin 0.5% ophthalmic ointment in each eye in a single
application at birth
Erythromycin ophthalmic ointment should be instilled
into both eyes of neonates as soon as possible after delivery,
regardless of whether they are delivered vaginally or by
cesarean delivery. Ideally, ointment should be applied by using
single-use tubes or ampules rather than multiple-use tubes. If
prophylaxis is delayed (i.e., not administered in the delivery
room), a monitoring system should be established to ensure
that all newborns receive prophylaxis <24 hours after delivery.
Diagnostic Considerations
Newborns at increased risk for gonococcal ophthalmia
include those who did not receive ophthalmic prophylaxis
and whose mothers had no prenatal care, have a history
of STIs during pregnancy, or have a history of substance
misuse. Gonococcal ophthalmia is strongly suspected when
intracellular gram-negative diplococci are identified on Gram
stain of conjunctival exudate, justifying presumptive treatment
for gonorrhea after appropriate cultures and antimicrobial
susceptibility testing for N. gonorrhoeae are performed.
Presumptive treatment for N. gonorrhoeae might be indicated
for newborns at increased risk for gonococcal ophthalmia who
have increased WBCs (no GNID) in a Gram-stained smear
of conjunctival exudate. Nongonococcal causes of neonatal
ophthalmia include Moraxella catarrhalis and other Neisseria
species, which are organisms that are indistinguishable from
N. gonorrhoeae on Gram-stained smear but can be differentiated
in the microbiology laboratory.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 79
US Department of Health and Human Services/Centers for Disease Control and Prevention
Treatment of Gonococcal Ophthalmia Neonatorum
Recommended Regimen for Gonococcal Ophthalmia
Neonatorum
Ceftriaxone 25–50 mg/kg body weight IV or IM in a single dose, not to
exceed 250 mg
One dose of ceftriaxone is adequate therapy for gonococcal
ophthalmia. Ceftriaxone should be administered cautiously
to neonates with hyperbilirubinemia, especially those born
prematurely. Cefotaxime 100 mg/kg body weight IV or IM as
a single dose can be administered for those neonates unable to
receive ceftriaxone because of simultaneous administration of
IV calcium. Topical antibiotic therapy alone is inadequate and
unnecessary if systemic treatment is administered.
Other Management Considerations
Chlamydial testing should be performed simultaneously from
the inverted eyelid specimen (see Ophthalmia Neonatorum
Caused by C. trachomatis). Newborns who have gonococcal
ophthalmia should be evaluated for signs of disseminated
infection (e.g., sepsis, arthritis, and meningitis). Newborns
who have gonococcal ophthalmia should be managed in
consultation with an infectious disease specialist.
Management of Mothers and Their Sex Partners
Mothers of newborns with ophthalmia neonatorum caused by
N. gonorrhoeae should be evaluated, tested, and presumptively
treated for gonorrhea, along with their sex partners (see
Gonococcal Infection Among Adolescents and Adults).
Disseminated Gonococcal Infection and
Gonococcal Scalp Abscesses Among Neonates
DGI might present as sepsis, arthritis, or meningitis and
is a rare complication of neonatal gonococcal infection.
Localized gonococcal infection of the scalp can result
from fetal monitoring through scalp electrodes. Detecting
gonococcal infection among neonates who have sepsis, arthritis,
meningitis, or scalp abscesses requires cultures of blood, CSF,
or joint aspirate. Specimens obtained from the conjunctiva,
vagina, oropharynx, and rectum are useful for identifying the
primary site or sites of infection. Antimicrobial susceptibility
testing of all isolates should be performed. Positive Gram-
stained smears of abscess exudate, CSF, or joint aspirate provide
a presumptive basis for initiating treatment for N. gonorrhoeae.
Treatment
Recommended Regimens for Disseminated Gonococcal
Infection Among Neonates
Ceftriaxone 25–50 mg/kg body weight/day IV or IM in a single
daily dose for 7 days, with a duration of 10–14 days if meningitis is
documented
or
Cefotaxime 25 mg/kg body weight/day IV or IM every 12 hours for
7 days, with a duration of 10–14 days if meningitis is documented
Ceftriaxone should be administered cautiously to neonates
with hyperbilirubinemia, especially those born prematurely.
Cefotaxime 100 mg/kg body weight IV or IM as a single
dose can be administered for those neonates unable to
receive ceftriaxone because of simultaneous administration
of IV calcium.
Other Management Considerations
Chlamydial testing should be performed simultaneously
among neonates with gonococcal infection (see Chlamydial
Infection Among Neonates). Neonates who have DGI should
be managed in consultation with an infectious disease specialist.
Management of Mothers and Their Sex Partners
Mothers of newborns who have DGI or scalp abscesses caused
by N. gonorrhoeae should be evaluated, tested, and presumptively
treated for gonorrhea, along with their sex partners (see
Gonococcal Infection Among Adolescents and Adults).
Neonates Born to Mothers Who Have Gonococcal
Infection
Neonates born to mothers who have untreated gonorrhea
are at high risk for infection. Neonates should be tested for
gonorrhea at exposed sites (e.g., conjunctiva, vagina, rectum,
and oropharynx) and treated presumptively for gonorrhea.
Treatment in the Absence of Signs of Gonococcal
Infection
Recommended Regimen for Neonates Without Signs of
Gonococcal Infection
Ceftriaxone 20–50 mg/kg body weight IV or IM in a single dose, not to
exceed 250 mg
Other Management Considerations
Ceftriaxone should be administered cautiously to neonates
with hyperbilirubinemia, especially those born prematurely.
Cefotaxime 100 mg/kg body weight IV or IM as a single
dose can be administered for those neonates unable to
receive ceftriaxone because of simultaneous administration
of IV calcium. Age-appropriate chlamydial testing should be
Recommendations and Reports
80 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
performed simultaneously among neonates with gonococcal
infection (see Chlamydial Infection Among Neonates).
Follow-up examination is not required.
Management of Mothers and Their Sex Partners
Mothers who have gonorrhea and their sex partners should
be evaluated, tested, and presumptively treated for gonorrhea
(see Gonococcal Infection Among Adolescents and Adults).
Gonococcal Infection Among
Infants and Children
Sexual abuse is the most frequent cause of gonococcal
infection among infants and children (see Sexual Assault or
Abuse of Children). For preadolescent girls, vaginitis is the
most common manifestation of this infection; gonococcal-
associated PID after vaginal infection can be less common
among preadolescents than adults. Among sexually abused
children, anorectal and pharyngeal infections with N.
gonorrhoeae are frequently asymptomatic.
Diagnostic Considerations
Culture can be used to test urogenital and extragenital sites for
girls and boys. NAAT can be used to test for N. gonorrhoeae from
vaginal and urine specimens from girls and urine for boys (see
Sexual Assault or Abuse of Children). Although data regarding
NAAT from extragenital sites (rectum and pharynx) among
children are more limited, and performance is test dependent,
no evidence supports that performance of NAAT for detection
of N. gonorrhoeae among children differs from that among adults
(553). Because of the implications of a N. gonorrhoeae diagnosis
in a child, only validated FDA-cleared NAAT assays should be
used with extragenital specimens. Consultation with an expert
is necessary before using NAAT to minimize the possibility of
cross-reaction with nongonococcal Neisseria species and other
commensals (e.g., N. meningitidis, Neisseria sicca, Neisseria
lactamica, Neisseria cinerea, or M. catarrhalis) and to ensure
correct interpretation of results.
Gram stains are inadequate for evaluating prepubertal children
for gonorrhea and should not be used to diagnose or exclude
gonorrhea. If evidence of DGI exists, gonorrhea culture and
antimicrobial susceptibility testing should be obtained from
relevant clinical sites (see Disseminated Gonococcal Infection).
Recommended Regimen for Uncomplicated Gonococcal
Vulvovaginitis, Cervicitis, Urethritis, Pharyngitis, or Proctitis
Among Infants and Children Weighing ≤45 kg
Ceftriaxone 25–50 mg/kg body weight IV or IM in a single dose, not to
exceed 250 mg IM
Recommended Regimen for Uncomplicated Gonococcal
Vulvovaginitis, Cervicitis, Urethritis, Pharyngitis, or Proctitis
Among Children Weighing >45 kg
Treat with the regimen recommended for adults
(see Gonococcal Infections)
Recommended Regimen for Bacteremia or Arthritis Among
Children Weighing ≤45 kg
Ceftriaxone 50 mg/kg body weight (maximum dose: 2 g) IM or IV in a
single dose daily every 24 hours for 7 days
Recommended Regimen for Bacteremia or Arthritis Among
Children Weighing >45 kg
Ceftriaxone 1 g IM or IV in a single dose daily every 24 hours for 7 days
Other Management Considerations
Follow-up cultures are unnecessary. Only parenteral
cephalosporins (i.e., ceftriaxone) are recommended for use
among children. All children identified as having gonococcal
infections should be tested for C. trachomatis, syphilis, and
HIV (see Sexual Assault or Abuse of Children).
Mycoplasma genitalium
M. genitalium causes symptomatic and asymptomatic
urethritis among men and is the etiology of approximately
15%–20% of NGU, 20%–25% of nonchlamydial NGU,
and 40% of persistent or recurrent urethritis (697,909,910).
Infection with C. trachomatis is common in selected geographic
areas (911913), although M. genitalium is often the sole
pathogen. Data are insufficient to implicate M. genitalium
infection with chronic complications among men (e.g.,
epididymitis, prostatitis, or infertility). The consequences
of asymptomatic infection with M. genitalium among men
are unknown.
Among women, M. genitalium has been associated with
cervicitis, PID, preterm delivery, spontaneous abortion, and
infertility, with an approximately twofold increase in the risk
for these outcomes among women infected with M. genitalium
(766). M. genitalium infections among women are also
frequently asymptomatic, and the consequences associated
with asymptomatic M. genitalium infection are unknown.
M. genitalium can be detected among 10%–30% of women
with clinical cervicitis (767,770,772,914916). The existing
evidence between M. genitalium and cervicitis is mostly
supportive of a causal association. Elevated proinflammatory
cytokines have been demonstrated among women with
M. genitalium, with return to baseline levels after clearance of
the pathogen (917).
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 81
US Department of Health and Human Services/Centers for Disease Control and Prevention
M. genitalium is identified in the cervix or endometrium
of women with PID more often than in women without PID
(918924). Prevalence of M. genitalium among women with
PID ranges from 4% to 22% (925,926) and was reported
as 60% in one study of women with postabortal PID (918).
The association with PID is supported by early studies among
nonhuman primates that determined that endosalpingitis
develops after inoculation with M. genitalium (927). Recent
studies evaluating the lower and upper genital tract using
highly sensitive M. genitalium NAAT assays or the role of
M. genitalium in histologically defined endometritis have
reported significantly elevated risk for PID (928). However,
most studies of M. genitalium and PID, even those that
controlled extensively for other infections and behavioral and
biologic risk, are cross-sectional. The few prospective studies
that have evaluated the role of M. genitalium in establishing
subsequent PID demonstrated increased PID risk; however,
these were not statistically significant associations, often
because of a lack of statistical power. No clinical trial data are
available that demonstrate that treating M. genitalium cervical
infection prevents development of PID or endometritis.
Although data regarding the benefits of testing women with
PID for M. genitalium and the importance of directing
treatment against this organism are limited, the associations of
M. genitalium with cervicitis and PID in cross-sectional studies
using NAAT testing are consistent (928).
Data from case-control serologic studies (929931)
and a meta-analysis of clinical studies (766) indicate a
potential role in causing infertility. However, seroassays are
suboptimal and inconclusive. Similarly, evidence for a role for
M. genitalium infection during pregnancy as a cause of perinatal
complications, including preterm delivery, spontaneous
abortion, or low birthweight, are conflicting because evidence is
insufficient to attribute cause (766,932934). Data are limited
regarding ectopic pregnancy and neonatal M. genitalium
infection (935,936).
Rectal infection with M. genitalium has been reported among
1%–26% of MSM (937940) and among 3% of women
(941). Rectal infections often are asymptomatic, although
higher prevalence of M. genitalium has been reported among
men with rectal symptoms. Similarly, although asymptomatic
M. genitalium has been detected in the pharynx, no evidence
exists of it causing oropharyngeal symptoms or systemic disease.
Urogenital M. genitalium infection is associated with HIV
among both men and women (942944); however, the data
are from case-control and cross-sectional studies. Risk for HIV
infection is increased among women with M. genitalium, and
evidence indicates that HIV shedding occurs more often among
persons with M. genitalium and HIV infection who are not taking
ART than among persons without M. genitalium (942,944).
Antimicrobial Resistance
Resistance to azithromycin has been rapidly increasing
and has been confirmed in multiple studies. Prevalence of
molecular markers for macrolide resistance, which highly
correlates with treatment failure, ranges from 44% to 90%
in the United States, Canada, Western Europe, and Australia
(697,702,945953). Treatment with azithromycin alone
has been reported to select for resistance (705,954,955),
with treatment of macrolide-susceptible infections with a
1-g dose of azithromycin resulting in selection of resistant-
strain populations in 10%–12% of cases. The prevalence of
quinolone resistance markers is much lower (697,956959).
The first clinical treatment failures after moxifloxacin were
associated specifically with the S83I mutation in the parC gene
(954,960). Prevalence of the S83I mutation in the United
States ranges from 0% to 15% (947); however, correlation
with fluoroquinolone treatment failure is less consistent than
that with mutations associated with macrolide resistance
(953,961,962). Clinically relevant quinolone resistance often
is associated with coexistent macrolide resistance (954).
Diagnostic Considerations
M. genitalium is an extremely slow-growing organism. Culture
can take up to 6 months, and technical laboratory capacity is
limited to research settings. NAAT for M. genitalium is FDA
cleared for use with urine and urethral, penile meatal, endocervical,
and vaginal swab samples (https://www.hologic.com/package-
inserts/diagnostic-products/aptima-mycoplasma-genitalium-
assay). Molecular tests for macrolide (i.e., azithromycin) or
quinolone (i.e., moxifloxacin) resistance markers are not
commercially available in the United States. However, molecular
assays that incorporate detection of mutations associated with
macrolide resistance are under evaluation.
Men with recurrent NGU should be tested for M. genitalium
using an FDA-cleared NAAT. If resistance testing is available,
it should be performed and the results used to guide therapy.
Women with recurrent cervicitis should be tested for
M. genitalium, and testing should be considered among women
with PID. Testing should be accompanied with resistance
testing, if available. Screening of asymptomatic M. genitalium
infection among women and men or extragenital testing
for M. genitalium is not recommended. In clinical practice,
if testing is unavailable, M. genitalium should be suspected
in cases of persistent or recurrent urethritis or cervicitis and
considered for PID.
Recommendations and Reports
82 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Treatment
M. genitalium lacks a cell wall, and thus antibiotics targeting
cell-wall biosynthesis (e.g., ß-lactams including penicillins and
cephalosporins) are ineffective against this organism. Because
of the high rates of macrolide resistance with treatment failures
(707) and efficient selection of additional resistance, a 1-g dose
of azithromycin should not be used.
Two-stage therapy approaches, ideally using resistance-
guided therapy, are recommended for treatment. Resistance-
guided therapy has demonstrated cure rates of >90% and
should be used whenever possible (759,963); however, it
requires access to macrolide-resistance testing. As part of this
approach, doxycycline is provided as initial empiric therapy,
which reduces the organism load and facilitates organism
clearance, followed by macrolide-sensitive M. genitalium
infections treated with high-dose azithromycin; macrolide-
resistant infections are treated with moxifloxacin (964,965).
Recommended Regimens if M. genitalium Resistance Testing Is
Available
If macrolide sensitive: Doxycycline 100 mg orally 2 times/day for
7 days, followed by azithromycin 1 g orally initial dose, followed by
500 mg orally daily for 3 additional days (2.5 g total)
If macrolide resistant: Doxycycline 100 mg orally 2 times/day for 7 days
followed by moxifloxacin 400 mg orally once daily for 7 days
Recommended Regimen if M. genitalium Resistance Testing Is
Not Available
If M. genitalium is detected by an FDA-cleared NAAT: Doxycycline
100 mg orally 2 times/day for 7 days, followed by moxifloxacin 400 mg
orally once daily for 7 days
Although the majority of M. genitalium strains are sensitive
to moxifloxacin, resistance has been reported, and adverse
side effects and cost should be considered with this regimen.
In settings without access to resistance testing and when
moxifloxacin cannot be used, an alternative regimen can be
considered, based on limited data: doxycycline 100 mg orally
2 times/day for 7 days, followed by azithromycin (1 g orally
on day 1 followed by 500 mg once daily for 3 days) and a test
of cure 21 days after completion of therapy (963). Because of
the high prevalence of macrolide resistance and high likelihood
of treatment failure, this regimen should be used only when
a test of cure is possible, and no other alternatives exist. If
symptomatic treatment failure or a positive test of cure occurs
after this regimen, expert consultation is recommended.
Data are limited regarding use of minocycline in instances of
treatment failure (966).
Recommended PID treatment regimens are not effective
against M. genitalium. Initial empiric therapy for PID, which
includes doxycycline 100 mg orally 2 times/day for 14 days,
should be provided at the time of presentation for care. If
M. genitalium is detected, a regimen of moxifloxacin 400 mg
orally once daily for 14 days has been effective in eradicating the
organism. Nevertheless, no data have been published that assess
the benefits of testing women with PID for M. genitalium, and
the importance of directing treatment against this organism
is unknown.
Follow-Up
Test of cure is not recommended for asymptomatic persons
who received treatment with a recommended regimen. In
settings in which M. genitalium testing is available, persons
with persistent urethritis, cervicitis, or PID accompanied by
detection of M. genitalium should be treated with moxifloxacin.
Management of Sex Partners
Recent studies report a high concordance of M. genitalium
among partners of males, females, and MSM; however, no
studies have determined whether reinfection is reduced with
partner treatment (940,967,968). Sex partners of patients
with symptomatic M. genitalium infection can be tested, and
those with a positive test can be treated to possibly reduce the
risk for reinfection. If testing the partner is not possible, the
antimicrobial regimen that was provided to the patient can
be provided.
Special Considerations
HIV Infection
Persons who have M. genitalium and HIV infection should
receive the same treatment regimen as those persons without HIV.
Diseases Characterized by
Vulvovaginal Itching, Burning,
Irritation, Odor, or Discharge
The majority of women will have a vaginal infection,
characterized by discharge, itching, burning, or odor, during
their lifetime. With the availability of complementary and
alternative therapies and over-the-counter medications for
candidiasis, symptomatic women often seek these products
before or in addition to an evaluation by a medical provider.
Obtaining a medical history alone has been reported to
be insufficient for accurate diagnosis of vaginitis and can
lead to inappropriate administration of medication (969).
Therefore, a careful history, examination, and laboratory
testing to determine the etiology of any vaginal symptoms
are warranted. Information regarding sexual behaviors and
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 83
US Department of Health and Human Services/Centers for Disease Control and Prevention
practices, sex of sex partners, menses, vaginal hygiene practices
(e.g., douching), and self-treatment with oral and intravaginal
medications or other products should be elicited. The
infections most frequently associated with vaginal symptoms
are BV (i.e., replacement of the vaginal flora by an overgrowth
of anaerobic bacteria including G. vaginalis, Prevotella bivia,
A. vaginae, Megasphaera type 1, and numerous other fastidious
or uncultivated anaerobes), trichomoniasis, and vulvovaginal
candidiasis (VVC). Cervicitis can also cause an abnormal
vaginal discharge. Although VVC is usually not sexually
transmitted, it is included in this section because it is frequently
diagnosed among women who have vaginal symptoms or are
being evaluated for an STI.
Multiple diagnostic methods are available for identifying the
etiology of vaginal symptoms. Clinical laboratory testing can
identify the vaginitis cause in the majority of women and is
discussed in detail in the sections of this report dedicated to
each condition. In the clinicians office, the cause of vaginal
symptoms can often be determined by pH, a potassium
hydroxide (KOH) test, and microscopic examination of a wet
mount of fresh samples of vaginal discharge. The pH of the
vaginal secretions can be measured by pH paper; an elevated
pH (i.e., >4.5) is common with BV or trichomoniasis (although
trichomoniasis can also be present with a normal vaginal pH).
Because pH testing is not highly specific, vaginal discharge
should be further examined microscopically by first diluting
one sample in 1 or 2 drops of 0.9% normal saline solution on
one slide and a second sample in 10% KOH solution (samples
that emit an amine odor immediately upon application of
KOH suggest BV or trichomoniasis). Coverslips are then
placed on the slides, and they are examined under a microscope
at low and high power. The saline-solution specimen might
display motile trichomonads or clue cells (i.e., epithelial cells
with borders obscured by small anaerobic bacteria), which
are characteristic of BV. The KOH specimen typically is used
to identify hyphae or blastospores observed with candidiasis.
However, absence of trichomonads in saline or fungal elements
in KOH samples does not rule out these infections because the
sensitivity of microscopy is approximately 50% compared with
NAAT (trichomoniasis) or culture (yeast) (670). Presence of
WBCs without evidence of trichomonads or yeast might also
indicate cervicitis (see Cervicitis).
In settings where pH paper, KOH, and microscopy
are unavailable, a broad range of clinical laboratory tests,
described in the diagnosis section for each disease, can be
used. Presence of objective signs of vulvovaginal inflammation
in the absence of vaginal pathogens after laboratory testing
indicates the possibility of mechanical, chemical, allergic, or
other noninfectious causes of vulvovaginal signs or symptoms.
For women with persistent symptoms and no clear etiology,
referral to a specialist should be considered.
Bacterial Vaginosis
BV is a vaginal dysbiosis resulting from replacement of normal
hydrogen peroxide and lactic-acid–producing Lactobacillus
species in the vagina with high concentrations of anaerobic
bacteria, including G. vaginalis, Prevotella species, Mobiluncus
species, A. vaginae, and other BV-associated bacteria. A notable
feature is the appearance of a polymicrobial biofilm on vaginal
epithelial cells (970). Certain women experience transient
vaginal microbial changes, whereas others experience them for
longer intervals (971). BV is a highly prevalent condition and
the most common cause of vaginal discharge worldwide (972).
However, in a nationally representative survey, the majority of
women with BV were asymptomatic (310).
BV is associated with having multiple male sex partners,
female partners, sexual relationships with more than one person
(973), a new sex partner, lack of condom use (974), douching
(975,976), and HSV-2 seropositivity (977). Male circumcision
reduces the risk for BV among women (978). In addition, BV
prevalence increases during menses (979,980). Women who
have never been sexually active are rarely affected (981). The
cause of the microbial alteration that precipitates BV is not
fully understood, and whether BV results from acquisition
of a single sexually transmitted pathogen is unknown. BV
prevalence has been reported to increase among women with
copper-containing IUDs (972,982). Hormonal contraception
does not increase risk for BV (983) and might protect against
BV development (983,984). Vitamin D deficiency has not
been reported to be a risk factor for BV (985).
Women with BV are at increased risk for STI acquisition,
such as HIV, N. gonorrhoeae, C. trachomatis, T. vaginalis
(977), M. genitalium (986), HPV (987), and HSV-2 (988);
complications after gynecologic surgery; complications of
pregnancy; and recurrence of BV (971,989991). BV also
increases HIV infection acquisition (992) because specific
BV-associated bacteria can increase susceptibility to HIV
(993,994) and the risk for HIV transmission to male
sex partners (187). Evaluation of short-term valacyclovir
suppression among women with HSV-2 did not decrease the
risk for BV, despite effective suppression of HSV-2 (995).
Although BV-associated bacteria can be identified on male
genitalia (996,997), treatment of male sex partners has not been
beneficial in preventing the recurrence of BV (998). Among
WSW, a high level of BV concordance occurs between sex
partners (292); however, no studies have evaluated treatment
of female sex partners of WSW to prevent BV recurrence.
Recommendations and Reports
84 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Diagnostic Considerations
BV can be diagnosed by using clinical criteria (i.e.,
Amsel’s diagnostic criteria) (999) or by determining the
Nugent score from a vaginal Gram stain (1000). Vaginal
Gram stain, considered the reference standard laboratory
method for diagnosing BV, is used to determine the relative
concentration of lactobacilli (i.e., long gram-positive rods),
small gram-negative and gram-variable rods (i.e., G. vaginalis or
Bacteroides), and curved gram-negative rods (i.e., Mobiluncus)
characteristic of BV. A Nugent score of 0–3 is consistent with
a Lactobacillus-predominant vaginal microbiota, 4–6 with
intermediate microbiota (emergence of G. vaginalis), and 7–10
with BV. Clinical diagnosis of BV by Amsel criteria requires at
least three of the following four symptoms or signs:
Homogeneous, thin discharge (milklike consistency) that
smoothly coats the vaginal walls
Clue cells (e.g., vaginal epithelial cells studded with
adherent bacteria) on microscopic examination
pH of vaginal fluid >4.5
A fishy odor of vaginal discharge before or after addition
of 10% KOH (i.e., the whiff test)
Detection of at least three Amsel criteria has been correlated
with results by Gram stain (1001). The sensitivity and
specificity of the Amsel criteria are 37%–70% and 94%–99%,
respectively, compared with the Nugent score (1002).
In addition to the Amsel criteria, multiple POC tests are
available for BV diagnosis. The Osom BV Blue test (Sekisui
Diagnostics) detects vaginal sialidase activity (1003,1004).
The Affirm VP III (Becton Dickinson) is an oligonucleotide
probe test that detects high concentrations of G. vaginalis
nucleic acids (>5 x 10
5
CFU of G. vaginalis/mL of vaginal
fluid) for diagnosing BV, Candida species, and T. vaginalis.
This test has been reported to be most useful for symptomatic
women in conjunction with vaginal pH measurement and
presence of amine odor (sensitivity of 97%); specificity is
81% compared with Nugent. Finally, the FemExam Test
Card (Cooper Surgical) measures vaginal pH, presence of
trimethylamine (a metabolic by-product of G. vaginalis),
and proline aminopeptidase (1005). Sensitivity is 91% and
specificity is 61%, compared with Nugent. This test has
primarily been studied in resource-poor settings (1005), and
although it has been reported to be beneficial compared with
syndromic management, it is not a preferred diagnostic method
for BV diagnosis.
Multiple BV NAATs are available for BV diagnosis among
symptomatic women (1002). These tests are based on detection
of specific bacterial nucleic acids and have high sensitivity
and specificity for BV (i.e., G. vaginalis, A. vaginae, BVAB2,
or Megasphaera type 1) (1006) and certain lactobacilli (i.e.,
Lactobacillus crispatus, Lactobacillus jensenii, and Lactobacillus
gasseri). They can be performed on either clinician- or self-
collected vaginal specimens with results available in <24 hours,
depending on the availability of the molecular diagnostic
platform (1002). Five quantitative multiplex PCR assays are
available: Max Vaginal Panel (Becton Dickinson) (1007),
Aptima BV (Hologic), NuSwab VG (LabCorp) (1008),
OneSwab BV Panel PCR with Lactobacillus Profiling by qPCR
(Medical Diagnostic Laboratories) (1009), and SureSwab BV
(Quest Diagnostics). Two of these assays are FDA cleared (BD
Max Vaginal Panel and Aptima BV), and the other three are
laboratory-developed tests.
The Max Vaginal Panel provides results by an algorithmic
analysis of molecular DNA detection of Lactobacillus species
(L. crispatus and L. jensenii) in addition to G. vaginalis,
A. vaginae, BVAB2, and Megasphaera type 1. This test has
90.5% sensitivity and 85.8% specificity for BV diagnosis,
compared with Amsel criteria and Nugent score. It also provides
results for Candida species and T. vaginalis. The Aptima BV
detects G. vaginalis, A. vaginae, and certain Lactobacillus species
including L. crispatus, L. jensenii, and L. gasseri, with sensitivity
and specificity ranging from 95.0% to 97.3% and 85.8% to
89.6%, respectively (using either clinician- or patient-collected
vaginal swabs). The three laboratory-developed tests (NuSwab
VG, OneSwab BV Panel PCR with Lactobacillus Profiling by
qPCR, and SureSwab BV) have to be internally validated before
use for patient care yet have good sensitivity and specificity,
similar to FDA-cleared assays. BV NAATs should be used
among symptomatic women only (e.g., women with vaginal
discharge, odor, or itch) because their accuracy is not well
defined for asymptomatic women. Despite the availability of
BV NAATs, traditional methods of BV diagnosis, including
the Amsel criteria, Nugent score, and the Affirm VP III assay,
remain useful for diagnosing symptomatic BV because of their
lower cost and ability to provide a rapid diagnosis. Culture of
G. vaginalis is not recommended as a diagnostic tool because
it is not specific. Cervical Pap tests have no clinical utility for
diagnosing BV because of their low sensitivity and specificity.
Treatment
Treatment for BV is recommended for women with
symptoms. Established benefits of therapy among nonpregnant
women are to relieve vaginal symptoms and signs of infection.
Other potential benefits of treatment include reduction
in the risk for acquiring C. trachomatis, N. gonorrhoeae,
T. vaginalis, M. genitalium, HIV, HPV, and HSV-2 (971,986
988,990,1010). No data are available that directly compare
the efficacy of oral and topical medications for treating BV.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 85
US Department of Health and Human Services/Centers for Disease Control and Prevention
Recommended Regimens for Bacterial Vaginosis
Metronidazole 500 mg orally 2 times/day for 7 days
or
Metronidazole gel 0.75% one full applicator (5 g) intravaginally, once
daily for 5 days
or
Clindamycin cream 2% one full applicator (5 g) intravaginally at
bedtime for 7 days
A review regarding alcohol consumption during
metronidazole treatment reported no in vitro studies, animal
models, reports of adverse effects, or clinical studies providing
convincing evidence of a disulfiram-like interaction between
alcohol and metronidazole (1011). The previous warning
against simultaneous use of alcohol and metronidazole was
based on laboratory experiments and individual case histories
in which the reported reactions were equally likely to have been
caused by alcohol alone or by adverse effects of metronidazole.
Metronidazole does not inhibit acetaldehyde dehydrogenase,
as occurs with disulfiram. Ethanol alone or ethanol-
independent side effects of metronidazole might explain the
suspicion of disulfiram-like effects. Thus, refraining from
alcohol use while taking metronidazole (or tinidazole) is
unnecessary. Clindamycin cream is oil based and might weaken
latex condoms and diaphragms for 5 days after use (refer to
clindamycin product labeling for additional information).
Women should be advised to refrain from sexual activity
or to use condoms consistently and correctly during the BV
treatment regimen. Douching might increase the risk for
relapse, and no data support use of douching for treatment
or symptom relief.
Alternative Regimens
Clindamycin 300 mg orally 2 times/day for 7 days
or
Clindamycin ovules 100 mg intravaginally once at bedtime for 3 days*
or
Secnidazole 2 g oral granules in a single dose
or
Tinidazole 2 g orally once daily for 2 days
or
Tinidazole 1 g orally once daily for 5 days
* Clindamycin ovules use an oleaginous base that might weaken latex or
rubber products (e.g., condoms and diaphragms). Use of such products
within 72 hours after treatment with clindamycin ovules is not
recommended.
Oral granules should be sprinkled onto unsweetened applesauce, yogurt,
or pudding before ingestion. A glass of water can be taken after
administration to aid in swallowing.
Alternative regimens include secnidazole oral granules
(10121014), multiple oral tinidazole regimens (1015), or
clindamycin (oral or intravaginal) (1016). In a phase 3 clinical
trial of secnidazole 2 g oral granules versus placebo, BV
clinical cure rates at days 21–30 were 53% in the secnidazole
arm compared with 19% in the placebo arm (p<0.001)
(1013). Secnidazole is listed as an alternative regimen, due
to its higher cost and lack of long-term outcomes compared
with recommended BV treatments. A patient savings card
for secnidazole is available at https://www.solosec.com/
savings-card.
Additional BV treatment regimens include metronidazole
1.3% vaginal gel in a single dose (1017,1018) and clindamycin
phosphate (Clindesse) 2% vaginal cream in a single dose
(1019). In a phase 3 clinical trial of metronidazole 1.3% vaginal
gel versus placebo, BV clinical cure rates at day 21 were 37.2%
in the metronidazole 1.3% vaginal gel arm, compared with
26.6% in the placebo arm (p = 0.01) (1018). A patient savings
card for metronidazole 1.3% vaginal gel is available at https://
nuvessa.com/nuvessa_files/19_Nuvessa_WEB_Card_032819.
pdf. In a multicenter, randomized, single-blind, parallel-
group study of Clindesse 2% vaginal cream single dose versus
clindamycin 2% vaginal cream at bedtime for 7 days among
540 women with BV, no statistically significant difference
existed between groups in clinical cure at days 21–30 (64.3%
versus 63.2%; p = 0.95) (1019); however, this study had
methodologic problems. A patient savings card for Clindesse
2% vaginal cream is available at https://www.clindesse.com/
pdf/CLINDESSE_SavingsCard.pdf.
BV biofilm disrupting agents (i.e., TOL-463) (1020) are
being investigated to determine their role in enhancing the
likelihood of BV cure relative to approved therapies. Studies
have evaluated the clinical and microbiologic efficacy of
intravaginal Lactobacillus and other probiotic formulations to
treat BV and restore normal vaginal microbiota (10211025);
overall, no studies support these products as an adjunctive or
replacement therapy for women with BV.
Other Management Considerations
All women with BV should be tested for HIV and other STIs.
Follow-Up
Follow-up visits are unnecessary if symptoms resolve. Because
persistent or recurrent BV is common, women should be advised
to return for evaluation if symptoms recur. Limited data are
available regarding optimal management strategies for women
with persistent or recurrent BV. Using a different recommended
treatment regimen can be considered for women who have a
recurrence; however, retreatment with the same recommended
regimen is an acceptable approach for treating persistent or
recurrent BV after the first occurrence (1026). For women with
multiple recurrences after completion of a recommended regimen,
either 0.75% metronidazole gel or 750 mg metronidazole vaginal
suppository twice weekly for >3 months has been reported to
reduce recurrences, although this benefit does not persist when
Recommendations and Reports
86 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
suppressive therapy is discontinued (1027,1028). Limited data
indicate that for women with multiple recurrences, an oral
nitroimidazole (metronidazole or tinidazole 500 mg 2 times/
day for 7 days), followed by intravaginal boric acid 600 mg
daily for 21 days and suppressive 0.75% metronidazole gel
twice weekly for 4–6 months, might be an option for women
with recurrent BV (1029). Monthly oral metronidazole 2 g
administered with fluconazole 150 mg has also been evaluated
as suppressive therapy; this regimen reduced the BV incidence
and promoted colonization with normal vaginal microbiota
(1030). A randomized controlled trial of a dendrimer-based
microbicide 1% vaginal gel (Astodrimer) also reported favorable
results in prolonging the time to BV recurrence, compared with
placebo (1031). In addition, a clinical trial of L. crispatus CTV-05
(Lactin-V), administered vaginally in 4 consecutive daily doses
for 4 days in week 1 followed by twice weekly doses for 10 weeks
(after initial treatment with 5 days of 0.75% vaginal metronidazole
gel), reported a substantially lower incidence of BV recurrence at
12 weeks in the Lactin-V arm, compared with placebo (1032);
however this medication is not yet FDA cleared or commercially
available. High-dose Vitamin D supplementation has not been
determined to decrease BV recurrence in randomized controlled
trials (1033) and is not recommended.
Management of Sex Partners
Data from earlier clinical trials indicate that a womans
response to therapy and the likelihood of relapse or
recurrence are not affected by treatment of her sex partner
(998). Therefore, routine treatment of sex partners is not
recommended. However, a pilot study reported that male
partner treatment (i.e., metronidazole 400 mg orally 2 times/
day in conjunction with 2% clindamycin cream applied
topically to the penile skin 2 times/day for 7 days) of women
with recurrent BV had an immediate and sustained effect on
the composition of the vaginal microbiota, with an overall
decrease in bacterial diversity at day 28 (1034). Male partner
treatment also had an immediate effect on the composition of
the penile microbiota; however, this was not as pronounced
at day 28, compared with that among women. A phase 3
multicenter randomized double-blinded trial evaluating the
efficacy of a 7-day oral metronidazole regimen versus placebo
for treatment of male sex partners of women with recurrent
BV did not find that male partner treatment reduced BV
recurrence in female partners, although women whose male
partners adhered to multidose metronidazole were less likely
to experience treatment failure (1035).
Special Considerations
Drug Allergy, Intolerance, or Adverse Reactions
Intravaginal clindamycin cream is preferred in case of allergy
or intolerance to metronidazole or tinidazole. Intravaginal
metronidazole gel can be considered for women who are not
allergic to metronidazole but do not tolerate oral metronidazole.
Pregnancy
BV treatment is recommended for all symptomatic pregnant
women because symptomatic BV has been associated with
adverse pregnancy outcomes, including premature rupture
of membranes, preterm birth, intra-amniotic infection, and
postpartum endometritis (989,991,1036). Studies have been
undertaken to determine the efficacy of BV treatment among
this population, including two trials demonstrating that oral
metronidazole was efficacious during pregnancy by using
the 250 mg 3 times/day regimen (1037,1038); however, oral
metronidazole administered as a 500 mg 2 times/day regimen
can also be used. One trial involving a limited number of
participants revealed treatment with oral metronidazole
500 mg 2 times/day for 7 days to be equally effective as
metronidazole gel 0.75% for 5 days, with cure rates of 70%
by using Amsel criteria to define cure (1039). Another trial
demonstrated a cure rate of 85% by using Gram-stain criteria
after treatment with oral clindamycin 300 mg 2 times/day for
7 days (10401043).
Although older studies indicated a possible link between
using vaginal clindamycin during pregnancy and adverse
outcomes for the newborn, newer data demonstrate that
this treatment approach is safe for pregnant women (1044).
Although metronidazole crosses the placenta, no evidence of
teratogenicity or mutagenic effects among infants has been
reported in multiple cross-sectional, case-control, and cohort
studies of pregnant women (10411043). These data indicate
that metronidazole therapy poses low risk during pregnancy.
Data from human studies are limited regarding the use of
tinidazole in pregnancy; however, animal data demonstrate
that such therapy poses moderate risk. Thus, tinidazole should
be avoided during pregnancy (431). Data are insufficient
regarding efficacy and adverse effects of secnidazole, Clindesse
2% vaginal cream, metronidazole 1.3% vaginal gel, and
750-mg vaginal metronidazole tablets during pregnancy; thus,
their use should be avoided.
Oral therapy has not been reported to be superior to topical
therapy for treating symptomatic BV in effecting cure or
preventing adverse outcomes of pregnancy. Pregnant women
can be treated with any of the recommended regimens for
nonpregnant women, in addition to the alternative regimens
of oral clindamycin and clindamycin ovules.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 87
US Department of Health and Human Services/Centers for Disease Control and Prevention
Treatment of asymptomatic BV among pregnant women
at high risk for preterm delivery (i.e., those with a previous
preterm birth or late miscarriage) has been evaluated by
multiple studies, which have yielded mixed results. Seven
trials have evaluated treatment of pregnant women with
asymptomatic BV at high risk for preterm delivery: one revealed
harm (1045), two reported no benefit (1046,1047), and four
demonstrated benefit (1037,1038,1048,1049).
Treatment of asymptomatic BV among pregnant women
at low risk for preterm delivery has not been reported to
reduce adverse outcomes of pregnancy in a large multicenter
randomized controlled trial (1050). Therefore, routine
screening for BV among asymptomatic pregnant women at
high or low risk for preterm delivery for preventing preterm
birth is not recommended.
Metronidazole is secreted in breast milk. With maternal
oral therapy, breastfed infants receive metronidazole in doses
that are less than those used to treat infections among infants,
although the active metabolite adds to the total infant exposure.
Plasma levels of the drug and metabolite are measurable
but remain less than maternal plasma levels (https://www.
ncbi.nlm.nih.gov/books/NBK501922/?report=classic).
Although multiple reported case series identified no evidence
of metronidazole-associated adverse effects for breastfed
infants, certain clinicians recommend deferring breastfeeding
for 12–24 hours after maternal treatment with a single 2-g
dose of metronidazole (1051). Lower doses produce a lower
concentration in breast milk and are considered compatible
with breastfeeding (1052,1053).
HIV Infection
BV appears to recur with higher frequency among women
who have HIV infection (1054). Women with HIV infection
and BV should receive the same treatment regimen as those
who do not have HIV.
Trichomoniasis
Trichomoniasis is estimated to be the most prevalent nonviral
STI worldwide, affecting approximately 3.7 million persons
in the United States (838,1055). Because trichomoniasis is
not a reportable disease (1056), and no recommendations are
available for general screening for T. vaginalis, the epidemiology
of trichomoniasis has largely come from population-based and
clinic-based surveillance studies. The U.S. population-based
T. vaginalis prevalence is 2.1% among females and 0.5% among
males, with the highest rates among Black females (9.6%)
and Black males (3.6%), compared with non-Hispanic White
women (0.8%) and Hispanic women (1.4%) (1057,1058).
Unlike chlamydia and gonorrhea, T. vaginalis prevalence
rates are as high among women aged >24 years as they are
for women aged <24 years (1057). Among persons attending
nine geographically diverse STD clinics, the trichomonas
prevalence was 14.6% among women (1059), and a study
of STD clinic attendees in Birmingham, Alabama, identified
a prevalence of 27% among women and 9.8% among men
(1060). Symptomatic women have a four times higher rate of
infection than asymptomatic women (26% versus 6.5%) (1061).
Rates are also high among incarcerated persons of both sexes at
9%–32% of incarcerated women (386,387,391,392,1062) and
3.2%–8% of incarcerated men (388). Women with a history of
incarceration are two to five times more likely to have T. vaginalis
(387,388,1063,1064). Other risk factors for T. vaginalis include
having two or more sex partners during the previous year, having
less than a high school education, and living below the national
poverty level (1065). Women with BV are at higher risk for
T. vaginalis (1066). Male partners of women with trichomoniasis
are likely to have infection (1067), although the prevalence of
trichomoniasis among MSM is low (179,1068).
The majority of persons who have trichomoniasis
(70%–85%) either have minimal or no genital symptoms,
and untreated infections might last from months to years
(137,1069,1070). Men with trichomoniasis sometimes have
symptoms of urethritis, epididymitis, or prostatitis, and women
with trichomoniasis sometimes have vaginal discharge, which
can be diffuse, malodorous, or yellow-green with or without
vulvar irritation, and might have a strawberry-appearing cervix,
which is observed more often on colposcopy than on physical
examination (1071). Although many persons might be unaware
of their infection, it is readily passed between sex partners
during penile-vaginal sex (1072) or through transmission of
infected vaginal fluids or fomites among women who have sex
with women (275,294).
Among persons who are sexually active, the best way to
prevent genital trichomoniasis is through consistent and correct
use of condoms (external or internal) (18). Partners of men
who have been circumcised might have a somewhat reduced
risk for T. vaginalis infection (1072,1073). Douching is not
recommended because it might increase the risk for vaginal
infections, including trichomoniasis (1074).
T. vaginalis causes reproductive morbidity and has been
reported to be associated with a 1.4-times greater likelihood
of preterm birth, premature rupture of membranes, and
infants who are small for gestational age (1075). T. vaginalis
was also determined to be associated with a 2.1-fold increased
risk for cervical cancer in a meta-analysis (1076). Another
meta-analysis of six studies reported a slightly elevated but
not statistically significant association between T. vaginalis and
prostate cancer (1077).
Recommendations and Reports
88 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
T. vaginalis infection is associated with a 1.5-fold increased risk
for HIV acquisition and is associated with an increase in HIV
vaginal shedding, which is reduced with T. vaginalis treatment
among women without viral suppression (1078,1079). Among
women with HIV infection, T. vaginalis infection is associated
with increased risk for PID (10801082).
Diagnostic testing for T. vaginalis should be performed for
women seeking care for vaginal discharge. Annual screening
might be considered for persons receiving care in high-
prevalence settings (e.g., STD clinics and correctional facilities)
and for asymptomatic women at high risk for infection (e.g.,
multiple sex partners, transactional sex, drug misuse, or a
history of STIs or incarceration). However, data are lacking
regarding whether screening and treatment for asymptomatic
trichomoniasis in high-prevalence settings for women at high
risk can reduce any adverse health events and health disparities
or reduce community infection burden. Decisions about
screening can be guided by local epidemiology of T. vaginalis
infection. Routine annual screening for T. vaginalis among
asymptomatic women with HIV infection is recommended
because of these adverse events associated with trichomoniasis
and HIV infection.
Extragenital T. vaginalis is possible but highly uncommon
compared with genital infections. A study of 500 men in San
Francisco, California, reported a 0.6% rate of rectal T. vaginalis
(1083); however, this might reflect deposition of T. vaginalis
DNA and not necessarily active infection. Few studies of
extragenital T. vaginalis among women have been published.
The efficacy, benefit, and cost-effectiveness of extragenital
screening are unknown, and no tests are FDA cleared for
extragenital testing; therefore, rectal and oral testing for
T. vaginalis is not recommended.
Diagnostic Considerations
Wet-mount microscopy traditionally has been used as
the preferred diagnostic test for T. vaginalis among women
because it is inexpensive and can be performed at the POC;
however, it has low sensitivity (44%–68%) compared with
culture (10841086). To improve detection, clinicians using
wet mounts should attempt to evaluate slides immediately
after specimen collection because sensitivity decreases quickly
to 20% within 1 hour after collection (1087). More highly
sensitive and specific molecular diagnostic options are available,
which should be used in conjunction with a negative wet
mount when possible.
NAATs are highly sensitive, detecting more T. vaginalis
infections than wet-mount microscopy among women
(1060). The Aptima T. vaginalis assay (Beckton Dickinson) is
FDA cleared for detection of T. vaginalis from symptomatic
or asymptomatic women. Reliable samples include
clinician-collected endocervical swabs, clinician-collected
vaginal swabs, female urine specimens, and liquid Pap
smear specimens collected in PreservCyt Solution (Hologic)
(698,1088). This assay detects RNA by transcription-
mediated amplification with a sensitivity of 95.3%–100%
and specificity of 95.2%–100%, compared with wet mount
and culture (1088,1089). Among women, vaginal swabs and
urine specimens have <100% concordance (1084). This assay
has not been FDA cleared for use among men and should be
internally validated in accordance with CLIA regulations before
use with urine or urethral swabs from men. The Probe Tec TV
Q
x
Amplified DNA Assay (Becton Dickinson) is FDA cleared
for detection of T. vaginalis from vaginal (patient-collected
or clinician-collected) swabs, endocervical swabs, or urine
specimens from women and has sensitivity of 98.3% and
specificity of 99.6%, compared with wet mount and culture
(1090). Similar to the Aptima T. vaginalis assay, this test is only
FDA cleared for use among women and should be internally
validated for use with men. The Max CTGCTV2 assay (Becton
Dickinson) is also FDA cleared for detection of T. vaginalis in
patient-collected or clinician-collected vaginal swab specimens
and male and female urine specimens, with sensitivity and
specificity of 96.2%–100% and 99.1%–100%, respectively,
depending on the specimen type, compared with wet mount
and culture (1091). GeneXpert TV (Cepheid) is a moderately
complex rapid test that can be performed in ≤1 hour and can be
used at the POC (1092). It has been FDA cleared for use with
female urine specimens, endocervical swabs, patient-collected
or clinician-collected vaginal specimens, and male urine
specimens, with sensitivity and specificity of 99.5%–100%
and 99.4%–99.9% (1007), respectively, compared with wet
mount and culture.
Multiple FDA-cleared rapid tests are available for detecting
T. vaginalis with improved sensitivities and specificities,
compared with wet mount. The Osom trichomonas rapid
test (Sekisui Diagnostics) is an antigen-detection test that uses
immunochromatographic capillary flow dipstick technology
that can be performed at the POC by using clinician-obtained
vaginal specimens. Results are available in approximately
10–15 minutes, with sensitivities of 82%–95% and specificity
of 97%–100%, compared with wet mount, culture, and
transcription-mediated amplification (1089,1093,1094). A
study of 209 women aged 14–22 years reported that >99%
could correctly perform and interpret a vaginal self-test by
using the Osom assay, with a high correlation with clinician
interpretation (96% agreement; κ=0.87) (1094). The Osom
test should not be used with men because of low sensitivity
(38% compared with Aptima) (1095). The Solana trichomonas
assay (Quidel) is another rapid test for the qualitative detection
of T. vaginalis DNA and can yield results <40 minutes after
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 89
US Department of Health and Human Services/Centers for Disease Control and Prevention
specimen collection. This assay is FDA cleared for diagnosing
T. vaginalis from female vaginal and urine specimens from
asymptomatic and symptomatic women with sensitivity >98%,
compared with NAAT for vaginal specimens, and >92% for
urine specimens (1096). The Amplivue trichomonas assay
(Quidel) is another rapid test providing qualitative detection
of T. vaginalis that has been FDA cleared for vaginal specimens
from symptomatic and asymptomatic women, with sensitivity
of 90.7% and specificity of 98.9%, compared with NAAT
(1097). Neither the Osom assay nor the Affirm VP III test is
FDA cleared for use with specimens from men.
Culture, such as the InPouch system (BioMed Diagnostics),
was considered the most sensitive method for diagnosing
T. vaginalis infection before molecular detection methods
became available. Culture has sensitivity of 44%–75% and
specificity of <100% (698,1086,1098). For women, vaginal
secretions are the preferred specimen type for culture because
urine culture is less sensitive (698,1099,1100). For men,
culture specimens require a urethral swab, urine sediment,
or semen. To improve diagnostic yield, multiple specimens
from men can be used to inoculate a single culture. Cultures
require an incubator and are necessary for T. vaginalis drug
susceptibility testing. The InPouch specimen should be
examined daily for 5 days over a 7-day period to reduce the
possibility of false negatives (1101).
Although T. vaginalis might be an incidental finding on a
Pap test, neither conventional nor liquid-based Pap smears
are considered diagnostic tests for trichomoniasis; however,
women with T. vaginalis identified on a Pap smear should be
retested with sensitive diagnostic tests and treated if infection
is confirmed (1102,1103).
Treatment
Treatment reduces symptoms and signs of T. vaginalis
infection and might reduce transmission. Treatment
recommendations for women are based on a meta-analysis
(1104) and a multicenter, randomized trial of mostly
symptomatic women without HIV infection (1105). The
study demonstrated that multidose metronidazole (500 mg
orally 2 times/day for 7 days) reduced the proportion of
women retesting positive at a 1-month test of cure visit by half,
compared with women who received the 2-g single dose.No
published randomized trials are available that compare these
doses among men.
Recommended Regimen for Trichomoniasis Among Women
Metronidazole 500 mg orally 2 times/day for 7 days
Recommended Regimen for Trichomoniasis Among Men
Metronidazole 2 g orally in a single dose
Alternative Regimen for Women and Men
Tinidazole 2 g orally in a single dose
The nitroimidazoles are the only class of medications with
clinically demonstrated efficacy against T. vaginalis infections.
Tinidazole is usually more expensive, reaches higher levels in
serum and the genitourinary tract, has a longer half-life than
metronidazole (12.5 hours versus 7.3 hours), and has fewer
gastrointestinal side effects (1106,1107). In randomized
clinical trials, recommended metronidazole regimens have
resulted in cure rates of approximately 84%–98% (1108),
and the recommended tinidazole regimen has resulted in cure
rates of approximately 92%–100% (11081112). Randomized
controlled trials comparing single 2-g doses of metronidazole
and tinidazole indicated that tinidazole is equivalent or superior
to metronidazole in achieving parasitologic cure and symptom
resolution (1110,1113,1114).
Metronidazole gel does not reach therapeutic levels in the
urethra and perivaginal glands. Because it is less efficacious
than oral metronidazole, it is not recommended.
Other Management Considerations
Providers should advise persons with T. vaginalis infections
to abstain from sex until they and their sex partners are treated
(i.e., when therapy has been completed and any symptoms
have resolved). Testing for other STIs, including HIV, syphilis,
gonorrhea, and chlamydia, should be performed for persons
with T. vaginalis.
Follow-Up
Because of the high rate of reinfection among women treated
for trichomoniasis, retesting for T. vaginalis is recommended
for all sexually active women <3 months after initial treatment
regardless of whether they believe their sex partners were
treated (137,1115). If retesting at 3 months is not possible,
clinicians should retest whenever persons next seek medical
care <12 months after initial treatment. Data are insufficient
to support retesting men after treatment.
Management of Sex Partners
Concurrent treatment of all sex partners is vital for
preventing reinfections. Current partners should be referred
for presumptive therapy. Partners also should be advised to
abstain from intercourse until they and their sex partners
have been treated and any symptoms have resolved. EPT
might have a role in partner management for trichomoniasis
(129,1116) and can be used in states where permissible by law
(https://www.cdc.gov/std/ept/legal/default.htm); however, no
partner management intervention has been demonstrated to
be superior in reducing reinfection rates (129,130). Although
Recommendations and Reports
90 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
no definitive data exist to guide treatment for partners of
persons with persistent or recurrent trichomoniasis among
whom nonadherence and reinfection are unlikely, partners
might benefit from being evaluated and receiving treatment
(see Recurrent Trichomoniasis).
Recurrent Trichomoniasis
A recurrent infection can result from treatment failure
(antimicrobial-resistant T. vaginalis or host-related problems),
lack of adherence, or reinfection from an untreated sex partner.
In the case of a recurrent infection, the origin of the repeat
infection should be assessed because most recurrent infections
likely result from reinfection. Retesting can be considered in
cases of persistent or recurrent trichomoniasis with culture, the
preferred test. If NAAT is used, it should not be conducted before
3 weeks after treatment completion because of possible detection
of residual nucleic acid that is not clinically relevant (1117).
The nitroimidazoles are the only class of antimicrobials
known to be effective against trichomonas infection.
Metronidazole resistance occurs in 4%–10% of cases of
vaginal trichomoniasis (1116,1118). Tinidazole resistance
is less well studied but was present in 1% of infections in
one study (1116). Overall, more T. vaginalis isolates have
reported susceptibility to tinidazole than metronidazole (1119).
Multidose oral metronidazole is more effective than single-dose
treatment, particularly for women who are symptomatic or
have a history of T. vaginalis (1120).
Nitroimidazole-resistant trichomoniasis is concerning
because few alternatives to standard therapy exist. If treatment
failure occurs in a woman after completing a regimen of
metronidazole 500 mg 2 times/day for 7 days and she has been
reexposed to an untreated partner, a repeat course of the same
regimen is recommended. If no reexposure has occurred, she
should be treated with metronidazole or tinidazole 2 g once
daily for 7 days. If a man has persistent T. vaginalis after a
single 2-g dose of metronidazole and has been reexposed to
an untreated partner, he should be retreated with a single 2-g
dose of metronidazole. If he has not been reexposed, he should
be administered a course of metronidazole 500 mg 2 times/
day for 7 days.
For persons who are experiencing persistent infection
not attributable to reexposure, clinicians should request a
kit from CDC to perform drug-resistance testing (https://
www.cdc.gov/laboratory/specimen-submission/detail.
html?CDCTestCode=CDC-10239). CDC is experienced with
susceptibility testing for nitroimidazole-resistant T. vaginalis
and can provide guidance regarding treatment in cases of
drug resistance. On the basis of drug resistance testing, an
alternative treatment regimen might be recommended.
Treatments for infections demonstrating in vitro resistance
can include metronidazole or tinidazole 2 g daily for 7 days.
If a patient has treatment failure after the 7-day regimen of
high-dose oral metronidazole or tinidazole, two additional
treatment options have been determined to have successful
results for women. The first is high-dose oral tinidazole
2 g daily plus intravaginal tinidazole 500 mg 2 times/day for
14 days (1121). If this regimen fails, high-dose oral tinidazole
(1 g 3 times/day) plus intravaginal paromomycin (4 g of 6.25%
intravaginal paromomycin cream nightly) for 14 days should
be considered (1122).
Alternative regimens might be effective but have not been
systemically evaluated; therefore, consultation with an infectious
disease specialist is recommended. Clinical improvement has
been reported with intravaginal boric acid (1123,1124) but
not with nitazoxanide (11231125). The following topically
applied agents have minimal success (<50%) and are not
recommended: intravaginal betadine (povidone-iodine),
clotrimazole, acetic acid, furazolidone, GV, nonoxynol-9, and
potassium permanganate (1126). No other topical microbicide
has been reported to be effective against trichomoniasis.
Special Considerations
Drug Allergy, Intolerance, and Adverse Reactions
Metronidazole and tinidazole are both nitroimidazoles.
Patients with an IgE-mediated-type hypersensitivity reaction
to 5-nitroimidazole antimicrobials should be managed by
metronidazole desensitization according to published regimens
(1127,1128) and in consultation with an allergy specialist. The
optimal treatment for patients with T. vaginalis who are unable
to be desensitized has not been systematically investigated and is
based on case reports, some of which report using paromomycin
or boric acid for treating T. vaginalis (1123,1129).
Pregnancy
T. vaginalis infection among pregnant women is associated
with adverse pregnancy outcomes, particularly premature
rupture of membranes, preterm delivery, and delivery of infants
who are small for gestational age (1075). One randomized
trial of pregnant women with asymptomatic trichomoniasis
reported no substantial difference in preterm birth after
treatment with 2 g of metronidazole 48 hours apart during
16–23 and 24–29 weeks’ gestation, compared with placebo
(1130). However, that trial had multiple limitations, including
use of an atypical metronidazole regimen. Another multicenter
observational study of asymptomatic pregnant women in sub-
Sahara African, the majority with HIV infection, reported
neither trichomoniasis nor its treatment appeared to influence
the risk for preterm birth or a low-birthweight infant (1131).
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 91
US Department of Health and Human Services/Centers for Disease Control and Prevention
Although metronidazole crosses the placenta, data indicate
that it poses a low risk to the developing fetus (1040,1042,1132).
No evidence of teratogenicity or mutagenic effects among
infants has been found in multiple cross-sectional and cohort
studies among pregnant women examining single-dose (2 g)
and multidose metronidazole regimens (1040,11311135).
Symptomatic pregnant women, regardless of pregnancy stage,
should be tested and treated. Treatment of T. vaginalis infection
can relieve symptoms of vaginal discharge for pregnant women
and reduce sexual transmission to partners. Although perinatal
transmission of trichomoniasis is uncommon, treatment
might also prevent respiratory or genital infection in the
newborn (1136,1137). Clinicians should counsel symptomatic
pregnant women with trichomoniasis about the potential
risks and benefits of treatment and about the importance of
partner treatment and condom use in the prevention of sexual
transmission. The benefit of routine screening for T. vaginalis
in asymptomatic pregnant women has not been established.
Metronidazole is secreted in breast milk. With maternal oral
therapy, breastfed infants receive metronidazole in doses that
are lower than those used to treat infections among infants,
although the active metabolite adds to the total infant exposure.
Plasma levels of the drug and metabolite are measurable but
remain less than maternal plasma levels (https://www.ncbi.nlm.
nih.gov/books/NBK501922). Although multiple reported case
series studies demonstrated no evidence of adverse effects among
infants exposed to metronidazole in breast milk, clinicians
sometimes advise deferring breastfeeding for 12–24 hours after
maternal treatment with metronidazole (1051). In one study,
maternal treatment with metronidazole (400 mg 3 times/day
for 7 days) produced a lower concentration in breast milk and
was considered compatible with breastfeeding over longer
periods (1052).
Data from studies involving human subjects are limited
regarding tinidazole use during pregnancy; however, animal data
indicate this drug poses moderate risk. Thus, tinidazole should
be avoided for pregnant women, and breastfeeding should be
deferred for 72 hours after a single 2-g oral dose of tinidazole
(https://www.ncbi.nlm.nih.gov/books/NBK501922).
HIV Infection
Up to 53% of women with HIV have T. vaginalis infection
(1115,1138). T. vaginalis infection among these women is
substantially associated with pelvic inflammatory disease
(1082). Among women who are not virally suppressed,
treatment of trichomoniasis is associated with decreases in
genital tract HIV viral load and viral shedding (1079,1139);
however, no difference might occur among women who are
virally suppressed (1140). Because of the high prevalence
of T. vaginalis among women with HIV and the potential
for adverse reproductive health, poor birth outcomes, and
possibly amplified HIV transmission, routine screening and
prompt treatment are recommended for all women with HIV
infection; screening should occur at entry to care and then at
least annually thereafter.
A randomized clinical trial involving women with HIV
and T. vaginalis infection demonstrated that a single dose
of metronidazole 2 g orally was less effective than 500 mg
2 times/day for 7 days (1105). Factors that might interfere
with standard single-dose treatment for trichomoniasis among
women with HIV include high rates of asymptomatic BV
infection, ART use, changes in vaginal ecology, and impaired
immunity (1141). Thus, to improve cure rates, women with
HIV who receive a diagnosis of T. vaginalis infection should
be treated with metronidazole 500 mg orally 2 times/day
for 7 days. For pregnant women with HIV, screening at
the first prenatal visit and prompt treatment, as needed, are
recommended because T. vaginalis infection is a risk factor for
vertical transmission of HIV (1142).
Treatment
Treatment reduces symptoms and signs of T. vaginalis
infection, cures infection, and might reduce transmission.
Likelihood of adverse outcomes among women with HIV
infection is also reduced with T. vaginalis therapy.
Recommended Regimen for Trichomonas and HIV Infection
Among Women
Metronidazole 500 mg orally 2 times/day for 7 days
If a woman with HIV infection experiences treatment failure, the
protocol outlined is recommended (see Recurrent Trichomonas).
Other management considerations, follow-up, and management
of sex partners should be performed as for women without HIV
infection. Treatment of men with HIV infection should follow
the same guidelines as for men without HIV.
For women with HIV who receive a diagnosis of T. vaginalis
infection, retesting is recommended 3 months after treatment;
NAAT is encouraged because of higher sensitivity of these
tests. Data are insufficient to support retesting of men with
trichomonas and HIV infection.
Vulvovaginal Candidiasis
VVC usually is caused by Candida albicans but can
occasionally be caused by other Candida species or yeasts.
Typical symptoms of VVC include pruritus, vaginal soreness,
dyspareunia, external dysuria, and abnormal vaginal discharge.
None of these symptoms is specific for VVC. An estimated
75% of women will have at least one episode of VVC, and
40%–45% will have two or more episodes. On the basis of
Recommendations and Reports
92 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
clinical presentation, microbiology, host factors, and response
to therapy, VVC can be classified as either uncomplicated or
complicated (Box 4). Approximately 10%–20% of women
will have complicated VVC, requiring special diagnostic and
therapeutic considerations.
Uncomplicated Vulvovaginal Candidiasis
Diagnostic Considerations
A diagnosis of Candida vaginitis is clinically indicated by the
presence of external dysuria and vulvar pruritus, pain, swelling,
and redness. Signs include vulvar edema, fissures, excoriations,
and thick curdy vaginal discharge. Most healthy women with
uncomplicated VVC have no identifiable precipitating factors.
The diagnosis can be made in a woman who has signs and
symptoms of vaginitis when either a wet preparation (saline,
10% KOH) of vaginal discharge demonstrates budding
yeasts, hyphae, or pseudohyphae, or a culture or other test
yields a positive result for a yeast species. Candida vaginitis is
associated with normal vaginal pH (<4.5). Use of 10% KOH
in wet preparations improves the visualization of yeast and
mycelia by disrupting cellular material that might obscure
the yeast or pseudohyphae. Examination of a wet mount with
KOH preparation should be performed for all women with
symptoms or signs of VVC, and women with a positive result
should be treated. For those with negative wet mounts but
existing signs or symptoms, vaginal cultures for Candida should
be considered. If Candida cultures cannot be performed for
these women, empiric treatment can be considered. Identifying
Candida by culture in the absence of symptoms or signs is not
an indication for treatment because approximately 10%–20%
of women harbor Candida species and other yeasts in the
vagina. The majority of PCR tests for yeast are not FDA
cleared, and providers who use these tests should be familiar
with the performance characteristics of the specific test used.
Yeast culture, which can identify a broad group of pathogenic
yeasts, remains the reference standard for diagnosis.
Treatment
Short-course topical formulations (i.e., single dose and
regimens of 1–3 days) effectively treat uncomplicated VVC.
Treatment with azoles results in relief of symptoms and negative
cultures in 80%–90% of patients who complete therapy.
Recommended Regimens for Vulvovaginal Candidiasis
Over-the-Counter Intravaginal Agents
Clotrimazole 1% cream 5 g intravaginally daily for 7–14 days
or
Clotrimazole 2% cream 5 g intravaginally daily for 3 days
or
Miconazole 2% cream 5 g intravaginally daily for 7 days
or
Miconazole 4% cream 5 g intravaginally daily for 3 days
or
Miconazole 100 mg vaginal suppository one suppository daily for 7 days
or
Miconazole 200 mg vaginal suppository one suppository for 3 days
or
Miconazole 1,200 mg vaginal suppository one suppository for 1 day
or
Tioconazole 6.5% ointment 5 g intravaginally in a single application
Prescription Intravaginal Agents
Butoconazole 2% cream (single-dose bioadhesive product) 5 g
intravaginally in a single application
or
Terconazole 0.4% cream 5 g intravaginally daily for 7 days
or
Terconazole 0.8% cream 5 g intravaginally daily for 3 days
or
Terconazole 80 mg vaginal suppository one suppository daily for 3 days
Oral Agent
Fluconazole 150 mg orally in a single dose
The creams and suppositories in these regimens are oil
based and might weaken latex condoms and diaphragms.
Patients should refer to condom product labeling for further
information. Even women who have previously received a
diagnosis of VVC by a clinician are not necessarily more
likely to be able to diagnose themselves; therefore, any woman
whose symptoms persist after using an over-the-counter
preparation or who has a recurrence of symptoms <2 months
BOX 4. Classification of vulvovaginal candidiasis
Uncomplicated vulvovaginal candidiasis (VVC)
Sporadic or infrequent VVC
and
Mild-to-moderate VVC
and
Likely to be Candida albicans
and
Nonimmunocompromised women
Complicated VVC
Recurrent VVC (three or more episodes of
symptomatic VVC in <1 year)
or
Severe VVC
or
Non–albicans candidiasis
or
Women with diabetes, immunocompromising
conditions (e.g., HIV infection), underlying
immunodeficiency, or immunosuppressive therapy
(e.g., corticosteroids)
Source: Sobel JD, Faro S, Force RW, et al. Vulvovaginal candidiasis:
epidemiologic, diagnostic, and therapeutic considerations.Am J Obstet Gynecol
1998;178:203–11.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 93
US Department of Health and Human Services/Centers for Disease Control and Prevention
after treatment for VVC should be evaluated clinically and
tested. Unnecessary or unapproved use of over-the-counter
preparations is common and can lead to a delay in treatment
of other vulvovaginitis etiologies, which can result in adverse
outcomes. No substantial evidence exists to support using
probiotics or homeopathic medications for treating VVC.
Follow-Up
Follow-up typically is not required. However, women with
persistent or recurrent symptoms after treatment should be
instructed to return for follow-up visits.
Management of Sex Partners
Uncomplicated VVC is not usually acquired through sexual
intercourse, and data do not support treatment of sex partners.
A minority of male sex partners have balanitis, characterized
by erythematous areas on the glans of the penis in conjunction
with pruritus or irritation. These men benefit from treatment
with topical antifungal agents to relieve symptoms.
Special Considerations
Drug Allergy, Intolerance, and Adverse Reactions
Topical agents usually cause no systemic side effects, although
local burning or irritation might occur. Oral azoles occasionally
cause nausea, abdominal pain, and headache. Therapy with the
oral azoles has rarely been associated with abnormal elevations
of liver enzymes. Clinically important interactions can occur
when oral azoles are administered with other drugs (1141).
Complicated Vulvovaginal Candidiasis
Diagnostic Considerations
Vaginal culture or PCR should be obtained from women
with complicated VVC to confirm clinical diagnosis and
identify non–albicans Candida. Candida glabrata does not
form pseudohyphae or hyphae and is not easily recognized
on microscopy. C. albicans azole resistance is becoming more
common in vaginal isolates (1144,1145), and non–albicans
Candida is intrinsically resistant to azoles; therefore, culture
and susceptibility testing should be considered for patients
who remain symptomatic.
Recurrent Vulvovaginal Candidiasis
Recurrent VVC, usually defined as three or more episodes
of symptomatic VVC in <1 year, affects <5% of women but
carries a substantial economic burden (1146). Recurrent VVC
can be either idiopathic or secondary (related to frequent
antibiotic use, diabetes, or other underlying host factors). The
pathogenesis of recurrent VVC is poorly understood, and the
majority of women with recurrent VVC have no apparent
predisposing or underlying conditions. C. glabrata and other
non–albicans Candida species are observed in 10%–20%
of women with recurrent VVC. Conventional antimycotic
therapies are not as effective against these non–albicans yeasts
as against C. albicans.
Treatment
Most episodes of recurrent VVC caused by C. albicans
respond well to short-duration oral or topical azole therapy.
However, to maintain clinical and mycologic control, a longer
duration of initial therapy (e.g., 7–14 days of topical therapy
or a 100-mg, 150-mg, or 200-mg oral dose of fluconazole
every third day for a total of 3 doses [days 1, 4, and 7])
is recommended, to attempt mycologic remission, before
initiating a maintenance antifungal regimen.
Oral fluconazole (i.e., a 100-mg, 150-mg, or 200-mg dose)
weekly for 6 months is the indicated maintenance regimen.
If this regimen is not feasible, topical treatments used
intermittently can also be considered. Suppressive maintenance
therapies are effective at controlling recurrent VVC but are
rarely curative long-term (1147). Because C. albicans azole
resistance is becoming more common, susceptibility tests, if
available, should be obtained among symptomatic patients who
remain culture positive despite maintenance therapy. These
women should be managed in consultation with a specialist.
Severe Vulvovaginal Candidiasis
Severe VVC (i.e., extensive vulvar erythema, edema,
excoriation, and fissure formation) is associated with lower
clinical response rates among patients treated with short courses
of topical or oral therapy. Either 7–14 days of topical azole or
150 mg of fluconazole in two sequential oral doses (second dose
72 hours after initial dose) is recommended.
Non–albicans Vulvovaginal Candidiasis
Because approximately 50% of women with a positive culture
for non–albicans Candida might be minimally symptomatic or
have no symptoms, and because successful treatment is often
difficult, clinicians should make every effort to exclude other
causes of vaginal symptoms for women with non–albicans
yeast (1148). The optimal treatment of non–albicans VVC
remains unknown; however, a longer duration of therapy
(7–14 days) with a nonfluconazole azole regimen (oral or
topical) is recommended. If recurrence occurs, 600 mg of boric
acid in a gelatin capsule administered vaginally once daily for
3 weeks is indicated. This regimen has clinical and mycologic
eradication rates of approximately 70% (1149). If symptoms
recur, referral to a specialist is advised.
Recommendations and Reports
94 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Management of Sex Partners
No data exist to support treating sex partners of patients with
complicated VVC. Therefore, no recommendation can be made.
Special Considerations
Compromised Host
Women with underlying immunodeficiency, those with
poorly controlled diabetes or other immunocompromising
conditions (e.g., HIV), and those receiving immunosuppression
therapy (e.g., corticosteroid treatment) might not respond as
well to short-term therapies. Efforts to correct modifiable
conditions should be made, and more prolonged (i.e.,
7–14 days) conventional treatment is necessary.
Pregnancy
VVC occurs frequently during pregnancy. Only topical
azole therapies, applied for 7 days, are recommended for use
among pregnant women. Epidemiologic studies indicate a
single 150-mg dose of fluconazole might be associated with
spontaneous abortion (1150) and congenital anomalies;
therefore, it should not be used (1151).
HIV Infection
Vaginal Candida colonization rates among women with
HIV infection are higher than among women without HIV
with similar demographic and risk behavior characteristics,
and the colonization rates correlate with increasing severity of
immunosuppression (1152). Symptomatic VVC is also more
frequent among women with HIV infection and similarly
correlates with severity of immunodeficiency (1153). In
addition, among women with HIV, systemic azole exposure
is associated with isolation of non–albicans Candida species
from the vagina.
Treatment for uncomplicated and complicated VVC
among women with HIV infection should not differ from
that for women who do not have HIV. Although long-
term prophylactic therapy with fluconazole 200 mg weekly
has been effective in reducing C. albicans colonization and
symptomatic VVC (1154), this regimen is not recommended
for women with HIV infection in the absence of complicated
VVC (98). Although VVC is associated with increased HIV
seroconversion among HIV-negative women and increased
HIV cervicovaginal levels among women with HIV infection,
the effect of treatment for VVC on HIV acquisition and
transmission remains unknown.
Pelvic Inflammatory Disease
PID comprises a spectrum of inflammatory disorders of
the upper female genital tract, including any combination
of endometritis, salpingitis, tubo-ovarian abscess, and pelvic
peritonitis (11551157). Sexually transmitted organisms,
especially N. gonorrhoeae and C. trachomatis, often are
implicated. Recent studies report that the proportion of
PID cases attributable to N. gonorrhoeae or C. trachomatis is
decreasing; of women who received a diagnosis of acute PID,
approximately 50% have a positive test for either of those
organisms (11581160). Micro-organisms that comprise the
vaginal flora, such as strict and facultative anaerobes (1160)
and G. vaginalis, H. influenzae, enteric gram-negative rods, and
Streptococcus agalactiae, have been associated with PID (1161).
In addition, cytomegalovirus (CMV), T. vaginalis, M. hominis,
and U. urealyticum might be associated with certain PID cases
(1072). Data also indicate that M. genitalium might have a role
in PID pathogenesis (765,928) and might be associated with
milder symptoms (919,923,928), although one study failed
to demonstrate a substantial increase in PID after detection of
M. genitalium in the lower genital tract (925).
Screening and treating sexually active women for chlamydia
and gonorrhea reduces their risk for PID (1162,1163). Although
BV is associated with PID, whether PID incidence can be
reduced by identifying and treating women with BV is unclear
(1161). Whether screening young women for M. genitalium is
associated with a reduction in PID is unknown.
Diagnostic Considerations
Acute PID is difficult to diagnose because of the considerable
variation in symptoms and signs associated with this condition.
Women with PID often have subtle or nonspecific symptoms or
are asymptomatic. Delay in diagnosis and treatment probably
contributes to inflammatory sequelae in the upper genital tract.
Laparoscopy can be used to obtain a more accurate diagnosis
of salpingitis and a more complete bacteriologic diagnosis.
However, this diagnostic tool frequently is not readily available,
and its use is not easily justifiable when symptoms are mild or
vague. Moreover, laparoscopy will not detect endometritis and
might not detect subtle inflammation of the fallopian tubes.
Consequently, a PID diagnosis usually is based on imprecise
clinical findings (11641166).
Data indicate that a clinical diagnosis of symptomatic PID
has a positive predictive value for salpingitis of 65%–90%,
compared with laparoscopy (11671170). The positive
predictive value of a clinical diagnosis of acute PID depends on
the epidemiologic characteristics of the population, with higher
positive predictive values among sexually active young women
(particularly adolescents), women attending STD clinics, and
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 95
US Department of Health and Human Services/Centers for Disease Control and Prevention
those who live in communities with high rates of gonorrhea
or chlamydia. Regardless of positive predictive value, no single
historical, physical, or laboratory finding is both sensitive
and specific for the diagnosis of acute PID. Combinations of
diagnostic findings that improve either sensitivity (i.e., detect
more women who have PID) or specificity (i.e., exclude more
women who do not have PID) do so only at the expense of the
other. For example, requiring two or more findings excludes
more women who do not have PID and reduces the number
of women with PID who are identified.
Episodes of PID often go unrecognized. Although certain
cases are asymptomatic, others are not diagnosed because
the patient or the health care provider do not recognize the
implications of mild or nonspecific symptoms or signs (e.g.,
abnormal bleeding, dyspareunia, and vaginal discharge). Even
women with mild or asymptomatic PID might be at risk for
infertility (1157). Because of the difficulty of diagnosis and
the potential for damage to the reproductive health of women,
health care providers should maintain a low threshold for the
clinical diagnosis of PID (1158). The recommendations for
diagnosing PID are intended to assist health care providers to
recognize when PID should be suspected and when additional
information should be obtained to increase diagnostic certainty.
Diagnosis and management of other causes of lower abdominal
pain (e.g., ectopic pregnancy, acute appendicitis, ovarian cyst,
ovarian torsion, or functional pain) are unlikely to be impaired
by initiating antimicrobial therapy for PID. Presumptive
treatment for PID should be initiated for sexually active
young women and other women at risk for STIs if they are
experiencing pelvic or lower abdominal pain, if no cause for
the illness other than PID can be identified, or if one or more
of the following three minimum clinical criteria are present
on pelvic examination: cervical motion tenderness, uterine
tenderness, or adnexal tenderness.
More specific criteria for diagnosing PID include endometrial
biopsy with histopathologic evidence of endometritis;
transvaginal sonography or magnetic resonance imaging
techniques demonstrating thickened, fluid-filled tubes with or
without free pelvic fluid or tubo-ovarian complex, or Doppler
studies indicating pelvic infection (e.g., tubal hyperemia);
and laparoscopic findings consistent with PID. A diagnostic
evaluation that includes some of these more extensive
procedures might be warranted in certain cases. Endometrial
biopsy is warranted for women undergoing laparoscopy who
do not have visual evidence of salpingitis because endometritis
is the only sign of PID for certain women.
Requiring that all three minimum criteria be present
before the initiation of empiric treatment can result in
insufficient sensitivity for a PID diagnosis. After deciding
whether to initiate empiric treatment, clinicians should also
consider the risk profile for STIs. More elaborate diagnostic
evaluation frequently is needed because incorrect diagnosis
and management of PID might cause unnecessary morbidity.
For example, the presence of signs of lower genital tract
inflammation (predominance of leukocytes in vaginal
secretions, cervical discharge, or cervical friability), in addition
to one of the three minimum criteria, increases the specificity of
the diagnosis. One or more of the following additional criteria
can be used to enhance the specificity of the minimum clinical
criteria and support a PID diagnosis:
Oral temperature >38.3°C (>101°F)
Abnormal cervical mucopurulent discharge or cervical
friability
Presence of abundant numbers of WBCs on saline
microscopy of vaginal fluid
Elevated erythrocyte sedimentation rate
Elevated C-reactive protein
Laboratory documentation of cervical infection with
N. gonorrhoeae or C. trachomatis
The majority of women with PID have either mucopurulent
cervical discharge or evidence of WBCs on a microscopic
evaluation of a saline preparation of vaginal fluid (i.e., wet
prep). If the cervical discharge appears normal and no WBCs
are observed on the wet prep of vaginal fluid, a PID diagnosis
is unlikely, and alternative causes of pain should be considered.
A wet prep of vaginal fluid also can detect the presence of
concomitant infections (e.g., BV or trichomoniasis).
Treatment
PID treatment regimens should provide empiric, broad-
spectrum coverage of likely pathogens. Multiple parenteral and
oral antimicrobial regimens have been effective in achieving
clinical and microbiologic cure in randomized clinical trials
with short-term follow-up (11711173). However, only
a limited number of studies have assessed and compared
these regimens with regard to infection elimination in the
endometrium and fallopian tubes or determined the incidence
of long-term complications (e.g., tubal infertility and ectopic
pregnancy) after antimicrobial regimens (1159,1164,1174).
The optimal treatment regimen and long-term outcome of
early treatment of women with subclinical PID are unknown.
All regimens used to treat PID should also be effective against
N. gonorrhoeae and C. trachomatis because negative endocervical
screening for these organisms does not rule out upper genital
tract infection. Anaerobic bacteria have been isolated from the
upper genital tract of women who have PID, and data from in
vitro studies have revealed that some anaerobes (e.g., Bacteroides
fragilis) can cause tubal and epithelial destruction. BV is often
present among women who have PID (22,1160,1161,1175).
Recommendations and Reports
96 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Addition of metronidazole to IM or oral PID regimens more
effectively eradicates anaerobic organisms from the upper
genital tract (1160). Until treatment regimens that do not
cover anaerobic microbes have been demonstrated to prevent
long-term sequelae (e.g., infertility and ectopic pregnancy)
as successfully as the regimens that are effective against these
microbes, using regimens with anaerobic activity should
be considered. Treatment should be initiated as soon as the
presumptive diagnosis has been made because prevention of
long-term sequelae is dependent on early administration of
recommended antimicrobials. For women with PID of mild or
moderate clinical severity, parenteral and oral regimens appear
to have similar efficacy. The decision of whether hospitalization
is necessary should be based on provider judgment and whether
the woman meets any of the following criteria:
Surgical emergencies (e.g., appendicitis) cannot be
excluded
Tubo-ovarian abscess
Pregnancy
Severe illness, nausea and vomiting, or oral temperature
>38.5°C (101°F)
Unable to follow or tolerate an outpatient oral regimen
No clinical response to oral antimicrobial therapy
No evidence is available to indicate that adolescents have
improved outcomes from hospitalization for treatment of PID,
and the clinical response to outpatient treatment is similar
among younger and older women. The decision to hospitalize
adolescents with acute PID should be based on the same criteria
used for older women.
Parenteral Treatment
Randomized trials have demonstrated the efficacy of
parenteral regimens (1160,1171,1172,1176). Clinical
experience should guide decisions regarding transition to oral
therapy, which usually can be initiated within 24–48 hours of
clinical improvement. For women with tubo-ovarian abscesses,
>24 hours of inpatient observation is recommended.
Recommended Parenteral Regimens for Pelvic Inflammatory
Disease
Ceftriaxone 1 g by every 24 hours
plus
Doxycycline 100 mg orally or IV every 12 hours
plus
Metronidazole 500 mg orally or IV every 12 hours
or
Cefotetan 2 g IV every 12 hours
plus
Doxycycline 100 mg orally or IV every 12 hours
or
Cefoxitin 2 g IV every 6 hours
plus
Doxycycline 100 mg orally or IV every 12 hours
Because of the pain associated with IV infusion, doxycycline
should be administered orally when possible. Oral and IV
administration of doxycycline and metronidazole provide
similar bioavailability. Oral metronidazole is well absorbed
and can be considered instead of IV for women without severe
illness or tubo-ovarian abscess when possible. After clinical
improvement with parenteral therapy, transition to oral therapy
with doxycycline 100 mg 2 times/day and metronidazole
500 mg 2 times/day is recommended to complete 14 days of
antimicrobial therapy.
Alternative Parenteral Regimens
Only limited data are available to support using other
parenteral second- or third- generation cephalosporins (e.g.,
ceftizoxime or cefotaxime). Because these cephalosporins are
less active than cefotetan or cefoxitin against anaerobic bacteria,
the addition of metronidazole should be considered.
Ampicillin-sulbactam plus doxycycline has been investigated
in at least one clinical trial and has broad-spectrum coverage
(1177). Ampicillin-sulbactam plus doxycycline is effective
against C. trachomatis, N. gonorrhoeae, and anaerobes for
women with tubo-ovarian abscess. Another trial demonstrated
short-term clinical cure rates with azithromycin monotherapy
or combined with metronidazole (1178).
When using the clindamycin and gentamicin alternative
parenteral regimen, women with clinical improvement after
24–28 hours can be transitioned to clindamycin (450 mg orally
4 times/day) or doxycycline (100 mg orally 2 times/day) to
complete the 14-day therapy. However, when tubo-ovarian
abscess is present, clindamycin (450 mg orally 4 times/day) or
metronidazole (500 mg orally 2 times/day) should be used to
complete 14 days of therapy with oral doxycycline to provide
more effective anaerobic coverage.
Alternative Parenteral Regimens
Ampicillin-sulbactam 3 g IV every 6 hours
plus
Doxycycline 100 mg orally or IV every 12 hours
or
Clindamycin 900 mg IV every 8 hours
plus
Gentamicin loading dose IV or IM (2 mg/kg body weight), followed by a
maintenance dose (1.5 mg/kg body weight) every 8 hours; single daily
dosing (3–5 mg/kg body weight) can be substituted
Intramuscular or Oral Treatment
IM or oral therapy can be considered for women with mild-
to-moderate acute PID because the clinical outcomes among
women treated with these regimens are similar to those treated
with IV therapy (1158). Women who do not respond to IM or
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 97
US Department of Health and Human Services/Centers for Disease Control and Prevention
oral therapy within 72 hours should be reevaluated to confirm
the diagnosis and be administered therapy IV.
Recommended Intramuscular or Oral Regimens for Pelvic
Inflammatory Disease
Ceftriaxone 500 mg* IM in a single dose
plus
Doxycycline 100 mg orally 2 times/day for 14 days with metronidazole
500 mg orally 2 times/day for 14 days
or
Cefoxitin 2 g IM in a single dose and probenecid 1 g orally administered
concurrently in a single dose
plus
Doxycycline 100 mg orally 2 times/day for 14 days with metronidazole
500 mg orally 2 times/day for 14 days
or
Other parenteral third-generation cephalosporin (e.g., ceftizoxime or
cefotaxime)
plus
Doxycycline 100 mg orally 2 times/day for 14 days with metronidazole
500 mg orally 2 times/day for 14 days
* For persons weighing ≥150 kg, 1 g of ceftriaxone should be administered.
These regimens provide coverage against frequent etiologic
agents of PID; however, the optimal choice of a cephalosporin
is unclear. Cefoxitin, a second-generation cephalosporin, has
better anaerobic coverage than ceftriaxone, and, in combination
with probenecid and doxycycline, has been effective in short-
term clinical response among women with PID. Ceftriaxone
has better coverage against N. gonorrhoeae. The addition of
metronidazole to these regimens provides extended coverage
against anaerobic organisms and will also effectively treat BV,
which is frequently associated with PID.
Alternative Intramuscular or Oral Regimens
No data have been published regarding use of oral
cephalosporins for treating PID. As a result of the emergence
of quinolone-resistant N. gonorrhoeae, regimens that include
a quinolone agent are not recommended for PID treatment.
However, if the patient has cephalosporin allergy, the
community prevalence and individual risk for gonorrhea
are low, and follow-up is likely, alternative therapy can be
considered. Use of either levofloxacin 500 mg orally once daily
or moxifloxacin 400 mg orally once daily with metronidazole
500 mg orally 2 times/day for 14 days (11791181) or
azithromycin 500 mg IV daily for 1–2 doses, followed by
250 mg orally daily in combination with metronidazole
500 mg 2 times/day for 12–14 days (1178), can be considered.
Moxifloxacin is the preferred quinolone antimicrobial for
M. genitalium infections; however, the importance of providing
coverage for M. genitalium is unknown. Diagnostic tests for
gonorrhea should be obtained before starting therapy, and
persons should be managed as follows:
If a culture for gonorrhea is positive, treatment should be
based on results of antimicrobial susceptibility testing.
If the isolate is determined to be quinolone-resistant
N. gonorrhoeae or if antimicrobial susceptibility cannot be
assessed (e.g., if only NAAT testing is available), consultation
with an infectious disease specialist is recommended.
Other Management Considerations
To minimize disease transmission, women should be
instructed to abstain from sexual intercourse until therapy is
complete, symptoms have resolved, and sex partners have been
treated (see Chlamydial Infections; Gonococcal Infections).
All women who receive a diagnosis of PID should be tested
for gonorrhea, chlamydia, HIV, and syphilis. The value of
testing women with PID for M. genitalium is unknown (see
Mycoplasma genitalium). All contraceptive methods can be
continued during treatment.
Follow-Up
Women should demonstrate clinical improvement (e.g.,
defervescence; reduction in direct or rebound abdominal
tenderness; and reduction in uterine, adnexal, and cervical
motion tenderness) <3 days after therapy initiation. If no
clinical improvement has occurred <72 hours after outpatient
IM or oral therapy, then hospitalization, assessment of the
antimicrobial regimen, and additional diagnostics, including
consideration of diagnostic laparoscopy for alternative
diagnoses, are recommended. All women who have received a
diagnosis of chlamydial or gonococcal PID should be retested
3 months after treatment, regardless of whether their sex
partners have been treated (753). If retesting at 3 months is
not possible, these women should be retested whenever they
next seek medical care <12 months after treatment.
Management of Sex Partners
Persons who have had sexual contact with a partner with
PID during the 60 days preceding symptom onset should be
evaluated, tested, and presumptively treated for chlamydia
and gonorrhea, regardless of the PID etiology or pathogens
isolated. If the last sexual intercourse was >60 days before
symptom onset or diagnosis, the most recent sex partner should
be treated. Sex partners of persons who have PID caused by
C. trachomatis or N. gonorrhoeae frequently are asymptomatic.
Arrangements should be made to link sex partners to care. If
linkage is delayed or unlikely, EPT is an alternative approach
to treating sex partners who have chlamydial or gonococcal
infection (125,126) (see Partner Services). Partners should
be instructed to abstain from sexual intercourse until they
Recommendations and Reports
98 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
and their sex partners have been treated (i.e., until therapy is
completed and symptoms have resolved, if originally present).
Special Considerations
Drug Allergy, Intolerance, and Adverse Reactions
The risk for penicillin cross-reactivity is highest with first-
generation cephalosporins but is negligible between the
majority of second-generation (e.g., cefoxitin) and all third-
generation (e.g., ceftriaxone) cephalosporins (619,631,653,656)
(see Management of Persons Who Have a History of
Penicillin Allergy).
Pregnancy
Pregnant women suspected of having PID are at high risk
for maternal morbidity and preterm delivery. These women
should be hospitalized and treated with IV antimicrobials in
consultation with an infectious disease specialist.
HIV Infection
Differences in PID clinical manifestations among women
with HIV infection and those without have not been well
delineated (1182). In early observational studies, women
with HIV infection and PID were more likely to require
surgical intervention. More comprehensive observational
and controlled studies have demonstrated that women with
HIV infection and PID coinfection have similar symptoms,
compared with women without HIV (11831185), except
they are more likely to have a tubo-ovarian abscess. Women
with HIV responded equally well to recommended parenteral
and IM or oral antibiotic regimens as women without HIV.
The microbiologic findings for women with HIV and women
without HIV were similar, except women with HIV had
higher rates of concomitant M. hominis and streptococcal
infections. These data are insufficient for determining whether
women with HIV infection and PID coinfection require
more aggressive management (e.g., hospitalization or IV
antimicrobial regimens).
Intrauterine Devices
IUDs are one of the most effective contraceptive methods.
Copper-containing and levonorgestrel-releasing IUDs are
available in the United States. The risk for PID associated with
IUD use is primarily confined to the first 3 weeks after insertion
(11861188). If an IUD user receives a diagnosis of PID,
the IUD does not need to be removed (59,1189). However,
the woman should receive treatment according to these
recommendations and should have close clinical follow-up.
If no clinical improvement occurs within 48–72 hours of
initiating treatment, providers should consider removing
the IUD. A systematic review of evidence demonstrated that
treatment outcomes did not differ between women with PID
who retained the IUD and those who had the IUD removed
(1190). These studies primarily included women using copper-
containing or other nonhormonal IUDs. No studies are
available regarding treatment outcomes among women using
levonorgestrel-releasing IUDs.
Epididymitis
Acute epididymitis is a clinical syndrome causing pain,
swelling, and inflammation of the epididymis and lasting
<6 weeks (1191). Sometimes a testicle is also involved, a
condition referred to as epididymo-orchitis. A high index
of suspicion for spermatic cord (testicular) torsion should
be maintained among men who have a sudden onset of
symptoms associated with epididymitis because this condition
is a surgical emergency.
Acute epididymitis can be caused by STIs (e.g., C. trachomatis,
N. gonorrhoeae, or M. genitalium) or enteric organisms (i.e.,
Escherichia coli) (1192). Acute epididymitis caused by an
STI is usually accompanied by urethritis, which is frequently
asymptomatic. Acute epididymitis caused by sexually
transmitted enteric organisms might also occur among men
who are the insertive partner during anal sex. Nonsexually
transmitted acute epididymitis caused by genitourinary
pathogens typically occurs with bacteriuria secondary to
bladder outlet obstruction (e.g., benign prostatic hyperplasia)
(1193). Among older men, nonsexually transmitted
acute epididymitis is also associated with prostate biopsy,
urinary tract instrumentation or surgery, systemic disease,
or immunosuppression. Uncommon infectious causes of
nonsexually transmitted acute epididymitis (e.g., Fournier’s
gangrene) should be managed in consultation with a urologist.
Chronic epididymitis is characterized by a ≥6-week history
of symptoms of discomfort or pain in the scrotum, testicle, or
epididymis. Chronic infectious epididymitis is most frequently
observed with conditions associated with a granulomatous
reaction. Mycobacterium tuberculosis (TB) is the most common
granulomatous disease affecting the epididymis and should be
suspected, especially among men with a known history of or recent
exposure to TB. The differential diagnosis of chronic noninfectious
epididymitis, sometimes termed orchialgia or epididymalgia, is
broad (e.g., trauma, cancer, autoimmune conditions, or idiopathic
conditions). Men with this diagnosis should be referred to a
urologist for clinical management (1191,1192).
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 99
US Department of Health and Human Services/Centers for Disease Control and Prevention
Diagnostic Considerations
Men who have acute epididymitis typically have unilateral
testicular pain and tenderness, hydrocele, and palpable swelling
of the epididymis. Although inflammation and swelling
usually begin in the tail of the epididymis, it can spread to
the rest of the epididymis and testicle. The spermatic cord
is usually tender and swollen. Spermatic cord (testicular)
torsion, a surgical emergency, should be considered in all cases;
however, it occurs more frequently among adolescents and
men without evidence of inflammation or infection. For men
with severe unilateral pain with sudden onset, those whose
test results do not support a diagnosis of urethritis or urinary
tract infection, or for whom diagnosis of acute epididymitis is
questionable, immediate referral to a urologist for evaluation
for testicular torsion is vital because testicular viability might
be compromised.
Bilateral symptoms should increase suspicion of other causes
of testicular pain. Radionuclide scanning of the scrotum is the
most accurate method for diagnosing epididymitis but it is
not routinely available. Ultrasound should be used primarily
for ruling out torsion of the spermatic cord in cases of acute,
unilateral, painful scrotal swelling. However, because partial
spermatic cord torsion can mimic epididymitis on scrotal
ultrasound, differentiation between spermatic cord torsion
and epididymitis when torsion is not ruled out by ultrasound
should be made on the basis of clinical evaluation. Although
ultrasound can demonstrate epididymal hyperemia and
swelling associated with epididymitis, it provides minimal
diagnostic usefulness for men with a clinical presentation
consistent with epididymitis. A negative ultrasound does
not rule out epididymitis and thus does not alter clinical
management. Ultrasound should be reserved for men if torsion
of the spermatic cord is suspected or for those with scrotal pain
who cannot receive an accurate diagnosis by history, physical
examination, and objective laboratory findings.
All suspected cases of acute epididymitis should be evaluated
for objective evidence of inflammation by one of the following
POC tests:
Gram, MB, or GV stain of urethral secretions demonstrating
≥2 WBCs per oil immersion field (737) (see Urethritis).
These stains are preferred POC diagnostic tests for
evaluating urethritis because they are highly sensitive and
specific for documenting both urethral inflammation and
presence or absence of gonococcal infection. Gonococcal
infection is established by documenting the presence of
WBC-containing intracellular gram-negative or purple
diplococci on urethral Gram, MB, or GV stain, respectively.
Positive leukocyte esterase test on first-void urine.
Microscopic examination of sediment from a spun first-
void urine demonstrating ≥10 WBCs/HPF.
All suspected cases of acute epididymitis should be tested
for C. trachomatis and N. gonorrhoeae by NAAT. Urine is
the preferred specimen for NAAT for men (553). Urine
cultures for chlamydial and gonococcal epididymitis are
insensitive and are not recommended. Urine bacterial
cultures should also be performed for all men to evaluate for
the presence of genitourinary organisms and to determine
antibiotic susceptibility.
Treatment
To prevent complications and transmission of STIs,
presumptive therapy for all sexually active men is indicated
at the time of the visit before all laboratory test results are
available. Selection of presumptive therapy is based on risk for
chlamydial and gonococcal infections or enteric organisms.
Treatment goals for acute epididymitis are 1) microbiologic
infection cure, 2) improvement of signs and symptoms,
3) prevention of transmission of chlamydia and gonorrhea to
others, and 4) decreased potential for chlamydial or gonococcal
epididymitis complications (e.g., infertility or chronic pain).
Although the majority of men with acute epididymitis can
be treated on an outpatient basis, referral to a specialist and
hospitalization should be considered when severe pain or fever
indicates other diagnoses (e.g., torsion, testicular infarction,
abscess, or necrotizing fasciitis) or when men are unable to
comply with an antimicrobial regimen. Age, history of diabetes,
fever, and elevated C-reactive protein can indicate more severe
disease requiring hospitalization (1193).
Recommended Regimens for Epididymitis
For acute epididymitis most likely caused by chlamydia or gonorrhea:
Ceftriaxone 500 mg* IM in a single dose
plus
Doxycycline 100 mg orally 2 times/day for 10 days
For acute epididymitis most likely caused by chlamydia, gonorrhea,
or enteric organisms (men who practice insertive anal sex):
Ceftriaxone 500 mg* IM in a single dose
plus
Levofloxacin 500 mg orally once daily for 10 days
For acute epididymitis most likely caused by enteric organisms only:
Levofloxacin 500 mg orally once daily for 10 days
* For persons weighing ≥150 kg, 1 g of ceftriaxone should be administered.
Levofloxacin monotherapy should be considered if the
infection is most likely caused by enteric organisms only, and
gonorrhea has been ruled out by Gram, MB, or GV stain. This
includes men who have undergone prostate biopsy, vasectomy,
and other urinary tract instrumentation procedures. Treatment
Recommendations and Reports
100 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
should be guided by bacterial cultures and antimicrobial
susceptibilities. As an adjunct to therapy, bed rest, scrotal
elevation, and nonsteroidal anti-inflammatory drugs are
recommended until fever and local inflammation have
subsided. Complete resolution of discomfort might not occur
for a few weeks after completion of the antibiotic regimen.
Other Management Considerations
Men who have acute epididymitis confirmed or suspected to
be caused by N. gonorrhoeae or C. trachomatis should be advised
to abstain from sexual intercourse until they and their partners
have been treated and symptoms have resolved. All men with
acute epididymitis should be tested for HIV and syphilis.
Follow-Up
Men should be instructed to return to their health care
providers if their symptoms do not improve <72 hours after
treatment. Signs and symptoms of epididymitis that do not
subside in <3 days require reevaluation of the diagnosis and
therapy. Men who experience swelling and tenderness that
persist after completion of antimicrobial therapy should be
evaluated for alternative diagnoses, including tumor, abscess,
infarction, testicular cancer, TB, and fungal epididymitis.
Management of Sex Partners
Men who have acute sexually transmitted epididymitis
confirmed or suspected to be caused by N. gonorrhoeae or
C. trachomatis should be instructed to refer all sex partners
during the previous 60 days before symptom onset for
evaluation, testing, and presumptive treatment (see Chlamydial
Infections; Gonococcal Infections). If the last sexual intercourse
was >60 days before onset of symptoms or diagnosis, the
most recent sex partner should be evaluated and treated.
Arrangements should be made to link sex partners to care. EPT
is an effective strategy for treating sex partners of men who
have or are suspected of having chlamydia or gonorrhea for
whom linkage to care is anticipated to be delayed (125,126)
(see Partner Services). Partners should be instructed to abstain
from sexual intercourse until they and their sex partners are
treated and symptoms have resolved.
Special Considerations
Drug Allergy, Intolerance, and Adverse Reactions
The risk for penicillin cross-reactivity is negligible between all
third-generation cephalosporins (e.g., ceftriaxone) (658,681)
(see Management of Persons Who Have a History of Penicillin
Allergy). Alternative regimens have not been studied; therefore,
clinicians should consult an infectious disease specialist if such
regimens are required.
HIV Infection
Men with HIV infection who have uncomplicated acute
epididymitis should receive the same treatment regimen as
those who do not have HIV. Other etiologic agents have
been implicated in acute epididymitis among men with HIV,
including CMV, salmonella, toxoplasmosis, U. urealyticum,
Corynebacterium species, Mycoplasma species, and Mima
polymorpha (1192).
Human Papillomavirus Infections
Approximately 150 types of HPV have been identified, at
least 40 of which infect the genital area (1194). The majority
of HPV infections are self-limited and are asymptomatic or
unrecognized. Sexually active persons are usually exposed to
HPV during their lifetime (838,1195,1196). Oncogenic,
high-risk HPV infection (e.g., HPV types 16 and 18) causes the
majority of cervical, penile, vulvar, vaginal, anal, and oropharyngeal
cancers and precancers (1197), whereas other HPV infection
(e.g., HPV types 6 and 11) causes genital warts and recurrent
respiratory papillomatosis. Persistent oncogenic HPV infection
is the strongest risk factor for development of HPV-attributable
precancers and cancers. A substantial proportion of cancers and
anogenital warts are attributable to HPV in the United States.
An estimated 34,800 new HPV-attributable cancers occurred
every year during 2012–2016 (1198). Before HPV vaccines were
introduced, approximately 355,000 new cases of anogenital warts
occurred every year (1199).
Prevention
HPV Vaccines
Three HPV vaccines are licensed in the United States:
Ceravrix, a 2-valent vaccine (2vHPV) that targets HPV types
16 and 18; Gardasil, a 4-valent vaccine (4vHPV) that targets
HPV types 6, 11, 16, and 18; and Gardasil 9, a 9-valent
vaccine (9vHPV) that targets HPV types 6, 11, 16, 18, 31,
33, 45, 52, and 58. Types 16 and 18 account for 66% of all
cervical cancers, whereas the five additional types targeted by
the 9-valent vaccine account for 15%. Types 6 and 11 cause
>90% of genital warts. Only 9vHPV vaccine is available in
the United States.
ACIP recommendations for HPV vaccination (https://www.
cdc.gov/vaccines/hcp/acip-recs/vacc-specific/hpv.html) include
the following:
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 101
US Department of Health and Human Services/Centers for Disease Control and Prevention
Routine HPV vaccination for all adolescents at age 11 or
12 years.
Administering vaccine starting at age 9 years.
Catch-up vaccination through age 26 years for those not
vaccinated previously.
Not using HPV vaccination for all adults aged >26 years.
Instead, shared clinical decision-making between a patient
and a provider regarding HPV vaccination is recommended
for certain adults aged 27–45 years not vaccinated previously.
A 2-dose vaccine schedule (at 0- and 6–12-month
intervals) is recommended for persons who initiate
vaccination before their 15th birthday.
A 3-dose vaccine schedule (at 0-, 1–2-, and 6-month
intervals) for immunocompromised persons regardless of
age of initiation.
HPV vaccines are not recommended for use in pregnant
women. HPV vaccines can be administered regardless of
history of anogenital warts, abnormal Pap test or HPV test, or
anogenital precancer. Women who have received HPV vaccine
should continue routine cervical cancer screening (see Cervical
Cancer). HPV vaccine is available for eligible children and
adolescents aged <19 years through the Vaccines for Children
(VFC) program (additional information is available at https://
www.cdc.gov/vaccines/programs/vfc/index.html or by calling
CDC INFO 800-232-4636). For uninsured persons aged
<19 years, patient assistance programs are available from the
vaccine manufacturers. Prelicensure and postlicensure safety
evaluations have determined that the vaccine is well tolerated.
With >120 million doses of HPV vaccines distributed in the
United States, robust data demonstrate that HPV vaccines are
safe (https://www.cdc.gov/vaccinesafety). Impact-monitoring
studies in the United States have demonstrated reductions
of genital warts as well as the HPV types contained within
the quadrivalent vaccine (12001203). Settings that provide
STI services should either administer the vaccine to eligible
clients within the routine and catch-up age groups through
age 26 years who have not started or completed the vaccine
series, or link these persons to another facility equipped to
provide the vaccine. Clinicians providing services to children,
adolescents, and young adults should be knowledgeable about
HPV and the vaccine (https://www.cdc.gov/vaccines/who/
teens/for-hcp/hpv-resources.html). HPV vaccination has not
been associated with initiation of sexual activity or sexual risk
behaviors (1204,1205).
Abstaining from sexual activity is the most reliable method
for preventing genital HPV infection. Persons can decrease
their chances of infection by practicing consistent and correct
condom use and limiting their number of sex partners. Although
these interventions might not fully protect against HPV, they
can decrease the chances of HPV acquisition and transmission.
Diagnostic Considerations
HPV tests are available for detecting oncogenic types of HPV
infection and are used in the context of cervical cancer screening
and management or follow-up of abnormal cervical cytology or
histology (see Cervical Cancer). These tests should not be used
for male partners of women with HPV or women aged <25 years,
for diagnosis of genital warts, or as a general STI test.
Application of 3%–5% acetic acid, which might cause
affected areas to turn white, has been used by certain providers
to detect genital mucosa infected with HPV. The routine use
of this procedure to detect mucosal changes attributed to
HPV infection is not recommended because the results do
not influence clinical management.
Treatment
Treatment is directed to the macroscopic (e.g., genital warts)
or pathologic precancerous lesions caused by HPV. Subclinical
genital HPV infection typically clears spontaneously; therefore,
specific antiviral therapy is not recommended to eradicate HPV
infection. Precancerous lesions are detected through cervical
cancer screening; HPV-related precancer should be managed
on the basis of existing guidance (see Cervical Cancer).
Counseling
Key Messages for Persons with Human
Papillomavirus Infection
When counseling persons with anogenital HPV infection,
the provider should discuss the following:
Anogenital HPV infection is common. It usually infects
the anogenital area but can infect other areas, including
the mouth and throat. The majority of sexually active
persons get HPV at some time during their lifetime,
although most never know it.
Partners tend to share HPV, and it is not possible to
determine which partner transmitted the original
infection. Having HPV does not mean that a person or
his or her partner is having sex outside the relationship.
Persons who acquire HPV usually clear the infection
spontaneously, meaning that HPV becomes undetectable
with no associated health problems.
If HPV infection persists, genital warts, precancers, and
cancers of the cervix, anus, penis, vulva, vagina, head, or
neck might develop.
Discussion of tobacco use, and provision of cessation
counseling, is important because of its contribution to the
progression of precancer and cancer.
The types of HPV that cause genital warts are different
from the types that can cause cancer.
Recommendations and Reports
102 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Many types of HPV are sexually transmitted through
anogenital contact, mainly during vaginal and anal sex. HPV
also might be transmitted during oral sex and genital-to-genital
contact without penetration. In rare cases, a pregnant woman
can transmit HPV to an infant during delivery.
Treatments are available for the conditions caused by HPV
but not for the virus itself.
Having HPV does not make it harder for a woman to get
pregnant or carry a pregnancy to term. However, certain
precancers or cancers that HPV can cause, and the surgical
procedures needed to treat them, can affect a womans
ability to get pregnant or carry a pregnancy to term.
No HPV test can determine which HPV infection will
become undetectable and which will persist or progress to
disease. However, in certain circumstances, HPV tests can
determine whether a woman is at increased risk for cervical
cancer. These tests are not for detecting other HPV-related
problems, nor are they useful for women aged<25 years
or men of any age.
Prevention
Three HPV vaccines can prevent diseases and cancers
caused by HPV. The 2vHPV, 4vHPV, and 9vHPV vaccines
protect against the majority of cervical cancer cases,
although the 4vHPV and 9vHPV vaccines also protect
against the majority of genital warts. Only 9vHPV vaccine
is available in the United States. HPV vaccines are safe
and effective and are recommended routinely for
adolescents aged 11–12 years. Catch-up vaccination is also
recommended for older adolescents and young adults
through age 26 years (https://www.cdc.gov/hpv/hcp/
index.html). Shared clinical decision-making is
recommended regarding HPV vaccination for certain
adults aged 27–45 years who are not adequately vaccinated
per guidance (https://www.cdc.gov/mmwr/volumes/68/
wr/pdfs/mm6832a3-H.pdf).
Condoms used consistently and correctly can lower the
chances of acquiring and transmitting HPV and
developing HPV-related diseases (e.g., genital warts or
cervical cancer). However, because HPV can infect areas
not covered by a condom, condoms might not fully protect
against HPV.
Limiting the number of sex partners can reduce the risk
for HPV. However, even persons with only one lifetime
sex partner can get HPV.
Abstaining from sexual activity is the most reliable method
for preventing genital HPV infection.
Anogenital Warts
Anogenital warts are a common disease, and 90% are caused
by nononcogenic HPV types 6 or 11. These types can be
commonly identified before or at the same time anogenital
warts are detected (1206). HPV types 16, 18, 31, 33, and 35
also are occasionally identified in anogenital warts (usually
as infections with HPV 6 or 11) and can be associated with
foci of high-grade squamous intraepithelial lesion (HSIL),
particularly among persons who have HIV infection. In
addition to anogenital warts, HPV types 6 and 11 have been
associated with conjunctival, nasal, oral, and laryngeal warts.
Anogenital warts are usually asymptomatic; however,
depending on the size and anatomic location, they can
be painful or pruritic. They are usually flat, papular, or
pedunculated growths on the genital mucosa. Anogenital warts
occur commonly at certain anatomic sites, including around
the vaginal introitus, under the foreskin of the uncircumcised
penis, and on the shaft of the circumcised penis. Warts can
also occur at multiple sites in the anogenital epithelium or
within the anogenital tract (e.g., cervix, vagina, urethra,
perineum, perianal skin, anus, or scrotum). Intra-anal warts
are observed predominantly in persons who have had receptive
anal intercourse; however, they also can occur among men and
women who have not had a history of anal sexual contact.
Prevention
Anogenital warts have decreased among adolescents,
young women, and heterosexual men with use of HPV
vaccination in multiple countries, including the United States
(1203,12071216).
Diagnostic Considerations
Diagnosis of anogenital warts is usually made by visual
inspection but can be confirmed by biopsy, which is indicated
if lesions are atypical (e.g., pigmented, indurated, affixed to
underlying tissue, bleeding, or ulcerated lesions). Biopsy might
also be indicated in the following circumstances, particularly
if the patient is immunocompromised (including those with
HIV infection): the diagnosis is uncertain, the lesions do not
respond to standard therapy, or the disease worsens during
therapy. HPV testing is not recommended for anogenital wart
diagnosis because test results are not confirmatory and do not
guide genital wart management. Some anogenital lesions can
resemble anogenital warts (condyloma accuminata), but do
not respond to anogenital wart treatment. Condyloma lata,
a manifestation of secondary syphilis, can be diagnosed by
serologic tests or through direct detection from serous fluid
from the lesions (see Syphilis, Diagnostic Considerations).
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 103
US Department of Health and Human Services/Centers for Disease Control and Prevention
Treatment
The aim of treatment is removal of the warts and amelioration
of symptoms, if present. The appearance of warts also can result
in considerable psychosocial distress, and removal can relieve
cosmetic concerns. For most patients, treatment results in
resolution of the warts. If left untreated, anogenital warts can
resolve spontaneously, remain unchanged, or increase in size or
number. Because warts might spontaneously resolve in <1 year,
an acceptable alternative for certain persons is to forego treatment
and wait for spontaneous resolution. Available therapies for
anogenital warts might reduce, but probably do not eradicate,
HPV infectivity. Whether reduction in HPV viral DNA resulting
from treatment reduces future transmission remains unknown.
Treatment of anogenital warts should be guided by wart size,
number, and anatomic site; patient preference; cost of treatment;
convenience; adverse effects; and provider experience. No
definitive evidence indicates that any one recommended treatment
is superior to another, and no single treatment is ideal for all
patients or all warts. Shared clinical decision-making between
a patient and a provider regarding treatment algorithms has
been associated with improved clinical outcomes and should be
encouraged. Because all available treatments have shortcomings,
clinicians sometimes use combination therapy (e.g., provider-
administered cryotherapy with patient-applied topical therapy
between visits to the provider). However, limited data exist
regarding the efficacy or risk for complications associated with
combination therapy. Treatment regimens are classified as either
patient-applied or provider-administered modalities. Patient-
applied modalities are preferred by certain persons because they
can be administered in the privacy of their home. To ensure
that patient-applied modalities are effective, instructions should
be provided to patients while in the clinic, and all anogenital
warts should be accessible and identified during the clinic visit.
Follow-up visits after weeks of therapy enable providers to answer
any questions about use of the medication, address any side effects
experienced, and facilitate assessment of the response to treatment.
Recommended Regimens for External Anogenital Warts (i.e.,
Penis, Groin, Scrotum, Vulva, Perineum, External Anus, or
Perianus)*
Patient-applied: Imiquimod 3.75% or 5% cream
or
Podofilox 0.5% solution or gel
or
Sinecatechins 15% ointment
Provider-administered: Cryotherapy with liquid nitrogen or cryoprobe
or
Surgical removal by tangential scissor excision, tangential shave
excision, curettage, laser, or electrosurgery
or
Trichloroacetic acid (TCA) or bichloroacetic acid (BCA)
80%–90% solution
* Persons with external anal or perianal warts might also have intra-anal
warts. Thus, persons with external anal warts might benefit from an
inspection of the anal canal by digital examination, standard anoscopy,
or high-resolution anoscopy.
Might weaken condoms and vaginal diaphragms.
Imiquimod is a patient-applied, topically active immune
enhancer that stimulates production of interferon and other
cytokines. Imiquimod 5% cream should be applied once
at bedtime, 3 times/week for <16 weeks (1217). Similarly,
imiquimod 3.75% cream should be applied once at bedtime
every night for <8 weeks (1218). With either formulation, the
treatment area should be washed with soap and water 6–10 hours
after the application. Local inflammatory reactions, including
redness, irritation, induration, ulceration or erosion, and vesicles
might occur with using imiquimod, and hypopigmentation has
also been described (1219). Limited case reports demonstrate
an association between treatment with imiquimod cream and
worsened inflammatory or autoimmune skin diseases (e.g.,
psoriasis, vitiligo, or lichenoid dermatoses) (12201222). Data
from studies of human participants are limited regarding use of
imiquimod during pregnancy; however, animal data indicate
that this therapy poses low risk (431).
Podofilox (podophyllotoxin) is a patient-applied antimitotic
drug that causes wart necrosis. Podofilox solution (using a
cotton swab) or podofilox gel (using a finger) should be applied
to anogenital warts 2 times/day for 3 days, followed by 4 days
of no therapy. This cycle can be repeated, as necessary, for up
to four cycles. The total wart area treated should not exceed
10 cm
2
, and the total volume of podofilox should be limited to
0.5 mL/day. If possible, the health care provider should apply the
initial treatment to demonstrate proper application technique
and identify which warts should be treated. Mild to moderate
pain or local irritation might develop after treatment. After each
treatment, the gel or solution should be allowed to dry. Patients
should wash their hands before and after each application.
Podofilox is contraindicated during pregnancy (431).
Sinecatechins is a patient-applied, green-tea extract with an
active product (catechins). Sinecatechins 15% ointment should
be applied 3 times/day (0.5-cm strand of ointment to each wart)
by using a finger to ensure coverage with a thin layer of ointment
until complete clearance of warts is achieved. This product should
not be continued for >16 weeks (12231225). The medication
should not be washed off after use. Genital, anal, and oral sexual
contact should be avoided while the ointment is on the skin. The
most common side effects of sinecatechins are erythema, pruritus
or burning, pain, ulceration, edema, induration, and vesicular
Recommendations and Reports
104 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
rash. This medication is not recommended for persons with HIV
infection, other immunocompromised conditions, or genital herpes
because the safety and efficacy of therapy has not been evaluated.
The safety of sinecatechins during pregnancy is unknown.
Cryotherapy is a provider-administered therapy that destroys
warts by thermal-induced cytolysis. Health care providers
should be trained on the correct use of this therapy because
overtreatment or undertreatment can result in complications
or low efficacy. Pain during and after application of the liquid
nitrogen, followed by necrosis and sometimes blistering, is
common. Local anesthesia (topical or injected) might facilitate
therapy if warts are present in many areas or if the area of warts
is large. Surgical therapy has the advantage of eliminating the
majority of warts at a single visit, although recurrence can
occur. Surgical removal requires substantial clinical training,
additional equipment, and sometimes a longer office visit. After
local anesthesia is applied, anogenital warts can be physically
destroyed by electrocautery, in which case no additional
hemostasis is required. Care should be taken to control the
depth of electrocautery to prevent scarring. Alternatively, the
warts can be removed either by tangential excision with a pair of
fine scissors or a scalpel, by CO
2
laser, or by curettage. Because
most warts are exophytic, this procedure can be accomplished
with a resulting wound that only extends into the upper dermis.
Hemostasis can be achieved with an electrocautery unit or, in
cases of minor bleeding, a chemical styptic (e.g., an aluminum
chloride solution). Suturing is neither required nor indicated
in the majority of cases. For patients with large or extensive
warts, surgical therapy, including CO
2
laser, might be most
beneficial; such therapy might also be useful for intraurethral
warts, particularly for those persons whose warts have not
responded to other treatments. Treatment of anogenital and
oral warts should be performed in a ventilated room by using
standard precautions (https://www.cdc.gov/infectioncontrol/
guidelines/isolation/index.html/Isolation2007.pdf#page) and
local exhaust ventilation (e.g., a smoke evacuator) (1226).
Trichloroacetic acid (TCA) and bichloroacetic acid (BCA)
are provider-administered caustic agents that destroy warts by
chemical coagulation of proteins. Although these preparations
are widely used, they have not been investigated thoroughly.
TCA solution has a low viscosity, comparable with that of
water, and can spread rapidly and damage adjacent tissues if
applied excessively. A small amount should be applied only
to the warts and allowed to dry (i.e., develop white frost on
tissue) before the patient sits or stands. If pain is intense or an
excess amount of acid is applied, the area can be covered with
sodium bicarbonate (i.e., baking soda), washed with liquid
soap preparations, or be powdered with talc to neutralize the
acid or remove unreacted acid. TCA or BCA treatment can be
repeated weekly if necessary.
Alternative Regimens for External Genital Warts
Fewer data are available regarding the efficacy of alternative
regimens for treating anogenital warts, which include
podophyllin resin, intralesional interferon, photodynamic
therapy, and topical cidofovir. Shared clinical decision-
making between the patient and provider regarding benefits
and risks of these regimens should be provided. In addition,
alternative regimens might be associated with more side
effects. Podophyllin resin is no longer a recommended regimen
because of the number of safer regimens available, and severe
systemic toxicity has been reported when podophyllin resin
was applied to large areas of friable tissue and was not washed
off within 4 hours (12271229). Podophyllin resin 10%–25%
in a compound tincture of benzoin might be considered for
provider-administered treatment under conditions of strict
adherence to recommendations. Podophyllin should be applied
to each wart and then allowed to air dry before the treated area
comes into contact with clothing. Overapplication or failure
to air dry can result in local irritation caused by spread of the
compound to adjacent areas and possible systemic toxicity. The
treatment can be repeated weekly, if necessary. To avoid the
possibility of complications associated with systemic absorption
and toxicity, application should be limited to <0.5 mL of
podophyllin or an area of <10 cm
2
of warts per session; the
area to which treatment is administered should not contain
any open lesions, wounds, or friable tissue; and the preparation
should be thoroughly washed off 1–4 hours after application.
Podophyllin resin preparations differ in the concentration of
active components and contaminants. Shelf life and stability
of podophyllin preparations are unknown. The safety of
podophyllin during pregnancy has not been established.
Recommended Regimens for Urethral Meatus Warts
Cryotherapy with liquid nitrogen
or
Surgical removal
Recommended Regimens for Vaginal Warts
Cryotherapy with liquid nitrogen
The use of a cryoprobe in the vagina is not recommended because of
the risk for vaginal perforation and fistula formation.
or
Surgical removal
or
Trichloracetic acid (TCA) or bichloroacetic acid (BCA) 80%–90%
solution
Recommended Regimens for Cervical Warts
Cryotherapy with liquid nitrogen
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 105
US Department of Health and Human Services/Centers for Disease Control and Prevention
or
Surgical removal
or
Trichloracetic acid (TCA) or bichloroacetic acid (BCA) 80%–90%
solution
Management of cervical warts should include consultation with a specialist.
For women who have exophytic cervical warts, a biopsy evaluation to
exclude HSIL should be performed before treatment is initiated.
Recommended Regimens for Intra-Anal Warts
Cryotherapy with liquid nitrogen
or
Surgical removal
or
Trichloracetic acid (TCA) or bichloroacetic acid (BCA) 80%–90%
solution
Management of intra-anal warts should include consultation with a
colorectal specialist.
Follow-Up
Anogenital warts typically respond within 3 months of
therapy. Factors that might affect response to therapy include
immunosuppression and treatment compliance. Warts
located on moist surfaces or in intertriginous areas respond
best to topical treatment. A new treatment modality should
be selected when no substantial improvement is observed
after a complete course of treatment or in the event of severe
side effects; treatment response and therapy-associated side
effects should be evaluated throughout the therapy course.
Complications occur rarely when treatment is administered
correctly. Persistent hypopigmentation or hyperpigmentation
can occur with ablative modalities (e.g., cryotherapy and
electrocautery) and have been described with immune
modulating therapies (e.g., imiquimod cream). Depressed or
hypertrophic scars are uncommon but can occur, especially
if patients have insufficient time to heal between treatments.
Rarely, treatment can result in chronic pain syndromes (e.g.,
vulvodynia and hyperesthesia of the treatment site) or, in the
case of anal warts, painful defecation or fistulas.
Counseling
When counseling persons with anogenital warts, the provider
should discuss the following:
If left untreated, genital warts might resolve, stay the same,
or increase in size or number. The types of HPV that cause
genital warts are different from the types that can cause cancer.
Women with genital warts do not need Pap tests more
often than other women.
Time of HPV acquisition cannot be definitively
determined. Genital warts can develop months or years
after acquiring HPV.
HPV types that cause genital warts can be passed on to
another person, even without visible signs of warts. Sex
partners tend to share HPV, even though signs of HPV
(e.g., warts) might occur in only one or neither partner.
Although genital warts are common and benign, certain
persons might experience considerable psychosocial impact
after receiving this diagnosis.
Although genital warts can be treated, such treatment does
not cure the virus itself. For this reason, genital warts often
recur after treatment, especially during the first 3 months.
Because genital warts can be sexually transmitted, persons
with genital warts benefit from testing for other STIs.
HPV might remain present and can still be transmitted
to partners even after the warts are gone.
Condoms might lower the chances of transmitting genital
warts if used consistently and correctly; however, HPV
can infect areas that are not covered by a condom and
might not fully protect against HPV.
A vaccine is available for males and females to prevent
genital warts (Gardasil 9) but it will not treat existing HPV
or genital warts. This vaccine can prevent the majority of
cases of genital warts among persons who have not yet
been exposed to wart-causing types of HPV.
Management of Sex Partners
Persons should inform current partners about having genital
warts because the types of HPV that cause warts can be passed
on to partners. Partners should be counseled that they might
already have HPV despite no visible signs of warts; therefore,
HPV testing of sex partners of persons with genital warts is
not recommended. Partners might benefit from a physical
examination to detect genital warts and tests for other STIs. No
recommendations can be made regarding informing future sex
partners about a diagnosis of genital warts because the duration
of viral persistence after warts have resolved is unknown.
Special Considerations
Pregnancy
Podofilox, podophyllin, and sinecatechins should not be
used during pregnancy. Imiquimod appears to pose low risk
but should be avoided until more data are available. Anogenital
warts can proliferate and become friable during pregnancy.
Although removal of warts during pregnancy can be considered,
resolution might be incomplete or poor until pregnancy is
complete. Rarely, HPV types 6 and 11 can cause respiratory
papillomatosis among infants and children, although the route
of transmission (i.e., transplacental, perinatal, or postnatal) is
not completely understood. Whether cesarean delivery prevents
respiratory papillomatosis among infants and children also
Recommendations and Reports
106 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
is unclear (1230); therefore, cesarean delivery should not be
performed solely to prevent transmission of HPV infection to
the newborn. Cesarean delivery is indicated for women with
anogenital warts if the pelvic outlet is obstructed or if vaginal
delivery would result in excessive bleeding. Pregnant women
with anogenital warts should be counseled about the low risk
for warts on the larynx of their infants or children (recurrent
respiratory papillomatosis).
HIV and Other Causes of Immunosuppression
Persons with HIV infection or who are otherwise
immunosuppressed are more likely to develop anogenital
warts than those who do not have HIV (1231). Moreover,
such persons can have larger or more numerous lesions,
might not respond to therapy as well as those who are
immunocompetent, and might have more frequent recurrences
after treatment (1231,12321234). Despite these factors, data
do not support altered approaches to treatment for persons
with HIV infection. Squamous cell carcinomas arising in or
resembling anogenital warts might occur more frequently
among immunosuppressed persons, therefore requiring biopsy
for confirmation of diagnosis for suspicious cases (12351237).
High-Grade Squamous Intraepithelial Lesions
Biopsy of an atypical wart might reveal HSIL or cancer of
the anogenital tract. In this instance, referral to a specialist for
treatment is recommended.
Cancers and Precancers Associated with Human
Papillomavirus
Persistent infection with high-risk (oncogenic) types of HPV
has a causal role in approximately all cervical cancers and in
certain vulvar, vaginal, penile, anal, and oropharyngeal cancers
(1238). However, cervical cancer is the only HPV-associated
cancer for which routine screening is recommended.
Cervical Cancer
Screening Recommendations
Recommendations for cervical cancer screening in the
United States are based on systematic evidence reviews
by major medical and advocacy organizations, including
USPSTF (174), ACS (177), and ACOG (175). Over time,
general alignment across these organizations has emerged as
to when to start and end cervical cancer screening as well as
the periodicity of screening. Although no single guideline
universally guides screening practices in the United States, the
Patient Protection and Affordable Care Act required Medicaid
and new private health insurance plans to provide coverage for
preventive services graded A or B by USPSTF, which includes
cervical cancer screening. In addition, the National Center for
Quality Assurance provides a set of measures (the Healthcare
Effectiveness Data and Information Set [HEDIS]) for up-to-
date cervical cancer screening that aligns with USPSTF
recommendations (https://www.ncqa.org/hedis/measures/
cervical-cancer-screening). The Center for Medicaid and
Medicare Services uses the same measure as HEDIS to measure
cervical cancer screening performance.
USPSTF screening recommendations apply to persons with
a cervix at average risk, defined as those with no previous
cervical cancer or high-grade precancer, not currently
under close follow-up for a recent abnormal result, not
immunocompromised (e.g., persons with HIV), and who
had no exposure to diethylstilbestrol in utero. Among these
persons, screening should be performed starting at age 21 years
and continue through age 65 years. Testing can be performed
using either conventional or liquid-based cytologic tests (i.e.,
Pap tests). For persons aged ≥30 years, screening can include
FDA-cleared tests for high-risk, oncogenic types of HPV.
For cytopathologic testing, clinics should use CLIA-certified
laboratories using acceptable terminology (Bethesda 2001 or
LAST terminology) (1239).
Annual cervical cancer screening is not recommended for
persons at average risk. Instead, cytology testing is recommended
every 3 years for persons aged 21–29 years. For persons aged
30–65 years, a cytology test every 3 years, an HPV test alone
every 5 years, or a cytology test plus an HPV test (cotest) every
5 years is recommended. Cotesting can be done by either
collecting one sample for the cytology test and another for the
HPV test or by using the remaining liquid cytology material
for the HPV test. Cervical screening programs should screen
those who have received HPV vaccination in the same manner
as those that are unvaccinated. Screening is not recommended
before age 21 years among those at average risk. For those aged
30–65 years, cytology alone or primary HPV testing is preferred
by USPSTF; however, cotesting can be used as an alternative
approach. ACOG (1240), ACS (177), and USPSTF (174) each
have screening recommendations (1241) (Table 1).
Clinics should weigh the benefits of each screening strategy
as well as their resources, such as time and cost, in deciding on
which of the three possible screening strategies to implement.
Decision analytic models (1242) estimating the benefits, harms,
and costs (1243) of several different strategies might be useful
in making this determination (174,1244,1245). Adopting
recommended screening and follow-up procedures, including
screening methods, results provision, and follow-up, can lead to
success in implementing cervical cancer screening in clinics (1246).
Patients should be provided a copy of their test results;
those with normal results should be provided information
on follow-up visits and the importance of continued cervical
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 107
US Department of Health and Human Services/Centers for Disease Control and Prevention
TABLE 1. Cervical cancer screening and surveillance recommendations
Population Screening specifics
Guideline group, yr of recommendation
USPSTF, 2018 ACOG, 2016 ACS, 2020
Persons at average risk Age to start screening 21 yrs 21 yrs 25 yrs
Age to end screening 65 yrs 65 yrs 65 yrs
If three consecutive negative cytology tests or two negative cytology plus HPV tests or two
negative HPV tests (ACS) with the most recent within the previous 5 yrs and no abnormal tests
within the previous 10 yrs (ACS) and no CIN 2 or CIN 3 within the previous 25 yrs
Screening test options
and intervals
Aged 21–65 yrs: Cytology alone every 3 yrs
or
Aged 21–29 yrs: Cytology alone every 3 yrs
Aged 30–65 yrs: Cytology plus HPV testing every 5 yrs
or
Aged 21–29 yrs: Cytology alone every 3 yrs
Aged 30–65: HPV testing alone every 5 yrs*
HPV testing alone every 5 yrs
or
Cytology plus HPV testing every
5 yrs
or
Cytology alone every 3 yrs
Preferred strategies Cytology alone every 3 yrs
and HPV testing alone every
5 yrs (equally preferred)
Cytology plus HPV testing every
5 yrs
HPV testing alone every 5 yrs
Previous hysterectomy
with removal of cervix
Screening not recommended after hysterectomy for benign indications
Surveillance testing recommended for previous diagnosis of high-grade precancer, AIS, or cancer
Persons with an
immunocompromising medical
condition
(e.g., HIV infection or
solid organ transplantation)
Age to start screening No specific
recommendation
Within 1 yr of onset of sexual activity or, if already sexually active,
within the first year after HIV or other immunocompromising
medical condition diagnosis but no later than age 21 yrs
Age to end screening None; lifelong screening recommended
Screening test options
and intervals
Aged 21–65 yrs: Cytology every year; after three consecutive annual
normal cytology test results, screening can be every 3 yrs
or
Aged 21–29 yrs: Cytology every year
Aged 30–65 yrs: Cytology plus HPV testing every 3 yrs
Previous hysterectomy
with removal of cervix
Not specified
Persons with in utero exposure to
diethylstilbestrol
§
Age to start screening No specific
recommendation
Not specified No specific recommendation
Age to end screening Not specified
Screening test options
and intervals
Cytology alone annually
Previous hysterectomy
with removal of cervix
Not specified
Persons who have received HPV
vaccination
No changes to the screening approaches above
ASCCP, 2019, and ACOG, 2020
Persons with a diagnosis of CIN 2
or CIN 3 (histologic HSIL
)
within the previous 25 yrs
Age to start screening Not applicable
Age to end screening May end at age 65 yrs if CIN diagnosis ≥25 yrs ago and criteria for ending screening met, otherwise
continue screening past age 65 yrs
Continued screening for ≥25 yrs after diagnosis is acceptable if patient is in good health
Screening test options
and intervals
Initial surveillance:
HPV testing alone or cytology plus HPV testing at 6, 18, and 30 mos
or
Cytology at 6, 12, 18, 24, and 30 mos
Long-term surveillance:
HPV testing alone or cytology plus HPV testing every 3 yrs
or
Cytology alone annually
Continue for ≥25 yrs from the initial CIN diagnosis, even if extends past age 65 yrs
Routine screening can resume after the posttreatment surveillance period
Previous hysterectomy
with removal of cervix
HPV testing alone or cytology plus HPV testing every 3 yrs
or
Cytology alone annually
Continue for ≥25 yrs from the initial CIN diagnosis, even if extends past age 65 yrs
Source: Perkins R, Guido R, Saraiya M, et al. Summary of current guidelines for cervical cancer screening and management of abnormal test results: 2016–2020. J
Womens Health (Larchmt) 2021;30:5–13.
Abbreviations: ACS = American Cancer Society; ACOG = American College of Obstetricians and Gynecologists; AIS = adenocarcinoma in situ; ASCCP = American
Society for Colposcopy and Cervical Pathology; CIN = cervical intraepithelial neoplasia; HPV = human papillomavirus; HSIL = high-grade squamous intraepithelial
lesion; USPSTF = U.S. Preventive Services Task Force.
* Considered an alternative screening strategy by ACOG.
Panel for Opportunistic Infections, ACOG, 2016.
§
ACOG, 2016.
Either by cytology or by histology; includes a persistent cytologic diagnosis of atypical squamous cells, cannot rule out HSIL.
Recommendations and Reports
108 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
cancer screening, if applicable. Those with abnormal screening
tests should be managed per published guidelines. National
consensus guidelines are available for the management of
abnormal cervical cancer screening tests (1247). HPV testing
or cotesting is preferred to cytology alone for surveillance
after an abnormal screening test result. These guidelines base
management recommendations on case-by-case assessment
of risk considering past screening history and current results
(see Follow-Up). Patients with abnormal cervical cancer
screening test results should be counseled about those results
(see Counseling Messages).
The following additional management considerations are
associated with performing Pap tests and HPV tests:
Cytology (Pap tests) and HPV tests should not be
considered screening tests for STIs.
All persons with a cervix should receive cervical cancer
screening, regardless of sexual orientation or gender
identity (i.e., those who identify as lesbian, bisexual,
heterosexual, or transgender).
A conventional cytology test (in which the sample is
smeared onto a dry slide) should ideally be scheduled for
10–20 days after the first day of menses. Liquid-based
cytology can be performed at any time during the
menstrual cycle.
If specific infections other than HPV (e.g., chlamydia or
gonorrhea) are identified at the visit, a repeat cytology test
after appropriate treatment for those infections might be
indicated. However, in most instances (even in the
presence of certain severe cervical infections), cytology
tests will be reported as satisfactory for evaluation, and
reliable final reports can be produced without the need to
repeat the cytology test after treatment.
The presence of a mucopurulent discharge should not
postpone cytology testing. The test can be performed after
removal of the discharge with a saline-soaked cotton swab.
HPV testing can be performed either as a separate test or
by using material from the liquid-based cytology specimen.
In the absence of other indications, the presence of external
genital warts does not warrant more frequent cervical
cancer screening.
The sequence of cytology testing in relation to collection
of other endocervical specimens does not influence Pap test
results or their interpretation (600). Typically, vaginal
specimens are preferred for chlamydia and gonorrhea
screening; however, during a pelvic examination, endocervical
specimens for STI testing can be collected first.
Persons who have had a total hysterectomy with removal
of the cervix do not require screening unless cervical
intraepithelial neoplasia (CIN) 2, CIN 3, or adenocarcinoma
in situ was diagnosed within the previous 20 years
(175,1247). If the cervix remains intact after a supracervical
hysterectomy, regularly scheduled Pap tests should be
performed as indicated (12481250).
Health care facilities that train providers on cytology test
collection and use simple quality assurance measures are
more likely to obtain satisfactory test results (as determined
by the laboratory).
The use of instruments designed to sample the cervical
transformation zone (e.g., cytobrushes) improves the
accuracy of cytology tests (1251).
Both liquid-based and conventional cytology are acceptable
because they have similar test-performance characteristics.
At an initial visit, providers should ask patients about their
recent cytology test and HPV results and any history of
evaluation and treatment (e.g., loop electrosurgical
excision procedure and colposcopy) to assist with
management; effort should be made to obtain copies of
recent results. The importance and frequency of screening
should be reinforced.
Counseling
Persons might believe the cytology (Pap test) or HPV test
screens for conditions other than cervical cancer, or they
might be confused by abnormal results (12521254). Health
care providers, as trusted sources of information about HPV
infections and abnormal cytology test results, have an important
role in educating persons about HPV and can moderate the
psychosocial impact of abnormal results (1255,1256). Persons
should be counseled on the risks, uncertainties, and benefits
of screening (174,1257).
An abnormal cytology test or a positive HPV test can
cause short-term anxiety, stress, fear, and confusion, possibly
decreasing the patients ability to absorb and retain information
and acting as a barrier to follow-up care (12581261). A
positive HPV test might elicit concerns about partners, worries
about disclosure, and feelings of guilt, anger, and stigmatization
(1260). Providers should frame HPV positivity in a neutral,
nonstigmatizing context and emphasize its common,
asymptomatic, and transient nature. Providers also should
emphasize that HPV infections often are shared between
partners but it is often not possible to know the origin of an
HPV infection; HPV tests might become positive many years
after initial exposure due to reactivation of latent infections
in both male and female partners. Having an HPV infection
should not raise concerns about a male partner’s health (1262).
Providers should communicate the meaning of both the
cytology and HPV test results to patients at screening.
Providers also should screen for tobacco use and
perform cessation counseling (www.acog.org/clinical/
clinical-guidance/committee-opinion/articles/2011/09/
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 109
US Department of Health and Human Services/Centers for Disease Control and Prevention
tobacco-use-and-womens-health). Smoking contributes to
the progression of CIN, with both active and passive smoking
associated with squamous cell carcinoma of the cervix in
women with HPV 16 or 18 infection (12631266).
Promoting Cervical Cancer Screening
Clinics can use the evidence-based interventions in the
Community Preventive Services Task Force guidelines to
promote cervical cancer screening in their communities
(https://www.thecommunityguide.org/findings/cancer-
screening-multicomponent-interventions-cervical-cancer).
Implementing interventions that increase community demand
for screening (1266) (e.g., client reminders, client incentives,
media, group education, or one-on-one education) together
with those that increase community access to screening (e.g.,
reducing structural barriers and reducing client out-of-pocket
costs) is effective in increasing cervical cancer screening
coverage. These interventions are more effective if they are
implemented with interventions to increase provider delivery
of screening services (e.g., provider assessment and feedback,
provider incentives, and provider reminders). Print materials
and online resources are available at https://www.cdc.gov/
cancer/cervical/basic_info/screening.htm and https://www.
cdc.gov/std/hpv/facts-brochures.htm. Patient navigators can
be effective in improving both screening and follow-up after
abnormal results (1267).
Key Messages About Cervical Cancer Screening
When counseling persons about cervical cancer screening,
the provider should discuss the following:
Cervical cancer can be prevented with regular screening tests,
like the Pap test (cytology) and HPV tests. Those at average
risk should start getting cytology tests at age 21 years.
The cytology test can find abnormal cervical cells, which
could lead to cervical cancer over time, and an HPV test
detects HPV infection of the cervix. The HPV test can be
used alone for cervical cancer screening or at the same time
as the cytology test (known as cotesting) for those aged
≥30 years to 65 years. The HPV test is also used after a
cytology test result of atypical squamous cells of
undetermined significance (ASC-US) among persons aged
>25 years (known as reflex HPV testing).
Positive cytology and HPV tests are markers of cervical
precancerous lesions, which often do not cause symptoms
until they become invasive. Appropriate follow-up is
essential to ensure that cervical cancer does not develop.
HPV is a common infection and is often controlled by
the body without any medical interventions. A positive
HPV test does not mean that a person has cancer.
Providers should emphasize that HPV infections often are
shared between partners, and it is often not possible to know
the origin of an HPV infection; HPV tests might become
positive many years after initial exposure due to reactivation
of latent infections in both male and female partners.
Management of Sex Partners
The benefit of disclosing a positive HPV test to current
and future sex partners is unclear. The following counseling
messages can be communicated to sex partners:
Sex partners do not need to be tested for HPV.
Sex partners tend to share HPV. Sex partners of persons
with HPV infection also are likely have an HPV infection.
Female sex partners of men who disclose they had a previous
female partner with HPV should be screened at the same
intervals as women with average risk. No data are available
to suggest that more frequent screening is of benefit.
When used correctly and consistently, condoms might
lower the risk for HPV infection and might decrease the
time to clear in those with HPV infection. However, HPV
can infect areas not covered by the condom, and condoms
might not fully protect against HPV (24,25).
Additional messages for partners include the messages for persons
with HPV (see Cervical Cancer Screening; Counseling Messages).
Screening Recommendations in Special
Populations
Pregnancy
Persons who are pregnant should be screened at the same
intervals as those who are not. A swab, Ayre’s spatula, or
cytobrush can be used for obtaining cytology test samples
during pregnancy (12681270).
HIV Infection
Several studies have documented an increased risk for cervical
precancers and cancers in individuals with HIV infection
(12711273). Adolescents with HIV should be screened 1 year
after onset of sexual activity but no later than age 21 years.
Sexually active persons should be screened at the time of the
initial HIV diagnosis. Conventional or liquid-based cytology
(Pap test) should be used as primary HPV testing and is not
recommended in individuals with HIV. Cotesting (cytology
and HPV test) can be done in individuals aged ≥30 years with
HIV. Annual screening is recommended for persons with HIV
infection; after 3 years of consecutive normal cytology results
or normal cotest (normal cytology and negative HPV test), the
screening interval can be increased to every 3 years. Lifelong
screening is recommended among persons with HIV infection.
Recommendations and Reports
110 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Providers should defer to existing Guidelines for the Prevention
and Treatment of Opportunistic Infections in Adults and
Adolescents with HIV for guidance on cervical cancer screening
and management of results in persons with HIV (98).
Adolescents
Prevalence of HPV infection is high among those aged
<21 years (174); however, HPV infections and squamous
intraepithelial lesions caused by HPV in adolescents are more
likely to regress than those in older persons. For these reasons,
cervical cancer screening and HPV testing are not recommended
in immunocompetent adolescents. However, for adolescents
with HIV infection, providers should screen 1 year after onset of
sexual activity, regardless of age or mode of HIV acquisition (e.g.,
perinatally acquired or sexually acquired) (98); such screening
is warranted because of the reported high rate of progression of
abnormal cytology in adolescents with HIV.
Human Papillomavirus Tests for Cervical
Cancer Screening
Clinical tests for HPV are used for the following: cervical
cancer screening as a primary test, cervical cancer screening
with a cytology test, triage of some abnormal cervical
cytology results, follow-up after abnormal screening test
results, follow-up after a colposcopy in which no CIN 2 or
CIN 3 is found, and follow-up after treatment of cervical
precancers. These tests are only FDA cleared for use with
cervical specimens, not oral or anal specimens. Testing
for nononcogenic HPV types (e.g., types 6 and 11) is not
recommended (https://www.asccp.org/guidelines).
FDA-cleared HPV tests detect viral DNA or messenger
RNA. Several FDA-cleared tests for HPV are available for
use in the United States. The Cobas 4800 HPV test (Roche
Molecular Diagnostics) and the Onclarity HPV test (Becton
Dickinson) can detect the presence of 14 oncogenic HPV types
(types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 66, and
68), as well as individual types 16 and 18, and are cleared for
primary cervical cancer screening.
Other HPV tests are cleared for use in conjunction with
a cytology test or to triage some abnormal cervical cytology
results; they should not be used for primary HPV testing
because they are not cleared for this purpose. These tests
include the Hybrid Capture 2 High-Risk HPV DNA test
(Qiagen), the Cervista HPV High-Risk DNA and HPV 16/18
DNA tests (Hologics), and the APTIMA HR HPV (Gen
Probe) test. All HPV assays should be FDA cleared and used
only for the appropriate indications (https://www.fda.gov/
media/122799/download) (158).
HPV testing should not be performed in the following situations:
Deciding whether to vaccinate against HPV
Conducting HPV tests for low-risk (nononcogenic) HPV
types (e.g., types 6 and 11)
Providing care to persons with genital warts or their partners
Testing persons aged <25 years as part of routine cervical
cancer screening
Testing oral or anal specimens
Unlike cytology, samples for HPV testing have the potential
to be collected by the patient and mailed to health programs
for analysis, thus self-collection might be one strategy for
increasing screening rates among populations where screening
rates are low. Self-collection for HPV testing is not cleared by
FDA or recommended by U.S. medical organizations (174).
Follow-Up of Abnormal Cytology and Human
Papillomavirus Test Results
If the result of the cytology (Pap test) is abnormal, follow-up
care should be provided according to the 2019 ASCCP Risk-
Based Management Consensus Guidelines for Abnormal Cervical
Cancer Screening Tests and Cancer Precursors (158). Clinics that
serve clients who might have difficulty adhering to follow-up
recommendations and for whom linkage to care is unlikely should
consider offering in-house colposcopy and biopsy services.
Consensus guidelines for management of abnormal
cervical cancer screening tests combine patient-level risk
data with clinical action thresholds to generate personalized
management recommendations (Table 2). This framework
allows management on the basis of risk for CIN 3, not specific
test results. The guidelines were designed to identify persons at
high risk who require colposcopy or expedited treatment and
persons at low risk who might be able to safely defer invasive
diagnostic procedures. The risk-based framework was designed
to easily incorporate future revisions, such as the inclusion of
new technologies for screening and management. Use of the
guidelines can be facilitated by electronic technology that is
continually updated, such as a smartphone application or the
website (https://www.asccp.org/Default.aspx).
The following are highlights of the new management guidelines:
Colposcopy can be deferred for patients at low risk.
ű
If a patient has a minimally abnormal test result (i.e.,
negative for intraepithelial lesion or malignancy HPV
positive, ASC-US HPV positive, LSIL, or HPV positive)
that was preceded by a negative screening HPV test or
cotest within the past 5 years, follow-up in 1 year instead
of colposcopy is recommended (a negative HPV test or
cotest performed during follow-up of abnormal results
would not similarly reduce risk).
ű
Referral to colposcopy is recommended if cytology test
results are abnormal or the HPV test is positive at the
1-year follow-up visit.
Treatment can be expedited for high-risk patients.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 111
US Department of Health and Human Services/Centers for Disease Control and Prevention
ű
If a patient has a high-grade cytology (Pap test) result
(i.e., HSIL) and an HPV test that is positive for HPV
type 16, then treatment with a loop electrosurgical
excision procedure (LEEP) is preferred. A colposcopy
with biopsy is not necessary to confirm the diagnosis first.
ű
If a patient who has not been screened in more than
5 years (i.e., rarely screened) has an HSIL cytology result
and a positive HPV test (regardless of type), then
treatment with LEEP is preferred. A colposcopy with
biopsy is not necessary to confirm the diagnosis first.
ű
When considering treatment without confirmatory
biopsy, shared decision-making with the patient is
important. Considerations include age, concern about
cancer, ability to follow up, financial concerns, and
concerns about the potential effect of treatment on a
future pregnancy.
When primary HPV testing is used for screening, cytology
testing should be performed for all positive HPV test
results to help determine the next steps in management.
ű
Ideally, cytology testing should be performed by the
laboratory as a reflex test from the same specimen so the
patient does not need to return to the clinic. Colposcopy
is recommended if HPV genotyping is positive for types
16 or 18, and it can be considered if it is infeasible for
the patient to return for cytology alone (1274).
ű
HPV 16 is the highest-risk HPV type. Expedited
treatment should be considered for HSIL cytology
results, and colposcopy is recommended in all other
cases, even if the cytology test is normal.
TABLE 2. Comparison of 2012 and 2019 consensus recommendations for management of common abnormalities — American Society for
Colposcopy and Cervical Pathology
Current HPV result
Current Pap
test result Previous result
Management by 2012
guidelines
Management by 2019
guidelines
Negative ASC-US Unknown or HPV negative* Repeat Pap plus HPV testing
in 3 yrs
Repeat HPV test with or without
concurrent Pap test in 3 yrs
Negative LSIL Unknown or HPV negative* Repeat Pap plus HPV testing
in 1 yr preferred, colposcopy
acceptable
Repeat HPV test with or without
concurrent Pap test in 1 yr
Negative ASC-H Noncontributory Colposcopy Colposcopy
Noncontributory AGC Noncontributory Colposcopy Colposcopy
Positive NILM Unknown or HPV negative* Repeat Pap plus HPV testing
in 1 yr
Repeat HPV test with or without
concurrent Pap test in 1 yr
Positive NILM HPV positive
Colposcopy Colposcopy
Positive for genotype HPV 16,
HPV 18, or both
NILM Noncontributory Colposcopy Colposcopy
Positive for genotype HPV 16,
HPV 18, or both
ASC-US or LSIL Noncontributory Not applicable, genotyping
not recommended for
ASC-US or LSIL in 2012
Colposcopy
Positive ASC-US or LSIL Unknown or HPV positive Colposcopy Colposcopy
Positive ASC-US or LSIL Negative screening results with
HPV testing or HPV plus Pap
testing within the previous 5 yrs
Colposcopy Repeat HPV test with or without
concurrent Pap test in 1 yr
§
Positive ASC-US or LSIL Colposcopy confirming the absence
of high-grade lesion within the
past yr
Colposcopy Repeat HPV test with or without
concurrent Pap test in 1 yr
§
Positive ASC-H Noncontributory Colposcopy Colposcopy or expedited
treatment
Positive untyped, positive for
genotype other than HPV 16,
or negative
HSIL Noncontributory Colposcopy or expedited
treatment
Colposcopy or expedited
treatment
Positive for genotype HPV 16 HSIL Noncontributory Colposcopy or expedited
treatment
Expedited treatment
Sources: Massad LS, Einstein MH, Huh WK, et al.; 2012 ASCCP Consensus Guidelines Conference. 2012 updated consensus guidelines for the management of abnormal
cervical cancer screening tests and cancer precursors. Obstet Gynecol 2013;121:829–46; Perkins RB, Guido RS, Castle PE, et al; 2019 ASCCP Risk-Based Management
Consensus Guidelines Committee. 2019 ASCCP risk-based management consensus guidelines for abnormal cervical cancer screening tests and cancer precursors.
J Low Genit Tract Dis 2020;24:102–31; Perkins R, Guido R, Saraiya M, et al. Summary of current guidelines for cervical cancer screening and management of abnormal
test results: 2016–2020. J Womens Health (Larchmt) 2021;30:5–13.
Abbreviations: AGC = atypical glandular cells; AIS = adenocarcinoma in situ; ASC-H = atypical squamous cells cannot exclude high-grade squamous intraepithelial
lesion; ASC-US = atypical squamous cells of undetermined significance; CIN = cervical intraepithelial neoplasia; HPV = human papillomavirus; HSIL = high-grade
squamous intraepithelial lesion; LSIL = low-grade squamous intraepithelial lesion; NILM = negative for intraepithelial lesion or malignancy; Pap = Papanicolaou.
* Colposcopy may be warranted for patients with a history of high-grade lesions (CIN 2 or CIN 3, histologic or cytologic HSIL, ASC-H, AGC, or AIS).
Previous Pap test results do not modify the recommendation; colposcopy is always recommended for two consecutive HPV-positive tests.
§
Negative HPV test or cotest (HPV plus Pap test) results only reduce risk sufficiently to defer colposcopy if performed for screening purposes within the last 5 years.
Colposcopy is still warranted if negative HPV test or cotest results occurred in the context of surveillance for a previous abnormal result.
Expedited treatment is preferred for nonpregnant patients aged ≥25 years. Colposcopy with biopsy is an acceptable option if desired by patient after shared
decision-making.
Recommendations and Reports
112 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
ű
HPV 18 has a relatively high association with cancer,
and colposcopy is recommended in all cases, even if the
cytology test is normal. Because of the association of
HPV 18 with adenocarcinoma, endocervical sampling
is acceptable at the time of colposcopy.
ű
If the HPV type is not HPV 16 or 18, and the cytology test
is normal, return in 1 year is recommended in most cases.
HPV testing or cotesting is preferred to cytology testing
alone for follow-up after an abnormal test result.
ű
Negative HPV testing or cotesting is less likely to miss
disease than normal cytology testing alone. Therefore,
cytology testing is recommended more often than HPV
testing or cotesting for follow-up of abnormal results.
Specifically, cytology testing is recommended annually
when HPV testing or cotesting is recommended at
3-year intervals, and cytology testing is recommended
at 6-month intervals when HPV testing or cotesting is
recommended annually.
After treatment for a high-grade precancer (moderate or
severe dysplasia), surveillance should continue for at least
25 years.
ű
Initial testing includes an HPV test or cotest at 6, 18,
and 30 months. If cytology alone is used, testing should
occur at 6, 12, 18, 24, and 30 months.
ű
After completing initial testing, long-term surveillance
includes testing at 3-year intervals if using HPV testing or
cotesting, or annual testing if using cytology testing alone.
ű
Surveillance should continue for at least 25 years after
the initial treatment, even if this extends beyond age
65 years. If a woman undergoes hysterectomy during the
surveillance period, vaginal screening should continue.
Anal Cancer
Anal cancer is rare in the general population (1–2 cases per
100,000 person-years); however, incidence is substantially
higher among specific populations, including MSM with HIV
infection (80–131 cases per 100,000 person-years), men with
HIV infection (40–60 cases per 100,000 person-years), women
with HIV infection (20–30 cases per 100,000 person-years),
and MSM without HIV infection (14 cases per 100,000
person-years) (12751279). Incidence is variable among
women with previous HPV-related gynecologic dysplasia and
cancer (6–63 cases per 100,000 person-years) (1280,1281).
Persistent HPV infection might be a risk factor for preventable
HPV-associated second primary cancers among survivors of
HPV-associated cancers (1282).
Data are insufficient to recommend routine anal cancer
screening with anal cytology in persons with HIV infection,
MSM without HIV infection, and the general population. An
annual digital anorectal examination (DARE) might be useful
to detect masses on palpation in persons with HIV infection
and possibly in MSM without HIV with a history of receptive
anal intercourse (98). More evidence is needed concerning
the natural history of anal intraepithelial neoplasia, the best
screening methods and target populations, the safety and
response to treatments, and other programmatic considerations
before screening can be routinely recommended.
Populations at High Risk and Digital Anorectal
Examination
Providers should discuss anal cancer risk with their patients
among specific populations to guide management. According
to the HIV Opportunistic Infection guidelines and the
International Anal Neoplasia Society, a DARE should be
performed to detect early anal cancer in persons with HIV
infection and MSM without HIV with a history of receptive
anal intercourse (98,1283). DARE is acceptable to patients and
has a low risk for adverse outcomes (1284,1285).
Data are insufficient to guide initiation of DARE at a defined
age or optimal intervals for examination. Whereas anal HSIL
is observed among young adults, cancer incidence begins
to increase after the early 30s and continues to increase as a
function of age.
Populations at High Risk and Anal Cytology
Data are insufficient to recommend routine anal cancer
screening with anal cytology among populations at risk for
anal cancer. Certain clinical centers perform anal cytology to
screen for anal cancer among populations at high risk (e.g.,
persons with HIV infection, MSM, and those having receptive
anal intercourse), followed by high-resolution anoscopy (HRA)
for those with abnormal cytologic results (e.g., ACS-US, LSIL,
or HSIL). Sensitivity and specificity of anal cytology to detect
HSIL are limited (sensitivity 55%–89% and specificity 40%–
67%) (12861291). Health centers that initiate a cytology-
based screening program should only do so if referrals to HRA
and biopsy are available.
HRA can be used for diagnosis of HSIL, to monitor response
to therapy, or to conduct surveillance of HSIL for evidence of
progression. HRA is the primary method used for diagnosis of
superficially invasive squamous carcinoma, a very early form of
anal cancer that is not palpable on DARE. However, data are
insufficient to conclude whether use of HRA leads to reductions
in anal cancer incidence or improves anal cancer morbidity and
mortality. An ongoing clinical trial is investigating whether
treatment of HSIL is effective in reducing the incidence of anal
cancer among persons with HIV infection (NCT02135419).
Human Papillomavirus Testing
HPV tests (using high-risk HPV types) are not clinically
useful for anal cancer screening because of a high prevalence
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 113
US Department of Health and Human Services/Centers for Disease Control and Prevention
of anal HPV infection among populations at high risk,
particularly MSM (1278,1289,1290). No standard HPV-based
algorithms exist for anal cancer screening due to the high
prevalence of high-risk HPV infection among groups at risk.
Treatment of Anal High-Grade Squamous
Intraepithelial Lesion
Multiple office-based treatments exist for anal HSIL,
including ablative methods (e.g., laser, electrocautery, or
infrared coagulation) and topical patient-applied therapies
(e.g., imiquimod). Recurrence rates with both provider-
applied and patient-applied treatments are high, ranging
from approximately 50% at 1 year to 77% after 3 years
(1289,1292,1293). In addition, evidence exists that HSIL
might spontaneously regress without treatment (1294,1295).
Shared decision-making about treatment for anal HSIL is
recommended because of limited data on the natural history
of anal HSIL, including factors related to progression or
regression of lesions.
Viral Hepatitis
Hepatitis A Virus Infection
HAV infection has an incubation period of approximately
28 days (range: 15–50 days) (1296). HAV replicates in the liver
and is shed in high concentrations in feces from 2–3 weeks
before to 1 week after the onset of clinical illness. HAV infection
produces a self-limited disease that does not result in chronic
infection or chronic liver disease. However, approximately
10% of patients experience a relapse of symptoms during
the 6 months after acute illness. Acute liver failure from
hepatitis A is rare (overall case-fatality rate: 0.5%). The risk
for symptomatic infection is directly related to age, with
approximately 70% of adults having symptoms compatible
with acute viral hepatitis and the majority of children having
either asymptomatic or unrecognized infection. Antibody
produced in response to HAV infection persists for life and
confers protection against reinfection (1297).
HAV infection is primarily transmitted by the fecal-oral route,
by either person-to-person contact or through consumption
of contaminated food or water (1298). Transmission of HAV
during sexual activity probably results from fecal-oral contact.
Although viremia occurs early during infection and can persist
for weeks after symptom onset, bloodborne transmission of
HAV is uncommon (1299). Transmission by saliva has not
been demonstrated.
In the United States, of the hepatitis A cases accompanied by
risk information, a particular risk was identified among only
23.8% (13,372). Among cases with a risk factor identified, a
recognized foodborne or waterborne outbreak was the most
commonly identified risk (49.6%). Other infection sources
identified in the United States include MSM; persons who
use injecting drugs; sexual and household contacts; those
experiencing homelessness; international travelers; those with
children attending a nursery, childcare, or preschool; and
persons working in such settings (13,372).
Diagnostic Considerations
Diagnosis of HAV infection cannot be made on a clinical
basis alone but requires serologic testing. Presence of IgM
antibody to HAV is diagnostic of acute HAV infection. A
positive test for total anti-HAV indicates immunity to HAV
infection but does not differentiate current from previous HAV
infection. Although usually not sensitive enough to detect the
low level of protective antibody after vaccination, anti-HAV
tests also might be positive after hepatitis A vaccination.
Treatment
Patients with acute HAV infection usually require only
supportive care, with no restrictions in diet or activity.
Hospitalization might be necessary for patients who become
dehydrated because of nausea and vomiting and is crucial
for patients with signs or symptoms of acute liver failure.
Medications that might cause liver damage or are metabolized
by the liver should be used with caution among persons with
HAV infection.
Prevention
Vaccination is the most effective means of preventing HAV
transmission among persons at risk for infection (e.g., MSM,
injecting drug users, and persons with chronic liver disease)
who did not receive hepatitis A vaccination during childhood.
Hepatitis A vaccines are prepared from formalin-inactivated,
cell-culture–derived HAV. Two monovalent vaccines (Havrix
and Vaqta are approved by FDA for persons aged ≥12 months
(Table 3). These vaccines are available for eligible children
and adolescents aged <19 years through the VFC program
(https://www.cdc.gov/vaccines/programs/vfc/index.html).
Administered IM in a 2-dose series at 0 and 6–12 months,
hepatitis A vaccines induce protective antibody levels among
virtually all adults. By 1 month after the first dose, 94%–100%
of adults have protective antibody levels, and after a second
dose, 100% achieve protective levels (1297,1300,1301).
Kinetic models of antibody decrease among adults indicate that
protective levels persist for >40 years (13021304). A study of
Alaska Natives demonstrated that seropositivity for hepatitis
A persists for >20 years after completing 2-dose vaccination at
age 12–21 months (1302). Anti-HAV persistence of >20 years
was demonstrated among immunocompetent adults vaccinated
Recommendations and Reports
114 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
with a 2-dose hepatitis A schedule as adults (1303,1305). A
combined hepatitis A and hepatitis B vaccine (Twinrix) has
been developed and licensed for use as a 3-dose series for
adults aged ≥18 years at risk for HAV or HBV infections.
When administered IM on a 0-, 1-, and 6-month schedule,
the vaccine has equivalent immunogenicity to that of the
monovalent hepatitis A vaccines.
Pre-Exposure Vaccination
Persons at risk for HAV infection (Box 5) (1297) should be
offered vaccine (Table 3). If persons are at risk for both HAV
and HBV, the combined vaccine can be considered.
Prevaccination Serologic Testing
Among U.S.-born adults aged >20 years, HAV susceptibility
prevalence (i.e., total antibody to HAV was negative) was
74.1% (95% CI:72.9%–75.3%) during 2007–2016 (1306).
Prevaccination serologic testing for HAV immunity before
vaccination is not routinely recommended; however, it can be
considered in specific settings to reduce costs by not vaccinating
persons who are already immune. Prevaccination serologic
testing should not be a barrier to vaccination of susceptible
persons, especially for populations that are difficult to access. If
prevaccination testing is performed, commercially available tests
for total anti-HAV or IgG anti-HAV should be used (1297).
BOX 5. Populations recommended for hepatitis A vaccination — Advisory Committee on Immunization Practices, 2020
Children
All children aged 12–23 months
Unvaccinated children and adolescents aged 2–18 years
Persons at increased risk for hepatitis A virus (HAV)
infection
International travelers
Men who have sex with men
Persons who use injecting or noninjecting drugs (i.e.,
all those who use illegal drugs)
Persons with occupational risk for exposure
Persons who anticipate close personal contact with an
international adoptee
Persons experiencing homelessness
Persons at increased risk for severe disease from
HAV infection
Persons with chronic liver disease
Persons with HIV infection
Other persons recommended for vaccination
Pregnant women at risk for HAV infection or severe
outcome from HAV infection
Any persons who requests a vaccine
Vaccination during outbreaks
Unvaccinated persons in outbreak settings who are at
risk for HAV infection or at risk for severe disease from
HAV
Implementation strategies for settings providing
services to adults
Persons in settings that provide services to adults where
a high proportion of those persons have risk factors for
HAV infection
Hepatitis A vaccination is no longer recommended by
the Advisory Committee on Immunization Practices
Persons who receive blood products for clotting disorders
(e.g., hemophilia)
Source: Nelson NP, Weng MK, Hofmeister MG, et al. Prevention of hepatitis A virus infection in the United States: recommendations of the Advisory Committee
on Immunization Practices, 2020. MMWR Recomm Rep 2020;69(No. RR-5).
TABLE 3. Vaccines for preventing hepatitis A infection
Vaccine Trade name (manufacturer) Age group (yrs) Dose Route Schedule Booster
Hep A inactivated
(2 doses)
Havrix (GlaxoSmithKline) 1–18 0.5 mL (720 ELISA units inactivated HAV) IM 0, 6–12 mos None
≥19 1 mL (1,440 ELISA units inactivated HAV IM 0, 6–12 mos None
Hep A inactivated
(2 doses)
Vaqta (Merck) 1–18 0.5 mL (25 units HAV antigen) IM 0, 6–18 mos None
≥19 1 mL (50 units HAV antigen) IM 0, 6–18 mos None
Combined Hep A
and Hep B*
(3 doses)
Twinrix (GlaxoSmithKline) ≥18 (primary) 1 mL (720 ELISA units inactivated plus
20 µg HBsAg
IM 0, 1, 6 mos None
≥18 (accelerated) 1 mL (720 ELISA units inactivated plus
20 µg HBsAg
IM 0, 7, 21–30 days 12 mos
Source: Nelson NP, Weng MK, Hofmeister MG, et al. Prevention of hepatitis A virus infection in the United States: recommendations of the Advisory Committee on
Immunization Practices, 2020. MMWR Recomm Rep 2020;69(No. RR-5).
Abbreviations: ELISA = enzyme-linked immunosorbent assay; HAV = hepatitis A virus; HBsAg = hepatitis B surface antigen; Hep A = hepatitis A; Hep B = hepatitis B;
IM= intramuscular.
* Combined Hep A and Hep B vaccine (Twinrix) should not be used as postexposure prophylaxis.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 115
US Department of Health and Human Services/Centers for Disease Control and Prevention
Persons for whom prevaccination testing will likely be most
cost-effective include adults who were either born in or lived for
extensive periods in geographic areas where HAV endemicity is
high or intermediate (1297). Prevaccination serologic testing
of children is not indicated because of the low prevalence of
infection among that age group.
For populations who are expected to have high rates of previous
HAV infection, vaccination history should be obtained when
feasible before testing or vaccination. Vaccination should not be
postponed if vaccination history cannot be obtained, records are
unavailable, or prevaccination testing is infeasible. Vaccinating
persons immune from natural infection carries no known risk, nor
does giving extra doses of hepatitis A vaccine (1307). Vaccination
of a person who is already immune is not harmful. Persons who
have a documented history of ≥2 doses of hepatitis A vaccine do
not need further vaccination or serologic testing.
Postvaccination Serologic Testing
Serologic testing for immunity is unnecessary after routine
vaccination of infants, children, or adults (1297). Testing
for anti-HAV antibody after vaccination is recommended
for persons whose subsequent clinical management depends
on knowledge of their immune status and persons for whom
revaccination might be indicated (e.g., persons with HIV
infection and other immunocompromising conditions).
Postexposure Prophylaxis
Persons who recently have been exposed to HAV and who
previously have not received hepatitis A vaccine should be
administered a single dose of monovalent hepatitis A vaccine
or immunoglobulin (IG) (0.1 mL/kg body weight) as soon as
possible, ideally <2 weeks after exposure because the efficacy
of vaccine or IG when administered >2 weeks after exposure
has not been established (1297). In most cases, monovalent
hepatitis A vaccine at the age-appropriate dose is preferred
over IG for PEP. Advantages of hepatitis A vaccine for PEP
include induction of active immunity, longer-term protection,
ease of administration, and better acceptability and availability.
Decisions to use vaccine versus IG should be guided by patient
characteristics associated with more severe manifestations
of HAV infection (e.g., older age, immunocompromising
conditions, and chronic liver disease) and the magnitude of the
risk for HAV transmission resulting from the exposure (1297).
IG should be used for children aged <6 months,
immunocompromised persons, persons with chronic liver
disease, and persons for whom vaccine is contraindicated. IG
can be administered to persons aged >40 years, in addition to
hepatitis A vaccine (1297).
IG administered IM can provide PEP against HAV (Table 4).
IG is a sterile solution of concentrated immunoglobulins prepared
TABLE 4. Recommendations for hepatitis A postexposure prophylaxis and pre-exposure protection, by age group and risk category — Advisory
Committee on Immunization Practices, 2020
Indication and age group Risk category and health status Hepatitis A vaccine IG*
Postexposure prophylaxis
0–11 mos Healthy No 0.1 mL/kg body weight
12 mos to 40 yrs Healthy 1 dose
None
>40 yrs Healthy 1 dose
0.1 mL/kg body weight
§
≥12 mos Immunocompromised or chronic liver disease 1 dose
0.1 mL/kg body weight
≥12 mos Vaccine contraindicated** No 0.1 mL/kg body weight
Pre-exposure protection (e.g., travel)
††
<6 mos Healthy No 0.1–0.2 mL/kg body weight
§§
6–11 mos Healthy 1 dose
¶¶
None
12 mos to 40 yrs Healthy 1 dose*** None
>40 yrs Healthy 1 dose*** 0.1–0.2 mL/kg body weight
§§,†††
>6 mos Immunocompromised or chronic liver disease 1 dose*** 0.1–0.2 mL/kg body weight
§§,†††
>6 mos Persons who elect not to receive vaccine or for whom
vaccine is contraindicated**
No 0.1–0.2 mL/kg body weight
§§
Source: Nelson NP, Weng MK, Hofmeister MG, et al. Prevention of hepatitis A virus infection in the United States: recommendations of the Advisory Committee on
Immunization Practices, 2020. MMWR Recomm Rep 2020;69(No. RR-5).
Abbreviations: HAV = hepatitis A virus; IG = immune globulin.
* Measles, mumps, and rubella vaccine should not be administered for ≥2 weeks before and 6 months after administration of IG.
A second dose of hepatitis A vaccine is not required for postexposure prophylaxis; however, for long-term immunity, the vaccination series should be completed
with a second dose ≥6 months after the first dose.
§
The provider’s risk assessment should determine the need for IG administration. If the provider’s risk assessment determines that both vaccine and IG are warranted,
hepatitis A vaccine and IG should be administered simultaneously at different anatomic sites (e.g., separate limbs).
Vaccine and IG should be administered simultaneously at different anatomic sites (e.g., separate limbs).
** Life-threatening allergic reaction to a previous dose of hepatitis A vaccine or allergy to any vaccine component.
††
IG should be considered before travel for persons with special risk factors for either HAV infection or severe disease from HAV infection.
§§
0.1 mL/kg body weight for travel ≤1 month; 0.2 mL/kg body weight for travel ≤2 months; 0.2 mL/kg every 2 months for travel of ≥2 months duration.
¶¶
This dose should not be counted toward the routine 2-dose series, which should be initiated at age 12 months.
*** For persons not previously vaccinated with hepatitis A vaccine, administer dose as soon as travel is considered and complete the series according to routine
schedule if the next dose is needed before travel.
†††
Can be administered on the basis of the providers risk assessment.
Recommendations and Reports
116 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
from pooled human plasma processed by cold ethanol
fractionation. In the United States, IG is produced only from
plasma that has tested negative for HBsAg, antibodies to HIV
and HCV, and HIV and HCV RNA. In addition, the process
used to manufacture IG inactivates viruses (e.g., HBV, HCV,
and HIV). When administered IM <2 weeks after exposure to
HAV, IG is >85% effective in preventing HAV infection (1308).
If IG is administered to persons for whom hepatitis A vaccine
also is recommended, a dose of vaccine should be provided
simultaneously with IG in different anatomic sites (e.g.,
different limbs) as soon as possible, and the second vaccine dose
should be administered according to the licensed schedule to
complete the series. The combined vaccine can be considered
for persons among whom both hepatitis A and hepatitis B
vaccine is recommended (13,1297,13021304).
Special Considerations
For persons with HIV infection, antibody response can be
directly related to CD4
+
T-cell levels. Although persons with
HIV who have lower CD4
+
T-cell counts or percentages might
have a weaker response to the vaccine, vaccination should
not be delayed for the CD4
+
T-cell count to exceed a certain
threshold because of the prolonged risk for HAV exposure
created by missed opportunities to vaccinate.
Hepatitis B Virus Infection
The incubation period for HBV infection from time of
exposure to symptom onset ranges from 6 weeks to 6 months.
The highest concentrations of HBV are located in blood, with
lower concentrations in other body fluids including wound
exudates, semen, vaginal secretions, and saliva (1309,1310).
HBV is more infectious and more stable in the environment
than other bloodborne pathogens (e.g., HCV or HIV).
HBV infection can be either self-limited or chronic.
Among adults, approximately half of newly acquired HBV
infections are symptomatic, and approximately 1% of reported
cases result in acute liver failure and death (1311). Risk for
chronic infection is inversely related to age at acquisition;
approximately 90% of infected infants and 30% of infected
children aged <5 years become chronically infected, compared
with 2%–6% of persons who become infected as adults (1312).
Among persons with chronic HBV infection, the risk for
premature death from cirrhosis or hepatocellular carcinoma
is 15%–25% (1313).
HBV is efficiently transmitted by percutaneous or mucous
membrane exposure to HBV-infected blood or body fluids
that contain HBV. The primary risk factors associated with
infection among adolescents and adults are unprotected sex
with an infected partner, having multiple partners, men having
sex with men, having history of other STIs, and injecting drug
use (233). In addition, studies have demonstrated other modes
of HBV transmission, including premastication and lapses
in health care infection control procedures, as less common
sources of transmission (13141317).
CDC’s national strategy for eliminating transmission of HBV
infection includes prevention of perinatal infection through
routine screening of all pregnant women for HBsAg and
immunoprophylaxis of infants born to mothers with HBsAg
or mothers whose HBsAg status is unknown, routine infant
vaccination, vaccination of previously unvaccinated children
and adolescents through age 18 years, and vaccination of
previously unvaccinated adults at increased risk for infection
(12). High vaccination coverage rates with subsequent
decreases in acute HBV infection incidence have been achieved
among infants and adolescents (1318). The vaccination of
persons as children and adolescents likely has led to improved
vaccination coverage among adults aged <30 years (1319) and
corresponding lower rates of acute HBV infection among this
group. In contrast, vaccination coverage among the majority
of adult populations at high risk aged ≥30 years (e.g., persons
with multiple sex partners, MSM, and injecting drug users)
has remained low (1320,1321); these groups account for the
highest rates of preventable acute infections (12,1319,1322).
STD clinics and other health care settings providing STI
services to adults at high risk for infection should administer
hepatitis B vaccine to those who are unvaccinated.
Diagnosis
Diagnosis of acute or chronic HBV infection requires
serologic testing (Table 5). Because HBsAg is present in both
acute and chronic infection, presence of IgM antibody to
hepatitis B core antigen (IgM anti-HBc) is diagnostic of acute
or recently acquired HBV infection. Antibody to HBsAg
(anti-HBs) is produced after a resolved infection and is the
only HBV antibody marker present after vaccination. The
presence of HBsAg and anti-HBc, with a negative test for IgM
anti-HBc, indicates chronic HBV infection. The presence of
total anti-HBc alone might indicate acute, resolved, or chronic
infection or a false-positive result.
Treatment
No specific therapy is available for persons with acute HBV
infection; treatment is supportive. Persons with chronic
HBV infection should be referred for evaluation to a provider
experienced in managing such infections. Therapeutic agents
approved by FDA for treatment of chronic HBV infection
can achieve sustained suppression of HBV replication and
remission of liver disease (1323).
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 117
US Department of Health and Human Services/Centers for Disease Control and Prevention
Prevention
Two products have been approved for HBV prevention:
hepatitis B immune globulin (HBIG) for PEP and hepatitis
B vaccine (12). HBIG provides temporary (i.e., 3–6 months)
protection from HBV infection and is typically used as
PEP as an adjunct to hepatitis B vaccination for previously
unvaccinated persons or for persons who have not responded to
vaccination. HBIG is prepared from plasma known to contain
high concentrations of anti-HBs. The recommended dose of
HBIG is 0.06 mL/kg body weight.
Hepatitis B vaccine contains HBsAg produced in yeast by
recombinant DNA technology and provides protection from
HBV infection when used for both pre-exposure vaccination
and PEP. The three available monovalent hepatitis B vaccines
for use in the United States are Recombivax HB, Engerix-B,
and Heplisav-B. A combination hepatitis A and hepatitis B
vaccine for use among persons aged ≥18 years, Twinrix, also
is available.
When selecting a hepatitis B vaccination schedule, health care
providers should consider the need to achieve completion of
the vaccine series. The recommended HBV dose and schedule
varies by product and age of recipient (Table 6). Three different
3-dose schedules for adolescents and adults have been approved
for both monovalent hepatitis B vaccines (i.e., Engerix-B and
Recombivax HB); these vaccines can be administered at 0, 1,
and 6 months; 0, 1, and 4 months; or 0, 2, and 4 months.
A 4-dose schedule of Engerix-B at 0, 1, 2, and 12 months is
licensed for all age groups. A 2-dose schedule of Recombivax
HB adult formulation (10 µg) is licensed for adolescents
aged 11–15 years, with a 4-month minimal interval between
doses. When scheduled to receive the second dose, adolescents
aged 16–19 years should be switched to a 3-dose series, with
doses 2 and 3 consisting of the pediatric formulation (5 µg)
administered on a recommended schedule. Heplisav-B is a
new single-antigen recombinant hepatitis B vaccine with a
novel cytosine-phosphate-guanine 1018 oligodeoxynucleotide
adjuvant for prevention of HBV infection among persons
aged ≥18 years, administered as a 2-dose series at 0 and
1 month (>4 weeks apart) (156). Twinrix is a 3-dose schedule
administered at 0, 1, and 6 months to persons aged ≥18 years
at risk for both HAV and HBV infections.
Hepatitis B vaccine should be administered IM in the deltoid
muscle and can be administered simultaneously with other
vaccines. If the vaccine series is interrupted after the first or
second dose of vaccine, the missed dose should be administered
as soon as possible. The series does not need to be restarted after
a missed dose. HBV vaccination is available for eligible children
and adolescents aged <19 years through the VFC program
(https://www.cdc.gov/vaccines/programs/vfc/contacts-state.
html). When feasible, the same manufacturer’s vaccines should
be used to complete the series; however, vaccination should
not be deferred when the manufacturer of the previously
administered vaccine is unknown or when the vaccine from
the same manufacturer is unavailable (1324).
Among adolescents and healthy adults aged <40 years,
approximately 30%–55% achieve a protective antibody response
(i.e., anti-HBs ≥10 mIU/mL) after the first single-antigen
vaccine dose, 75% after the second, and >90% after the third.
Recent clinical trials reported a protective antibody response
achieved among approximately 90% of participants receiving
Heplisav-B, compared with 70.5%–90.2% of participants
receiving Engerix-B (12). Vaccine-induced immune memory
has been demonstrated to persist for >30 years (13251327).
TABLE 5. Interpretation of serologic test results* for hepatitis B virus infection
Serologic marker
InterpretationHBSAG Total anti-HBc IgM anti-HBc Anti-HBs
Never infected
+
Early acute infection; transient (≤18 days) after vaccination
+ + + Acute infection
+ + Acute resolving infection
+ + Recovered from past infection and immune
+ + Chronic infection
+ Past infection; low-level chronic infection
§
; passive transfer to infant born to HBsAg-
positive mother; false positive (no infection)
+ Immune if concentration is >10 mIU/mL after vaccination, passive transfer after HBIG
administration
Source: Adapted from Schillie S, Vellozzi C, Reingold A, et al. Prevention of hepatitis B virus infection in the United States: recommendations of the Advisory Committee
on Immunization Practices. MMWR Recomm Rep 2018;67(No. RR-1).
Abbreviations: anti-HBc = antibody to hepatitis B core antigen; anti-HBs = antibody to hepatitis B surface antigen; HBIG = hepatitis B immune globulin; HBsAg =
hepatitis B surface antigen; IgM = immunoglobulin M.
* − = negative test result; + = positive test result.
To ensure that an HBsAg-positive test result is not false positive, samples with repeatedly reactive HBsAg results should be tested with a neutralizing confirmatory
test cleared by the Food and Drug Administration.
§
Persons positive for only anti-HBc are unlikely to be infectious, except under unusual circumstances involving direct percutaneous exposure to large quantities of
blood (e.g., blood transfusion or organ transplantation) or mutant HBsAg-related infection.
Recommendations and Reports
118 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Periodic testing to determine antibody levels after routine
vaccination among immunocompetent persons is unnecessary,
and booster doses of vaccine are not recommended.
Hepatitis B vaccination is usually well tolerated by the
majority of recipients. Pain at the injection site and low-grade
fever are reported by a minority of recipients. For children
and adolescents, a causal association exists between receipt of
hepatitis B vaccination and anaphylaxis. For each 1.1 million
doses of vaccine administered, approximately one recipient will
experience this type of reaction (1328); however, no deaths have
been reported among these patients (1318,1328). Vaccine is
contraindicated for persons with a history of anaphylaxis after a
previous dose of hepatitis B vaccine and persons with a known
anaphylactic reaction to any vaccine component (1329). No
other adverse events after administration of hepatitis B vaccine
have been demonstrated.
Pre-Exposure Vaccination
Hepatitis B vaccination is recommended for all unvaccinated
children and adolescents; all unvaccinated adults at risk for HBV
infection, especially injecting drug users; MSM; adults with
multiple sex partners; sex partners, needle-sharing contacts,
or household contacts of persons with chronic hepatitis B;
and persons with diabetes and all adults seeking protection
from HBV infection (1318). For adults, acknowledgment of
a specific risk factor is not a requirement for vaccination.
Hepatitis B vaccine should be routinely offered to all
unvaccinated persons attending STD clinics and to all
unvaccinated persons seeking evaluation or treatment for STIs
in other settings, especially correctional facilities, facilities
providing substance misuse treatment and prevention services,
Federally Qualified Health Centers, and settings serving MSM
(e.g., HIV infection care and prevention settings). If hepatitis
B vaccine is unavailable at a particular facility, persons should
be linked to a setting where they can receive vaccine. Persons
with a reliable vaccination history (i.e., a written, dated record
of each dose of a complete series) or reliable history of hepatitis
B infection (i.e., a written record of infection and serologic
results providing evidence of previous infection) do not require
vaccination. In all settings, vaccination should be initiated
at the initial visit, even if concerns about completion of the
vaccine series exist.
Prevaccination Serologic Testing
Conducting prevaccination serologic testing for susceptibility
just before the initial vaccine dose is administered can
be considered for identifying persons with chronic HBV
infection and, potentially, reducing the cost of completing the
vaccination series for adult populations that have an expected
high prevalence (20%–30%) of HBV infection (e.g., injecting
drug users and MSM, especially those among older age groups,
or persons born where HBV endemicity is moderate to
high). In addition, prevaccination testing for susceptibility is
TABLE 6. Recommended doses of licensed formulations of hepatitis B vaccines
Age group (yrs)
Single-antigen vaccine Combination vaccine
Recombivax HB Engerix-B Heplisav-B* Twinrix
Dose (μg)
§
Volume (mL) Dose (μg)
§
Volume (mL) Dose (μg)
§
Volume (mL) Dose (μg)
§
Volume (mL)
Infants (<1) 5 0.5 10 0.5
NA NA
Children (1–10) 5 0.5 10 0.5
NA NA
Adolescents (11–15) 10** 1.0 NA NA
NA NA
Adolescents (11–19) 5 0.5 10 0.5
NA NA
Adults (≥18)
††
††
††
††
20* 0.5 20
1
Adults (≥20) 10 1.0 20 1.0 20
0.5 20
1
Hemodialysis patients and other
immunocompromised persons (<20
§§
)
5 0.5 10 0.5 20 0.5 NA NA
Hemodialysis patients and other
immunocompromised persons (≥20)
40
¶¶
1.0 40*** 2.0 20 0.5 NA NA
Source: Adapted from Schillie S, Vellozzi C, Reingold A, et al. Prevention of hepatitis B virus infection in the United States: recommendations of the Advisory Committee
on Immunization Practices. MMWR Recomm Rep 2018;67(No. RR-1).
Abbreviation: NA = not applicable.
*
Administered on a 2-dose schedule.
Combined hepatitis A and B vaccines. This vaccine is recommended for persons aged ≥18 years who are at increased risk for both hepatitis B and hepatitis A virus
infections.
§
Recombinant hepatitis B surface antigen protein dose.
Heplisav-B should not be used for vaccination of infants, children, or adolescents because the safety and effectiveness of Heplisav-B has not been established in
persons aged <8 years and is not approved for use in these populations.
**
Adult formulation administered on a 2-dose schedule.
††
Engerix-B and Recombivax HB are approved for use in persons of all ages.
§§
Higher doses might be more immunogenic; however, no specific recommendations have been made.
¶¶
Dialysis formulation administered on a 3-dose schedule at 0, 1, and 6 months.
*** Two 1.0-mL doses administered at one site, on a 4-dose schedule at 0, 1, 2, and 6 months.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 119
US Department of Health and Human Services/Centers for Disease Control and Prevention
recommended for unvaccinated household, sexual, and needle-
sharing contacts of HBsAg-positive persons (1318). Serologic
testing should not be a barrier to vaccination. The first vaccine
dose should be administered immediately after collection of the
blood sample for serologic testing. Vaccination of persons who
are immune to HBV infection because of current or previous
infection or vaccination is not harmful and does not increase
the risk for adverse events.
Prevaccination testing should be performed with HBsAg,
anti-HBs, and total anti-HBc to define patients’ HBV clinical
status and deliver recommended care (1330). Persons who
test HBsAg positive should receive prevention counseling and
evaluation for antiviral treatment (see Management of Persons
Who Are HBsAg Positive). Persons who test total anti-HBc
positive and anti-HBs positive should be counseled that they
have had previous HBV infection and are immune. Those
persons with isolated anti-HBc (i.e., negative HBsAg and
anti-HBs) need further assessment to rule out occult HBV
infection, and they are at higher risk for reactivation if exposed
to immunosuppressants. Persons who test negative to all three
HBV seromarkers should receive the complete vaccination
series, with the first vaccine dose administered immediately.
Postvaccination Serologic Testing for Response
Postvaccination serologic testing for immunity is unnecessary
after routine vaccination of adolescents or adults. However,
such testing is recommended for persons whose subsequent
clinical management depends on knowledge of their immune
status. Persons recommended to receive postvaccination
serologic testing include health care personnel and public safety
workers, persons with HIV infection, sex and needle-sharing
partners of HBsAg-positive persons, hemodialysis patients
and others who might require outpatient hemodialysis (e.g.,
predialysis, peritoneal dialysis, or home dialysis), and other
immunocompromised persons (e.g., hematopoietic stem-cell
transplant recipients or persons receiving chemotherapy) (1318).
If indicated, anti-HBs testing should be performed
1–2 months after administration of the last dose of the
vaccine series. Persons determined to have anti-HBs levels
of <10 mIU/mL after the primary vaccine series should be
revaccinated with a 3-dose series and tested again for anti-HBs
1–2 months after the third dose. Persons who do not respond to
revaccination should be tested for HBsAg and HBc. If HBsAg
positive, persons should receive recommended management
(see Management of Persons Who Are HBsAg Positive). If
HBsAg negative, persons should be considered susceptible to
HBV infection and counseled about precautions for preventing
HBV infection and the need for HBIG PEP for any known
exposure. If isolated anti-HBc positive (i.e., negative HBsAg
and anti-HBs), persons will need further assessment to rule out
occult HBV infection and are at higher risk for reactivation if
exposed to immunosuppressants.
Postexposure Prophylaxis
Both passive and active PEP (simultaneous administration of
HBIG [i.e., 0.06 mL/kg body weight] and hepatitis B vaccine
at separate anatomic sites) and active PEP (administration of
hepatitis B vaccination alone) have been demonstrated to be
highly effective in preventing transmission after exposure to
HBV (12). HBIG alone also has been demonstrated to be
effective in preventing HBV transmission; however, with the
availability of hepatitis B vaccine, HBIG typically is used as
an adjunct to vaccination.
Exposure to a Source Who Is HBsAg Positive
Unvaccinated persons or persons known not to have
responded to a complete hepatitis B vaccine series should
receive both HBIG and hepatitis vaccine as soon as possible
(preferably ≤24 hours) after a discrete, identifiable exposure
to blood or body fluids that contain blood from a person with
HBsAg (Table 7). Hepatitis B vaccine should be administered
simultaneously with HBIG at a separate anatomic site, and the
vaccine series should be completed by using the age-appropriate
vaccine dose and schedule (Table 6). Exposed persons who
are not fully vaccinated because they have not completed the
vaccine series should receive HBIG (i.e., 0.06 mL/kg body
weight) and complete the vaccine series. Persons who have
written documentation of a complete hepatitis B vaccine series
who did not receive postvaccination testing should receive a
single vaccine booster dose. Exposed persons who are known
to have responded to vaccination by postvaccination testing
are considered protected; therefore, they need no additional
doses of vaccine or HBIG. All persons with an occupational
exposure to blood or body fluids that contain HBV should be
managed according to guidelines (12).
Exposure to a Source with Unknown HBsAg Status
Unvaccinated persons and persons with previous nonresponse
to hepatitis B vaccination who have a discrete, identifiable
exposure to blood or body fluids containing blood from
a person with unknown HBsAg status should receive the
hepatitis B vaccine series, with the first dose initiated as soon
as possible after exposure (preferably <24 hours) and the series
completed according to the age-appropriate dose and schedule.
Exposed persons who are not fully vaccinated but started the
series should complete the vaccine series. Exposed persons
with written documentation of a complete hepatitis B vaccine
series who did not receive postvaccination testing require no
further treatment.
Recommendations and Reports
120 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Other Management Considerations
All persons with HBV infection should be tested for HIV,
syphilis, gonorrhea, and chlamydia.
Management of Persons Who Are HBsAg Positive
Recommendations for management of all persons with
HBsAg include the following:
All persons with HBsAg documented on laboratory results
should be reported to the state or local health department.
To verify the presence of chronic HBV infection, persons
with HBsAg should be retested. The absence of IgM anti-
HBc or the persistence of HBsAg for ≥6 months indicates
chronic HBV infection.
Persons with chronic HBV infection should be referred
for evaluation to a specialist experienced in managing
chronic hepatitis B infection.
Household, sexual, and needle-sharing contacts of persons
with chronic infection should be evaluated. Unvaccinated
sex partners and household and needle-sharing contacts
should be tested for susceptibility to HBV infection and
receive the first dose of hepatitis B vaccine immediately
after collection of the blood sample for serologic testing
(see Prevaccination Serologic Testing). Susceptible persons
should complete the vaccine series by using an age-
appropriate vaccine dose and schedule.
Sex partners of persons with HBsAg should be counseled
to use latex condoms (1331) to protect themselves from
sexual exposure to infectious body fluids (e.g., semen and
vaginal secretions), unless they have been demonstrated
to be immune after vaccination (anti-HBs ≥10 mIU/mL)
or previously infected (anti-HBc positive).
To prevent or reduce the risk for transmission to others in
addition to vaccination, persons with HBsAg also should
be advised to
ű
use methods (e.g., condoms) to protect nonimmune sex
partners from acquiring HBV infection from sexual
activity until the partner can be vaccinated and
immunity documented;
ű
cover cuts and skin lesions to prevent spread by
infectious secretions or blood;
ű
refrain from donating blood, plasma, body organs, other
tissue, or semen; and
ű
refrain from sharing household articles (e.g.,
toothbrushes, razors, or personal injecting equipment)
that could become contaminated with blood, and refrain
from premastication of food.
To protect the liver from further harm, persons with
HBsAg should be advised to
TABLE 7. Guidelines for postexposure prophylaxis* of persons with nonoccupational exposure
to blood or body fluids that contain blood, by
exposure type and hepatitis B vaccination status
Source of exposure Unvaccinated person
§
Previously vaccinated person
HBsAg-positive source
Percutaneous (e.g., bite or needlestick) or mucosal
exposure to HBsAg-positive blood or body fluids
or
Sex or needle-sharing contact with an HBsAg-
positive person
or
Victim of sexual assault or abuse by an assailant
who is HBsAg positive
Administer hepatitis B vaccine series and HBIG Complete hepatitis B vaccine series and HBIG, if
vaccine series not completed
or
Administer hepatitis B vaccine booster dose, if
previous vaccination without testing**
Source with unknown HBsAg status
Percutaneous (e.g., bite or needlestick) or mucosal
exposure to potentially infectious blood or body
fluids from a source with unknown HBsAg status
or
Sex or needle-sharing contact with person with
unknown HBsAg status
or
Victim of sexual assault or abuse by a perpetrator
with unknown HBsAg status
Administer hepatitis B vaccine series Complete hepatitis B vaccine series
Sources: CDC. CDC guidance for evaluating health-care personnel for hepatitis B virus protection and for administering postexposure management. MMWR Recomm
Rep 2013;62(No. RR-10); CDC. Postexposure prophylaxis to prevent hepatitis B virus infection. MMWR Recomm Rep 2006;55(No. RR-16).
Abbreviations: HBIG = hepatitis B immune globulin; HBsAg = hepatitis B surface antigen.
* When indicated, immunoprophylaxis should be initiated as soon as possible, preferably within 24 hours. Studies are limited regarding the maximum interval after
exposure during which postexposure prophylaxis is effective, but the interval is unlikely to exceed 7 days for percutaneous exposures or 14 days for sexual exposures.
The hepatitis B vaccine series should be completed. These guidelines apply to nonoccupational exposures.
These guidelines apply to nonoccupational exposures.
§
A person who is in the process of being vaccinated but who has not completed the vaccine series should complete the series and receive treatment for hepatitis
B as indicated.
A person who has written documentation of a complete hepatitis B vaccine series and who did not receive postvaccination testing.
** No booster dose is needed for persons who have written documentation of hepatitis B vaccine series with serologic response.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 121
US Department of Health and Human Services/Centers for Disease Control and Prevention
ű
avoid or limit alcohol consumption because of the effects
of alcohol on the liver;
ű
refrain from starting any new medicines, including over-
the-counter and herbal medicines, without checking
with their health care provider; and
ű
obtain vaccination against hepatitis A.
When seeking medical or dental care, persons who are
HBsAg positive should be advised to inform their health care
providers of their HBsAg status so that they can be evaluated
and managed. The following are key counseling messages for
persons with HBsAg:
HBV is not usually spread by hugging, coughing, food or water,
sharing eating utensils or drinking glasses, or casual contact.
Persons should not be excluded from work, school, play,
childcare, or other settings because they are infected with HBV.
Involvement with a support group might help patients
cope with chronic HBV infection.
HBV infection is a chronic condition that can be treated,
and patients should receive prevention counseling and be
evaluated for antiviral treatment.
Special Considerations
Pregnancy
Regardless of whether they have been previously tested or
vaccinated, all pregnant women should be tested for HBsAg
at the first prenatal visit and again at delivery if at high
risk for HBV infection (see STI Detection Among Special
Populations). Pregnant women at risk for HBV infection and
without documentation of a complete hepatitis B vaccine series
should receive hepatitis B vaccination. All pregnant women
with HBsAg should be reported to state and local perinatal
hepatitis B prevention programs and referred to a specialist.
Information about management of pregnant women with
HBsAg and their infants is available at https://www.cdc.gov/
hepatitis/hbv/perinatalxmtn.htm.
HIV Infection
HIV infection can impair the response to hepatitis B
vaccination. Persons with HIV should be tested for anti-HBs
1–2 months after the third vaccine dose (see Postvaccination
Serologic Testing). Modified dosing regimens, including a
doubling of the standard antigen dose and administration of
additional doses, might increase the response rate and should be
managed in consultation with an infectious disease specialist.
Additional recommendations for management of persons with
HBsAg and HIV infection are available (98).
Hepatitis C Virus Infection
HCV infection is the most common chronic bloodborne
infection in the United States, with an estimated 2.4 million
persons living with chronic infection (1332). HCV is not
efficiently transmitted through sex (13331335). Studies of
HCV transmission between heterosexual couples and MSM
have yielded mixed results; however, studies have reported
either no or minimally increased rates of HCV infection
among partners of persons with HCV infection compared
with partners of those without HCV (1334,13361338).
However, data indicate that sexual transmission of HCV
can occur, especially among persons with HIV infection.
Increasing incidence of acute HCV infection among MSM
with HIV infection has been reported in multiple U.S.
(96,236,239,1339) and European cities (237,13401342). A
recent systematic review reported an HCV incidence of 6.35
per 1,000 person years among MSM with HIV infection
(1343). An association exists with high-risk and traumatic
sexual practices (e.g., condomless receptive anal intercourse
or receptive fisting) and concurrent genital ulcerative disease
or STI-related proctitis (237,1342). HCV transmission among
MSM with HIV infection has also been associated with group
sex and chemsex (i.e., using recreational drugs in a sexual
context) (13441348). Shedding of HCV in the semen and in
the rectum of men with HIV infection has been documented
(1349,1350). Certain studies have revealed that risk increases
commensurate with increasing numbers of sex partners among
heterosexual persons (1337,1338,13511353) and MSM with
HIV infection (1349,13541357), especially if their partners are
also coinfected with HIV (237,1340,13541356,1358). More
recently, acute HCV infections have been reported among MSM
on PrEP, increasing concerns that certain MSM might be at
increased risk for incident HCV infection through condomless
sexual intercourse with MSM with HCV infection (1359,1360).
Persons newly infected with HCV typically are either
asymptomatic or have a mild clinical illness. HCV RNA can
be detected in blood within 1–3 weeks after exposure. The
average time from exposure to antibody to HCV (anti-HCV)
seroconversion is 4−10 weeks, and anti-HCV can be detected
among approximately 97% of persons by 6 months after
exposure (13611364) (https://www.cdc.gov/hepatitis/hcv/
hcvfaq.htm#section3).
Chronic HCV infection develops among 75%–85% of
persons with HCV infection (1365,1366), and 10%–20% of
persons with chronic infection develop cirrhosis in 20–30 years
of active liver disease (1367). The majority of infected persons
remain unaware of their infection because they are not clinically
Recommendations and Reports
122 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
ill. However, infected persons are a source of transmission to
others and are at risk for cirrhosis and hepatocellular carcinoma
decades after infection.
HCV is primarily transmitted parenterally, usually through
shared drug-injecting needles and paraphernalia. HCV also
can be transmitted through exposures in health care settings
as a consequence of inadequate infection control practices
(1314). Transmission after receipt of blood from donors and
from transplantation of tissues and organs with HCV infection
has occurred only rarely since 1992, when routine screening
of these donated products was mandated in the United
States (1367,1369). Tattoos applied in regulated settings
have not been associated with HCV transmission, although
those obtained in certain settings have been linked to such
transmission (1336). Occupational and perinatal exposures also
can result in transmission of HCV; however, such transmission
is uncommon.
Acute HCV infection is a reportable condition in 49 states.
Matching viral hepatitis and HIV surveillance registries, and
molecular epidemiologic assessments, can facilitate early
detection of social networks of HCV transmission among
MSM with HIV infection.
CDC recommends hepatitis C screening at least once in a
lifetime for all adults aged ≥18 years and for all women during
each pregnancy, except in settings where the prevalence of
HCV infection is <0.1% (156). One-time hepatitis C testing
is also recommended regardless of age, setting, or recognized
conditions or exposures (e.g., HIV infection, history of
injecting drug use, or children born to women with HCV
infection). Routine periodic HCV testing is recommended
for persons with ongoing risk factors (e.g., injecting drug use
or hemodialysis).
Diagnosis
Testing for HCV infection should include use of an FDA-
cleared test for antibody to HCV (i.e., immunoassay, EIA, or
enhanced CIA and, if recommended, a supplemental antibody
test) followed by NAAT to detect HCV RNA for those with a
positive antibody result (1370). Persons with HIV infection with
low CD4
+
T-cell count might require further testing by NAAT
because of the potential for a false-negative antibody assay.
Persons determined to have HCV infection (i.e., positive
for HCV RNA) should be evaluated for treatment. Antibody
to HCV remains positive after spontaneously resolving or
successful treatment; therefore, subsequent testing for HCV
reinfection among persons with ongoing risk factors should
be limited to HCV RNA. Persons who have spontaneous
resolution or who have undergone successful treatment are
not immune to reinfection.
Treatment
HCV infection is curable, and persons with diagnosed HCV
infection should be linked to care and treatment. Providers
should consult existing guidelines to learn about the latest
advances in treating HCV infection (https://www.hcvguidelines.
org) and with hepatitis specialists, as needed. Persons at high
risk for transmitting HCV to others should be treated both for
individual benefit and to prevent HCV transmission.
Management of Sex Partners
Because incident HCV has not been demonstrated to
occur among heterosexual couples followed over time
(1334,13711373), condom use might not be necessary in
such circumstances. Persons with HCV infection with one
long-term, steady sex partner do not need to change their
sexual practices. However, they should discuss the risk for
transmission with their partner and discuss the need for
testing (234) (https://www.cdc.gov/hepatitis/hcv/index.htm).
Heterosexual persons and MSM with HCV infection and
more than one partner, especially those with concurrent HIV
infection, should protect their partners against HCV and HIV
acquisition by using external latex condoms (237,1358,1374)
and HIV PrEP. Partners of persons with HCV and HIV should
be tested for both infections.
Other Management Considerations
All persons with HCV infection for whom HIV and
HBV infection status is unknown should be tested for these
infections. Those who have HIV or HBV infection should be
referred for or provided with recommended care and treatment.
Persons without previous exposure to HAV or HBV should
be vaccinated.
Prevention
Reducing the burden of HCV infection and disease in
the United States requires implementing both primary and
secondary prevention activities. Primary prevention reduces or
eliminates HCV transmission, whereas secondary prevention
identifies persons through screening and then provides
treatment to reduce chronic liver disease and other chronic
diseases and HCV transmission. No vaccine for hepatitis
C is available, and prophylaxis with IG is not effective in
preventing HCV infection after exposure. PEP using direct-
acting antivirals is not recommended.
Persons with HCV infection should be provided information
about how to protect their liver from further harm (i.e.,
hepatotoxic agents); for instance, persons with HCV infection
should be advised to avoid drinking alcohol and taking any new
medicines, including over-the-counter or herbal medications,
without checking with their clinician. In addition, a need for
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 123
US Department of Health and Human Services/Centers for Disease Control and Prevention
hepatitis A and B vaccination should be determined; persons
who are not immune should be vaccinated.
To reduce the risk for transmission to others, persons with HCV
infection should be advised not to donate blood, body organs,
other tissue, or semen; not to share any personal items that might
have blood on them (e.g., toothbrushes or razors); and to cover
cuts and sores on the skin to keep the virus from spreading by
blood or secretions. Women with HCV infection do not need
to avoid pregnancy or breastfeeding, although children born to
women with HCV also should be tested for HCV.
Persons who use or inject drugs should be counseled about
the importance of prevention and provided access to substance
misuse treatment, including medication-assisted treatment,
if indicated. Persons who inject drugs should be encouraged
to take the following additional steps to reduce personal and
public health risks:
Never reuse or share syringes, water, or drug preparation
equipment.
Only use syringes obtained from a reliable source (e.g., a
syringe services program or a pharmacy).
Use a new, sterile syringe to prepare and inject drugs each time.
If possible, use sterile water to prepare drugs; otherwise,
use clean water from a reliable source (e.g., fresh tap water).
Use a new or disinfected container (i.e., cooker) and a new
filter (i.e., cotton) to prepare drugs.
Clean the injection site with a new alcohol swab before
injection.
Safely dispose of syringes after one use.
Postexposure Follow-Up
No PEP has been demonstrated to be effective against HCV
infection. Testing for HCV is recommended for health care
workers after percutaneous or perimucosal exposures to HCV-
positive blood. Prompt identification of acute infection is vital
because outcomes are improved when treatment is initiated
early during the illness course.
Special Considerations
Pregnancy
All pregnant women should be screened with each pregnancy
for HCV antibodies at the first prenatal visit in settings
where the HCV prevalence is >0.1% (https://www.cdc.gov/
hepatitis/hcv/index.htm) (154,155). Although the rate of
transmission is highly variable, more than six of every 100
infants born to women with HCV infection become infected;
this infection occurs predominantly during or near delivery,
and no treatment or delivery method (e.g., cesarean delivery)
has been demonstrated to decrease this risk (1375). However,
the risk is increased by the presence of maternal HCV viremia
at delivery and is twofold to threefold greater if the woman has
HIV infection. Although no recommendations are available
for HCV treatment during pregnancy, discussion about the
individual risks and benefits of postpartum treatment can be
considered in accordance with existing guidance (https://www.
hcvguidelines.org/unique-populations/pregnancy).
HCV has not been reported to be transmitted through breast
milk, although mothers with HCV infection should consider
abstaining from breastfeeding if their nipples are cracked or
bleeding. Infants born to mothers with HCV infection should
be tested for HCV infection; children should be tested for
anti-HCV no sooner than age 18 months because anti-HCV
from the mother might last until that age. If diagnosis is desired
before the child reaches age 18 months, testing for HCV
RNA can be performed at or after the infant’s first well-child
visit at age 1–2 months. HCV RNA testing can be repeated
at a subsequent visit, independent of the initial HCV RNA
test result (1376) (https://www.cdc.gov/hepatitis/hcv/hcvfaq.
htm#section3).
HIV Infection
All persons with HIV infection should undergo serologic
screening for HCV at initial evaluation (98) (https://www.
hcvguidelines.org). Providers should be aware of the likelihood
that MSM with HIV infection can acquire HCV after initial
screening. Because acute HCV infection acquisition among
persons with HIV infection can occur, especially among
MSM, and regular screening of those with HIV is cost-
effective (238,239,1377), periodic HCV screening should be
conducted (13781380). For persons with HIV infection,
hepatitis C screening with HCV antibody assays (followed
by HCV RNA if antibody positive) can be considered at least
yearly, for those at high risk for infection, and more frequently
depending on specific circumstances (e.g., community HCV
infection prevalence and incidence, high-risk sexual behavior,
and concomitant ulcerative STIs and proctitis). Antibody to
HCV remains positive after spontaneously resolved infection
or successful treatment; therefore, subsequent testing for
potential HCV reinfection among persons with ongoing risk
should be limited to HCV RNA testing only. Indirect testing
(e.g., alanine aminotransferase [ALT]) is not recommended
for detecting incident HCV infections because such testing,
especially if performed once a year, can miss persons who have
reverted after acute HCV infection to a normal ALT level at
the time of testing (239) (https://www.hcvguidelines.org).
Conversely, ALT can be elevated by antiretroviral and other
medications, alcohol, and toxins. If ALT levels are being
monitored, persons with HIV infection who experience new
or unexplained increases in ALT should be tested for acute
Recommendations and Reports
124 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
HCV infection and evaluated for possible medication toxicity
or excessive alcohol use.
Continued unprotected sexual contact between partners
with HIV can facilitate spread of HCV infection because
the virus can be recovered from the semen of men with HIV
infection (1349,1381). Specific prevention practices (e.g.,
barrier precautions that limit contact with body fluids during
sexual contact with other MSM) should be discussed.
Because a minimal percentage of persons with HIV infection
do not develop HCV antibodies, HCV RNA testing should
be performed for persons with HIV infection and unexplained
liver disease who are anti-HCV negative. The course of liver
disease is more rapid among persons with HIV and HCV, and
the risk for cirrhosis is higher than that for persons with HCV
infection alone.
Proctitis, Proctocolitis, and Enteritis
Sexually transmitted gastrointestinal syndromes include
proctitis, proctocolitis, and enteritis. Evaluation for these
syndromes should include recommended diagnostic
procedures, including anoscopy or sigmoidoscopy, stool
examination for WBCs, and microbiologic workup (e.g.,
gonorrhea, chlamydia [LGV PCR if available], herpes simplex
NAAT, and syphilis serology). For those with enteritis, stool
culture or LGV PCR also is recommended.
Proctitis is inflammation of the rectum (i.e., the distal
10–12 cm) that can be associated with anorectal pain,
tenesmus, or rectal discharge. Fecal leukocytes are common.
Proctitis occurs predominantly among persons who have
receptive anal exposures (oral-anal, digital-anal, or genital-
anal). N. gonorrhoeae, C. trachomatis (including LGV serovars),
HSV, and T. pallidum are the most common STI pathogens.
Genital HSV and LGV proctitis are more prevalent among
persons with HIV infection (545,556,1382). M. genitalium
has been detected in certain cases of proctitis and might be
more common among persons with HIV infection (937,1382).
N. meningitidis has been identified as an etiology of proctitis
among MSM with HIV infection (1383).
Proctocolitis is associated with symptoms of proctitis,
diarrhea or abdominal cramps, and inflammation of the colonic
mucosa extending to 12 cm above the anus. Fecal leukocytes
might be detected on stool examination, depending on the
pathogen. Proctocolitis can be acquired through receptive anal
intercourse or by oral-anal contact, depending on the pathogen.
Pathogenic organisms include Campylobacter species,
Shigella species, E. histolytica, LGV serovars of C. trachomatis,
and T. pallidum. Among immunosuppressed persons with
HIV infection, CMV or other opportunistic agents should
be considered. The clinical presentation can be mistaken for
inflammatory bowel disease or malignancy, resulting in a
delayed diagnosis (1384,1385).
Enteritis usually results in diarrhea and abdominal cramping
without signs of proctitis or proctocolitis. Fecal leukocytes
might be detected on stool examination, depending on the
pathogen. When outbreaks of gastrointestinal illness occur
among social or sexual networks of MSM, clinicians should
consider sexual transmission as a mode of spread and provide
counseling accordingly. Sexual practices that can facilitate
transmission of enteric pathogens include oral-anal contact or,
in certain instances, direct genital-anal contact. G. lamblia is the
most frequently implicated parasite, and bacterial pathogens
include Shigella species, Salmonella, E. coli, Campylobacter
species, and Cryptosporidium. Outbreaks of Shigella species,
Campylobacter, Cryptosporidium, and microsporidiosis have
been reported among MSM (259,274,1386,1387). Multiple
enteric pathogens and concurrent STIs have also been reported.
Among immunosuppressed persons with HIV infection, CMV
or other opportunistic pathogens should be considered.
Diagnostic and Treatment Considerations
for Acute Proctitis
Diagnosis
Persons with symptoms of acute proctitis should be examined
by anoscopy. A Gram-stained smear of any anorectal exudate
from anoscopic or anal examination should be examined for
polymorphonuclear leukocytes. All persons should be evaluated
for herpes simplex (preferably by NAAT of rectal lesions),
N. gonorrhoeae (NAAT or culture), C. trachomatis (NAAT),
and T. pallidum (darkfield of lesion if available and serologic
testing). If the C. trachomatis NAAT test is positive on a rectal
swab and severe symptoms associated with LGV are present
(including rectal ulcers, anal discharge, bleeding, ≥10 WBCs
on Gram stain, and tenesmus), patients should be treated
empirically for LGV. Molecular testing for LGV is not widely
available or not FDA cleared, and results are not typically
available in time for clinical decision-making. However, if
available, molecular PCR testing for C. trachomatis serovars
L1, L2, or L3 can be considered for confirming LGV (553).
The pathogenic role of M. genitalium in proctitis is unclear.
For persons with persistent symptoms after standard treatment,
providers should consider testing for M. genitalium with NAAT
and treat if positive (see Mycoplasma genitalium).
Treatment
Acute proctitis among persons who have anal exposure
through oral, genital, or digital contact is usually sexually
acquired (1382,1388). Presumptive therapy should be
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 125
US Department of Health and Human Services/Centers for Disease Control and Prevention
initiated while awaiting results of laboratory tests for
persons with anorectal exudate detected on examination or
polymorphonuclear leukocytes detected on a Gram-stained
smear of anorectal exudate or secretions. Such therapy also
should be initiated when anoscopy or Gram stain is not
available and the clinical presentation is consistent with acute
proctitis for persons reporting receptive anal exposures.
Recommended Regimen for Acute Proctitis
Ceftriaxone 500 mg* IM in a single dose
plus
Doxycycline 100 mg orally 2 times/day for 7 days
* For persons weighing ≥150 kg, 1 g of ceftriaxone should be administered.
Doxycycline course should be extended to 100 mg orally 2 times/day for
21 days in the presence of bloody discharge, perianal or mucosal ulcers,
or tenesmus and a positive rectal chlamydia test.
Bloody discharge, perianal ulcers, or mucosal ulcers among
persons with acute proctitis and rectal chlamydia (NAAT)
should receive presumptive treatment for LGV with an
extended course of doxycycline 100 mg orally 2 times/day for
3 weeks (1389,1390) (see Lymphogranuloma Venereum). If
painful perianal ulcers are present or mucosal ulcers are
detected on anoscopy, presumptive therapy should also include
a regimen for genital herpes (see Genital Herpes).
Diagnostic and Treatment Considerations
for Proctocolitis or Enteritis
Treatment for proctocolitis or enteritis should be directed
to the specific enteric pathogen identified. Multiple stool
examinations might be necessary for detecting Giardia,
and special stool preparations are required for diagnosing
cryptosporidiosis and microsporidiosis. Diagnostic and
treatment recommendations for all enteric infections are beyond
the scope of these guidelines. Providers should be aware of the
potential for antimicrobial-resistant pathogens, particularly
during outbreaks of Shigella and Campylobacter among sexual
networks of MSM where increased resistance to azithromycin,
fluoroquinolones, and isolates resistant to multiple antibiotics
have been described (266,272,273,1391,1392).
Other Management Considerations
To minimize transmission and reinfection, patients treated
for acute proctitis should be instructed to abstain from sexual
intercourse until they and their partners have been treated
(i.e., until completion of a 7-day regimen and symptoms
have resolved). Studies have reported that behaviors that
facilitate enteric pathogen transmission might be associated
with acquisition of other STIs, including HIV infection.
All persons with acute proctitis and concern for sexually
transmitted proctocolitis or enteritis should be tested for HIV,
syphilis, gonorrhea, and chlamydia (at other exposed sites).
PEP should be considered for exposures that present a risk for
HIV acquisition. For ongoing risk for HIV acquisition, PrEP
should be considered.
Evidence-based interventions for preventing acquisition
of sexually transmitted enteric pathogens are not available.
However, extrapolating from general infection control practices
for communicable diseases and established STI prevention
practices, recommendations include avoiding contact with
feces during sex, using barriers, and washing hands after
handing materials that have been in contact with the anal
area (i.e., barriers and sex toys) and after touching the anus
or rectal area.
Follow-Up
Follow-up should be based on specific etiology and severity
of clinical symptoms. For proctitis associated with gonorrhea
or chlamydia, retesting for the respective pathogen should be
performed 3 months after treatment.
Management of Sex Partners
Partners who have had sexual contact with persons treated
for gonorrhea or chlamydia <60 days before the onset of
the persons symptoms should be evaluated, tested, and
presumptively treated for the respective infection. Partners
of persons with proctitis should be evaluated for any diseases
diagnosed in the index partner. Sex partners should abstain
from sexual contact until they and their partners are treated.
No specific recommendations are available for screening
or treating sex partners of persons with diagnosed sexually
transmitted enteric pathogens; however, partners should seek
care if symptomatic.
Special Considerations
Drug Allergy, Intolerance, and Adverse Reactions
Allergic reactions with third-generation cephalosporins (e.g.,
ceftriaxone) are uncommon among persons with a history of
penicillin allergy (620,631,658,896).
HIV Infection
Persons with HIV infection and acute proctitis might present
with bloody discharge, painful perianal ulcers, or mucosal
ulcers and LGV and herpes proctitis are more prevalent among
this population. Presumptive treatment in such cases should
include a regimen for genital herpes and LGV.
Recommendations and Reports
126 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Ectoparasitic Infections
Pediculosis Pubis
Persons who have pediculosis pubis (i.e., pubic lice) usually
seek medical attention because of pruritus or because they
notice lice or nits on their pubic hair. Pediculosis pubis is
caused by the parasite Phthirus pubis and is usually transmitted
by sexual contact (1393).
Diagnosis
The clinical diagnosis is based on typical symptoms of itching
in the pubic region. Lice and nits can be observed on pubic hair.
Treatment
Recommended Regimens for Pediculosis Pubis
Permethrin 1% cream rinse applied to affected areas and washed off
after 10 minutes
or
Pyrethrin with piperonyl butoxide applied to the affected area and
washed off after 10 minutes
Alternative Regimens
Malathion 0.5% lotion applied to affected areas and washed off after
8–12 hours
or
Ivermectin 250 µg/kg body weight orally, repeated in 7–14 days
Reported resistance to pediculcides (permethrin and
pyrethrin) has been increasing and is widespread (1394,1395).
Malathion can be used when treatment failure is believed to
have occurred as a result of resistance. The odor and longer
duration of application associated with malathion therapy
make it a less attractive alternative compared with the
recommended pediculicides. Ivermectin has limited ovicidal
activity (1396). Ivermectin might not prevent recurrences
from eggs at the time of treatment, and therefore treatment
should be repeated in 7–14 days (1397,1398). Ivermectin
should be taken with food because bioavailability is increased,
thus increasing penetration of the drug into the epidermis.
Adjustment of ivermectin dosage is not required for persons
with renal impairment; however, the safety of multiple doses
among persons with severe liver disease is unknown. Lindane
is not recommended for treatment of pediculosis because of
toxicity, contraindications for certain populations (pregnant
and breastfeeding women, children aged <10 years, and those
with extensive dermatitis), and complexity of administration.
Other Management Considerations
The recommended regimens should not be applied to the
eyes. Pediculosis of the eyelashes should be treated by applying
occlusive ophthalmic ointment or petroleum jelly to the eyelid
margins 2 times/day for 10 days. Bedding and clothing should
be decontaminated (i.e., machine washed and dried by using
the heat cycle or dry cleaned) or removed from body contact
for at least 72 hours. Fumigation of living areas is unnecessary.
Pubic hair removal has been associated with atypical patterns
of pubic lice infestation and decreasing incidence of infection
(537,1399). Persons with pediculosis pubis should be evaluated
for HIV, syphilis, chlamydia, and gonorrhea.
Follow-Up
Evaluation should be performed after 1 week if symptoms
persist. Retreatment might be necessary if lice are found or
if eggs are observed at the hair-skin junction. If no clinical
response is achieved to one of the recommended regimens,
retreatment with an alternative regimen is recommended.
Management of Sex Partners
Sex partners within the previous month should be treated.
Sexual contact should be avoided until patients and partners
have been treated, bedding and clothing decontaminated, and
reevaluation performed to rule out persistent infection.
Special Considerations
Pregnancy
Existing data from human participants demonstrate that
pregnant and lactating women should be treated with either
permethrin or pyrethrin with piperonyl butoxide. Because no
teratogenicity or toxicity attributable to ivermectin has been
observed during human pregnancy experience, ivermectin is
classified as “human data suggest low risk” during pregnancy
and probably compatible with breastfeeding (431).
HIV Infection
Persons who have pediculosis pubis and HIV infection
should receive the same treatment regimen as those who do
not have HIV.
Scabies
Scabies is a skin infestation caused by the mite Sarcoptes
scabiei, which causes pruritus. Sensitization to S. scabiei occurs
before pruritus begins. The first time a person is infested with
S. scabiei, sensitization takes weeks to develop. However,
pruritus might occur <24 hours after a subsequent reinfestation.
Scabies among adults frequently is sexually acquired, although
scabies among children usually is not (14001402).
Diagnosis
Scabies diagnosis is made by identifying burrows, mites, eggs,
or the mites’ feces from affected areas. Skin scrapings can be
examined under the microscope to identify organisms, although
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 127
US Department of Health and Human Services/Centers for Disease Control and Prevention
this method has low sensitivity and is time consuming (1403).
Alternatively, noninvasive examination of the affected skin by
using videodermatoscopy, videomicroscopy, or dermoscopy
can be used, each of which has high sensitivity and specificity,
particularly when performed by experienced operators (1404).
Low-technology strategies include the burrow ink test and the
adhesive tape test.
Treatment
Recommended Regimens for Scabies
Permethrin 5% cream applied to all areas of the body from the neck
down and washed off after 8–14 hours
or
Ivermectin 200 ug/kg body weight orally, repeated in 14 days*
or
Ivermectin 1% lotion applied to all areas of the body from the neck
down and washed off after 8–14 hours; repeat treatment in 1 week if
symptoms persist
* Oral ivermectin has limited ovicidal activity; a second dose is required
for eradication.
Alternative Regimen
Lindane 1% 1 oz of lotion or 30 g of cream applied in a thin layer to all
areas of the body from the neck down and thoroughly washed off after
8 hours*
*
Infants and children aged <10 years should not be treated with lindane.
Topical permethrin and oral and topical ivermectin have
similar efficacy for cure of scabies (14051410). Choice of
treatment might be based on patient preference for topical
versus oral therapy, drug interactions with ivermectin (e.g.,
azithromycin, trimethoprim/sulfamethoxazole [Bactrim], or
cetirizine [Zytrec]), and cost. Permethrin is safe and effective
with a single application (1411). Ivermectin has limited ovicidal
activity and might not prevent recurrences of eggs at the time
of treatment; therefore, a second dose of ivermectin should be
administered 14 days after the first dose (1412). Ivermectin
should be taken with food because bioavailability is increased,
thereby increasing penetration of the drug into the epidermis.
Adjustments to ivermectin dosing are not required for patients
with renal impairment; however, the safety of multiple doses
among patients with severe liver disease is unknown.
Lindane is an alternative regimen because it can cause toxicity
(1413); it should be used only if the patient cannot tolerate
the recommended therapies or if these therapies have failed
(14141416). Lindane is not recommended for pregnant and
breastfeeding women, children aged <10 years, and persons
with extensive dermatitis. Seizures have occurred when lindane
was applied after a bath or used by patients who had extensive
dermatitis. Aplastic anemia after lindane use also has been
reported (1413). Lindane resistance has been reported in some
areas of the world, including parts of the United States (1413).
Other Management Considerations
Bedding and clothing should be decontaminated (i.e.,
either machine washed and dried by using the heat cycle or
dry cleaned) or removed from body contact for >72 hours.
Fumigation of living areas is unnecessary. Persons with scabies
should be advised to keep fingernails closely trimmed to reduce
injury from excessive scratching (1417).
Crusted Scabies
Crusted scabies is an aggressive infestation that usually
occurs among immunodeficient, debilitated, or malnourished
persons, including persons receiving systemic or potent topical
glucocorticoids, organ transplant recipients, persons with HIV
infection or human T-lymphotrophic virus-1 infection, and
persons with hematologic malignancies. Crusted scabies is
transmitted more easily than scabies (1418). No controlled
therapeutic studies for crusted scabies have been conducted,
and a recommended treatment remains unclear. Substantial
treatment failure might occur with a single-dose topical
scabicide or with oral ivermectin treatment. Combination
treatment is recommended with a topical scabicide, either 5%
topical permethrin cream (full-body application to be repeated
daily for 7 days then 2 times/week until cure) or 25% topical
benzyl benzoate, and oral ivermectin 200 ug/kg body weight on
days 1, 2, 8, 9, and 15. Additional ivermectin treatment on days
22 and 29 might be required for severe cases (1419). Lindane
should be avoided because of the risks for neurotoxicity with
heavy applications on denuded skin.
Follow-Up
The rash and pruritus of scabies might persist for <2 weeks
after treatment. Symptoms or signs persisting for >2 weeks
can be attributed to multiple factors. Treatment failure
can occur as a result of resistance to medication or faulty
application of topical scabicides. These medications do not
easily penetrate into thick, scaly skin of persons with crusted
scabies, perpetuating the harboring of mites in these difficult-
to-penetrate layers. In the absence of recommended contact
treatment and decontamination of bedding and clothing,
persisting symptoms can be attributed to reinfection by family
members or fomites. Finally, other household mites can cause
symptoms to persist as a result of cross-reactivity between
antigens. Even when treatment is successful, reinfection is
avoided, and cross-reactivity does not occur, symptoms can
persist or worsen as a result of allergic dermatitis.
Retreatment 2 weeks after the initial treatment regimen can
be considered for those persons who are still symptomatic or
when live mites are observed. Use of an alternative regimen is
recommended for those persons who do not respond initially
to the recommended treatment.
Recommendations and Reports
128 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
Management of Sex Partners and
Household Contacts
Persons who have had sexual, close personal, or household
contact with the patient within the month preceding scabies
infestation should be examined. Those identified as being
infested should be provided treatment.
Management of Outbreaks in Communities,
Nursing Homes, and Other Institutional Settings
Scabies epidemics frequently occur in nursing homes,
hospitals, residential facilities, and other communities
(1420,1421). Control of an epidemic can only be achieved
by treating the entire population at risk. Ivermectin can be
considered in these settings, especially if treatment with topical
scabicides fails. Mass treatment with oral ivermectin is highly
effective in decreasing prevalence in settings where scabies is
endemic (1422). Epidemics should be managed in consultation
with a specialist.
Special Considerations
Infants, Young Children, and Pregnant or
Lactating Women
Infants and young children should be treated with
permethrin; the safety of ivermectin for children weighing
<15 kg has not been determined. Infants and children aged
<10 years should not be treated with lindane. Ivermectin likely
poses a low risk to pregnant women and is likely compatible
with breastfeeding; however, because of limited data regarding
ivermectin use for pregnant and lactating women, permethrin
is the preferred treatment (431) (see Pediculosis Pubis).
HIV Infection
Persons with HIV infection who have uncomplicated scabies
should receive the same treatment regimens as those who do
not have HIV. Persons with HIV infection and others who are
immunosuppressed are at increased risk for crusted scabies and
should be managed in consultation with a specialist.
Sexual Assault and Abuse and STIs
Adolescents and Adults
These guidelines are primarily limited to the identification,
prophylaxis, and treatment of STIs and conditions among
adolescent and adult female sexual assault survivors. However,
some of the following guidelines might still apply to male
sexual assault survivors. Documentation of findings, collection
of nonmicrobiologic specimens for forensic purposes,
and management of potential pregnancy or physical and
psychological trauma are beyond the scope of these guidelines.
Examinations of survivors of sexual assault should be
conducted by an experienced clinician in a way that minimizes
further trauma to the person. The decision to obtain genital
or other specimens for STI diagnosis should be made on an
individual basis. Care systems for survivors should be designed
to ensure continuity, including timely review of test results,
support adherence, and monitoring for adverse reactions to
any prescribed therapeutic or prophylactic regimens. Laws in
all 50 states limit the evidentiary use of a survivor’s previous
sexual history, including evidence of previously acquired STIs,
as part of an effort to undermine the credibility of the survivors
testimony. Evidentiary privilege against revealing any aspect of
the examination or treatment also is enforced in most states.
Although it rarely occurs, STI diagnoses might later be accessed,
and the survivor and clinician might opt to defer testing for this
reason. Although collection of specimens at initial examination
for laboratory STI diagnosis gives the survivor and clinician
the option of deferring empiric prophylactic antimicrobial
treatment, compliance with follow-up visits is typically poor
(14231425). Among sexually active adults, identification
of an STI might represent an infection acquired before the
assault, and therefore might be more important for the medical
management of the patient than for legal purposes.
Trichomoniasis, BV, gonorrhea, and chlamydia are the
most frequently diagnosed infections among women who
have been sexually assaulted. Such conditions are prevalent
among the population, and detection of these infections
after an assault does not necessarily imply acquisition during
the assault. However, a postassault examination presents an
important opportunity for identifying or preventing an STI.
Chlamydial and gonococcal infections among women are
of particular concern because of the possibility of ascending
infection. In addition, HBV infection can be prevented
through postexposure vaccination (see Hepatitis B Virus
Infection). Because persons who have been sexually assaulted
also are at risk for acquiring HPV infection, and the efficacy
of the HPV vaccine is high (1426,1427), HPV vaccination is
also recommended for females and males through age 26 years
(https://www.cdc.gov/vaccines/hcp/acip-recs/vacc-specific/
hpv.html) (11). Reproductive-aged female survivors should be
evaluated for pregnancy and offered emergency contraception.
Evaluating Adolescents and Adults for STIs
Initial Examination
Decisions to perform the following tests should be made on
an individual basis. An initial examination after a sexual assault
might include the following:
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 129
US Department of Health and Human Services/Centers for Disease Control and Prevention
NAATs for C. trachomatis and N. gonorrhoeae at the sites
of penetration or attempted penetration should be
performed (553). These tests are preferred for diagnostic
evaluation of adolescent or adult sexual assault survivors.
Females should be offered NAAT testing for T. vaginalis
from a urine or vaginal specimen. POC or wet mount with
measurement of vaginal pH and KOH application for the
whiff test from vaginal secretions should be performed for
evidence of BV and candidiasis, especially if vaginal
discharge, malodor, or itching is present.
MSM should be offered screening for C. trachomatis and
N. gonorrhoeae if they report receptive oral or anal sex
during the preceding year, regardless of whether sexual
contact occurred at these anatomic sites during the assault.
Anoscopy should be considered in instances of reported
anal penetration.
A serum sample should be performed for HIV, HBV, and
syphilis infection.
Treatment
Compliance with follow-up visits is poor among survivors
of sexual assault (14231425). Consequently, the following
routine presumptive treatments after a sexual assault are
recommended:
An empiric antimicrobial regimen for chlamydia,
gonorrhea, and trichomonas for women and chlamydia
and gonorrhea for men.
Emergency contraception should be considered when the
assault could result in pregnancy (see Emergency
Contraception).
Postexposure hepatitis B vaccination (without HBIG) if
the hepatitis status of the assailant is unknown and the
survivor has not been previously vaccinated. If the assailant
is known to be HBsAg positive, unvaccinated survivors
should receive both hepatitis B vaccine and HBIG. The
vaccine and HBIG, if indicated, should be administered
to sexual assault survivors at the time of the initial
examination, and follow-up doses of vaccine should be
administered 1–2 and 4–6 months after the first dose.
Survivors who were previously vaccinated but did not
receive postvaccination testing should receive a single
vaccine booster dose (see Hepatitis B Virus Infection).
HPV vaccination for female and male survivors aged
9–26 years who have not been vaccinated or are
incompletely vaccinated (11) (https://www.cdc.gov/
vaccines/hcp/acip-recs/vacc-specific/hpv.html). The
vaccine should be administered to sexual assault survivors
at the time of the initial examination, and follow-up doses
should be administered at 1–2 months and 6 months after
the first dose. A 2-dose schedule (0 and 6–12 months) is
recommended for persons initiating vaccination before
age 15 years.
Recommendations for HIV PEP are made on a case-by-
case basis according to risk (see Risk for Acquiring HIV
Infection; Recommendations for Postexposure HIV Risk
Assessment of Adolescents and Adults <72 Hours After
Sexual Assault).
Recommended Regimen for Adolescent and Adult Female
Sexual Assault Survivors
Ceftriaxone 500 mg* IM in a single dose
plus
Doxycycline 100 mg 2 times/day orally for 7 days
plus
Metronidazole 500 mg 2 times/day orally for 7 days
* For persons weighing ≥150 kg, 1 g of ceftriaxone should be administered.
Recommended Regimen for Adolescent and Adult Male Sexual
Assault Survivors
Ceftriaxone 500 mg* IM in a single dose
plus
Doxycycline 100 mg 2 times/day orally for 7 days
* For persons weighing ≥150 kg, 1 g of ceftriaxone should be administered.
Clinicians should counsel persons regarding the possible
benefits and toxicities associated with these treatment regimens;
gastrointestinal side effects can occur with this combination.
The efficacy of these regimens in preventing infections after
sexual assault has not been evaluated. For those requiring
alternative treatments, refer to the specific sections in this
report relevant to the specific organisms.
Other Management Considerations
At the initial examination and, if indicated, at follow-up
examinations, patients should be counseled regarding
symptoms of STIs and the need for immediate examination if
symptoms occur. Further, they should be instructed to abstain
from sexual intercourse until STI prophylactic treatment
is completed.
Follow-Up
After the initial postassault examination, follow-up
examinations provide an opportunity to detect new infections
acquired during or after the assault, complete hepatitis B
and HPV vaccinations, if indicated, complete counseling
and treatment for other STIs, and monitor side effects and
adherence to PEP, if prescribed. If initial testing was performed,
follow-up evaluation should be conducted in <1 week to ensure
that results of positive tests can be discussed promptly with
the survivor, treatment is provided if not administered at the
initial visit, and any follow-up for infections can be arranged.
If initial tests are negative and treatment was not provided,
Recommendations and Reports
130 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
examination for STIs can be repeated 1–2 weeks after the
assault; repeat testing detects infectious organisms that might
not have reached sufficient concentrations to produce positive
test results at the time of initial examination. For survivors who
are treated during the initial visit, regardless of whether testing
was performed, posttreatment testing should be conducted
only if the person reports having symptoms. If initial test results
were negative and infection in the assailant cannot be ruled
out, serologic tests for syphilis can be repeated at 4–6 weeks
and 3 months; HIV testing can be repeated at 6 weeks and at
3 months by using methods to identify acute HIV infection.
Risk for Acquiring HIV Infection
HIV seroconversion has occurred among persons whose only
known risk factor was sexual assault or sexual abuse; however,
the frequency of this occurrence likely is low (1428,1429). In
consensual sex, the per-act risk for HIV transmission from
vaginal intercourse is 0.08%, and for receptive anal intercourse,
1.38% (192). The per-act risk for HIV transmission from
oral sex is substantially lower. Specific circumstances of an
assault (e.g., bleeding, which often accompanies trauma)
might increase risk for HIV transmission in cases involving
vaginal, anal, or oral penetration. Site of exposure to ejaculate,
viral load in ejaculate, and the presence of an STI or genital
lesions in the assailant or survivor also might increase risk for
HIV acquisition.
PEP with a 28-day course of zidovudine was associated with
an 81% reduction in risk for acquiring HIV in a study of health
care workers who had percutaneous exposures to HIV-infected
blood (1430). On the basis of these results and results from
animal studies, PEP has been recommended for health care
workers who have occupational exposures to HIV (1431).
These findings have been extrapolated to nonoccupational
injecting drug and sexual HIV exposures, including sexual
assault. The possibility of HIV exposure from the assault should
be assessed at the initial examination; survivors determined
to be at risk for acquiring HIV should be informed about the
possible benefit of PEP in preventing HIV infection. Initiation
of PEP as soon as possible after the exposure increases the
likelihood of prophylactic benefit.
Multiple factors affect the medical recommendation for
PEP and affect the assault survivor’s acceptance of that
recommendation. These factors include the likelihood of the
assailant having HIV, any exposure characteristics that might
increase the risk for HIV transmission, the time elapsed after
the event, and the potential benefits and risks associated
with PEP (1431). Determination of the assailants HIV
status at the time of the postassault examination is usually
not possible. Therefore, health care providers should assess
any available information concerning the characteristics and
HIV risk behaviors of the assailant (e.g., being an MSM or
using injecting drugs), local epidemiology of HIV/AIDS, and
exposure characteristics of the assault. When an assailant’s
HIV status is unknown, determinations about risk for HIV
transmission to the survivor should be based on whether vaginal
or anal penetration occurred; whether ejaculation occurred on
mucous membranes; whether multiple assailants were involved;
whether mucosal lesions were present in the assailant or
survivor; and any other characteristics of the assault, survivor,
or assailant that might increase risk for HIV transmission.
If PEP is offered, the following information should be
discussed with the survivor: the necessity of early initiation
of PEP to optimize potential benefits (i.e., as soon as possible
after and <72 hours after the assault), the importance of close
follow-up, the benefit of adherence to recommended dosing,
and potential adverse effects of antiretroviral medications.
Providers should emphasize that severe adverse effects are
rare from PEP (14311435). Clinical management of the
survivor should be implemented according to the HIV PEP
guidelines and in collaboration with specialists (1436). Health
care providers should provide an initial course of 3–7 days of
medication (i.e., a starter pack) with a prescription for the
remainder of the course, or, if starter packs are unavailable,
they should provide a prescription for an entire 28-day
course. Provision of the entire 28-day PEP medication supply
at the initial visit has been reported to increase likelihood of
adherence, especially when patients have difficulty returning
for multiple follow-up visits (1437). Routinely providing
starter packs or the entire 28-day course requires that health
care providers stock PEP drugs in their practice setting or have
an established agreement with a pharmacy to stock, package,
and urgently dispense PEP drugs with required administration
instructions. Uninsured patients or those with high copayments
can be enrolled in a patient-assistance program to ensure
access to PEP medications. An early follow-up visit should be
scheduled at which health care providers can discuss the results
of HIV and STI testing, provide additional counseling and
support, provide indicated vaccines not administered at the
initial evaluation, assess medication side effects and adherence,
or provide an altered PEP medication regimen if indicated by
side effects or laboratory test results.
Recommendations for Postexposure HIV Risk
Assessment of Adolescents and Adults <72 Hours
After Sexual Assault
Health care providers should do the following:
Assess risk for HIV infection in the assailant, and test that
person for HIV whenever possible.
Use the algorithm to evaluate the survivor for the need for
HIV PEP (Figure) (1436).
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 131
US Department of Health and Human Services/Centers for Disease Control and Prevention
Consult with a specialist in HIV treatment if PEP is being
considered.
If the survivor appears to be at risk for acquiring HIV from
the assault, discuss PEP, including benefits and risks.
If the survivor chooses to start PEP, provide an initial
course of 3–7 days of medication (i.e., a starter pack) with
a prescription for the remainder of the course or provide
a prescription for an entire 28-day course. Schedule an
early follow-up visit to discuss test results and provide
additional counseling (1438).
If PEP is started, obtain serum creatinine, AST, and alanine
aminotransferase at baseline.
Perform an HIV antibody test at original assessment;
repeat at 6 weeks and 3 months.
Counsel the survivor regarding ongoing risk for HIV
acquisition and about HIV PrEP, and provide referrals to
a PrEP provider.
Assistance with PEP-related decisions can be obtained by
calling the National Clinicians Post Exposure Prophylaxis
Hotline (PEP Line) (telephone: 888-448-4911).
Sexual Assault or Abuse of Children
These guidelines are limited to the identification and
treatment of STIs in prepubertal children. Management of
the psychosocial or legal aspects of the sexual assault or abuse
of children is beyond the scope of these guidelines.
Identification of STIs in children past the neonatal period
strongly indicates sexual abuse (1438). The importance of
FIGURE. Algorithm to evaluate the need for nonoccupational HIV postexposure prophylaxis among adult and adolescent survivors of sexual assault
Substantial
exposure risk
<72 hours
since exposure
Source patient
known to be
HIV positive
PEP
recommended
Source patient
of unknown
HIV status
Case-by-case
determination
PEP not
recommended
≥72 hours
since exposure
Negligible
exposure risk
Substantial Risk for HIV Acquisition
Exposure of
vagina, rectum, eye, mouth, or other mucous membrane, nonintact skin, or
percutaneous contact
With
blood, semen, vaginal secretions, rectal secretions, breast milk, or any body
uid that is visibly contaminated with blood
When
the source is known to be HIV positive
Negligible Risk for HIV Acquisition
Exposure of
vagina, rectum, eye, mouth, or other mucous
membrane, intact or nonintact skin, or
percutaneous contact
With
urine, nasal secretions, saliva, sweat, or tears if
not visibly contaminated with blood
Regardless
of the known or suspected HIV status of
the source
Source: Adapted from Announcement: updated guidelines for antiretroviral postexposure prophylaxis after sexual, injection-drug use, or other nonoccupational
exposure to HIV—United States, 2016. MMWR Morb Mortal Wkly Rep 2016;65:458.
Abbreviation: PEP = postexposure prophylaxis.
Recommendations and Reports
132 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
identifying a sexually transmitted organism for such children
as evidence of possible child sexual abuse varies by pathogen.
Postnatally acquired gonorrhea, syphilis, chlamydia, and
T. vaginalis infection and nontransfusion, nonperinatally
acquired HIV infection are indicative of sexual abuse.
Sexual abuse should be suspected when anogenital herpes or
anogenital warts are diagnosed. Investigation of sexual abuse
among children who have an infection that might have been
transmitted sexually should be conducted in compliance with
recommendations by clinicians who have experience and
training in all elements of the evaluation of child abuse, neglect,
and assault. The social significance of an infection that might
have been acquired sexually varies by the specific organism, as
does the threshold for reporting suspected child sexual abuse
(Table 8). When any STI has been diagnosed in a child, efforts
should be made in consultation with a specialist to evaluate
the possibility of sexual abuse, including conducting a history
and physical examination for evidence of abuse and diagnostic
testing for other commonly occurring STIs (14391441).
The general rule that STIs beyond the neonatal period are
evidence of sexual abuse has exceptions. For example, genital
infection with T. vaginalis (1442) or rectal or genital infection
with C. trachomatis among young children might be the result
of perinatally acquired infection and has, in certain cases
of chlamydial infection, persisted for as long as 2–3 years
(14431445), although perinatal chlamydial infection is now
uncommon because of prenatal screening and treatment of
pregnant women. Genital warts have been diagnosed among
children who have been sexually abused (1426) but also
among children who have no other evidence of sexual abuse
(1446,1447); lesions appearing for the first time in a child
aged >5 years are more likely to have been caused by sexual
transmission (1448). BV has been diagnosed among children
who have been abused but its presence alone does not prove
sexual abuse. The majority of HBV infections among children
result from household exposure to persons who have chronic
HBV infection rather than sexual abuse.
Reporting
All U.S. states and territories have laws that require reporting
of child abuse. Although the exact requirements differ by state
or territory, if a health care provider has reasonable cause to
suspect child abuse, a report must be made (1448). Health care
providers should contact their state or local child protection
service agency regarding child abuse reporting requirements.
Evaluating Children for STIs
Evaluating children for sexual assault or abuse should be
conducted in a manner designed to minimize pain and trauma
to the child. Examinations and collection of vaginal specimens
in prepubertal girls can be extremely uncomfortable and should
be performed by an experienced clinician to avoid psychological
and physical trauma to the child. The decision to obtain genital
or other specimens from a child to evaluate for STIs should be
made on an individual basis. However, children who received
a diagnosis of one STI should be screened for other STIs.
History and reported type of sexual contact might not be a
reliable indicator, and urogenital, pharyngeal, and rectal testing
should be considered for preverbal children and children who
cannot verbalize details of the assault (1438,1449). Factors that
should lead the physician to consider testing for STIs include
the following (1449):
The child has experienced penetration or has evidence of
recent or healed penetrative injury to the genitals, anus,
or oropharynx.
The child has been abused by a stranger.
TABLE 8. Implications of commonly encountered sexually transmitted or sexually associated infections for diagnosis and reporting of sexual
abuse among infants and prepubertal children
Infection Evidence for sexual abuse Recommended action
Gonorrhea* Diagnostic Report
Syphilis* Diagnostic Report
HIV
§
Diagnostic Report
Chlamydia trachomatis* Diagnostic Report
Trichomonas vaginalis* Diagnostic Report
Anogenital herpes Suspicious Consider report
†,¶
Condylomata acuminata (anogenital warts)* Suspicious Consider report
†,¶,
**
Anogenital molluscum contagiosum Inconclusive Medical follow-up
Bacterial vaginosis Inconclusive Medical follow-up
Sources: Adapted from Kellogg N; American Academy of Pediatrics Committee on Child Abuse and Neglect. The evaluation of child abuse in children. Pediatrics
2005;16:506–12; Adams JA, Farst KJ, Kellogg ND. Interpretation of medical findings in suspected child abuse: an update for 2018. J Pediatr Adolesc Gynecol
2018;31:225–31.
* If unlikely to have been perinatally acquired and vertical transmission, which is rare, is excluded.
Reports should be made to the local or state agency mandated to receive reports of suspected child abuse or neglect.
§
If unlikely to have been acquired perinatally or through transfusion.
Unless a clear history of autoinoculation exists.
** Report if evidence exists to suspect abuse, including history, physical examination, or other identified infections. Lesions appearing for the first time in a child aged
>5 years are more likely to have been caused by sexual transmission.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 133
US Department of Health and Human Services/Centers for Disease Control and Prevention
The child has been abused by an assailant known to be
infected with an STI or at high risk for STIs (e.g., injecting
drug user, MSM, person with multiple sex partners, or
person with a history of STIs).
The child has a sibling, other relative, or another person
in the household with an STI.
The child lives in an area with a high rate of STIs in the
community.
The child has signs or symptoms of STIs (e.g., vaginal
discharge or pain, genital itching or odor, urinary
symptoms, or genital lesions or ulcers).
The child or parent requests STI testing.
The child is unable to verbalize details of the assault.
If a child has symptoms, signs, or evidence of an infection
that might be sexually transmitted, the child should be tested
for common STIs before initiation of any treatment that might
interfere with diagnosing other STIs. Because of the legal and
psychosocial consequences of a false-positive diagnosis, only
tests with high specificities should be used. The potential
benefit to the child of a reliable STI diagnosis justifies deferring
presumptive treatment until specimens for highly specific tests
are obtained by providers with experience in evaluating sexually
abused and assaulted children.
Evaluations should be performed on a case-by-case basis,
according to history of assault or abuse and in a manner
that minimizes the possibility for psychological trauma and
social stigma. If the initial exposure was recent, the infectious
organisms acquired through the exposure might not have
produced sufficient concentrations to result in positive test
results or examination findings (1450). Alternatively, positive
test results after a recent exposure might represent the assailant’s
secretions (but would nonetheless be an indication for
treatment of the child). A second visit approximately 2–6 weeks
after the most recent sexual exposure should be scheduled
to include a repeat physical examination and collection of
additional specimens to identify any infection that might not
have been detected at the time of initial evaluation. A single
evaluation might be sufficient if the child was abused for an
extended period and if a substantial amount of time elapsed
between the last suspected episode of abuse and the medical
evaluation. Compliance with follow-up appointments might be
improved when law enforcement personnel or child protective
services are involved.
Initial Examination
Visual inspection of the genital, perianal, and oral areas
for genital discharge, odor, bleeding, irritation, warts,
and ulcerative lesions should be performed during initial
examination. The clinical manifestations of certain STIs are
different for children than for adults. For example, typical
vesicular lesions might be absent even in the presence of HSV
infection. The following should be performed during the initial
examination, if STI testing is indicated:
Testing for N. gonorrhoeae and C. trachomatis can be
performed from specimens collected from the pharynx
and rectum, as well as the vagina for girls and urine for
boys. Cervical specimens are not recommended for
prepubertal girls. For boys with a urethral discharge, a
meatal specimen discharge is an adequate substitute for
an intraurethral swab specimen. Culture or NAAT can be
used to test for N. gonorrhoeae and C. trachomatis.
Although data regarding NAAT for children are more
limited and performance is test dependent (553), no
evidence demonstrates that performance of NAAT for
detection of N. gonorrhoea or C. trachomatis among
children differs from that among adults. Only FDA-cleared
NAAT assays should be used. Consultation with an expert
is necessary before using NAAT in this context, both to
minimize the possibility of cross-reaction with
nongonococcal Neisseria species and other commensals
(e.g., N. meningitidis, N. sicca, N. lactamica, N. cinerea,
or M. catarrhalis) and to ensure correct interpretation of
results. Because of the implications of a diagnosis of
N. gonorrhoeae or C. trachomatis infection in a child, only
CLIA-validated, FDA-cleared NAATs should be used
(837). If culture for the isolation of N. gonorrhoeae or
C. trachomatis is performed, only standard culture
procedures should be followed. Specimens from the vagina,
urethra, pharynx, or rectum should be streaked onto
selective media for isolation of N. gonorrhoeae, and all
presumptive isolates of N. gonorrhoeae should be identified
definitively by at least two tests that involve different
approaches (e.g., biochemical, enzyme substrate, or
molecular probes). Gram stains are inadequate for
evaluating prepubertal children for gonorrhea and should
not be used to diagnose or exclude gonorrhea. Specimens
(either NAAT or culture, including any isolates) obtained
before treatment should be preserved for further validation
if needed. When a specimen is positive, the result should
be confirmed either by retesting the original specimen or
obtaining another. Because of the overall low prevalence
of N. gonorrhoeae and C. trachomatis among children,
false-positive results can occur, and all specimens that are
initially positive should be confirmed.
Testing for T. vaginalis should not be limited to girls with
vaginal discharge if other indications for vaginal testing
exist because evidence indicates that asymptomatic sexually
abused children might be infected with T. vaginalis and
might benefit from treatment (1451,1452). NAAT can
be used as an alternative or in addition to culture and wet
Recommendations and Reports
134 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
mount, especially in settings where culture and wet mount
of vaginal swab specimens are not obtainable. Data
regarding use of NAATs for detection of T. vaginalis among
children are limited; however, no evidence indicates that
performance of NAAT for detection of T. vaginalis for
children would differ from that for adults. Consultation
with an expert is necessary before using NAAT in this
context to ensure correct interpretation of results. Because
of the implications of a diagnosis of T. vaginalis infection in
a child, only CLIA-validated, FDA-cleared NAATs should
be used (837). POC tests for T. vaginalis have not been
validated for prepubertal children and should not be used.
In the case of a positive specimen, the result should be
confirmed either by retesting the original specimen or
obtaining another. Because of the overall low prevalence of
T. vaginalis among children, false-positive results can occur,
and all specimens that are initially positive should
be confirmed.
HSV can be indicative of sexual abuse; therefore,
specimens should be obtained from all vesicular or
ulcerative genital or perianal lesions and sent for NAAT
or viral culture.
Wet mount can be used for a vaginal swab specimen for
BV if discharge is present.
Collection of serum samples should be evaluated, preserved
for subsequent analysis, and used as a baseline for
comparison with follow-up serologic tests. Sera can be tested
for antibodies to T. pallidum, HIV, and HBV. Decisions
regarding the infectious agents for which to perform
serologic tests should be made on a case-by-case basis.
Treatment
The risk for a child acquiring an STI as a result of sexual
abuse or assault has not been well studied. Presumptive
treatment for children who have been sexually assaulted or
abused is not recommended because the incidence of most
STIs among children is low after abuse or assault, prepubertal
girls appear to be at lower risk for ascending infection than
adolescent or adult women, and regular follow-up of children
usually can be ensured. However, certain children or their
parent or guardian might be concerned about the possibility
of infection with an STI, even if the health care provider
has perceived the risk to be low. Such concerns might be an
indication for presumptive treatment in certain settings and
might be considered after all relevant specimens for diagnostic
tests have been collected.
Other Management Considerations
Children who are survivors of sexual assault or abuse are at
increased risk for future unsafe sexual practices that have been
linked to higher risk for HPV acquisition (1426,1453) and
are more likely to engage in these behaviors at an earlier age;
therefore, ACIP recommends vaccination of these children
at age ≥9 years if they have not initiated or completed HPV
vaccination (see Human Papillomavirus Infections, Prevention)
(https://www.cdc.gov/vaccines/hcp/acip-recs/vacc-specific/
hpv.html). Although HPV vaccine will not protect against
progression of infection already acquired or promote clearance
of the infection, the vaccine protects against HPV types not
yet acquired.
Follow-Up
If no infections were identified at the initial examination
after the last suspected sexual exposure, and if this exposure
was recent, a follow-up evaluation approximately 2 weeks
after the last exposure can be considered. Likewise, if no
physical examination or diagnostic testing was performed
at the initial visit, a complete examination can be scheduled
approximately 2 weeks after the last exposure to identify any
evidence of STIs. In circumstances in which transmission of
syphilis, HIV, HBV, or HPV is a concern but baseline tests
for syphilis, HIV, and HBV are negative and examinations
for genital warts are negative, follow-up serologic testing and
examination approximately 6 weeks and <3 months after the
last suspected sexual exposure is recommended to allow time
for antibodies to develop and signs of infection to appear.
In addition, results of HBsAg testing should be interpreted
carefully because HBV can be transmitted nonsexually.
Decisions regarding which tests should be performed should
be made on a case-by-case basis.
Risk for Acquiring HIV Infection
HIV has been reported among children for whom sexual
abuse was the only known risk factor. Serologic testing for HIV
should be considered for sexually abused children. The decision
to test for HIV should involve the family, if possible, and be
made on a case-by-case basis depending on the likelihood
of infection in the assailant (1448,1454). Although data are
insufficient concerning the efficacy of PEP among children,
treatment is well tolerated by infants and children with and
without HIV, and children have a minimal risk for serious
adverse reactions because of the short period recommended
for prophylaxis (1455).
Recommendations for Postexposure HIV Risk
Assessment of Children <72 Hours After
Sexual Assault
Providers should do the following:
Review local HIV epidemiology, assess risk for HIV in the
assailant, and test for HIV.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 135
US Department of Health and Human Services/Centers for Disease Control and Prevention
Evaluate the circumstances of the assault or abuse that
might affect risk for HIV transmission.
Perform HIV antigen or antibody testing (or antibody
testing, if antigen or antibody testing is unavailable) during
the original assessment and again at follow-up visits, in
accordance with CDC guidelines (https://stacks.cdc.gov/
view/cdc/38856). In considering whether to offer PEP,
health care providers should consider whether the child
can be treated soon after the sexual exposure (i.e.,
<72 hours), the likelihood that the assailant has HIV
infection, and the likelihood of high compliance with the
prophylactic regimen (1436). Potential benefit of treating
a sexually abused child should be weighed against the risk
for adverse reactions.
Consult with a provider specializing in evaluating or
treating children with HIV infection to determine age-
appropriate dosing and regimens and baseline laboratory
testing, if PEP is being considered.
Discuss PEP with the caregivers, including its toxicity,
unknown efficacy, and possible benefits, for children
determined to be at risk for HIV transmission from the
assault or abuse.
Provided adequate doses of medication, if PEP is begun,
to last until the follow-up visit 3–7 days after the initial
assessment, at which time the child should be reevaluated
and tolerance of medication assessed (139).
Conflicts of Interest
All authors have completed and submitted the International
Committee of Medical Journal Editors form for disclosure of
potential conflicts of interest. Christina Muzny reports other support
from CDC, during the conduct of the study; grants from the National
Institutes of Health/National Institute of Allergy and Infectious
Diseases and Lupin Pharmaceuticals; personal fees from Lupin
Pharmaceuticals, PhagoMed, Cepheid, and Beckton Dickinson; and
personal fees and other support from Roche Diagnostics, Abbott
Molecular, and BioFire Diagnostics, outside the submitted work.
Hilary Reno reports grants from Hologic, outside the submitted
work. Christine Johnston reports other support from CDC, during
the conduct of the study; received research funding from Sanofi-
Pasteur; royalties from UpToDate; and personal fees from MedPace,
Gilead, AbbVie, and UpToDate, outside the submitted work.
References
1. Workowski KA, Bolan GA; CDC. Sexually transmitted diseases
treatment guidelines, 2015. MMWR Recomm Rep 2015;64(No. RR-3).
PMID:26042815
2. Barrow RY, Ahmed F, Bolan GA, Workowski KA. Recommendations
for providing quality sexually transmitted diseases clinical services, 2020.
MMWR Recomm Rep 2020;68(No. RR-5). PMID:31899459 https://
doi.org/10.15585/mmwr.rr6805a1
3. CDC. A guide to taking a sexual history. Atlanta, GA: US Department
of Health and Human Services, CDC. https://www.cdc.gov/std/
treatment/sexualhistory.pdf
4. Henderson JT, Senger CA, Henninger M, Bean SI, Redmond N,
O’Connor EA. Behavioral counseling interventions to prevent sexually
transmitted infections: updated evidence report and systematic review
for the US Preventive Services Task Force. JAMA 2020;324:682–99.
PMID:32809007 https://doi.org/10.1001/jama.2020.10371
5. Kamb ML, Fishbein M, Douglas JM Jr, et al.; Project RESPECT Study
Group. Efficacy of risk-reduction counseling to prevent human
immunodeficiency virus and sexually transmitted diseases: a randomized
controlled trial. JAMA 1998;280:1161–7. PMID:9777816 https://doi.
org/10.1001/jama.280.13.1161
6. Metsch LR, Feaster DJ, Gooden L, et al. Effect of risk-reduction
counseling with rapid HIV testing on risk of acquiring sexually
transmitted infections: the AWARE randomized clinical trial. JAMA
2013;310:170110. PMID:24150466 https://doi.org/10.1001/
jama.2013.280034
7. Brookmeyer KA, Hogben M, Kinsey J. The role of behavioral counseling
in sexually transmitted disease prevention program settings. Sex Transm
Dis 2016;43(Suppl 1):S10212. PMID:26779681 https://doi.
org/10.1097/OLQ.0000000000000327
8. Patel P, Bush T, Mayer K, et al.; SUN Study Investigators. Routine brief
risk-reduction counseling with biannual STD testing reduces STD
incidence among HIV-infected men who have sex with men in care. Sex
Transm Dis 2012;39:470–4. PMID:22592834 https://doi.org/10.1097/
OLQ.0b013e31824b3110
9. Warner L, Klausner JD, Rietmeijer CA, et al.; Safe in the City Study
Group. Effect of a brief video intervention on incident infection among
patients attending sexually transmitted disease clinics. PLoS Med
2008;5:e135. PMID:18578564 https://doi.org/10.1371/journal.
pmed.0050135
10. Mustanski B, Parsons JT, Sullivan PS, Madkins K, Rosenberg E, Swann
G. Biomedical and behavioral outcomes of Keep It Up!: an ehealth HIV
prevention program RCT. Am J Prev Med 2018;55:151–8.
PMID:29937115 https://doi.org/10.1016/j.amepre.2018.04.026
11. Meites E, Szilagyi PG, Chesson HW, Unger ER, Romero JR, Markowitz
LE. Human papillomavirus vaccination for adults: updated
recommendations of the Advisory Committee on Immunization
Practices. MMWR Morb Mortal Wkly Rep 2019;68:698702.
PMID:31415491 https://doi.org/10.15585/mmwr.mm6832a3
12. Schillie S, Vellozzi C, Reingold A, et al. Prevention of hepatitis B virus
infection in the United States: recommendations of the Advisory
Committee on Immunization Practices. MMWR Recomm Rep
2018;67(No. RR-1). PMID:29939980 https://doi.org/10.15585/mmwr.
rr6701a1
13. Doshani M, Weng M, Moore KL, Romero JR, Nelson NP.
Recommendations of the Advisory Committee on Immunization
Practices for use of hepatitis A vaccine for persons experiencing
homelessness. MMWR Morb Mortal Wkly Rep 2019;68:1536.
PMID:30763295 https://doi.org/10.15585/mmwr.mm6806a6
14. Weller S, Davis K. Condom effectiveness in reducing heterosexual HIV
transmission. Cochrane Database Syst Rev 2002;(1):CD003255.
PMID:11869658 https://doi.org/10.1002/14651858.CD003255
15. Giannou FK, Tsiara CG, Nikolopoulos GK, et al. Condom effectiveness
in reducing heterosexual HIV transmission: a systematic review and
meta-analysis of studies on HIV serodiscordant couples. Expert Rev
Pharmacoecon Outcomes Res 2016;16:48999. PMID:26488070
https://doi.org/10.1586/14737167.2016.1102635
16. Smith DK, Herbst JH, Zhang X, Rose CE. Condom effectiveness for
HIV prevention by consistency of use among men who have sex with
men in the United States. J Acquir Immune Defic Syndr 2015;68:337–44.
PMID:25469526 https://doi.org/10.1097/QAI.0000000000000461
Recommendations and Reports
136 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
17. Johnson WD, O’Leary A, Flores SA. Per-partner condom effectiveness
against HIV for men who have sex with men. AIDS 2018;32:1499505.
PMID:29794493 https://doi.org/10.1097/QAD.0000000000001832
18. Crosby RA, Charnigo RA, Weathers C, Caliendo AM, Shrier LA.
Condom effectiveness against non-viral sexually transmitted infections:
a prospective study using electronic daily diaries. Sex Transm Infect
2012;88:4849. PMID:23002192 https://doi.org/10.1136/
sextrans-2012-050618
19. Holmes KK, Levine R, Weaver M. Effectiveness of condoms in
preventing sexually transmitted infections. Bull World Health Organ
2004;82:454–61. PMID:15356939
20. Warner L, Stone KM, Macaluso M, Buehler JW, Austin HD. Condom
use and risk of gonorrhea and chlamydia: a systematic review of design
and measurement factors assessed in epidemiologic studies. Sex Transm
Dis 2006;33:3651. PMID:16385221 https://doi.org/10.1097/01.
olq.0000187908.42622.fd
21. Bernabe-Ortiz A, Carcamo CP, Scott JD, Hughes JP, Garcia PJ, Holmes
KK. HBV infection in relation to consistent condom use: a population-
based study in Peru. PLoS One 2011;6:e24721. PMID:21931828
https://doi.org/10.1371/journal.pone.0024721
22. Ness RB, Hillier SL, Kip KE, et al. Bacterial vaginosis and risk of pelvic
inflammatory disease. Obstet Gynecol 2004;104:7619. PMID:15458899
https://doi.org/10.1097/01.AOG.0000139512.37582.17
23. Martin IE, Gu W, Yang Y, Tsang RS. Macrolide resistance and molecular
types of Treponema pallidum causing primary syphilis in Shanghai, China.
Clin Infect Dis 2009;49:51521. PMID:19583516 https://doi.
org/10.1086/600878
24. Winer RL, Hughes JP, Feng Q, et al. Condom use and the risk of genital
human papillomavirus infection in young women. N Engl J Med
2006;354:264554. PMID:16790697 https://doi.org/10.1056/
NEJMoa053284
25. Bleeker MC, Hogewoning CJ, Voorhorst FJ, et al. Condom use promotes
regression of human papillomavirus-associated penile lesions in male
sexual partners of women with cervical intraepithelial neoplasia. Int
J Cancer 2003;107:80410. PMID:14566831 https://doi.org/10.1002/
ijc.11473
26. Hogewoning CJ, Bleeker MC, van den Brule AJ, et al. Condom use
promotes regression of cervical intraepithelial neoplasia and clearance
of human papillomavirus: a randomized clinical trial. Int J Cancer
2003;107:811–6. PMID:14566832 https://doi.org/10.1002/ijc.11474
27. Koss CA, Dunne EF, Warner L. A systematic review of epidemiologic
studies assessing condom use and risk of syphilis. Sex Transm Dis
2009;36:4015. PMID:19455075 https://doi.org/10.1097/
OLQ.0b013e3181a396eb
28. Hernández-Romieu AC, Siegler AJ, Sullivan PS, Crosby R, Rosenberg
ES. How often do condoms fail? A cross-sectional study exploring
incomplete use of condoms, condom failures and other condom
problems among black and white MSM in southern U.S.A. Sex Transm
Infect 2014;90:6027. PMID:25080511 https://doi.org/10.1136/
sextrans-2014-051581
29. D’Anna LH, Margolis AD, Warner L, et al.; Safe City Study Group.
Condom use problems during anal sex among men who have sex with
men (MSM): findings from the Safe in the City study. AIDS Care
2012;24:1028–38. PMID:22519680 https://doi.org/10.1080/095401
21.2012.668285
30. Steiner MJ, Cates W Jr, Warner L. The real problem with male condoms
is nonuse. Sex Transm Dis 1999;26:459–62. PMID:10494937 https://
doi.org/10.1097/00007435-199909000-00007
31. Kowal D, Hatcher RA, Nelson AL, et al., eds. Contraceptive technology.
21st ed. Atlanta, GA: Managing Contraception; 2017.
32. Gallo MF, Kilbourne-Brook M, Coffey PS. A review of the effectiveness
and acceptability of the female condom for dual protection. Sex Health
2012;9:18–26. PMID:22348629 https://doi.org/10.1071/SH11037
33. Mantell JE, Kelvin EA, Exner TM, Hoffman S, Needham S, Stein ZA.
Anal use of the female condom: does uncertainty justify provider
inaction? AIDS Care 2009;21:1185–94. PMID:20024779 https://doi.
org/10.1080/09540120902730005
34. Rosenberg MJ, Davidson AJ, Chen JH, Judson FN, Douglas JM. Barrier
contraceptives and sexually transmitted diseases in women: a comparison
of female-dependent methods and condoms. Am J Public Health
1992;82:66974. PMID:1566944 https://doi.org/10.2105/
AJPH.82.5.669
35. de Bruyn G, Shiboski S, van der Straten A, et al.; MIRA Team. The
effect of the vaginal diaphragm and lubricant gel on acquisition of
HSV-2. Sex Transm Infect 2011;87:301–5. PMID:21447515 https://
doi.org/10.1136/sti.2010.047142
36. Ramjee G, van der Straten A, Chipato T, et al.; MIRA team. The
diaphragm and lubricant gel for prevention of cervical sexually
transmitted infections: results of a randomized controlled trial. PLoS
One 2008;3:e3488. PMID:18941533 https://doi.org/10.1371/journal.
pone.0003488
37. Lusti-Narasimhan M, Merialdi M, Holt B. Multipurpose prevention
technologies: maximising positive synergies. BJOG 2014;121:251.
PMID:24393212 https://doi.org/10.1111/1471-0528.12606
38. Ahmed K, Baeten JM, Beksinska M, et al.; Evidence for Contraceptive
Options and HIV Outcomes (ECHO) Trial Consortium. HIV incidence
among women using intramuscular depot medroxyprogesterone acetate,
a copper intrauterine device, or a levonorgestrel implant for
contraception: a randomised, multicentre, open-label trial. Lancet
2019;394:303–13. PMID:31204114 https://doi.org/10.1016/
S0140-6736(19)31288-7
39. Young Holt B, Dellplain L, Creinin MD, Peine KJ, Romano J,
Hemmerling A. A strategic action framework for multipurpose
prevention technologies combining contraceptive hormones and
antiretroviral drugs to prevent pregnancy and HIV. Eur J Contracept
Reprod Health Care 2018;23:326–34. PMID:30247084 https://doi.
org/10.1080/13625187.2018.1508650
40. Wilkinson D, Tholandi M, Ramjee G, Rutherford GW. Nonoxynol-9
spermicide for prevention of vaginally acquired HIV and other sexually
transmitted infections: systematic review and meta-analysis of
randomised controlled trials including more than 5000 women. Lancet
Infect Dis 2002;2:613–7. PMID:12383611 https://doi.org/10.1016/
S1473-3099(02)00396-1
41. McCormack S, Ramjee G, Kamali A, et al. PRO2000 vaginal gel for
prevention of HIV-1 infection (Microbicides Development Programme
301): a phase 3, randomised, double-blind, parallel-group trial. Lancet
2010;376:132937. PMID:20851460 https://doi.org/10.1016/
S0140-6736(10)61086-0
42. Skoler-Karpoff S, Ramjee G, Ahmed K, et al. Efficacy of Carraguard for
prevention of HIV infection in women in South Africa: a randomised,
double-blind, placebo-controlled trial. Lancet 2008;372:197787.
PMID:19059048 https://doi.org/10.1016/S0140-6736(08)61842-5
43. Van Damme L, Govinden R, Mirembe FM, et al.; CS Study Group.
Lack of effectiveness of cellulose sulfate gel for the prevention of vaginal
HIV transmission. N Engl J Med 2008;359:463–72. PMID:18669425
https://doi.org/10.1056/NEJMoa0707957
44. Feldblum PJ, Adeiga A, Bakare R, et al. SAVVY vaginal gel (C31G) for
prevention of HIV infection: a randomized controlled trial in Nigeria.
PLoS One 2008;3:e1474. PMID:18213382 https://doi.org/10.1371/
journal.pone.0001474
45. Cottrell ML, Kashuba AD. Topical microbicides and HIV prevention
in the female genital tract. J Clin Pharmacol 2014;54:60315.
PMID:24664786 https://doi.org/10.1002/jcph.292
46. Abdool Karim SS, Abdool Karim Q, Kharsany ABM, et al.; CAPRISA
004 Trial Group. Tenofovir gel for the prevention of herpes simplex virus
Type 2 infection. N Engl J Med 2015;373:530–9. PMID:26244306
https://doi.org/10.1056/NEJMoa1410649
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 137
US Department of Health and Human Services/Centers for Disease Control and Prevention
47. Abdool Karim Q, Abdool Karim SS, Frohlich JA, et al.; CAPRISA 004
Trial Group. Effectiveness and safety of tenofovir gel, an antiretroviral
microbicide, for the prevention of HIV infection in women. Science
2010;329:116874. Erratum in: Science 2011;333:524. PMID:20643915
https://doi.org/10.1126/science.1193748
48. Marrazzo JM, Ramjee G, Richardson BA, et al.; VOICE Study Team.
Tenofovir-based preexposure prophylaxis for HIV infection among
African women. N Engl J Med 2015;372:509–18. PMID:25651245
https://doi.org/10.1056/NEJMoa1402269
49. Delany-Moretlwe S, Lombard C, Baron D, et al. Tenofovir 1% vaginal
gel for prevention of HIV-1 infection in women in South Africa
(FACTS-001): a phase 3, randomised, double-blind, placebo-controlled
trial. Lancet Infect Dis 2018;18:1241–50. PMID:30507409 https://
doi.org/10.1016/S1473-3099(18)30428-6
50. Baeten JM, Palanee-Phillips T, Brown ER, et al.; MTN-020–ASPIRE
Study Team. Use of a vaginal ring containing dapivirine for HIV-1
prevention in women. N Engl J Med 2016;375:212132.
PMID:26900902 https://doi.org/10.1056/NEJMoa1506110
51. Nel A, van Niekerk N, Kapiga S, et al.; Ring Study Team. Safety and
efficacy of a dapivirine vaginal ring for HIV prevention in women. N
Engl J Med 2016;375:213343. PMID:27959766 https://doi.
org/10.1056/NEJMoa1602046
52. Cranston RD, Lama JR, Richardson BA, et al.; MTN-017 Protocol
Team. MTN-017: a rectal phase 2 extended safety and acceptability
study of tenofovir reduced-glycerin 1% gel. Clin Infect Dis
2017;64:614–20. PMID:27986684
53. Hooton TM, Roberts PL, Stamm WE. Effects of recent sexual activity
and use of a diaphragm on the vaginal microflora. Clin Infect Dis
1994;19:2748. PMID:7986899 https://doi.org/10.1093/
clinids/19.2.274
54. Fihn SD, Boyko EJ, Normand EH, et al. Association between use of
spermicide-coated condoms and Escherichia coli urinary tract infection
in young women. Am J Epidemiol 1996;144:512–20. PMID:8781467
https://doi.org/10.1093/oxfordjournals.aje.a008958
55. Polis CB, Curtis KM, Hannaford PC, et al. An updated systematic review
of epidemiological evidence on hormonal contraceptive methods and
HIV acquisition in women. AIDS 2016;30:2665–83. PMID:27500670
https://doi.org/10.1097/QAD.0000000000001228
56. Kiweewa FM, Brown E, Mishra A, et al.; MTN-020/ASPIRE Study
Team. Acquisition of sexually transmitted infections among women
using a variety of contraceptive options: a prospective study among
high-risk African women. J Int AIDS Soc 2019;22:e25257.
PMID:30816632 https://doi.org/10.1002/jia2.25257
57. McCarthy KJ, Gollub EL, Ralph L, van de Wijgert J, Jones HE.
Hormonal contraceptives and the acquisition of sexually transmitted
infections: an updated systematic review. Sex Transm Dis 2019;46:290–6.
PMID:30628946 https://doi.org/10.1097/OLQ.0000000000000975
58. Curtis KM, Tepper NK, Jatlaoui TC, et al. U.S. medical eligibility criteria
for contraceptive use, 2016. MMWR Recomm Rep 2016;65(No. RR-3).
PMID:27467196 https://doi.org/10.15585/mmwr.rr6503a1
59. Curtis KM, Jatlaoui TC, Tepper NK, et al. U.S. selected practice
recommendations for contraceptive use, 2016. MMWR Recomm Rep
2016;65(No. RR-4). PMID:27467319 https://doi.org/10.15585/mmwr.
rr6504a1
60. Cleland K, Zhu H, Goldstuck N, Cheng L, Trussell J. The efficacy of
intrauterine devices for emergency contraception: a systematic review
of 35 years of experience. Hum Reprod 2012;27:19942000.
PMID:22570193 https://doi.org/10.1093/humrep/des140
61. Shen J, Che Y, Showell E, Chen K, Cheng L. Interventions for emergency
contraception. Cochrane Database Syst Rev 2019;1:CD001324.
PMID:30661244
62. Marcell AV, Waks AB, Rutkow L, McKenna R, Rompalo A, Hogan MT.
What do we know about males and emergency contraception? A synthesis
of the literature. Perspect Sex Reprod Health 2012;44:18493.
PMID:22958663 https://doi.org/10.1363/4418412
63. Gray RH, Kigozi G, Serwadda D, et al. Male circumcision for HIV
prevention in men in Rakai, Uganda: a randomised trial. Lancet
2007;369:65766. PMID:17321311 https://doi.org/10.1016/
S0140-6736(07)60313-4
64. Bailey RC, Moses S, Parker CB, et al. Male circumcision for HIV
prevention in young men in Kisumu, Kenya: a randomised controlled
trial. Lancet 2007;369:64356. PMID:17321310 https://doi.
org/10.1016/S0140-6736(07)60312-2
65. Auvert B, Taljaard D, Lagarde E, Sobngwi-Tambekou J, Sitta R, Puren
A. Randomized, controlled intervention trial of male circumcision for
reduction of HIV infection risk: the ANRS 1265 Trial. PLoS Med
2005;2:e298. Erratum in: PLoS Med 2006;3:298. PMID:16231970
https://doi.org/10.1371/journal.pmed.0020298
66. Tobian AA, Serwadda D, Quinn TC, et al. Male circumcision for the
prevention of HSV-2 and HPV infections and syphilis. N Engl J Med
2009;360:1298309. PMID:19321868 https://doi.org/10.1056/
NEJMoa0802556
67. Auvert B, Sobngwi-Tambekou J, Cutler E, et al. Effect of male
circumcision on the prevalence of high-risk human papillomavirus in
young men: results of a randomized controlled trial conducted in Orange
Farm, South Africa. J Infect Dis 2009;199:14–9. PMID:19086814
https://doi.org/10.1086/595566
68. Sobngwi-Tambekou J, Taljaard D, Lissouba P, et al. Effect of HSV-2
serostatus on acquisition of HIV by young men: results of a longitudinal
study in Orange Farm, South Africa. J Infect Dis 2009;199:958–64.
PMID:19220143 https://doi.org/10.1086/597208
69. Gray R, Kigozi G, Kong X, et al. The effectiveness of male circumcision
for HIV prevention and effects on risk behaviors in a posttrial follow-up
study. AIDS 2012;26:60915. PMID:22210632 https://doi.
org/10.1097/QAD.0b013e3283504a3f
70. Mehta SD, Moses S, Parker CB, Agot K, Maclean I, Bailey RC.
Circumcision status and incident herpes simplex virus type 2 infection,
genital ulcer disease, and HIV infection. AIDS 2012;26:11419.
PMID:22382150 https://doi.org/10.1097/QAD.0b013e328352d116
71. World Health Organization/UNAIDS. New data on male circumcision
and HIV prevention: policy and programme implications [Internet].
Geneva, Switzerland: WHO/UNAIDS Technical Consultation on Male
Circumcision and HIV Prevention: Research Implications for Policy
and Programming; 2007. https://www.who.int/hiv/pub/
malecircumcision/research_implications/en/
72. American Urological Association. Circumcision policy statement
[Internet]. Linthicum, MD: American Urological Association; 2017.
https://www.auanet.org/guidelines/guidelines/circumcision
73. Yuan T, Fitzpatrick T, Ko NY, et al. Circumcision to prevent HIV and
other sexually transmitted infections in men who have sex with men: a
systematic review and meta-analysis of global data. Lancet Glob Health
2019;7:e43647. PMID:30879508 https://doi.org/10.1016/
S2214-109X(18)30567-9
74. Grohskopf LA, Chillag KL, Gvetadze R, et al. Randomized trial of
clinical safety of daily oral tenofovir disoproxil fumarate among HIV-
uninfected men who have sex with men in the United States. J Acquir
Immune Defic Syndr 2013;64:7986. PMID:23466649 https://doi.
org/10.1097/QAI.0b013e31828ece33
75. Grant RM, Lama JR, Anderson PL, et al.; iPrEx Study Team. Preexposure
chemoprophylaxis for HIV prevention in men who have sex with men.
N Engl J Med 2010;363:258799. PMID:21091279 https://doi.
org/10.1056/NEJMoa1011205
76. Baeten JM, Donnell D, Ndase P, et al.; Partners PrEP Study Team.
Antiretroviral prophylaxis for HIV prevention in heterosexual men and
women. N Engl J Med 2012;367:399–410. PMID:22784037 https://
doi.org/10.1056/NEJMoa1108524
77. Thigpen MC, Kebaabetswe PM, Paxton LA, et al.; TDF2 Study Group.
Antiretroviral preexposure prophylaxis for heterosexual HIV transmission
in Botswana. N Engl J Med 2012;367:42334. PMID:22784038 https://
doi.org/10.1056/NEJMoa1110711
Recommendations and Reports
138 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
78. Choopanya K, Martin M, Suntharasamai P, et al.; Bangkok Tenofovir
Study Group. Antiretroviral prophylaxis for HIV infection in injecting
drug users in Bangkok, Thailand (the Bangkok Tenofovir Study): a
randomised, double-blind, placebo-controlled phase 3 trial. Lancet
2013;381:208390. PMID:23769234 https://doi.org/10.1016/
S0140-6736(13)61127-7
79. Molina JM, Charreau I, Spire B, et al.; ANRS IPERGAY Study Group.
Efficacy, safety, and effect on sexual behaviour of on-demand pre-
exposure prophylaxis for HIV in men who have sex with men: an
observational cohort study. Lancet HIV 2017;4:e40210.
PMID:28747274 https://doi.org/10.1016/S2352-3018(17)30089-9
80. CDC. Preexposure prophylaxis for the prevention of HIV infection in the
United States—2017 update: a clinical practice guideline. Atlanta, GA:
US Department of Health and Human Services, CDC; 2018. https://
www.cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-guidelines-2017.pdf
81. Jones J, Weiss K, Mermin J, et al. Proportion of incident human
immunodeficiency virus cases among men who have sex with men
attributable to gonorrhea and chlamydia: a modeling analysis. Sex Transm
Dis 2019;46:35763. PMID:31095100 https://doi.org/10.1097/
OLQ.0000000000000980
82. Pathela P, Braunstein SL, Blank S, Schillinger JA. HIV incidence among
men with and those without sexually transmitted rectal infections:
estimates from matching against an HIV case registry. Clin Infect Dis
2013;57:1203–9. PMID:23800942 https://doi.org/10.1093/cid/cit437
83. Pathela P, Braunstein SL, Blank S, Shepard C, Schillinger JA. The high
risk of an HIV diagnosis following a diagnosis of syphilis: a population-
level analysis of New York City men. Clin Infect Dis 2015;61:281–7.
PMID:25870333 https://doi.org/10.1093/cid/civ289
84. Chou R, Evans C, Hoverman A, et al. Preexposure prophylaxis for the
prevention of HIV infection: evidence report and systematic review for
the US Preventive Services Task Force. JAMA 2019;321:221430.
PMID:31184746 https://doi.org/10.1001/jama.2019.2591
85. Liu AY, Cohen SE, Vittinghoff E, et al. Preexposure prophylaxis for HIV
infection integrated with municipal- and community-based sexual health
services. JAMA Intern Med 2016;176:7584. PMID:26571482 https://
doi.org/10.1001/jamainternmed.2015.4683
86. McCormack S, Dunn DT, Desai M, et al. Pre-exposure prophylaxis to
prevent the acquisition of HIV-1 infection (PROUD): effectiveness
results from the pilot phase of a pragmatic open-label randomised trial.
Lancet 2016;387:5360. PMID:26364263 https://doi.org/10.1016/
S0140-6736(15)00056-2
87. Volk JE, Marcus JL, Phengrasamy T, et al. No new HIV infections with
increasing use of HIV preexposure prophylaxis in a clinical practice
setting. Clin Infect Dis 2015;61:1601–3. PMID:26334052 https://doi.
org/10.1093/cid/civ778
88. Celum C, Wald A, Lingappa JR, et al.; Partners in Prevention HSV/
HIV Transmission Study Team. Acyclovir and transmission of HIV-1
from persons infected with HIV-1 and HSV-2. N Engl J Med
2010;362:42739. PMID:20089951 https://doi.org/10.1056/
NEJMoa0904849
89. Celum C, Wald A, Hughes J, et al.; HPTN 039 Protocol Team. Effect
of aciclovir on HIV-1 acquisition in herpes simplex virus 2 seropositive
women and men who have sex with men: a randomised, double-blind,
placebo-controlled trial. Lancet 2008;371:2109–19. PMID:18572080
https://doi.org/10.1016/S0140-6736(08)60920-4
90. Bolan RK, Beymer MR, Weiss RE, Flynn RP, Leibowitz AA, Klausner
JD. Doxycycline prophylaxis to reduce incident syphilis among HIV-
infected men who have sex with men who continue to engage in high-risk
sex: a randomized, controlled pilot study. Sex Transm Dis 2015;42:98–103.
PMID:25585069 https://doi.org/10.1097/OLQ.0000000000000216
91. Grant JS, Stafylis C, Celum C, et al. Doxycycline prophylaxis for bacterial
sexually transmitted infections. Clin Infect Dis 2020;70:124753.
PMID:31504345 https://doi.org/10.1093/cid/ciz866
92. Myer L, Kuhn L, Stein ZA, Wright TC Jr, Denny L. Intravaginal
practices, bacterial vaginosis, and womens susceptibility to HIV
infection: epidemiological evidence and biological mechanisms. Lancet
Infect Dis 2005;5:786–94. PMID:16310150 https://doi.org/10.1016/
S1473-3099(05)70298-X
93. Molina JM, Charreau I, Chidiac C, et al.; ANRS IPERGAY Study Group.
Post-exposure prophylaxis with doxycycline to prevent sexually
transmitted infections in men who have sex with men: an open-label
randomised substudy of the ANRS IPERGAY trial. Lancet Infect Dis
2018;18:30817. PMID:29229440 https://doi.org/10.1016/
S1473-3099(17)30725-9
94. Cohen MS, Chen YQ, McCauley M, et al.; HPTN 052 Study Team.
Prevention of HIV-1 infection with early antiretroviral therapy. N Engl
J Med 2011;365:493–505. PMID:21767103 https://doi.org/10.1056/
NEJMoa1105243
95. Rodger AJ, Cambiano V, Bruun T, et al.; PARTNER Study Group.
Sexual activity without condoms and risk of HIV transmission in
serodifferent couples when the HIV-positive partner is using suppressive
antiretroviral therapy. JAMA 2016;316:17181. PMID:27404185
https://doi.org/10.1001/jama.2016.5148
96. Bavinton BR, Pinto AN, Phanuphak N, et al.; Opposites Attract Study
Group. Viral suppression and HIV transmission in serodiscordant male
couples: an international, prospective, observational, cohort study. Lancet
HIV 2018;5:e43847. PMID:30025681 https://doi.org/10.1016/
S2352-3018(18)30132-2
97. Rodger AJ, Cambiano V, Bruun T, et al.; PARTNER Study Group. Risk
of HIV transmission through condomless sex in serodifferent gay couples
with the HIV-positive partner taking suppressive antiretroviral therapy
(PARTNER): final results of a multicentre, prospective, observational
study. Lancet 2019;393:242838. PMID:31056293 https://doi.
org/10.1016/S0140-6736(19)30418-0
98. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines
for the use of antiretroviral agents in adults and adolescents with HIV.
Bethesda, MD: US Department of Health and Human Services, National
Institutes of Health, AIDSinfo. https://clinicalinfo.hiv.gov/sites/default/
files/inline-files/AdultandAdolescentGL.pdf
99. Golden MR, Kerani RP, Stenger M, et al. Effect of expedited partner
therapy (EPT) on chlamydial prevalence: the Washington State
Community-Level Trial. Presented at the STD Prevention Conference,
Minneapolis, MN; March 12–15, 2012.
100. Philip SS, Yu X, Donnell D, Vittinghoff E, Buchbinder S. Serosorting
is associated with a decreased risk of HIV seroconversion in the
EXPLORE Study Cohort. PLoS One 2010;5:e12662. PMID:20844744
https://doi.org/10.1371/journal.pone.0012662
101. Vallabhaneni S, Li X, Vittinghoff E, Donnell D, Pilcher CD,
Buchbinder SP. Seroadaptive practices: association with HIV acquisition
among HIV-negative men who have sex with men. PLoS One
2012;7:e45718. PMID:23056215 https://doi.org/10.1371/journal.
pone.0045718
102. Jin F, Prestage GP, Templeton DJ, et al. The impact of HIV seroadaptive
behaviors on sexually transmissible infections in HIV-negative
homosexual men in Sydney, Australia. Sex Transm Dis 2012;39:191–4.
PMID:22337105 https://doi.org/10.1097/OLQ.0b013e3182401a2f
103. Hotton AL, Gratzer B, Mehta SD. Association between serosorting
and bacterial sexually transmitted infection among HIV-negative men
who have sex with men at an urban lesbian, gay, bisexual, and
transgender health center. Sex Transm Dis 2012;39:95964.
PMID:23191950 https://doi.org/10.1097/OLQ.0b013e31826e870d
104. Anderson C, Gallo MF, Hylton-Kong T, et al. Randomized controlled
trial on the effectiveness of counseling messages for avoiding
unprotected sexual intercourse during sexually transmitted infection
and reproductive tract infection treatment among female sexually
transmitted infection clinic patients. Sex Transm Dis 2013;40:10510.
PMID:23321990 https://doi.org/10.1097/OLQ.0b013e31827938a1
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 139
US Department of Health and Human Services/Centers for Disease Control and Prevention
105. Golden MR, Hogben M, Handsfield HH, St Lawrence JS, Potterat JJ,
Holmes KK. Partner notification for HIV and STD in the United
States: low coverage for gonorrhea, chlamydial infection, and HIV. Sex
Transm Dis 2003;30:4906. PMID:12782949 https://doi.
org/10.1097/00007435-200306000-00004
106. Katz DA, Dombrowski JC, Kerani RP, et al. Integrating HIV testing
as an outcome of STD partner services for men who have sex with
men. AIDS Patient Care STDS 2016;30:208–14. PMID:27158848
https://doi.org/10.1089/apc.2016.0027
107. Katz DA, Dombrowski JC, Barry M, Spellman D, Bell TR, Golden
MR. STD partner services to monitor and promote HIV pre-exposure
prophylaxis use among men who have sex with men. J Acquir Immune
Defic Syndr 2019;80:53341. PMID:30649032 https://doi.
org/10.1097/QAI.0000000000001952
108. Bocour A, Renaud TC, Udeagu CC, Shepard CW. HIV partner services
are associated with timely linkage to HIV medical care. AIDS
2013;27:29613. PMID:24189585 https://doi.org/10.1097/
QAD.0000000000000031
109. Tesoriero JM, Johnson BL, Hart-Malloy R, et al. Improving retention
in HIV care through New Yorks expanded partner services Data-to-
Care pilot. J Public Health Manag Pract 2017;23:25563.
PMID:27902561 https://doi.org/10.1097/PHH.0000000000000483
110. Trelle S, Shang A, Nartey L, Cassell JA, Low N. Improved effectiveness
of partner notification for patients with sexually transmitted infections:
systematic review. BMJ 2007;334:354. PMID:17237298 https://doi.
org/10.1136/bmj.39079.460741.7C
111. CDC. Recommendations for partner services programs for HIV
infection, syphilis, gonorrhea, and chlamydial infection. MMWR
Recomm Rep 2008;57(No. RR-9). PMID:18987617
112. Thurman AR, Shain RN, Holden AE, Champion JD, Perdue ST, Piper
JM. Partner notification of sexually transmitted infections: a large
cohort of Mexican American and African American women. Sex Transm
Dis 2008;35:13640. PMID:17898679 https://doi.org/10.1097/
OLQ.0b013e318151498f
113. Kissinger PJ, Niccolai LM, Magnus M, et al. Partner notification for
HIV and syphilis: effects on sexual behaviors and relationship stability.
Sex Transm Dis 2003;30:7582. PMID:12514447 https://doi.
org/10.1097/00007435-200301000-00015
114. Smith SG, Zhang X, Basile KC, et al. The National Intimate Partner
and Sexual Violence Survey: 2015 data brief—updated release. Atlanta
GA: US Department of Health and Human Services, CDC, National
Center for Injury Prevention and Control; 2018. https://www.cdc.gov/
violenceprevention/pdf/2015data-brief508.pdf
115. Wilson TE, Hogben M, Malka ES, et al. A randomized controlled trial
for reducing risks for sexually transmitted infections through enhanced
patient-based partner notification. Am J Public Health 2009;99(Suppl
1):S10410. PMID:18556619 https://doi.org/10.2105/
AJPH.2007.112128
116. Yu YY, Frasure-Williams JA, Dunne EF, Bolan G, Markowitz L, Bauer
HM. Chlamydia partner services for females in California family
planning clinics. Sex Transm Dis 2011;38:913–8. PMID:21934563
https://doi.org/10.1097/OLQ.0b013e3182240366
117. Mickiewicz T, Al-Tayyib A, Thrun M, Rietmeijer C. Implementation
and effectiveness of an expedited partner therapy program in an urban
clinic. Sex Transm Dis 2012;39:923–9. PMID:23169171 https://doi.
org/10.1097/OLQ.0b013e3182756f20
118. Kachur R, Strona FV, Kinsey J, Collins D. Introducing technology
into partner services: a toolkit for programs. Atlanta, GA: US
Department of Health and Human Services, CDC; 2015. https://www.
cdc.gov/std/program/ips/ips-toolkit-12-28-2015.pdf
119. Kachur R, Hall W, Coor A, Kinsey J, Collins D, Strona FV. The use
of technology for sexually transmitted disease partner services in the
United States: a structured review. Sex Transm Dis 2018;45:707–12.
PMID:29771868 https://doi.org/10.1097/OLQ.0000000000000864
120. Pellowski J, Mathews C, Kalichman MO, Dewing S, Lurie MN,
Kalichman SC. Advancing partner notification through electronic
communication technology: a review of acceptability and utilization
research. J Health Commun 2016;21:62937. PMID:27144318
https://doi.org/10.1080/10810730.2015.1128020
121. Borchardt LN, Pickett ML, Tan KT, Visotcky AM, Drendel AL.
Expedited partner therapy: pharmacist refusal of legal prescriptions.
Sex Transm Dis 2018;45:3503. PMID:29465689 https://doi.
org/10.1097/OLQ.0000000000000751
122. Qin JZ, Diniz CP, Coleman JS. Pharmacy-level barriers to implementing
expedited partner therapy in Baltimore, Maryland. Am J Obstet
Gynecol 2018;218:504.e16. PMID:29410060 https://doi.
org/10.1016/j.ajog.2018.01.036
123. Schillinger J, Slutsker J, Tsang L, et al. Do prescriptions for expedited
partner therapy get filled? Findings from a multi-jurisdictional
evaluation, US, 2017–2018. Sex Transm Infect 2019;95(Suppl
1):A107.
124. Slutsker JS, Tsang LB, Schillinger JA. Do prescriptions for expedited
partner therapy for chlamydia get filled? Findings from a multi-
jurisdictional evaluation, United States, 2017–2019. Sex Transm Dis
2020;47:37682. PMID:32149956 https://doi.org/10.1097/
OLQ.0000000000001163
125. Golden MR, Whittington WL, Handsfield HH, et al. Effect of
expedited treatment of sex partners on recurrent or persistent gonorrhea
or chlamydial infection. N Engl J Med 2005;352:67685.
PMID:15716561 https://doi.org/10.1056/NEJMoa041681
126. Schillinger JA, Kissinger P, Calvet H, et al. Patient-delivered partner
treatment with azithromycin to prevent repeated Chlamydia trachomatis
infection among women: a randomized, controlled trial. Sex Transm
Dis 2003; 30: 49 56. PMID:12514443 https://doi.
org/10.1097/00007435-200301000-00011
127. Kissinger P, Mohammed H, Richardson-Alston G, et al. Patient-
delivered partner treatment for male urethritis: a randomized, controlled
trial. Clin Infect Dis 2005;41:623–9. PMID:16080084 https://doi.
org/10.1086/432476
128. Cameron ST, Glasier A, Scott G, et al. Novel interventions to reduce
re-infection in women with chlamydia: a randomized controlled trial.
Hum Reprod 2009;24:88895. PMID:19136481 https://doi.
org/10.1093/humrep/den475
129. Kissinger P, Schmidt N, Mohammed H, et al. Patient-delivered partner
treatment for Trichomonas vaginalis infection: a randomized controlled
trial. Sex Transm Dis 2006;33:445–50. PMID:16531939 https://doi.
org/10.1097/01.olq.0000204511.84485.4c
130. Schwebke JR, Desmond RA. A randomized controlled trial of partner
notification methods for prevention of trichomoniasis in women. Sex
Transm Dis 2010;37:3926. PMID:20453720 https://doi.org/10.1097/
OLQ.0b013e3181dd1691
131. Stephens SC, Bernstein KT, Katz MH, Philip SS, Klausner JD. The
effectiveness of patient-delivered partner therapy and chlamydial and
gonococcal reinfection in San Francisco. Sex Transm Dis 2010;37:525
9. PMID:20502392 https://doi.org/10.1097/OLQ.0b013e3181d8920f
132. Kerani RP, Fleming M, DeYoung B, Golden MR. A randomized,
controlled trial of inSPOT and patient-delivered partner therapy for
gonorrhea and chlamydial infection among men who have sex with
men. Sex Transm Dis 2011;38:941–6. PMID:21934569 https://doi.
org/10.1097/OLQ.0b013e318223fcbc
133. Stekler J, Bachmann L, Brotman RM, et al. Concurrent sexually
transmitted infections (STIs) in sex partners of patients with selected
STIs: implications for patient-delivered partner therapy. Clin Infect
Dis 2005;40:78793. PMID:15736009 https://doi.
org/10.1086/428043
134. McNulty A, Te h MF, Freedman E. Patient delivered partner therapy
for chlamydial infection—what would be missed? Sex Transm Dis
2008;35:8346. PMID:18580822 https://doi.org/10.1097/
OLQ.0b013e3181761993
Recommendations and Reports
140 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
135. Schillinger J, Jamison K, Slutsker J, et al. STI and HIV infections
among MSM reporting exposure to gonorrhea or chlamydia:
implications for expedited partner therapy. Sex Transm Infect
2019;95(Suppl 1):A107. http://dx.doi.org/10.1136/
sextrans-2019-sti.272
136. Turner AN, Feldblum PJ, Hoke TH. Baseline infection with a sexually
transmitted disease is highly predictive of reinfection during follow-up
in Malagasy sex workers. Sex Transm Dis 2010;37:55962.
PMID:20716996 https://doi.org/10.1097/OLQ.0b013e3181d70a03
137. Peterman TA, Tian LH, Metcalf CA, et al.; RESPECT-2 Study Group.
High incidence of new sexually transmitted infections in the year
following a sexually transmitted infection: a case for rescreening. Ann
Intern Med 2006;145:56472. PMID:17043338 https://doi.
org/10.7326/0003-4819-145-8-200610170-00005
138. Owens DK, Davidson KW, Krist AH, et al.; US Preventive Services
Task Force. Screening for HIV infection: US Preventive Services Task
Force recommendation statement. JAMA 2019;321:232636.
PMID:31184701 https://doi.org/10.1001/jama.2019.6587
139. Health and Human Services Panel on Treatment of HIV-Infected
Pregnant Women and Prevention of Perinatal Transmission.
Recommendations for use of antiretroviral drugs in pregnant HIV-1-
infected women for maternal health and interventions to reduce
perinatal HIV transmission in the United States. Bethesda, MD: US
Department of Health and Human Services, National Institutes of
Health, AIDSinfo; 2014. https://npin.cdc.gov/publication/
recommendations-use-antiretroviral-drugs-pregnant-hiv-1-infected-
women-maternal-health
140. Committee on Obstetric Practice HIV Expert Work Group. ACOG
Committee opinion no. 752: prenatal and perinatal human
immunodeficiency virus testing. Obstet Gynecol 2018;132:e138–42.
PMID:30134428 https://doi.org/10.1097/AOG.0000000000002825
141. CDC. Sexually transmitted disease surveillance 2019 [Internet]. Atlanta
GA: US Department of Health and Human Services, CDC; 2021.
https://www.cdc.gov/std/statistics/2019/default.htm
142. Warren HP, Cramer R, Kidd S, Leichliter JS. State requirements for
prenatal syphilis screening in the United States, 2016. Matern Child
Health J 2018;22:122732. PMID:30019155 https://doi.org/10.1007/
s10995-018-2592-0
143. Lin JS, Eder M, Bean S. Screening for syphilis infection in pregnant
women: a reaffirmation evidence update for the U.S. Preventive Services
Task Force. Evidence Synthesis No. 167. AHRQ Publication No.
18-05238-EF-1. Rockville, MD: Agency for Healthcare Research and
Quality; 2018.
144. Neblett Fanfair R, Tao G, Owusu-Edusei K, Gift TL, Bernstein KT.
Suboptimal prenatal syphilis testing among commercially insured
women in the United States, 2013. Sex Transm Dis 2017;44:219–21.
PMID:28282647 https://doi.org/10.1097/OLQ.0000000000000569
145. Patel CG, Huppert JS, Tao G. Provider adherence to syphilis testing
recommendations for women delivering a stillbirth. Sex Transm Dis
2017;44:68590. PMID:28876321 https://doi.org/10.1097/
OLQ.0000000000000656
146. Matthias JM, Rahman MM, Newman DR, Peterman TA. Effectiveness
of prenatal screening and treatment to prevent congenital syphilis,
Louisiana and Florida, 2013–2014. Sex Transm Dis 2017;44:498502.
PMID:28703731 https://doi.org/10.1097/OLQ.0000000000000638
147. Albright CM, Emerson JB, Werner EF, Hughes BL. Third-trimester
prenatal syphilis screening: a cost-effectiveness analysis. Obstet Gynecol
2015;126:47985. PMID:26244531 https://doi.org/10.1097/
AOG.0000000000000997
148. Owens DK, Davidson KW, Krist AH, et al.; US Preventive Services
Task Force. Screening for hepatitis B virus infection in pregnant women:
US Preventive Services Task Force reaffirmation recommendation
statement. JAMA 2019;322:34954. PMID:31334800 https://doi.
org/10.1001/jama.2019.9365
149. LeFevre ML; US Preventive Services Task Force. Screening for
chlamydia and gonorrhea: U.S. Preventive Services Task Force
recommendation statement. Ann Intern Med 2014;161:90210.
PMID:25243785 https://doi.org/10.7326/M14-1981
150. Watts T, Stockman L, Martin J, Guilfoyle S, Vergeront JM. Increased
risk for mother-to-infant transmission of hepatitis C virus among
Medicaid recipients—Wisconsin, 2011–2015. MMWR Morb Mortal
Wkly Rep 2017;66:11369. PMID:29072864 https://doi.
org/10.15585/mmwr.mm6642a3
151. Patrick SW, Bauer AM, Warren MD, Jones TF, Wester C. Hepatitis C
virus infection among women giving birth—Tennessee and United
States, 2009–2014. MMWR Morb Mortal Wkly Rep 2017;66:470–3.
PMID:28493860 https://doi.org/10.15585/mmwr.mm6618a3
152. Chappell CA, Hillier SL, Crowe D, Meyn LA, Bogen DL, Krans EE.
Hepatitis C virus screening among children exposed during pregnancy.
Pediatrics 2018;141:e20173273. PMID:29720535 https://doi.
org/10.1542/peds.2017-3273
153. Gowda C, Kennedy S, Glover C, Prasad MR, Wang L, Honegger JR.
Enhanced identification of maternal hepatitis C virus infection using
existing public health surveillance systems. Paediatr Perinat Epidemiol
2018;32:40110. PMID:29972246 https://doi.org/10.1111/
ppe.12481
154. Waruingi W, Mhanna MJ, Kumar D, Abughali N. Hepatitis C virus
universal screening versus risk based selective screening during
pregnancy. J Neonatal Perinatal Med 2015;8:371–8. PMID:26836823
https://doi.org/10.3233/NPM-15915024
155. Boudova S, Mark K, El-Kamary SS. Risk-based hepatitis C screening
in pregnancy is less reliable than universal screening: a retrospective
chart review. Open Forum Infect Dis 2018;5:ofy043. PMID:29564364
https://doi.org/10.1093/ofid/ofy043
156. Schillie S, Wester C, Osborne M, Wesolowski L, Ryerson AB. CDC
recommendations for hepatitis C screening among adults—United
States, 2020. MMWR Recomm Rep 2020;69(No. RR-2).
PMID:32271723 https://doi.org/10.15585/mmwr.rr6902a1
157. Moyer VA; US Preventive Services Task Force. Screening for hepatitis C virus
infection in adults: U.S. Preventive Services Task Force recommendation
statement. Ann Intern Med 2013;159:34957. PMID:23798026
https://doi.org/10.7326/0003-4819-159-5-201309030-00672
158. Perkins RB, Guido RS, Castle PE, et al.; 2019 ASCCP Risk-Based
Management Consensus Guidelines Committee. 2019 ASCCP risk-
based management consensus guidelines for abnormal cervical cancer
screening tests and cancer precursors. J Low Genit Tract Dis
2020;24:10231. PMID:32243307 https://doi.org/10.1097/
LGT.0000000000000525
159. Owens DK, Davidson KW, Krist AH, et al.; US Preventive Services
Task Force. Screening for bacterial vaginosis in pregnant persons to
prevent preterm delivery: US Preventive Services Task Force
recommendation statement. JAMA 2020;323:128692.
PMID:32259236 https://doi.org/10.1001/jama.2020.2684
160. American Academy of Pediatrics Committee on Fetus and Newborn;
American College of Obstetrics and Gynecology Committee on Obstetric
Practice. Guidelines for perinatal care. Kilpatrick SJ, Papile LA, eds. 8th
ed. Itasca, IL: American Academy of Pediatrics and Washington, DC:
American College of Obstetrics and Gynecology; 2017.
161. Curry SJ, Krist AH, Owens DK, et al.; US Preventive Services Task
Force. Screening for syphilis infection in pregnant women: US
Preventive Services Task Force reaffirmation recommendation
statement. JAMA 2018;320:9117. PMID:30193283 https://doi.
org/10.1001/jama.2018.11785
162. Bibbins-Domingo K, Grossman DC, Curry SJ, et al.; US Preventive
Services Task Force. Serologic screening for genital herpes infection:
US Preventive Services Task Force recommendation statement. JAMA
2016;316:252530. PMID:27997659 https://doi.org/10.1001/
jama.2016.16776
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 141
US Department of Health and Human Services/Centers for Disease Control and Prevention
163. Selph SS, Bougatsos C, Dana T, Grusing S, Chou R. Screening for
HIV infection in pregnant women: updated evidence report and
systematic review for the US Preventive Services Task Force. JAMA
2019;321:234960. PMID:31184704 https://doi.org/10.1001/
jama.2019.2593
164. Henderson JT, Webber EM, Bean SI. Screening for Hepatitis B
infection in pregnant women: updated evidence report and systematic
review for the US Preventive Services Task Force. JAMA 2019;322:360–2.
PMID:31334780 https://doi.org/10.1001/jama.2019.1655
165. Leichliter JS, Dittus PJ, Copen CE, Aral SO. Trends in factors indicating
increased risk for STI among key subpopulations in the United States,
20022015. Sex Transm Infect 2020;96:121–3. PMID:31350378
https://doi.org/10.1136/sextrans-2019-054045
166. Committee on Adolescence; Council on Clinical and Information
Technology; Blythe MJ, Del Beccaro MA. Standards for health
information technology to ensure adolescent privacy. Pediatrics
2012;130:98790. PMID:23109684 https://doi.org/10.1542/
peds.2012-2580
167. ACOG Committee Opinion no. 599: Committee on Adolescent Health
Care: adolescent confidentiality and electronic health records. Obstet
Gynecol 2014;123:114850. PMID:24785881 https://doi.
org/10.1097/01.AOG.0000446825.08715.98
168. Thompson LA, Martinko T, Budd P, Mercado R, Schentrup AM.
Meaningful use of a confidential adolescent patient portal. J Adolesc
Health 2016;58:13440. PMID:26802988 https://doi.org/10.1016/j.
jadohealth.2015.10.015
169. Society for Adolescent Health and Medicine; American Academy of
Pediatrics. Confidentiality protections for adolescents and young adults
in the health care billing and insurance claims process. J Adolesc Health
2016;58:3747. PMID:26903437 https://doi.org/10.1016/j.
jadohealth.2015.12.009
170. Bamberger DM, Graham G, Dennis L, Gerkovich MM. Extragenital
gonorrhea and chlamydia among men and women according to type
of sexual exposure. Sex Transm Dis 2019;46:32934. PMID:30676485
https://doi.org/10.1097/OLQ.0000000000000967
171. Chan PA, Robinette A, Montgomery M, et al. Extragenital infections
caused by Chlamydia trachomatis and Neisseria gonorrhoeae: a review of
the literature. Infect Dis Obstet Gynecol 2016;2016:5758387.
PMID:27366021 https://doi.org/10.1155/2016/5758387
172. Owusu-Edusei K Jr, Hoover KW, Gift TL. Cost-effectiveness of opt-out
chlamydia testing for high-risk young women in the U.S. Am J Prev
Med 2016;51:21624. PMID:26952078 https://doi.org/10.1016/j.
amepre.2016.01.007
173. DiClemente RJ, Sales JM, Danner F, Crosby RA. Association between
sexually transmitted diseases and young adults’ self-reported abstinence.
Pediatrics 2011;127:20813. PMID:21199852 https://doi.
org/10.1542/peds.2009-0892
174. Curry SJ, Krist AH, Owens DK, et al.; US Preventive Services Task
Force. Screening for cervical cancer: US Preventive Services Task Force
recommendation statement. JAMA 2018;320:67486.
PMID:30140884 https://doi.org/10.1001/jama.2018.10897
175. Committee on Practice Bulletins—Gynecology. Practice bulletin no.
157: cervical cancer screening and prevention. Obstet Gynecol
2016;127:e120. PMID:26695583 https://doi.org/10.1097/
AOG.0000000000001263
176. Benard VB, Watson M, Castle PE, Saraiya M. Cervical carcinoma rates
among young females in the United States. Obstet Gynecol
2012;120:111723. PMID:23090530 https://doi.org/10.1097/
AOG.0b013e31826e4609
177. Fontham ETH, Wolf AMD, Church TR, et al. Cervical cancer screening
for individuals at average risk: 2020 guideline update from the American
Cancer Society. CA Cancer J Clin 2020;70:32146. PMID:32729638
https://doi.org/10.3322/caac.21628
178. Owens DK, Davidson KW, Krist AH, et al.; US Preventive Services
Task Force. Preexposure prophylaxis for the prevention of HIV
infection: US Preventive Services Task Force recommendation
statement. JAMA 2019;321:2203–13. PMID:31184747 https://doi.
org/10.1001/jama.2019.6390
179. Mayer KH, Bekker LG, Stall R, Grulich AE, Colfax G, Lama JR.
Comprehensive clinical care for men who have sex with men: an
integrated approach. Lancet 2012;380:37887. PMID:22819653
https://doi.org/10.1016/S0140-6736(12)60835-6
180. Buchbinder SP, Vittinghoff E, Heagerty PJ, et al. Sexual risk, nitrite
inhalant use, and lack of circumcision associated with HIV
seroconversion in men who have sex with men in the United States.
J Acquir Immune Defic Syndr 2005;39:829. PMID:15851918
https://doi.org/10.1097/01.qai.0000134740.41585.f4
181. Paz-Bailey G, Mendoza MC, Finlayson T, et al.; NHBS Study Group.
Trends in condom use among MSM in the United States: the role of
antiretroviral therapy and seroadaptive strategies. AIDS
2016;30:1985–90. PMID:27149088 https://doi.org/10.1097/
QAD.0000000000001139
182. Spicknall IH, Gift TL, Bernstein KT, Aral SO. Sexual networks and
infection transmission networks among men who have sex with men
as causes of disparity and targets of prevention. Sex Transm Infect
2017;93:3078. PMID:28389442 https://doi.org/10.1136/
sextrans-2016-052676
183. Glick SN, Morris M, Foxman B, et al. A comparison of sexual behavior
patterns among men who have sex with men and heterosexual men
and women. J Acquir Immune Defic Syndr 2012;60:8390.
PMID:22522237 https://doi.org/10.1097/QAI.0b013e318247925e
184. Goodreau SM, Golden MR. Biological and demographic causes of
high HIV and sexually transmitted disease prevalence in men who have
sex with men. Sex Transm Infect 2007;83:458–62. PMID:17855487
https://doi.org/10.1136/sti.2007.025627
185. Chew Ng RA, Samuel MC, Lo T, et al. Sex, drugs (methamphetamines),
and the Internet: increasing syphilis among men who have sex with
men in California, 2004–2008. Am J Public Health 2013;103:1450–6.
PMID:23153138 https://doi.org/10.2105/AJPH.2012.300808
186. Bernstein KT, Stephens SC, Strona FV, Kohn RP, Philip SS.
Epidemiologic characteristics of an ongoing syphilis epidemic among
men who have sex with men, San Francisco. Sex Transm Dis
2013;40:117. PMID:23254114 https://doi.org/10.1097/
OLQ.0b013e31827763ea
187. Cohen SE, Chew Ng RA, Katz KA, et al. Repeat syphilis among men
who have sex with men in California, 2002–2006: implications for
syphilis elimination efforts. Am J Public Health 2012;102:e18.
PMID:22095364 https://doi.org/10.2105/AJPH.2011.300383
188. Kirkcaldy RD, Harvey A, Papp JR, et al. Neisseria gonorrhoeae
antimicrobial susceptibility surveillance—The Gonococcal Isolate
Surveillance Project, 27 sites, United States, 2014. MMWR Surveill
Summ 2016;65(No. SS-7). PMID:27414503 https://doi.org/10.15585/
mmwr.ss6507a1
189. Kirkcaldy RD, Zaidi A, Hook EW 3rd, et al. Neisseria
gonorrhoeae antimicrobial resistance among men who have sex
with men and men who have sex exclusively with women:
the Gonococcal Isolate Surveillance Project, 2005–2010. Ann
Intern Med 2013;158:3218. PMID:23460055 https://doi.
org/10.7326/0003-4819-158-5-201303050-00004
190. Newman LM, Dowell D, Bernstein K, et al. A tale of two gonorrhea
epidemics: results from the STD Surveillance Network. Public Health
Rep 2012;127:28292. PMID:22547859 https://doi.
org/10.1177/003335491212700308
191. Hess KL, Hu X, Lansky A, Mermin J, Hall HI. Lifetime risk of a
diagnosis of HIV infection in the United States. Ann Epidemiol
2017;27:23843. PMID:28325538 https://doi.org/10.1016/j.
annepidem.2017.02.003
Recommendations and Reports
142 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
192. Patel P, Borkowf CB, Brooks JT, Lasry A, Lansky A, Mermin J.
Estimating per-act HIV transmission risk: a systematic review. AIDS
2014;28:150919. PMID:24809629 https://doi.org/10.1097/
QAD.0000000000000298
193. Koblin BA, Husnik MJ, Colfax G, et al. Risk factors for
HIV infection among men who have sex with men. AIDS
2006;20:7319. PMID:16514304 https://doi.org/10.1097/01.
aids.0000216374.61442.55
194. Ackers ML, Greenberg AE, Lin CY, et al. High and persistent HIV
seroincidence in men who have sex with men across 47 U.S. cities.
PLoS One 2012;7:e34972. PMID:22529964 https://doi.org/10.1371/
journal.pone.0034972
195. Zetola NM, Bernstein KT, Wong E, Louie B, Klausner JD. Exploring
the relationship between sexually transmitted diseases and HIV
acquisition by using different study designs. J Acquir Immune Defic
Syndr 2009;50:54651. PMID:19367993 https://doi.org/10.1097/
QAI.0b013e318195bd2b
196. Solomon MM, Mayer KH, Glidden DV, et al.; iPrEx Study Team.
Syphilis predicts HIV incidence among men and transgender women
who have sex with men in a preexposure prophylaxis trial. Clin Infect
Dis 2014;59:1020–6. PMID:24928295 https://doi.org/10.1093/cid/
ciu450
197. Fleming DT, Wasserheit JN. From epidemiological synergy to public
health policy and practice: the contribution of other sexually
transmitted diseases to sexual transmission of HIV infection. Sex
Transm Infect 1999;75:317. PMID:10448335 https://doi.
org/10.1136/sti.75.1.3
198. Freeman EE, Weiss HA, Glynn JR, Cross PL, Whitworth JA, Hayes
RJ. Herpes simplex virus 2 infection increases HIV acquisition in men
and women: systematic review and meta-analysis of longitudinal studies.
AIDS 2006;20:7383. PMID:16327322 https://doi.org/10.1097/01.
aids.0000198081.09337.a7
199. Reynolds SJ, Risbud AR, Shepherd ME, et al. High rates of syphilis
among STI patients are contributing to the spread of HIV-1 in India.
Sex Transm Infect 2006;82:1216. PMID:16581736 https://doi.
org/10.1136/sti.2005.015040
200. Hoots BE, Wejnert C, Martin A, et al.; NHBS Study Group.
Undisclosed HIV infection among MSM in a behavioral surveillance
study. AIDS 2019;33:9138. PMID:30649053 https://doi.
org/10.1097/QAD.0000000000002147
201. Dolling DI, Desai M, McOwan A, et al.; PROUD Study Group. An
analysis of baseline data from the PROUD study: an open-label
randomised trial of pre-exposure prophylaxis. Trials 2016;17:163.
PMID:27013513 https://doi.org/10.1186/s13063-016-1286-4
202. Oldenburg CE, Nunn AS, Montgomery M, et al. Behavioral changes
following uptake of HIV pre-exposure prophylaxis among men who
have sex with men in a clinical setting. AIDS Behav 2018;22:1075–9.
PMID:28150120 https://doi.org/10.1007/s10461-017-1701-1
203. Montaño MA, Dombrowski JC, Dasgupta S, et al. Changes in sexual
behavior and STI diagnoses among MSM initiating PrEP in a clinic
setting. AIDS Behav 2019;23:548–55. PMID:30117076 https://doi.
org/10.1007/s10461-018-2252-9
204. Traeger MW, Schroeder SE, Wright EJ, et al. Effects of pre-exposure
prophylaxis for the prevention of human immunodeficiency virus
infection on sexual risk behavior in men who have sex with men: a
systematic review and meta-analysis. Clin Infect Dis 2018;67:676–86.
PMID:29509889 https://doi.org/10.1093/cid/ciy182
205. Jenness SM, Weiss KM, Goodreau SM, et al. Incidence of gonorrhea
and chlamydia following human immunodeficiency virus preexposure
prophylaxis among men who have sex with men: a modeling study.
Clin Infect Dis 2017;65:7128. PMID:28505240 https://doi.
org/10.1093/cid/cix439
206. Tang EC, Vittinghoff E, Philip SS, et al. Quarterly screening optimizes
detection of sexually transmitted infections when prescribing HIV
preexposure prophylaxis. AIDS 2020;34:1181–6. PMID:32205724
https://doi.org/10.1097/QAD.0000000000002522
207. Barbee LA, Khosropour CM, Dombrowksi JC, Golden MR. New
human immunodeficiency virus diagnosis independently associated
with rectal gonorrhea and chlamydia in men who have sex with men.
Sex Transm Dis 2017;44:3859. PMID:28608786 https://doi.
org/10.1097/OLQ.0000000000000614
208. Bernstein KT, Marcus JL, Nieri G, Philip SS, Klausner JD. Rectal
gonorrhea and chlamydia reinfection is associated with increased risk
of HIV seroconversion. J Acquir Immune Defic Syndr 2010;53:537–43.
PMID:19935075 https://doi.org/10.1097/QAI.0b013e3181c3ef29
209. Barbee LA, Khosropour CM, Dombrowski JC, Manhart LE, Golden
MR. An estimate of the proportion of symptomatic gonococcal,
chlamydial and non-gonococcal non-chlamydial urethritis attributable
to oral sex among men who have sex with men: a case-control study.
Sex Transm Infect 2016;92:15560. PMID:26297719 https://doi.
org/10.1136/sextrans-2015-052214
210. Lafferty WE, Hughes JP, Handsfield HH. Sexually transmitted diseases
in men who have sex with men. Acquisition of gonorrhea and
nongonococcal urethritis by fellatio and implications for STD/HIV
prevention. Sex Transm Dis 1997;24:272–8. PMID:9153736 https://
doi.org/10.1097/00007435-199705000-00007
211. Bernstein KT, Stephens SC, Barry PM, et al. Chlamydia trachomatis
and Neisseria gonorrhoeae transmission from the oropharynx to the
urethra among men who have sex with men. Clin Infect Dis
2009;49:1793–7. PMID:19911970 https://doi.org/10.1086/648427
212. Patton ME, Kidd S, Llata E, et al. Extragenital gonorrhea and chlamydia
testing and infection among men who have sex with men—STD
Surveillance Network, United States, 2010–2012. Clin Infect Dis
2014;58:156470. PMID:24647015 https://doi.org/10.1093/cid/
ciu184
213. Kent CK, Chaw JK, Wong W, et al. Prevalence of rectal, urethral, and
pharyngeal chlamydia and gonorrhea detected in 2 clinical settings
among men who have sex with men: San Francisco, California, 2003.
Clin Infect Dis 2005;41:6774. PMID:15937765 https://doi.
org/10.1086/430704
214. Koedijk FD, van Bergen JE, Dukers-Muijrers NH, van Leeuwen AP,
Hoebe CJ, van der Sande MA; Dutch STI centres. The value of testing
multiple anatomic sites for gonorrhoea and chlamydia in sexually
transmitted infection centres in the Netherlands, 2006–2010. Int J STD
AIDS 2012;23:62631. PMID:23033514 https://doi.org/10.1258/
ijsa.2012.011378
215. Barbee LA, Dombrowski JC, Kerani R, Golden MR. Effect of nucleic
acid amplification testing on detection of extragenital gonorrhea and
chlamydial infections in men who have sex with men sexually
transmitted disease clinic patients. Sex Transm Dis 2014;41:168–72.
PMID:24521722 https://doi.org/10.1097/OLQ.0000000000000093
216. Danby CS, Cosentino LA, Rabe LK, et al. Patterns of extragenital
chlamydia and gonorrhea in women and men who have sex with men
reporting a history of receptive anal intercourse. Sex Transm Dis
2016;43:1059. PMID:26766527 https://doi.org/10.1097/
OLQ.0000000000000384
217. van der Helm JJ, Hoebe CJ, van Rooijen MS, et al. High performance
and acceptability of self-collected rectal swabs for diagnosis of
Chlamydia trachomatis and Neisseria gonorrhoeae in men who have sex
with men and women. Sex Transm Dis 2009;36:493–7. PMID:19617869
https://doi.org/10.1097/OLQ.0b013e3181a44b8c
218. Alexander S, Ison C, Parry J, et al.; Brighton Home Sampling Kits
Steering Group. Self-taken pharyngeal and rectal swabs are appropriate
for the detection of Chlamydia trachomatis and Neisseria gonorrhoeae
in asymptomatic men who have sex with men. Sex Transm Infect
2008;84:48892. PMID:19028953 https://doi.org/10.1136/
sti.2008.031443
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 143
US Department of Health and Human Services/Centers for Disease Control and Prevention
219. Freeman AH, Bernstein KT, Kohn RP, Philip S, Rauch LM, Klausner
JD. Evaluation of self-collected versus clinician-collected swabs for the
detection of Chlamydia trachomatis and Neisseria gonorrhoeae pharyngeal
infection among men who have sex with men. Sex Transm Dis
2011;38:10369. PMID:21992980 https://doi.org/10.1097/
OLQ.0b013e318227713e
220. Chesson HW, Bernstein KT, Gift TL, Marcus JL, Pipkin S, Kent CK.
The cost-effectiveness of screening men who have sex with men for
rectal chlamydial and gonococcal infection to prevent HIV Infection.
Sex Transm Dis 2013;40:36671. PMID:23588125 https://doi.
org/10.1097/OLQ.0b013e318284e544
221. Jenness SM, Weiss KM, Prasad P, Zlotorzynska M, Sanchez T. Bacterial
sexually transmitted infection screening rates by symptomatic status
among men who have sex with men in the United States: a hierarchical
Bayesian analysis. Sex Transm Dis 2019;46:25–30. PMID:30044334
https://doi.org/10.1097/OLQ.0000000000000896
222. Hoover KW, Butler M, Workowski K, et al.; Evaluation Group for
Adherence to STD and Hepatitis Screening. STD screening of HIV-
infected MSM in HIV clinics. Sex Transm Dis 2010;37:7716.
PMID:20585275 https://doi.org/10.1097/OLQ.0b013e3181e50058
223. de Voux A, Bernstein KT, Bradley H, Kirkcaldy RD, Tie Y, Shouse RL;
Medical Monitoring Project. Syphilis testing among sexually active
men who have sex with men and who are receiving medical care for
human immunodeficiency virus in the United States: Medical
Monitoring Project, 2013–2014. Clin Infect Dis 2019;68:934–9.
PMID:29985985 https://doi.org/10.1093/cid/ciy571
224. Gray RT, Hoare A, Prestage GP, Donovan B, Kaldor JM, Wilson DP.
Frequent testing of highly sexually active gay men is required to control
syphilis. Sex Transm Dis 2010;37:298305. PMID:20393383 https://
doi.org/10.1097/OLQ.0b013e3181ca3c0a
225. Tuite AR, Fisman DN, Mishra S. Screen more or screen more often?
Using mathematical models to inform syphilis control strategies. BMC
Public Health 2013;13:606. PMID:23800206 https://doi.
org/10.1186/1471-2458-13-606
226. Tuite A, Fisman D. Go big or go home: impact of screening coverage
on syphilis infection dynamics. Sex Transm Infect 2016;92:4954.
PMID:25954016 https://doi.org/10.1136/sextrans-2014-052001
227. Tuite AR, Shaw S, Reimer JN, Ross CP, Fisman DN, Mishra S. Can
enhanced screening of men with a history of prior syphilis infection
stem the epidemic in men who have sex with men? A mathematical
modelling study. Sex Transm Infect 2018;94:10510. PMID:28705938
https://doi.org/10.1136/sextrans-2017-053201
228. Raifman JR, Gebo KA, Mathews WC, et al.; HIV Research Network.
Gonorrhea and chlamydia case detection increased when testing
increased in a multisite US HIV cohort, 2004–2014. J Acquir Immune
Defic Syndr 2017;76:40916. PMID:28777262 https://doi.
org/10.1097/QAI.0000000000001514
229. Barbee LA, Dhanireddy S, Tat SA, Marrazzo JM. Barriers to bacterial
sexually transmitted infection testing of HIV-infected men who have
sex with men engaged in HIV primary care. Sex Transm Dis
2015;42:5904. PMID:26372931 https://doi.org/10.1097/
OLQ.0000000000000320
230. McMillan A, Young H, Moyes A. Rectal gonorrhoea in homosexual
men: source of infection. Int J STD AIDS 2000;11:2847.
PMID:10824935 https://doi.org/10.1177/095646240001100502
231. Chow EP, Cornelisse VJ, Read TR, Chen MY, Bradshaw CS, Fairley
CK. Saliva use in sex: associations with use of smartphone dating
applications in men who have sex with men. Int J STD AIDS
2018; 29: 362 6 . PMID:28835197 https://doi.
org/10.1177/0956462417727669
232. Cornelisse VJ, Priest D, Fairley CK, et al. The frequency of kissing as
part of sexual activity differs depending on how men meet their male
casual sexual partners. Int J STD AIDS 2018;29:598602.
PMID:29256822 https://doi.org/10.1177/0956462417748717
233. Krist AH, Davidson KW, Mangione CM, et al.; US Preventive Services
Task Force. Screening for hepatitis B virus infection in adolescents and
adults: US Preventive Services Task Force recommendation statement.
JAMA 2020;324:241522. PMID:33320230 https://doi.org/10.1001/
jama.2020.22980
234. Tohme RA, Holmberg SD. Is sexual contact a major mode of hepatitis
C virus transmission? Hepatology 2010;52:1497505. PMID:20635398
https://doi.org/10.1002/hep.23808
235. Wandeler G, Gsponer T, Bregenzer A, et al.; Swiss HIV Cohort Study.
Hepatitis C virus infections in the Swiss HIV Cohort Study: a rapidly
evolving epidemic. Clin Infect Dis 2012;55:140816. PMID:22893583
https://doi.org/10.1093/cid/cis694
236. Garg S, Taylor LE, Grasso C, Mayer KH. Prevalent and incident
hepatitis C virus infection among HIV-infected men who have sex with
men engaged in primary care in a Boston community health center.
Clin Infect Dis 2013;56:14807. PMID:23386630 https://doi.
org/10.1093/cid/cit054
237. Urbanus AT, van de Laar TJ, Stolte IG, et al. Hepatitis C virus infections
among HIV-infected men who have sex with men: an expanding
epidemic. AIDS 2009;23:F17. PMID:19542864 https://doi.
org/10.1097/QAD.0b013e32832e5631
238. Linas BP, Wong AY, Schackman BR, Kim AY, Freedberg KA. Cost-
effective screening for acute hepatitis C virus infection in HIV-infected
men who have sex with men. Clin Infect Dis 2012;55:27990.
PMID:22491339 https://doi.org/10.1093/cid/cis382
239. Taylor LE, DeLong AK, Maynard MA, et al. Acute hepatitis C virus
in an HIV clinic: a screening strategy, risk factors, and perception of
risk. AIDS Patient Care STDS 2011;25:571–7. PMID:21859307
https://doi.org/10.1089/apc.2011.0106
240. Kaul R, Kimani J, Nagelkerke NJ, et al.; Kibera HIV Study Group.
Monthly antibiotic chemoprophylaxis and incidence of sexually
transmitted infections and HIV-1 infection in Kenyan sex workers: a
randomized controlled trial. JAMA 2004;291:255562.
PMID:15173146 https://doi.org/10.1001/jama.291.21.2555
241. Ong JJ, Baggaley RC, Wi TE, et al. Global epidemiologic characteristics
of sexually transmitted infections among individuals using preexposure
prophylaxis for the prevention of HIV infection: a systematic review and
meta-analysis. JAMA Netw Open 2019;2:e1917134. PMID:31825501
https://doi.org/10.1001/jamanetworkopen.2019.17134
242. Paz-Bailey G, Hoots BE, Xia M, Finlayson T, Prejean J, Purcell DW;
NHBS Study Group. Trends in Internet use among men who have sex
with men in the United States. J Acquir Immune Defic Syndr
2017;75(Suppl 3):S28895. PMID:28604430 https://doi.org/10.1097/
QAI.0000000000001404
243. Badal HJ, Stryker JE, DeLuca N, Purcell DW. Swipe right: dating
website and app use among men who have sex with men. AIDS Behav
2018;22:126572. PMID:28884248 https://doi.org/10.1007/
s10461-017-1882-7
244. Chan PA, Crowley C, Rose JS, et al. A network analysis of sexually
transmitted diseases and online hookup sites among men who have sex
with men. Sex Transm Dis 2018;45:462–8. PMID:29465663 https://
doi.org/10.1097/OLQ.0000000000000784
245. Beymer MR, Weiss RE, Bolan RK, et al. Sex on demand: geosocial
networking phone apps and risk of sexually transmitted infections
among a cross-sectional sample of men who have sex with men in Los
Angeles County. Sex Transm Infect 2014;90:56772. PMID:24926041
https://doi.org/10.1136/sextrans-2013-051494
246. Medina MM, Crowley C, Montgomery MC, et al. Disclosure of HIV
serostatus and pre-exposure prophylaxis use on internet hookup sites
among men who have sex with men. AIDS Behav 2019;23:1681–8.
PMID:30267365 https://doi.org/10.1007/s10461-018-2286-z
247. Chan PA, Towe y C, Poceta J, et al. Online hookup sites for meeting
sexual partners among men who have sex with men in Rhode Island,
2013: a call for public health action. Public Health Rep 2016;131:264–71.
PMID:26957661 https://doi.org/10.1177/003335491613100210
Recommendations and Reports
144 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
248. Lampkin D, Crawley A, Lopez TP, Mejia CM, Yuen W, Levy V.
Reaching suburban men who have sex with men for STD and HIV
services through online social networking outreach: a public health
approach. J Acquir Immune Defic Syndr 2016;72:738.
PMID:27097365 https://doi.org/10.1097/QAI.0000000000000930
249. Sun CJ, Stowers J, Miller C, Bachmann LH, Rhodes SD. Acceptability
and feasibility of using established geosocial and sexual networking
mobile applications to promote HIV and STD testing among men
who have sex with men. AIDS Behav 2015;19:54352. PMID:25381563
https://doi.org/10.1007/s10461-014-0942-5
250. Dritz SK, Back AF. Letter: Shigella enteritis venereally transmitted.
N Engl J Med 1974;291:1194. PMID:4608062 https://doi.
org/10.1056/NEJM197411282912223
251. Aragón TJ, Vugia DJ, Shallow S, et al. Case-control study of shigellosis
in San Francisco: the role of sexual transmission and HIV infection.
Clin Infect Dis 2007;44:32734. PMID:17205436 https://doi.
org/10.1086/510593
252. Simms I, Field N, Jenkins C, et al. Intensified shigellosis epidemic
associated with sexual transmission in men who have sex with men—
Shigella flexneri and S. sonnei in England, 2004 to end of February
2015. Euro Surveill 2015;20:21097. PMID:25953129 https://doi.
org/10.2807/1560-7917.ES2015.20.15.21097
253. Gilbart VL, Simms I, Jenkins C, et al. Sex, drugs and smart phone
applications: findings from semistructured interviews with men who
have sex with men diagnosed with Shigella flexneri 3a in England and
Wales. Sex Transm Infect 2015;91:598602. PMID:25921020 https://
doi.org/10.1136/sextrans-2015-052014
254. Narayan S, Galanis E; BC STEI Group. Are enteric infections sexually
transmitted in British Columbia? Can Commun Dis Rep 2016;42:24–9.
PMID:29770000 https://doi.org/10.14745/ccdr.v42i02a01
255. Mohan K, Hibbert M, Rooney G, et al. What is the overlap between
HIV and shigellosis epidemics in England: further evidence of MSM
transmission? Sex Transm Infect 2018;94:6771. PMID:28490580
https://doi.org/10.1136/sextrans-2016-052962
256. Hughes G, Silalang P, Were J, et al. Prevalence and characteristics of
gastrointestinal infections in men who have sex with men diagnosed
with rectal chlamydia infection in the UK: an ‘unlinked anonymous
cross-sectional study. Sex Transm Infect 2018;94:51821.
PMID:28360379 https://doi.org/10.1136/sextrans-2016-053057
257. O’Sullivan B, Delpech V, Pontivivo G, et al. Shigellosis linked to sex
venues, Australia. Emerg Infect Dis 2002;8:862–4. PMID:12141976
https://doi.org/10.3201/eid0808.010534
258. Marcus U, Zucs P, Bremer V, et al. Shigellosis—a re-emerging sexually
transmitted infection: outbreak in men having sex with men in Berlin.
Int J STD AIDS 2004;15:5337. PMID:15307964 https://doi.
org/10.1258/0956462041558221
259. Danila RN, Eikmeier DL, Robinson TJ, La Pointe A, DeVries AS. Two
concurrent enteric disease outbreaks among men who have sex with
men, Minneapolis-St Paul area. Clin Infect Dis 2014;59:9879.
PMID:24944234 https://doi.org/10.1093/cid/ciu478
260. Okame M, Adachi E, Sato H, et al. Shigella sonnei outbreak among
men who have sex with men in Tokyo. Jpn J Infect Dis 2012;65:277–8.
PMID:22627317 https://doi.org/10.7883/yoken.65.277
261. Wilmer A, Romney MG, Gustafson R, et al. Shigella flexneri serotype
1 infections in men who have sex with men in Vancouver, Canada.
HIV Med 2015;16:16875. PMID:25656740 https://doi.org/10.1111/
hiv.12191
262. CDC. Shigella sonnei outbreak among men who have sex with men—
San Francisco, California, 2000–2001. MMWR Morb Mortal Wkly
Rep 2001;50:922–6. PMID:11699845
263. Baer JT, Vugia DJ, Reingold AL, Aragon T, Angulo FJ, Bradford WZ.
HIV infection as a risk factor for shigellosis. Emerg Infect Dis
1999;5:8203. PMID:10603219 https://doi.org/10.3201/
eid0506.990614
264. Simms I, Gilbart VL, Byrne L, et al. Identification of verocytotoxin-
producing Escherichia coli O117:H7 in men who have sex with men,
England, November 2013 to August 2014. Euro Surveill 2014;19:20946.
PMID:25375900 https://doi.org/10.2807/1560-7917.
ES2014.19.43.20946
265. Quinn TC, Goodell SE, Fennell C, et al. Infections with Campylobacter
jejuni and Campylobacter-like organisms in homosexual men. Ann
Intern Med 1984;101:18792. PMID:6547580 https://doi.
org/10.7326/0003-4819-101-2-187
266. Gaudreau C, Pilon PA, Sylvestre J-L, Boucher F, Bekal S. Multidrug-
resistant Campylobacter coli in men who have sex with men, Quebec,
Canada, 2015. Emerg Infect Dis 2016;22:1661–3. PMID:27533504
https://doi.org/10.3201/eid2209.151695
267. Chen GJ, Lin KY, Hung CC, Chang SC. Hepatitis A outbreak among
men who have sex with men in a country of low endemicity of hepatitis
A infection. J Infect Dis 2017;215:133940. PMID:28329351 https://
doi.org/10.1093/infdis/jix123
268. Lo YC, Ji DD, Hung CC. Prevalent and incident HIV diagnoses among
Entamoeba histolytica-infected adult males: a changing epidemiology
associated with sexual transmission—Taiwan, 2006–2013. PLoS Negl
Trop Dis 2014;8:e3222. PMID:25299178 https://doi.org/10.1371/
journal.pntd.0003222
269. Stark D, van Hal SJ, Matthews G, Harkness J, Marriott D. Invasive
amebiasis in men who have sex with men, Australia. Emerg Infect Dis
2008;14:11413. PMID:18598643 https://doi.org/10.3201/
eid1407.080017
270. Mitchell H, Hughes G. Recent epidemiology of sexually transmissible
enteric infections in men who have sex with men. Curr Opin Infect
Dis 2018;31:506. PMID:29251673 https://doi.org/10.1097/
QCO.0000000000000423
271. Weatherburn P, Hickson F, Reid D, Torres-Rueda S, Bourne A.
Motivations and values associated with combining sex and illicit drugs
(‘chemsex’) among gay men in South London: findings from a
qualitative study. Sex Transm Infect 2017;93:203–6. PMID:27519259
https://doi.org/10.1136/sextrans-2016-052695
272. Baker KS, Dallman TJ, Ashton PM, et al. Intercontinental dissemination
of azithromycin-resistant shigellosis through sexual transmission: a
cross-sectional study. Lancet Infect Dis 2015;15:91321.
PMID:25936611 https://doi.org/10.1016/S1473-3099(15)00002-X
273. Bowen A, Grass J, Bicknese A, Campbell D, Hurd J, Kirkcaldy RD.
Elevated risk for antimicrobial drug-resistant Shigella infection among
men who have sex with men, United States, 2011–2015. Emerg Infect
Dis 2016;22:1613–6. PMID:27533624 https://doi.org/10.3201/
eid2209.160624
274. Gaudreau C, Rodrigues-Coutlée S, Pilon PA, Coutlée F, Bekal S. Long-
lasting outbreak of erythromycin- and ciprofloxacin-resistant
Campylobacter jejuni subspecies jejuni from 2003 to 2013 in men who
have sex with men, Quebec, Canada. Clin Infect Dis 2015;61:154952.
PMID:26187024 https://doi.org/10.1093/cid/civ570
275. Muzny CA, Sunesara IR, Martin DH, Mena LA. Sexually transmitted
infections and risk behaviors among African American women who
have sex with women: does sex with men make a difference? Sex Transm
Dis 2011;38:111825. PMID:22082722 https://doi.org/10.1097/
OLQ.0b013e31822e6179
276. Eisenberg M. Differences in sexual risk behaviors between college
students with same-sex and opposite-sex experience: results from a
national survey. Arch Sex Behav 2001;30:575–89. PMID:11725456
https://doi.org/10.1023/A:1011958816438
277. Koh AS, Gómez CA, Shade S, Rowley E. Sexual risk factors among
self-identified lesbians, bisexual women, and heterosexual women
accessing primary care settings. Sex Transm Dis 2005;32:5639.
PMID:16118605 https://doi.org/10.1097/01.
olq.0000175417.17078.21
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 145
US Department of Health and Human Services/Centers for Disease Control and Prevention
278. Goodenow C, Szalacha LA, Robin LE, Westheimer K. Dimensions of
sexual orientation and HIV-related risk among adolescent females:
evidence from a statewide survey. Am J Public Health 2008;98:1051–8.
PMID:18445809 https://doi.org/10.2105/AJPH.2005.080531
279. Muzny CA, Austin EL, Harbison HS, Hook EW 3rd. Sexual
partnership characteristics of African American women who have sex
with women; impact on sexually transmitted infection risk. Sex Transm
Dis 2014;41:6117. PMID:25211257 https://doi.org/10.1097/
OLQ.0000000000000194
280. Riskind RG, Tornello SL, Younger BC, Patterson CJ. Sexual identity,
partner gender, and sexual health among adolescent girls in the United
States. Am J Public Health 2014;104:195763. PMID:25121821
https://doi.org/10.2105/AJPH.2014.302037
281. Schick V, Rosenberger JG, Herbenick D, Reece M. Sexual behaviour
and risk reduction strategies among a multinational sample of women
who have sex with women. Sex Transm Infect 2012;88:40712.
PMID:22563015 https://doi.org/10.1136/sextrans-2011-050404
282. Richters J, Prestage G, Schneider K, Clayton S. Do women use dental
dams? Safer sex practices of lesbians and other women who have sex
with women. Sex Health 2010;7:1659. PMID:20465981 https://doi.
org/10.1071/SH09072
283. Rowen TS, Breyer BN, Lin TC, Li CS, Robertson PA, Shindel AW.
Use of barrier protection for sexual activity among women who have
sex with women. Int J Gynaecol Obstet 2013;120:425.
PMID:23106842 https://doi.org/10.1016/j.ijgo.2012.08.011
284. Lindley LL, Friedman DB, Struble C. Becoming visible: assessing the
availability of online sexual health information for lesbians. Health
Promot Pract 2012;13:47280. PMID:21677116 https://doi.
org/10.1177/1524839910390314
285. Chetcuti N, Beltzer N, Methy N, Laborde C, Velter A, Bajos N; CSF
Group. Preventive care’s forgotten women: life course, sexuality, and
sexual health among homosexually and bisexually active women in
France. J Sex Res 2013;50:587–97. PMID:22497621 https://doi.org
/10.1080/00224499.2012.657264
286. Logie CH, Navia D, Loutfy MR. Correlates of a lifetime history of
sexually transmitted infections among women who have sex with
women in Toronto, Canada: results from a cross-sectional internet-
based survey. Sex Transm Infect 2015;91:278–83. PMID:25477474
https://doi.org/10.1136/sextrans-2014-051745
287. Muzny CA, Kapil R, Austin EL, Hook EW, Geisler WM. Lower sexually
transmissible infection prevalence among lifetime exclusive women
who have sex with women compared with women who have sex with
women and men. Sex Health 2014;11:592–3. PMID:25435197
https://doi.org/10.1071/SH14181
288. Muzny CA, Harbison HS, Pembleton ES, Austin EL. Sexual behaviors,
perception of sexually transmitted infection risk, and practice of safe
sex among southern African American women who have sex with
women. Sex Transm Dis 2013;40:395400. PMID:23588129 https://
doi.org/10.1097/OLQ.0b013e31828caf34
289. Przedworski JM, McAlpine DD, Karaca-Mandic P, VanKim NA. Health
and health risks among sexual minority women: an examination of 3
subgroups. Am J Public Health 2014;104:1045–7. PMID:24825204
https://doi.org/10.2105/AJPH.2013.301733
290. Brenick A, Romano K, Kegler C, Eaton LA. Understanding the
influence of stigma and medical mistrust on engagement in routine
healthcare among Black women who have sex with women. LGBT
Health 2017;4:410. PMID:28113005 https://doi.org/10.1089/
lgbt.2016.0083
291. Fethers K, Marks C, Mindel A, Estcourt CS. Sexually transmitted
infections and risk behaviours in women who have sex with women.
Sex Transm Infect 2000;76:3459. PMID:11141849 https://doi.
org/10.1136/sti.76.5.345
292. Marrazzo JM, Koutsky LA, Eschenbach DA, Agnew K, Stine K, Hillier
SL. Characterization of vaginal flora and bacterial vaginosis in women
who have sex with women. J Infect Dis 2002;185:130713.
PMID:12001048 https://doi.org/10.1086/339884
293. Kellock D, O’Mahony CP. Sexually acquired metronidazole-resistant
trichomoniasis in a lesbian couple. Genitourin Med 1996;72:60–1.
PMID:8655171 https://doi.org/10.1136/sti.72.1.60
294. Muzny CA, Rivers CA, Mena LA, Schwebke JR. Genotypic
characterization of Trichomonas vaginalis isolates among women who
have sex with women in sexual partnerships. Sex Transm Dis
2012;39:5568. PMID:22706219 https://doi.org/10.1097/
OLQ.0b013e31824f1c49
295. Chan SK, Thornton LR, Chronister KJ, et al.; CDC. Likely female-
to-female sexual transmission of HIV—Texas, 2012. MMWR Morb
Mortal Wkly Rep 2014;63:209–12. PMID:24622284
296. Kwakwa HA, Ghobrial MW. Female-to-female transmission of human
immunodeficiency virus. Clin Infect Dis 2003;36:e401.
PMID:12539088 https://doi.org/10.1086/345462
297. Marrazzo JM, Handsfield HH, Whittington WL. Predicting chlamydial
and gonococcal cervical infection: implications for management of
cervicitis. Obstet Gynecol 2002;100:57984. PMID:12220782 https://
doi.org/10.1097/00006250-200209000-00029
298. Diamant AL, Schuster MA, McGuigan K, Lever J. Lesbians’ sexual
history with men: implications for taking a sexual history. Arch Intern
Med 1999;159:2730–6. PMID:10597764 https://doi.org/10.1001/
archinte.159.22.2730
299. Xu F, Sternberg MR, Markowitz LE. Women who have sex with women
in the United States: prevalence, sexual behavior and prevalence of
herpes simplex virus type 2 infection—results from national health and
nutrition examination survey 2001–2006. Sex Transm Dis
2010;37:40713. PMID:20531032 https://doi.org/10.1097/
OLQ.0b013e3181db2e18
300. Everett BG, Higgins JA, Haider S, Carpenter E. Do sexual minorities
receive appropriate sexual and reproductive health care and counseling?
J Womens Health (Larchmt) 2019;28:5362. PMID:30372369 https://
doi.org/10.1089/jwh.2017.6866
301. Marrazzo JM, Koutsky LA, Stine KL, et al. Genital human
papillomavirus infection in women who have sex with women. J Infect
Dis 1998;178:16049 . PMID:9815211 https://doi.
org/10.1086/314494
302. Marrazzo JM, Koutsky LA, Kiviat NB, Kuypers JM, Stine K.
Papanicolaou test screening and prevalence of genital human
papillomavirus among women who have sex with women. Am J Public
Health 2001;91:947–52. PMID:11392939 https://doi.org/10.2105/
AJPH.91.6.947
303. Bailey JV, Kavanagh J, Owen C, McLean KA, Skinner CJ. Lesbians
and cervical screening. Br J Gen Pract 2000;50:481–2. PMID:10962789
304. Anderson TA, Schick V, Herbenick D, Dodge B, Fortenberry JD. A
study of human papillomavirus on vaginally inserted sex toys, before
and after cleaning, among women who have sex with women and men.
Sex Transm Infect 2014;90:52931. PMID:24739872 https://doi.
org/10.1136/sextrans-2014-051558
305. Marrazzo JM, Stine K, Wald A. Prevalence and risk factors for infection
with herpes simplex virus type-1 and -2 among lesbians. Sex Transm
Dis 2003;30:890–5. PMID:14646636 https://doi.org/10.1097/01.
OLQ.0000091151.52656.E5
306. Muzny CA, Schwebke JR. The clinical spectrum of Trichomonas
vaginalis infection and challenges to management. Sex Transm Infect
2013;89:4235. PMID:23543252 https://doi.org/10.1136/
sextrans-2012-050893
307. Muzny CA, Blackburn RJ, Sinsky RJ, Austin EL, Schwebke JR. Added
benefit of nucleic acid amplification testing for the diagnosis of
Trichomonas vaginalis among men and women attending a sexually
transmitted diseases clinic. Clin Infect Dis 2014;59:83441.
PMID:24928292 https://doi.org/10.1093/cid/ciu446
Recommendations and Reports
146 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
308. Singh D, Fine DN, Marrazzo JM. Chlamydia trachomatis infection
among women reporting sexual activity with women screened in family
planning clinics in the Pacific Northwest, 1997 to 2005. Am J Public
Health 2011;101:128490. PMID:20724697 https://doi.org/10.2105/
AJPH.2009.169631
309. Muzny CA, Kapil R, Austin EL, Brown L, Hook EW 3rd, Geisler WM.
Chlamydia trachomatis infection in African American women who
exclusively have sex with women. Int J STD AIDS 2016;27:978–83.
PMID:26384942 https://doi.org/10.1177/0956462415604092
310. Koumans EH, Sternberg M, Bruce C, et al. The prevalence of bacterial
vaginosis in the United States, 2001–2004; associations with symptoms,
sexual behaviors, and reproductive health. Sex Transm Dis
2007;34:864–9. PMID:17621244 https://doi.org/10.1097/
OLQ.0b013e318074e565
311. Evans AL, Scally AJ, Wellard SJ, Wilson JD. Prevalence of bacterial
vaginosis in lesbians and heterosexual women in a community setting.
Sex Transm Infect 2007;83:4705. PMID:17611235 https://doi.
org/10.1136/sti.2006.022277
312. Olson KM, Boohaker LJ, Schwebke JR, Aslibekyan S, Muzny CA.
Comparisons of vaginal flora patterns among sexual behaviour groups
of women: implications for the pathogenesis of bacterial vaginosis. Sex
Health 2018;15:617. PMID:29212588 https://doi.org/10.1071/
SH17087
313. Muzny CA, Lensing SY, Aaron KJ, Schwebke JR. Incubation period
and risk factors support sexual transmission of bacterial vaginosis in
women who have sex with women. Sex Transm Infect 2019;95:511–5.
PMID:30872415 https://doi.org/10.1136/sextrans-2018-053824
314. Bradshaw CS, Walker SM, Vodstrcil LA, et al. The influence of behaviors
and relationships on the vaginal microbiota of women and their female
partners: the WOW Health Study. J Infect Dis 2014;209:1562–72.
PMID:24285846 https://doi.org/10.1093/infdis/jit664
315. Marrazzo JM, Antonio M, Agnew K, Hillier SL. Distribution of genital
Lactobacillus strains shared by female sex partners. J Infect Dis
2009;199:680–3. PMID:19199538 https://doi.org/10.1086/596632
316. Marrazzo JM, Fiedler TL, Srinivasan S, et al. Extravaginal reservoirs of
vaginal bacteria as risk factors for incident bacterial vaginosis. J Infect
Dis 2012;205:1580–8. PMID:22448002 https://doi.org/10.1093/
infdis/jis242
317. Mitchell C, Manhart LE, Thomas K, Fiedler T, Fredricks DN, Marrazzo
J. Behavioral predictors of colonization with Lactobacillus crispatus or
Lactobacillus jensenii after treatment for bacterial vaginosis: a cohort
study. Infect Dis Obstet Gynecol 2012;2012:706540. Epub May 30,
2012. PMID:22693410 https://doi.org/10.1155/2012/706540
318. Mitchell C, Manhart LE, Thomas KK, Agnew K, Marrazzo JM. Effect
of sexual activity on vaginal colonization with hydrogen peroxide-
producing Lactobacilli and Gardnerella vaginalis. Sex Transm Dis
2011;38:113744. PMID:22082725 https://doi.org/10.1097/
OLQ.0b013e31822e6121
319. Fethers K, Twin J, Fairley CK, et al. Bacterial vaginosis (BV) candidate
bacteria: associations with BV and behavioural practices in sexually-
experienced and inexperienced women. PLoS One 2012;7:e30633.
PMID:22363457 https://doi.org/10.1371/journal.pone.0030633
320. Bradshaw CS, Vodstrcil LA, Hocking JS, et al. Recurrence of bacterial
vaginosis is significantly associated with posttreatment sexual activities
and hormonal contraceptive use. Clin Infect Dis 2013;56:77786.
PMID:23243173 https://doi.org/10.1093/cid/cis1030
321. Marrazzo JM, Thomas KK, Fiedler TL, Ringwood K, Fredricks DN.
Risks for acquisition of bacterial vaginosis among women who report
sex with women: a cohort study. PLoS One 2010;5:e11139.
PMID:20559445 https://doi.org/10.1371/journal.pone.0011139
322. Vodstrcil LA, Walker SM, Hocking JS, et al. Incident bacterial vaginosis
(BV) in women who have sex with women is associated with behaviors
that suggest sexual transmission of BV. Clin Infect Dis 2015;60:1042–53.
PMID:25516188 https://doi.org/10.1093/cid/ciu1130
323. Muzny CA, Blanchard E, Taylor CM, et al. Identification of key bacteria
involved in the induction of incident bacterial vaginosis: a prospective
study. J Infect Dis 2018;218:966–78. PMID:29718358 https://doi.
org/10.1093/infdis/jiy243
324. Marrazzo JM, Thomas KK, Ringwood K. A behavioural intervention
to reduce persistence of bacterial vaginosis among women who report
sex with women: results of a randomised trial. Sex Transm Infect
2011;87:399405. PMID:21653935 https://doi.org/10.1136/
sti.2011.049213
325. Bradshaw CS, Walker J, Fairley CK, et al. Prevalent and incident
bacterial vaginosis are associated with sexual and contraceptive
behaviours in young Australian women. PLoS One 2013;8:e57688.
PMID:23472099 https://doi.org/10.1371/journal.pone.0057688
326. Poteat T, Reisner SL, Radix A. HIV epidemics among transgender
women. Curr Opin HIV AIDS 2014;9:16873. PMID:24322537
https://doi.org/10.1097/COH.0000000000000030
327. White Hughto JM, Reisner SL, Pachankis JE. Transgender stigma and
health: a critical review of stigma determinants, mechanisms, and
interventions. Soc Sci Med 2015;147:22231. PMID:26599625
https://doi.org/10.1016/j.socscimed.2015.11.010
328. Radix AE, Lelutiu-Weinberger C, Gamarel KE. Satisfaction and
healthcare utilization of transgender and gender non-conforming
individuals in NYC: a community-based participatory study. LGBT
Health 2014;1:302–8. PMID:26789858 https://doi.org/10.1089/
lgbt.2013.0042
329. Rapues J, Wilson EC, Packer T, Colfax GN, Raymond HF. Correlates
of HIV infection among transfemales, San Francisco, 2010: results
from a respondent-driven sampling study. Am J Public Health
2013;103:148592. PMID:23763398 https://doi.org/10.2105/
AJPH.2012.301109
330. Sevelius JM, Patouhas E, Keatley JG, Johnson MO. Barriers and
facilitators to engagement and retention in care among transgender
women living with human immunodeficiency virus. Ann Behav Med
2014;47:516. PMID:24317955 https://doi.org/10.1007/
s12160-013-9565-8
331. Sevelius JM. Gender affirmation: a framework for conceptualizing risk
behavior among transgender women of color. Sex Roles 2013;68:675–89.
PMID:23729971 https://doi.org/10.1007/s11199-012-0216-5
332. Reisner SL, White Hughto JM, Pardee D, Sevelius J. Syndemics and
gender affirmation: HIV sexual risk in female-to-male trans masculine
adults reporting sexual contact with cisgender males. Int J STD AIDS
2016; 27: 955 66. PMID:26384946 https://doi.
org/10.1177/0956462415602418
333. Coleman E, Bockting W, Botzer M, et al. Standards of care for the
health of transsexual, transgender, and gender-nonconforming people,
version 7. Int J Transgenderism 2012;13:165–232. https://doi.org/10
.1080/15532739.2011.700873
334. Winter S, Diamond M, Green J, et al. Transgender people: health at
the margins of society. Lancet 2016;388:390–400. PMID:27323925
https://doi.org/10.1016/S0140-6736(16)00683-8
335. Cahill S, Singal R, Grasso C, et al. Do ask, do tell: high levels of
acceptability by patients of routine collection of sexual orientation and
gender identity data in four diverse American community health
centers. PLoS One 2014;9:e107104. PMID:25198577 https://doi.
org/10.1371/journal.pone.0107104
336. Cahill SR, Baker K, Deutsch MB, Keatley J, Makadon HJ. Inclusion
of sexual orientation and gender identity in Stage 3 Meaningful Use
Guidelines: a huge step forward for LGBT health. LGBT Health
2016;3:1002. PMID:26698386 https://doi.org/10.1089/
lgbt.2015.0136
337. Tordoff DM, Morgan J, Dombrowski JC, Golden MR, Barbee LA.
Increased ascertainment of transgender and non-binary patients using
a 2-step versus 1-step gender identity intake question in an STD clinic
setting. Sex Transm Dis 2019;46:2549. PMID:30516726 https://doi.
org/10.1097/OLQ.0000000000000952
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 147
US Department of Health and Human Services/Centers for Disease Control and Prevention
338. Grant JM, Mottet LA, Tanis J. National transgender discrimination
survey report on health and health care. Washington, DC: National
Center for Transgender Equality and the National Gay and Lesbian
Task Force; 2010. https://cancer-network.org/wp-content/
uploads/2017/02/National_Transgender_Discrimination_Survey_
Report_on_health_and_health_care.pdf
339. Jaffee KD, Shires DA, Stroumsa D. Discrimination and delayed health
care among transgender women and men: implications for improving
medical education and health care delivery. Med Care 2016;54:10106.
PMID:27314263 https://doi.org/10.1097/MLR.0000000000000583
340. Glick JL, Theall KP, Andrinopoulos KM, Kendall C. The role of
discrimination in care postponement among trans-feminine individuals
in the U.S. National Transgender Discrimination Survey. LGBT Health
2018;5:1719. PMID:29589995 https://doi.org/10.1089/
lgbt.2017.0093
341. Callander D, Cook T, Cornelisse V, et al. Trans and gender diverse
peoples experiences of sexual health care are associated with sexual
health screening uptake. Sex Transm Infect 2019;95(Suppl 1):A64.
https://sti.bmj.com/content/sextrans/95/Suppl_1.toc.pdf
342. Casey LS, Reisner SL, Findling MG, et al. Discrimination in the United
States: experiences of lesbian, gay, bisexual, transgender, and queer
Americans. Health Serv Res 2019;54(Suppl 2):1454–66.
PMID:31659745 https://doi.org/10.1111/1475-6773.13229
343. Potter J, Peitzmeier SM, Bernstein I, et al. Cervical cancer screening
for patients on the female-to-male spectrum: a narrative review and
guide for clinicians. J Gen Intern Med 2015;30:185764.
PMID:26160483 https://doi.org/10.1007/s11606-015-3462-8
344. Becasen JS, Denard CL, Mullins MM, Higa DH, Sipe TA. Estimating
the prevalence of HIV and sexual behaviors among the US transgender
population: a systematic review and meta-analysis, 2006–2017. Am J
Public Health 2019;109:e18. PMID:30496000 https://doi.
org/10.2105/AJPH.2018.304727
345. Baral SD, Poteat T, Strömdahl S, Wirtz AL, Guadamuz TE, Beyrer C.
Worldwide burden of HIV in transgender women: a systematic review
and meta-analysis. Lancet Infect Dis 2013;13:21422. PMID:23260128
https://doi.org/10.1016/S1473-3099(12)70315-8
346. Allan-Blitz LT, Konda KA, Calvo GM, et al. High incidence of extra-
genital gonorrheal and chlamydial infections among high-risk men
who have sex with men and transgender women in Peru. Int J STD
AIDS 2018;29:56876. PMID:29183269 https://doi.
org/10.1177/0956462417744098
347. Hiransuthikul A, Janamnuaysook R, Sungsing T, et al. High burden
of chlamydia and gonorrhoea in pharyngeal, rectal and urethral sites
among Thai transgender women: implications for anatomical site
selection for the screening of STI. Sex Transm Infect 2019;95:534–9.
PMID:30982000 https://doi.org/10.1136/sextrans-2018-053835
348. Kojima N, Park H, Konda KA, et al. The PICASSO Cohort: baseline
characteristics of a cohort of men who have sex with men and male-
to-female transgender women at high risk for syphilis infection in Lima,
Peru. BMC Infect Dis 2017;17:255. PMID:28399798 https://doi.
org/10.1186/s12879-017-2332-x
349. Pitasi MA, Kerani RP, Kohn R, et al. Chlamydia, gonorrhea, and human
immunodeficiency virus infection among transgender women and
transgender men attending clinics that provide sexually transmitted
disease services in six US cities: results from the Sexually Transmitted
Disease Surveillance Network. Sex Transm Dis 2019;46:1127.
PMID:30278030 https://doi.org/10.1097/OLQ.0000000000000917
350. James S, Herman JL, Rankin S, Keisling M, Mottet L, Anafi M. The
report of the 2015 US transgender survey. Washington, DC: National
Center for Transgender Equality; 2016. https://transequality.org/sites/
default/files/docs/usts/USTS-Full-Report-Dec17.pdf
351. Hadj-Moussa M, Ohl DA, Kuzon WM Jr. Feminizing genital gender-
confirmation surgery. Sex Med Rev 2018;6:457468.e2.
PMID:29454634 https://doi.org/10.1016/j.sxmr.2017.11.005
352. Salgado CJ, Nugent A, Kuhn J, Janette M, Bahna H. Primary sigmoid
vaginoplasty in transwomen: technique and outcomes. BioMed Res
Int 2018;2018:4907208. PMID:29862275 https://doi.
org/10.1155/2018/4907208
353. Radix AE, Harris AB, Belkind U, Ting J, Goldstein ZG. Chlamydia
trachomatis infection of the neovagina in transgender women. Open
Forum Infect Dis 2019;6:ofz470. PMID:32395566 https://doi.
org/10.1093/ofid/ofz470
354. Elfering L, van der Sluis WB, Mermans JF, Buncamper ME. Herpes
neolabialis: herpes simplex virus type 1 infection of the neolabia in a
transgender woman. Int J STD AIDS 2017;28:841–3. PMID:28632111
https://doi.org/10.1177/0956462416685658
355. van der Sluis WB, Buncamper ME, Bouman MB, et al. Prevalence of
neovaginal high-risk human papillomavirus among transgender women
in the Netherlands. Sex Transm Dis 2016;43:503–5. PMID:27414682
https://doi.org/10.1097/OLQ.0000000000000476
356. Yang C, Liu S, Xu K, Xiang Q, Yang S, Zhang X. Condylomata gigantea
in a male transsexual. Int J STD AIDS 2009;20:211–2. PMID:19255276
https://doi.org/10.1258/ijsa.2008.008213
357. Matsuki S, Kusatake K, Hein KZ, Anraku K, Morita E. Condylomata
acuminata in the neovagina after male-to-female reassignment treated
with CO
2
laser and imiquimod. Int J STD AIDS 2015;26:509–11.
PMID:24970474 https://doi.org/10.1177/0956462414542476
358. Bodsworth NJ, Price R, Davies SC. Gonococcal infection of the
neovagina in a male-to-female transsexual. Sex Transm Dis
1994; 21: 211 2 . PMID:7974071 https://doi.
org/10.1097/00007435-199407000-00005
359. Haustein UF. Pruritus of the artificial vagina of a transsexual patient
caused by gonococcal infection [German]. Hautarzt 1995;46:858–9.
PMID:8567271 https://doi.org/10.1007/s001050050354
360. Yamada K, Shida D, Kato T, Yoshida H, Yoshinaga S, Kanemitsu Y.
Adenocarcinoma arising in sigmoid colon neovagina 53years after
construction. World J Surg Oncol 2018;16:88. PMID:29703260
https://doi.org/10.1186/s12957-018-1372-z
361. Hiroi H, Yasugi T, Matsumoto K, et al. Mucinous adenocarcinoma
arising in a neovagina using the sigmoid colon thirty years after
operation: a case report. J Surg Oncol 2001;77:614. PMID:11344485
https://doi.org/10.1002/jso.1067
362. Heller DS. Lesions of the neovagina—a review. J Low Genit Tract Dis
2015;19:26770. PMID:26111041 https://doi.org/10.1097/
LGT.0000000000000110
363. Scheim AI, Bauer GR, Travers R. HIV-related sexual risk among
transgender men who are gay, bisexual, or have sex with men. J Acquir
Immune Defic Syndr 2017;74:e89–96. PMID:27798432 https://doi.
org/10.1097/QAI.0000000000001222
364. Sevelius J. “There’s no pamphlet for the kind of sex I have”: HIV-related
risk factors and protective behaviors among transgender men who have
sex with nontransgender men. J Assoc Nurses AIDS Care
2009;20:398–410. PMID:19732698 https://doi.org/10.1016/j.
jana.2009.06.001
365. Pitasi MA, Oraka E, Clark H, Tow n M, DiNenno EA. HIV testing
among transgender women and men—27 states and Guam, 2014–
2015. MMWR Morb Mortal Wkly Rep 2017;66:8837.
PMID:28837547 https://doi.org/10.15585/mmwr.mm6633a3
366. Reisner SL, Perkovich B, Mimiaga MJ. A mixed methods study of the
sexual health needs of New England transmen who have sex with
nontransgender men. AIDS Patient Care STDS 2010;24:50113.
PMID:20666586 https://doi.org/10.1089/apc.2010.0059
367. Hembree WC, Cohen-Kettenis PT, Gooren L, et al. Endocrine
treatment of gender-dysphoric/gender-incongruent persons: an
Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab
2017;102:3869903. PMID:28945902 https://doi.org/10.1210/
jc.2017-01658
Recommendations and Reports
148 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
368. Deutsch M, ed. Guidelines for the primary and gender-affirming care
of transgender and gender nonbinary people. San Francisco, CA:
University of California San Francisco, Department of Family and
Community Medicine, Center of Excellence for Transgender Care;
2016. https://transcare.ucsf.edu/sites/transcare.ucsf.edu/files/
Transgender-PGACG-6-17-16.pdf
369. Peitzmeier SM, Reisner SL, Harigopal P, Potter J. Female-to-male
patients have high prevalence of unsatisfactory Paps compared to non-
transgender females: implications for cervical cancer screening. J Gen
Intern Med 2014;29:77884. PMID:24424775 https://doi.
org/10.1007/s11606-013-2753-1
370. Peitzmeier SM, Khullar K, Reisner SL, Potter J. Pap test use is lower
among female-to-male patients than non-transgender women. Am J
Prev Med 2014;47:80812. PMID:25455121 https://doi.
org/10.1016/j.amepre.2014.07.031
371. Reisner SL, Deutsch MB, Peitzmeier SM, et al. Test performance and
acceptability of self- versus provider-collected swabs for high-risk HPV
DNA testing in female-to-male trans masculine patients. PLoS One
2018;13:e0190172. PMID:29538411 https://doi.org/10.1371/journal.
pone.0190172
372. CDC. Surveillance for viral hepatitis—United States, 2017. Atlanta,
GA: US Department of Health and Human Services, CDC; 2019.
https://www.cdc.gov/hepatitis/statistics/2017surveillance/
pdfs/2017HepSurveillanceRpt.pdf
373. Kouyoumdjian FG, Leto D, John S, Henein H, Bondy S. A systematic
review and meta-analysis of the prevalence of chlamydia, gonorrhoea
and syphilis in incarcerated persons. Int J STD AIDS 2012;23:24854.
PMID:22581947 https://doi.org/10.1258/ijsa.2011.011194
374. CDC. Evaluation of large jail STD screening programs, 2008–2009.
Atlanta, GA: US Department of Health and Human Services, CDC;
2011. https://www.cdc.gov/std/publications/JailScreening2011.pdf
375. Pathela P, Hennessy RR, Blank S, Parvez F, Franklin W, Schillinger JA.
The contribution of a urine-based jail screening program to citywide
male chlamydia and gonorrhea case rates in New York City. Sex Transm
Dis 2009;36(Suppl):S5861. PMID:17989586 https://doi.
org/10.1097/OLQ.0b013e31815615bb
376. Joesoef MR, Weinstock HS, Kent CK, et al.; Corrections STD
Prevalence Monitoring Group. Sex and age correlates of chlamydia
prevalence in adolescents and adults entering correctional facilities,
2005: implications for screening policy. Sex Transm Dis
2009;36(Suppl):S67–71. PMID:19125147 https://doi.org/10.1097/
OLQ.0b013e31815d6de8
377. Blank S, McDonnell DD, Rubin SR, et al. New approaches to syphilis
control. Finding opportunities for syphilis treatment and congenital
syphilis prevention in a womens correctional setting. Sex Transm Dis
1997; 24: 218 26. PMID:9101633 https://doi.
org/10.1097/00007435-199704000-00006
378. Owusu-Edusei K Jr, Gift TL, Chesson HW, Kent CK. Investigating
the potential public health benefit of jail-based screening and treatment
programs for chlamydia. Am J Epidemiol 2013;177:46373.
PMID:23403986 https://doi.org/10.1093/aje/kws240
379. Spaulding AC, Miller J, Trigg BG, et al. Screening for sexually
transmitted diseases in short-term correctional institutions: summary
of evidence reviewed for the 2010 Centers for Disease Control and
Prevention Sexually Transmitted Diseases Treatment Guidelines. Sex
Transm Dis 2013;40:67984. PMID:23945422 https://doi.
org/10.1097/01.olq.0000431353.88464.ab
380. CDC. Male chlamydia screening consultation, March 28–29, 2006,
meeting report. Atlanta, GA: US Department of Health and Human
Services, CDC; 2007. https://www.cdc.gov/std/chlamydia/
chlamydiascreening-males.pdf
381. Cole J, Hotton A, Zawitz C, Kessler H. Opt-out screening for
Chlamydia trachomatis and Neisseria gonorrhoeae in female detainees at
Cook County jail in Chicago, IL. Sex Transm Dis 2014;41:161–5.
PMID:24521720 https://doi.org/10.1097/OLQ.0000000000000106
382. Shaikh RA, Simonsen KA, O’Keefe A, et al. Comparison of opt-in
versus opt-out testing for sexually transmitted infections among inmates
in a county jail. J Correct Health Care 2015;21:40816.
PMID:26285597 https://doi.org/10.1177/1078345815600447
383. Spaulding AC, Kim MJ, Corpening KT, Carpenter T, Watlington P,
Bowden CJ. Establishing an HIV screening program led by staff nurses
in a county jail. J Public Health Manag Pract 2015;21:53845.
PMID:25427254 https://doi.org/10.1097/PHH.0000000000000183
384. Rosen DL, Wohl DA, Golin CE, et al. Comparing HIV case detection
in prison during opt-in vs. opt-out testing policies. J Acquir Immune
Defic Syndr 2016;71:e858. PMID:26536318 https://doi.
org/10.1097/QAI.0000000000000889
385. Gratrix J, Smyczek P, Bertholet L, et al. A cross-sectional evaluation of
opt-in testing for sexually transmitted and blood-borne infections in
three Canadian provincial correctional facilities: a missed opportunity
for public health? Int J Prison Health 2019;15:27381. PMID:31329036
https://doi.org/10.1108/IJPH-07-2018-0043
386. Sutcliffe S, Newman SB, Hardick A, Gaydos CA. Prevalence and
correlates of Trichomonas vaginalis infection among female US federal
prison inmates. Sex Transm Dis 2010;37:585–90. PMID:20803782
https://doi.org/10.1097/OLQ.0b013e3181de4113
387. Freeman AH, Katz KA, Pandori MW, et al. Prevalence and correlates
of Trichomonas vaginalis among incarcerated persons assessed using a
highly sensitive molecular assay. Sex Transm Dis 2010;37:165–8.
PMID:20023598 https://doi.org/10.1097/OLQ.0b013e3181bcd3fc
388. Sosman J, Macgowan R, Margolis A, et al.; Project START Biologics
Study Group. Sexually transmitted infections and hepatitis in men with
a history of incarceration. Sex Transm Dis 2011;38:6349.
PMID:21844713 https://doi.org/10.1097/OLQ.0b013e31820bc86c
389. Javanbakht M, Stirland A, Stahlman S, et al. Prevalence and factors
associated with Trichomonas vaginalis infection among high-risk women
in Los Angeles. Sex Transm Dis 2013;40:804–7. PMID:24275733
https://doi.org/10.1097/OLQ.0000000000000026
390. Nijhawan AE, Chapin KC, Salloway R, et al. Prevalence and predictors
of Trichomonas infection in newly incarcerated women. Sex Transm
Dis 2012;39:9738. PMID:23191953 https://doi.org/10.1097/
OLQ.0b013e31826e8847
391. Nijhawan AE, DeLong AK, Celentano DD, et al. The association
between Trichomonas infection and incarceration in HIV-seropositive
and at-risk HIV-seronegative women. Sex Transm Dis
2011;38:1094–100. PMID:22082718 https://doi.org/10.1097/
OLQ.0b013e31822ea147
392. Willers DM, Peipert JF, Allsworth JE, Stein MD, Rose JS, Clarke JG.
Prevalence and predictors of sexually transmitted infection among
newly incarcerated females. Sex Transm Dis 2008;35:6872.
PMID:18090178 https://doi.org/10.1097/OLQ.0b013e318154bdb2
393. Nijhawan AE, Salloway R, Nunn AS, Poshkus M, Clarke JG. Preventive
healthcare for underserved women: results of a prison survey. J Womens
Health (Larchmt) 2010;19:1722. PMID:20088654 https://doi.
org/10.1089/jwh.2009.1469
394. Binswanger IA, White MC, Pérez-Stable EJ, Goldenson J, Tulsky JP.
Cancer screening among jail inmates: frequency, knowledge, and
willingness. Am J Public Health 2005;95:1781–7. PMID:16186455
https://doi.org/10.2105/AJPH.2004.052498
395. Brinkley-Rubinstein L, Peterson M, Arnold T, et al. Knowledge, interest,
and anticipated barriers of pre-exposure prophylaxis uptake and
adherence among gay, bisexual, and men who have sex with men who
are incarcerated. PLoS One 2018;13:e0205593. PMID:30532275
https://doi.org/10.1371/journal.pone.0205593
396. Brinkley-Rubinstein L, Dauria E, Tolou-Shams M, et al. The path to
implementation of HIV pre-exposure prophylaxis for people involved
in criminal justice systems. Curr HIV/AIDS Rep 2018;15:935.
PMID:29516265 https://doi.org/10.1007/s11904-018-0389-9
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 149
US Department of Health and Human Services/Centers for Disease Control and Prevention
397. Morrow KM; Project START Study Group. HIV, STD, and hepatitis
risk behaviors of young men before and after incarceration. AIDS Care
2009; 21: 235 43. PMID:19229694 https://doi.
org/10.1080/09540120802017586
398. Bryan AD, Magnan RE, Gillman AS, et al. Effect of including alcohol
and cannabis content in a sexual risk-reduction intervention on the
incidence of sexually transmitted infections in adolescents: a cluster
randomized clinical trial. JAMA Pediatr 2018;172:e175621.
PMID:29435591 https://doi.org/10.1001/jamapediatrics.2017.5621
399. DiClemente RJ, Davis TL, Swartzendruber A, et al. Efficacy of an
HIV/STI sexual risk-reduction intervention for African American
adolescent girls in juvenile detention centers: a randomized controlled
trial. Women Health 2014;54:726–49. PMID:25190056 https://doi.
org/10.1080/03630242.2014.932893
400. Fogel CI, Crandell JL, Neevel AM, et al. Efficacy of an adapted HIV
and sexually transmitted infection prevention intervention for
incarcerated women: a randomized controlled trial. Am J Public Health
2015;105:8029. PMID:25211714 https://doi.org/10.2105/
AJPH.2014.302105
401. Son J, Miller WM, Tossone K, Butcher F, Kuo K. The effect of
interprofessional student-led reproductive health education on youths
in juvenile detention. J Pediatr Adolesc Gynecol 2017;30:370–5.
PMID:27871918 https://doi.org/10.1016/j.jpag.2016.11.002
402. Costumbrado J, Stirland A, Cox G, et al. Implementation of a hepatitis
A/B vaccination program using an accelerated schedule among high-risk
inmates, Los Angeles County Jail, 2007–2010. Vaccine
2012;30:6878–82. PMID:22989688 https://doi.org/10.1016/j.
vaccine.2012.09.006
403. Allison M, Musser B, Satterwhite C, Ault K, Kelly P, Ramaswamy M.
Human papillomavirus vaccine knowledge and intention among adult
inmates in Kansas, 2016–2017. Am J Public Health 2018;108:10002.
PMID:29927651 https://doi.org/10.2105/AJPH.2018.304499
404. Lucas KD, Miller JL, Eckert V, Horne RL, Samuel MC, Mohle-Boetani
JC. Risk, feasibility, and cost evaluation of a prisoner condom access
pilot program in one California state prison. J Correct Health Care
2014; 20: 184 94. PMID:24934836 https://doi.
org/10.1177/1078345814530869
405. Scott N, McBryde E, Kirwan A, Stoové M. Modelling the impact of
condom distribution on the incidence and prevalence of sexually
transmitted infections in an adult male prison system. PLoS One
2015;10:e0144869. PMID:26658518 https://doi.org/10.1371/journal.
pone.0144869
406. Schacker T, Collier AC, Hughes J, Shea T, Corey L. Clinical and
epidemiologic features of primary HIV infection. Ann Intern Med
1996; 125: 257 64. PMID:8678387 https://doi.
org/10.7326/0003-4819-125-4-199608150-00001
407. Henn A, Flateau C, Gallien S. Primary HIV infection: clinical
presentation, testing, and treatment. Curr Infect Dis Rep 2017;19:37.
PMID:28884279 https://doi.org/10.1007/s11908-017-0588-3
408. Robb ML, Eller LA, Kibuuka H, et al.; RV 217 Study Team. Prospective
study of acute HIV-1 infection in adults in East Africa and Thailand.
N Engl J Med 2016;374:212030. PMID:27192360 https://doi.
org/10.1056/NEJMoa1508952
409. Hoenigl M, Green N, Camacho M, et al. Signs or symptoms of acute
HIV infection in a cohort undergoing community-based screening.
Emerg Infect Dis 2016;22:5324. PMID:26890854 https://doi.
org/10.3201/eid2203.151607
410. Legarth RA, Ahlström MG, Kronborg G, et al. Long-term mortality
in HIV-infected individuals 50 years or older: a nationwide, population-
based cohort study. J Acquir Immune Defic Syndr 2016;71:213–8.
PMID:26334734 https://doi.org/10.1097/QAI.0000000000000825
411. Marcus JL, Chao CR, Leyden WA, et al. Narrowing the gap in life
expectancy between HIV-infected and HIV-uninfected individuals
with access to care. J Acquir Immune Defic Syndr 2016;73:39–46.
PMID:27028501 https://doi.org/10.1097/QAI.0000000000001014
412. Cohen MS, Chen YQ, McCauley M, et al.; HPTN 052 Study Team.
Antiretroviral therapy for the prevention of HIV-1 transmission. N Engl
J Med 2016;375:830–9. PMID:27424812 https://doi.org/10.1056/
NEJMoa1600693
413. Saag MS, Benson CA, Gandhi RT, et al. Antiretroviral drugs for
treatment and prevention of HIV infection in adults: 2018
recommendations of the International Antiviral Society-USA Panel.
JAMA 2018;320:37996. PMID:30043070 https://doi.org/10.1001/
jama.2018.8431
414. Seth P, Wang G, Sizemore E, Hogben M. HIV testing and HIV service
delivery to populations at high risk attending sexually transmitted
disease clinics in the United States, 2011–2013. Am J Public Health
2015;105:237481. PMID:26378854 https://doi.org/10.2105/
AJPH.2015.302778
415. Benton S, Smith J, Wang F, Heitgerd J, Belcher L, Patel H. HIV testing,
diagnosis, and linkage to care among persons tested in select CDC-
funded health care and non-health care settings, 2012–2017. Presented
at the National HIV Prevention Conference, Atlanta, GA: March
18–21, 2019.
416. Pathela P, Braunstein SL, Schillinger JA, Shepard C, Sweeney M, Blank
S. Men who have sex with men have a 140-fold higher risk for newly
diagnosed HIV and syphilis compared with heterosexual men in New
York City. J Acquir Immune Defic Syndr 2011;58:40816.
PMID:21857351 https://doi.org/10.1097/QAI.0b013e318230e1ca
417. Chou R, Selph S, Dana T, et al. Screening for HIV: systematic review
to update the 2005 U.S. Preventive Services Task Force recommendation.
Ann Intern Med 2012;157:70618. PMID:23165662 https://doi.
org/10.7326/0003-4819-157-10-201211200-00007
418. Branson BM, Handsfield HH, Lampe MA, et al.; CDC. Revised
recommendations for HIV testing of adults, adolescents, and pregnant
women in health-care settings. MMWR Recomm Rep 2006;55(No.
RR-14). PMID:16988643
419. DiNenno EA, Prejean J, Irwin K, et al. Recommendations for HIV
screening of gay, bisexual, and other men who have sex with men—
United States, 2017. MMWR Morb Mortal Wkly Rep 2017;66:830–2.
PMID:28796758 https://doi.org/10.15585/mmwr.mm6631a3
420. CDC. HIV-2 infection surveillance—United States, 1987–2009.
MMWR Morb Mortal Wkly Rep 2011;60:985–8. PMID:21796096
421. Wawer MJ, Gray RH, Sewankambo NK, et al. Rates of HIV-1
transmission per coital act, by stage of HIV-1 infection, in Rakai,
Uganda. J Infect Dis 2005;191:1403–9. PMID:15809897 https://doi.
org/10.1086/429411
422. Pilcher CD, Eron JJ Jr, Vemazza PL, et al. Sexual transmission during
the incubation period of primary HIV infection. JAMA
2001;286:1713–4. PMID:11594895 https://doi.org/10.1001/
jama.286.14.1713
423. Calabrese SK, Mayer KH. Providers should discuss U=U with all
patients living with HIV. Lancet HIV 2019;6:e211–3. PMID:30772420
https://doi.org/10.1016/S2352-3018(19)30030-X
424. Gilbert P, Ciccarone D, Gansky SA, et al. Interactive “Video Doctor
counseling reduces drug and sexual risk behaviors among HIV-positive
patients in diverse outpatient settings. PLoS One 2008;3:e1988.
PMID:18431475 https://doi.org/10.1371/journal.pone.0001988
425. Aberg JA, Gallant JE, Ghanem KG, Emmanuel P, Zingman BS, Horberg
MA; Infectious Diseases Society of America. Primary care guidelines
for the management of persons infected with HIV: 2013 update by
the HIV medicine association of the Infectious Diseases Society of
America. Clin Infect Dis 2014;58:e1–34. PMID:24235263 https://
doi.org/10.1093/cid/cit665
426. DiCarlo RP, Martin DH. The clinical diagnosis of genital ulcer disease
in men. Clin Infect Dis 1997;25:292–8. PMID:9332527 https://doi.
org/10.1086/514548
427. Lockett AE, Dance DA, Mabey DC, Drasar BS. Serum-free media for
isolation of Haemophilus ducreyi. Lancet 1991;338:326. PMID:1677152
https://doi.org/10.1016/0140-6736(91)90473-3
Recommendations and Reports
150 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
428. Lewis DA, Mitjà O. Haemophilus ducreyi: from sexually transmitted
infection to skin ulcer pathogen. Curr Opin Infect Dis 2016;29:52–7.
PMID:26658654 https://doi.org/10.1097/QCO.0000000000000226
429. Romero L, Huerfano C, Grillo-Ardila CF. Macrolides for treatment of
Haemophilus ducreyi infection in sexually active adults. Cochrane
Database Syst Rev 2017;12:CD012492. PMID:29226307 https://doi.
org/10.1002/14651858.CD012492.pub2
430. Jessamine PG, Plummer FA, Ndinya Achola JO, et al. Human
immunodeficiency virus, genital ulcers and the male foreskin: synergism
in HIV-1 transmission. Scand J Infect Dis Suppl 1990;69:181–6.
PMID:2263893
431. Briggs GC. Drugs in pregnancy and lactation: a reference guide to fetal
and neonatal risk. 11th ed. Philadelphia, PA: Wolters Kluwer; 2017.
432. Lewis DA. Epidemiology, clinical features, diagnosis and treatment of
Haemophilus ducreyi—a disappearing pathogen? Expert Rev Anti Infect
Ther 2014;12:687–96. PMID:24597521 https://doi.org/10.1586/14
787210.2014.892414
433. Mitjà O, Lukehart SA, Pokowas G, et al. Haemophilus ducreyi as a
cause of skin ulcers in children from a yaws-endemic area of Papua
New Guinea: a prospective cohort study. Lancet Glob Health
2014;2:e23541. PMID:25103064 https://doi.org/10.1016/
S2214-109X(14)70019-1
434. Marks M, Chi KH, Vahi V, et al. Haemophilus ducreyi associated with
skin ulcers among children, Solomon Islands. Emerg Infect Dis
2014;20:17057. PMID:25271477 https://doi.org/10.3201/
eid2010.140573
435. Ghinai R, El-Duah P, Chi KH, et al. A cross-sectional study of ‘yaws
in districts of Ghana which have previously undertaken azithromycin
mass drug administration for trachoma control. PLoS Negl Trop Dis
2015;9:e0003496. PMID:25632942 https://doi.org/10.1371/journal.
pntd.0003496
436. McQuillan G, Kruszon-Moran D, Flagg EW, Paulose-Ram R.
Prevalence of herpes simplex virus type 1 and type 2 in persons aged
14–49: United States, 2015–2016. NCHS Data Brief 2018;304:1–8.
PMID:29442994
437. Ryder N, Jin F, McNulty AM, Grulich AE, Donovan B. Increasing role
of herpes simplex virus type 1 in first-episode anogenital herpes in
heterosexual women and younger men who have sex with men,
1992–2006. Sex Transm Infect 2009;85:416–9. PMID:19273479
https://doi.org/10.1136/sti.2008.033902
438. Roberts CM, Pfister JR, Spear SJ. Increasing proportion of herpes
simplex virus type 1 as a cause of genital herpes infection in college
students. Sex Transm Dis 2003;30:797800. PMID:14520181 https://
doi.org/10.1097/01.OLQ.0000092387.58746.C7
439. Benedetti J, Corey L, Ashley R. Recurrence rates in genital
herpes after symptomatic first-episode infection. Ann
Intern Med 1994;121:84754. PMID:7978697 https://doi.
org/10.7326/0003-4819-121-11-199412010-00004
440. Engelberg R, Carrell D, Krantz E, Corey L, Wald A. Natural
history of genital herpes simplex virus type 1 infection. Sex
Transm Dis 2003;30:1747. PMID:12567178 https://doi.
org/10.1097/00007435-200302000-00015
441. Masese L, Baeten JM, Richardson BA, et al. Changes in the contribution
of genital tract infections to HIV acquisition among Kenyan high-risk
women from 1993 to 2012. AIDS 2015;29:107785. PMID:26125141
https://doi.org/10.1097/QAD.0000000000000646
442. Sam SS, Caliendo AM, Ingersoll J, Abdul-Ali D, Kraft CS. Performance
evaluation of the Aptima HSV-1 and 2 assay for the detection of HSV
in cutaneous and mucocutaneous lesion specimens. J Clin Virol
2018;99-100:14. PMID:29253834 https://doi.org/10.1016/j.
jcv.2017.12.006
443. Wald A, Huang ML, Carrell D, Selke S, Corey L. Polymerase chain
reaction for detection of herpes simplex virus (HSV) DNA on mucosal
surfaces: comparison with HSV isolation in cell culture. J Infect Dis
2003;188:134551. PMID:14593592 https://doi.org/10.1086/379043
444. Van Der Pol B, Warren T, Taylor SN, et al. Type-specific identification
of anogenital herpes simplex virus infections by use of a commercially
available nucleic acid amplification test. J Clin Microbiol 2012;50:3466
71. PMID:22875892 https://doi.org/10.1128/JCM.01685-12
445. Binnicker MJ, Espy MJ, Duresko B, Irish C, Mandrekar J. Automated
processing, extraction and detection of herpes simplex virus types 1
and 2: a comparative evaluation of three commercial platforms using
clinical specimens. J Clin Virol 2017;89:30–3. PMID:28226272
https://doi.org/10.1016/j.jcv.2017.02.006
446. Teo JW, Chiang D, Jureen R, Lin RT. Clinical evaluation of a helicase-
dependant amplification (HDA)-based commercial assay for the
simultaneous detection of HSV-1 and HSV-2. Diagn Microbiol Infect
Dis 2015;83:2612. PMID:26302856 https://doi.org/10.1016/j.
diagmicrobio.2015.07.018
447. Gitman MR, Ferguson D, Landry ML. Comparison of Simplexa HSV 1
& 2 PCR with culture, immunofluorescence, and laboratory-developed
TaqMan PCR for detection of herpes simplex virus in swab specimens.
J Clin Microbiol 2013;51:37659. PMID:24006008 https://doi.
org/10.1128/JCM.01413-13
448. Corey L, Holmes KK. Genital herpes simplex virus infections:
current concepts in diagnosis, therapy, and prevention. Ann
Intern Med 1983;98:97383. PMID:6344713 https://doi.
org/10.7326/0003-4819-98-6-958
449. Caviness AC, Oelze LL, Saz UE, Greer JM, Demmler-Harrison GJ.
Direct immunofluorescence assay compared to cell culture for the
diagnosis of mucocutaneous herpes simplex virus infections in children.
J Clin Virol 2010;49:5860. PMID:20620099 https://doi.
org/10.1016/j.jcv.2010.06.006
450. Song B, Dwyer DE, Mindel A. HSV type specific serology in sexual
health clinics: use, benefits, and who gets tested. Sex Transm Infect
2004;80:1137. PMID:15054171 https://doi.org/10.1136/
sti.2003.006783
451. Whittington WL, Celum CL, Cent A, Ashley RL. Use of a glycoprotein
G-based type-specific assay to detect antibodies to herpes simplex virus
type 2 among persons attending sexually transmitted disease clinics.
Sex Transm Dis 2001;28:99104. PMID:11234793 https://doi.
org/10.1097/00007435-200102000-00007
452. Zimet GD, Rosenthal SL, Fortenberry JD, et al. Factors predicting
the acceptance of herpes simplex virus type 2 antibody testing among
adolescents and young adults. Sex Transm Dis 2004;31:665–9.
PMID:15502674 https://doi.org/10.1097/01.olq.0000143089.77493.c2
453. Turner KR, Wong EH, Kent CK, Klausner JD. Serologic herpes
testing in the real world: validation of new type-specific serologic
herpes simplex virus tests in a public health laboratory. Sex
Transm Dis 2002;29:4225. PMID:12170133 https://doi.
org/10.1097/00007435-200207000-00011
454. Eing BR, Lippelt L, Lorentzen EU, et al. Evaluation of confirmatory
strategies for detection of type-specific antibodies against herpes simplex
virus type 2. J Clin Microbiol 2002;40:40713. PMID:11825950
https://doi.org/10.1128/JCM.40.2.407-413.2002
455. Golden MR, Ashley-Morrow R, Swenson P, Hogrefe WR, Handsfield
HH, Wald A. Herpes simplex virus type 2 (HSV-2) Western blot
confirmatory testing among men testing positive for HSV-2 using the
focus enzyme-linked immunosorbent assay in a sexually transmitted
disease clinic. Sex Transm Dis 2005;32:771–7. PMID:16314775
https://doi.org/10.1097/01.olq.0000175377.88358.f3
456. Morrow RA, Friedrich D, Meier A, Corey L. Use of “biokit HSV-2
Rapid Assay” to improve the positive predictive value of Focus
HerpeSelect HSV-2 ELISA. BMC Infect Dis 2005;5:84.
PMID:16225691 https://doi.org/10.1186/1471-2334-5-84
457. Ngo TD, Laeyendecker O, La H, Hogrefe W, Morrow RA, Quinn TC.
Use of commercial enzyme immunoassays to detect antibodies to the
herpes simplex virus type 2 glycoprotein G in a low-risk population in
Hanoi, Vietnam. Clin Vaccine Immunol 2008;15:3824.
PMID:18077617 https://doi.org/10.1128/CVI.00437-06
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 151
US Department of Health and Human Services/Centers for Disease Control and Prevention
458. Agyemang E, Le QA, Warren T, et al. Performance of commercial
enzyme-linked immunoassays for diagnosis of herpes simplex virus-1
and herpes simplex virus-2 infection in a clinical setting. Sex Transm
Dis 2017;44:7637. PMID:28876290 https://doi.org/10.1097/
OLQ.0000000000000689
459. Morrow R, Friedrich D. Performance of a novel test for IgM and
IgG antibodies in subjects with culture-documented genital herpes
simplex virus-1 or -2 infection. Clin Microbiol Infect 2006;12:463–9.
PMID:16643524 https://doi.org/10.1111/j.1469-0691.2006.01370.x
460. Ameli N, Bacchetti P, Morrow RA, et al. Herpes simplex virus infection
in women in the WIHS: epidemiology and effect of antiretroviral therapy
on clinical manifestations. AIDS 2006;20:1051–8. PMID:16603858
https://doi.org/10.1097/01.aids.0000222078.75867.77
461. Bradley H, Markowitz LE, Gibson T, McQuillan GM. Seroprevalence of
herpes simplex virus types 1 and 2—United States, 1999–2010. J Infect
Dis 2014;209:32533. PMID:24136792 https://doi.org/10.1093/
infdis/jit458
462. Bernstein DI, Bellamy AR, Hook EW 3rd, et al. Epidemiology, clinical
presentation, and antibody response to primary infection with herpes
simplex virus type 1 and type 2 in young women. Clin Infect Dis
2013;56:34451. PMID:23087395 https://doi.org/10.1093/cid/
cis891
463. Leone PA, Trottier S, Miller JM. Valacyclovir for episodic treatment of
genital herpes: a shorter 3-day treatment course compared with 5-day
treatment. Clin Infect Dis 2002;34:95862. PMID:11880962 https://
doi.org/10.1086/339326
464. Wald A, Carrell D, Remington M, Kexel E, Zeh J, Corey L. Two-
day regimen of acyclovir for treatment of recurrent genital herpes
simplex virus type 2 infection. Clin Infect Dis 2002;34:944–8.
PMID:11880960 https://doi.org/10.1086/339325
465. Aoki FY, Tyring S, Diaz-Mitoma F, Gross G, Gao J, Hamed K. Single-
day, patient-initiated famciclovir therapy for recurrent genital herpes:
a randomized, double-blind, placebo-controlled trial. Clin Infect Dis
2006;42:8–13. PMID:16323085 https://doi.org/10.1086/498521
466. Chosidow O, Drouault Y, Leconte-Veyriac F, et al. Famciclovir vs.
aciclovir in immunocompetent patients with recurrent genital herpes
infections: a parallel-groups, randomized, double-blind clinical trial.
Br J Dermatol 2001;144:81824. PMID:11298543 https://doi.
org/10.1046/j.1365-2133.2001.04139.x
467. Bodsworth NJ, Crooks RJ, Borelli S, et al.; International Valaciclovir
HSV Study Group. Valaciclovir versus aciclovir in patient initiated
treatment of recurrent genital herpes: a randomised, double blind
clinical trial. Genitourin Med 1997;73:110–6. PMID:9215092
468. Fife KH, Barbarash RA, Rudolph T, Degregorio B, Roth R; The
Valaciclovir International Herpes Simplex Virus Study Group. Valaciclovir
versus acyclovir in the treatment of first-episode genital herpes infection.
Results of an international, multicenter, double-blind, randomized
clinical trial. Sex Transm Dis 1997;24:481–6. PMID:9293612 https://
doi.org/10.1097/00007435-199709000-00007
469. Diaz-Mitoma F, Sibbald RG, Shafran SD, Boon R, Saltzman RL;
Collaborative Famciclovir Genital Herpes Research Group. Oral
famciclovir for the suppression of recurrent genital herpes: a randomized
controlled trial. JAMA 1998;280:88792. PMID:9739972 https://doi.
org/10.1001/jama.280.10.887
470. Mertz GJ, Loveless MO, Levin MJ, et al.; Collaborative Famciclovir
Genital Herpes Research Group. Oral famciclovir for suppression
of recurrent genital herpes simplex virus infection in women. A
multicenter, double-blind, placebo-controlled trial. Arch Intern
Med 1997;157:3439. PMID:9040303 https://doi.org/10.1001/
archinte.1997.00440240109016
471. Reitano M, Tyring S, Lang W, et al.; International Valaciclovir HSV
Study Group. Valaciclovir for the suppression of recurrent genital herpes
simplex virus infection: a large-scale dose range-finding study. J Infect
Dis 1998;178:60310. PMID:9728526 https://doi.
org/10.1086/515385
472. Romanowski B, Marina RB, Roberts JN; Valtrex HS230017 Study
Group. Patients’ preference of valacyclovir once-daily suppressive
therapy versus twice-daily episodic therapy for recurrent genital herpes: a
randomized study. Sex Transm Dis 2003;30:22631. PMID:12616141
https://doi.org/10.1097/00007435-200303000-00010
473. Corey L, Wald A, Patel R, et al.; Valacyclovir HSV Transmission Study
Group. Once-daily valacyclovir to reduce the risk of transmission of
genital herpes. N Engl J Med 2004;350:1120. PMID:14702423
https://doi.org/10.1056/NEJMoa035144
474. Tyring SK, Baker D, Snowden W. Valacyclovir for herpes simplex
virus infection: long-term safety and sustained efficacy after 20 years
experience with acyclovir. J Infect Dis 2002;186(Suppl 1):S40–6.
PMID:12353186 https://doi.org/10.1086/342966
475. Bartlett BL, Tyring SK, Fife K, et al. Famciclovir treatment options
for patients with frequent outbreaks of recurrent genital herpes: the
RELIEF trial. J Clin Virol 2008;43:190–5. PMID:18621575 https://
doi.org/10.1016/j.jcv.2008.06.004
476. Tronstein E, Johnston C, Huang ML, et al. Genital shedding of herpes
simplex virus among symptomatic and asymptomatic persons with
HSV-2 infection. JAMA 2011;305:1441–9. PMID:21486977 https://
doi.org/10.1001/jama.2011.420
477. Bender Ignacio RA, Perti T, Magaret AS, et al. Oral and vaginal tenofovir
for genital herpes simplex virus type 2 shedding in immunocompetent
women: a double-blind, randomized, cross-over trial. J Infect Dis
2015;212:194956. PMID:26044291 https://doi.org/10.1093/infdis/
jiv317
478. Wald A, Selke S, Warren T, et al. Comparative efficacy of famciclovir
and valacyclovir for suppression of recurrent genital herpes and viral
shedding. Sex Transm Dis 2006;33:52933. PMID:16540883 https://
doi.org/10.1097/01.olq.0000204723.15765.91
479. Johnston C, Magaret A, Stern M, et al. Natural history of genital and
oral herpes simplex virus-1 (HSV-1) shedding after first episode genital
HSV-1 infection. Sex Transm Infect 2019;95:A42.
480. Tang YW, Cleavinger PJ, Li H, Mitchell PS, Smith TF, Persing DH.
Analysis of candidate-host immunogenetic determinants in herpes
simplex virus-associated Mollarets meningitis. Clin Infect Dis
2000;30:176–8. PMID:10619748 https://doi.org/10.1086/313616
481. Shalabi M, Whitley RJ. Recurrent benign lymphocytic meningitis.
Clin Infect Dis 2006;43:11947. PMID:17029141 https://doi.
org/10.1086/508281
482. Landry ML, Greenwold J, Vikram HR. Herpes simplex type-2
meningitis: presentation and lack of standardized therapy. Am J Med
2009;122:68891. PMID:19559173 https://doi.org/10.1016/j.
amjmed.2009.02.017
483. Aurelius E, Franzen-Röhl E, Glimåker M, et al.; HSV-2 Meningitis
Study Group. Long-term valacyclovir suppressive treatment after herpes
simplex virus type 2 meningitis: a double-blind, randomized controlled
trial. Clin Infect Dis 2012;54:130413. PMID:22460966 https://doi.
org/10.1093/cid/cis031
484. Magawa S, Tanaka H, Furuhashi F, et al. A literature review of herpes
simplex virus hepatitis in pregnancy. J Matern Fetal Neonatal Med
2020;33:1774–9. PMID:30235956 https://doi.org/10.1080/147670
58.2018.1527311
485. Masadeh M, Shen H, Lee Y, et al. A fatal case of herpes simplex virus
hepatitis in a pregnant patient. Intractable Rare Dis Res 2017;6:124–7.
PMID:28580213 https://doi.org/10.5582/irdr.2017.01013
486. Martin ET, Krantz E, Gottlieb SL, et al. A pooled analysis of the
effect of condoms in preventing HSV-2 acquisition. Arch Intern
Med 2009;169:1233–40. PMID:19597073 https://doi.org/10.1001/
archinternmed.2009.177
487. Wald A, Langenberg AG, Krantz E, et al. The relationship
between condom use and herpes simplex virus acquisition. Ann
Intern Med 2005;143:70713. PMID:16287791 https://doi.
org/10.7326/0003-4819-143-10-200511150-00007
Recommendations and Reports
152 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
488. Wald A, Langenberg AG, Link K, et al. Effect of condoms on reducing
the transmission of herpes simplex virus type 2 from men to women.
JAMA 2001;285:3100–6. PMID:11427138 https://doi.org/10.1001/
jama.285.24.3100
489. Magaret AS, Mujugira A, Hughes JP, et al.; Partners in Prevention
HSV/HIV Transmission Study Team. Effect of condom use on per-act
HSV-2 transmission risk in HIV-1, HSV-2-discordant couples. Clin
Infect Dis 2016;62:456–61. PMID:26578538
490. Mehta SD, Moses S, Agot K, et al. Medical male circumcision and
herpes simplex virus 2 acquisition: posttrial surveillance in Kisumu,
Kenya. J Infect Dis 2013;208:186976. PMID:23901089 https://doi.
org/10.1093/infdis/jit371
491. Grund JM, Bryant TS, Jackson I, et al. Association between male
circumcision and womens biomedical health outcomes: a systematic
review. Lancet Glob Health 2017;5:e111322. PMID:29025633
https://doi.org/10.1016/S2214-109X(17)30369-8
492. Celum C, Morrow RA, Donnell D, et al.; Partners PrEP Study Team.
Daily oral tenofovir and emtricitabine-tenofovir preexposure
prophylaxis reduces herpes simplex virus type 2 acquisition among
heterosexual HIV-1-uninfected men and women: a subgroup analysis
of a randomized trial. Ann Intern Med 2014;161:119. PMID:24979446
https://doi.org/10.7326/M13-2471
493. Abdool Karim SS, Abdool Karim Q, Gengiah TN. Tenofovir gel to
prevent HSV-2 infection. N Engl J Med 2015;373:19801.
PMID:26559584 https://doi.org/10.1056/NEJMoa1410649
494. Marcus JL, Glidden DV, McMahan V, et al. Daily oral emtricitabine/
tenofovir preexposure prophylaxis and herpes simplex virus type 2
among men who have sex with men. PLoS One 2014;9:e91513.
PMID:24637511 https://doi.org/10.1371/journal.pone.0091513
495. Celum C, Hong T, Cent A, et al.; ACTG PEARLS/A5175 Team.
Herpes simplex virus type 2 acquisition among HIV-1-infected adults
treated with tenofovir disoproxyl fumarate as part of combination
antiretroviral therapy: results from the ACTG A5175 PEARLS Study.
J Infect Dis 2017;215:90710. PMID:28453835 https://doi.
org/10.1093/infdis/jix029
496. Gilbert LK, Wyand F. Genital herpes education and counselling: testing
a one-page ‘FAQ’ intervention. Herpes 2009;15:51–6. PMID:19306603
497. Rosenthal SL, Zimet GD, Leichliter JS, et al. The psychosocial impact
of serological diagnosis of asymptomatic herpes simplex virus type 2
infection. Sex Transm Infect 2006;82:1547, discussion 157–8.
PMID:16581745 https://doi.org/10.1136/sti.2005.016311
498. Miyai T, Turner KR, Kent CK, Klausner J. The psychosocial impact
of testing individuals with no history of genital herpes for herpes simplex
virus type 2. Sex Transm Dis 2004;31:51721. PMID:15480111
https://doi.org/10.1097/01.olq.0000137901.71284.6b
499. Ross K, Johnston C, Wald A. Herpes simplex virus type 2 serological
testing and psychosocial harm: a systematic review. Sex Transm Infect
2011;87:594600. PMID:21903980 https://doi.org/10.1136/
sextrans-2011-050099
500. Henry RE, Wegmann JA, Hartle JE, Christopher GW. Successful oral
acyclovir desensitization. Ann Allergy 1993;70:3868. PMID:8498729
501. Leeyaphan C, Surawan TM, Chirachanakul P, et al. Clinical
characteristics of hypertrophic herpes simplex genitalis and treatment
outcomes of imiquimod: a retrospective observational study. Int J Infect
Dis 2015;33:16570. PMID:25660091 https://doi.org/10.1016/j.
ijid.2015.02.002
502. Keller MJ, Huber A, Espinoza L, et al. Impact of herpes simplex virus
type 2 and human immunodeficiency virus dual infection on female
genital tract mucosal immunity and the vaginal microbiome. J Infect
Dis 2019;220:85261. PMID:31111902 https://doi.org/10.1093/
infdis/jiz203
503. Posavad CM, Wald A, Kuntz S, et al. Frequent reactivation of herpes
simplex virus among HIV-1-infected patients treated with highly active
antiretroviral therapy. J Infect Dis 2004;190:693–6. PMID:15272395
https://doi.org/10.1086/422755
504. Tobian AA, Grabowski MK, Serwadda D, et al.; Rakai Health Sciences
Program. Reactivation of herpes simplex virus type 2 after initiation
of antiretroviral therapy. J Infect Dis 2013;208:83946.
PMID:23812240 https://doi.org/10.1093/infdis/jit252
505. Mujugira A, Magaret AS, Celum C, et al.; Partners in Prevention HSV/
HIV Transmission Study Team. Daily acyclovir to decrease herpes simplex
virus type 2 (HSV-2) transmission from HSV-2/HIV-1 coinfected
persons: a randomized controlled trial. J Infect Dis 2013;208:1366–74.
PMID:23901094 https://doi.org/10.1093/infdis/jit333
506. Van Wagoner N, Geisler WM, Bachmann LH, Hook EW. The effect
of valacyclovir on HIV and HSV-2 in HIV-infected persons on
antiretroviral therapy with previously unrecognised HSV-2. Int J STD
AIDS 2015;26:57481. PMID:25147236 https://doi.
org/10.1177/0956462414546504
507. Reyes M, Shaik NS, Graber JM, et al.; Task Force on Herpes Simplex
Virus Resistance. Acyclovir-resistant genital herpes among persons
attending sexually transmitted disease and human immunodeficiency
virus clinics. Arch Intern Med 2003;163:7680. PMID:12523920
https://doi.org/10.1001/archinte.163.1.76
508. Safrin S, Crumpacker C, Chatis P, et al.; The AIDS Clinical Trials
Group. A controlled trial comparing foscarnet with vidarabine for
acyclovir-resistant mucocutaneous herpes simplex in the acquired
immunodeficiency syndrome. N Engl J Med 1991;325:5515.
PMID:1649971 https://doi.org/10.1056/NEJM199108223250805
509. Levin MJ, Bacon TH, Leary JJ. Resistance of herpes simplex virus
infections to nucleoside analogues in HIV-infected patients. Clin Infect
Dis 2004;39(Suppl 5):S24857. PMID:15494896 https://doi.
org/10.1086/422364
510. Tandon S, Singh J, Sinha S, Sharma DP. Recalcitrant hypertrophic
herpes genitalis in HIV-infected patient successfully treated with topical
imiquimod. Dermatol Ther (Heidelb) 2017;30:e12479.
PMID:28261899 https://doi.org/10.1111/dth.12479
511. Perkins N, Nisbet M, Thomas M. Topical imiquimod treatment of
aciclovir-resistant herpes simplex disease: case series and literature
review. Sex Transm Infect 2011;87:292–5. PMID:21406577 https://
doi.org/10.1136/sti.2010.047431
512. McElhiney LF. Topical cidofovir for treatment of resistant viral
infections. Int J Pharm Compd 2006;10:324–8. PMID:23974309
513. Erard V, Wald A, Corey L, Leisenring WM, Boeckh M. Use of long-
term suppressive acyclovir after hematopoietic stem-cell transplantation:
impact on herpes simplex virus (HSV) disease and drug-resistant HSV
disease. J Infect Dis 2007;196:266–70. PMID:17570114 https://doi.
org/10.1086/518938
514. Brown ZA, Selke S, Zeh J, et al. The acquisition of herpes simplex virus
during pregnancy. N Engl J Med 1997;337:509–15. PMID:9262493
https://doi.org/10.1056/NEJM199708213370801
515. Pinninti SG, Kimberlin DW. Maternal and neonatal herpes simplex
virus infections. Am J Perinatol 2013;30:113–9. PMID:23303485
https://doi.org/10.1055/s-0032-1332802
516. Brown ZA, Benedetti J, Ashley R, et al. Neonatal herpes simplex virus
infection in relation to asymptomatic maternal infection at the time
of labor. N Engl J Med 1991;324:1247–52. PMID:1849612 https://
doi.org/10.1056/NEJM199105023241804
517. Brown ZA, Wald A, Morrow RA, Selke S, Zeh J, Corey L. Effect of
serologic status and cesarean delivery on transmission rates of herpes
simplex virus from mother to infant. JAMA 2003;289:203–9.
PMID:12517231 https://doi.org/10.1001/jama.289.2.203
518. Ahrens KA, Anderka MT, Feldkamp ML, Canfield MA, Mitchell AA,
Werler MM; National Birth Defects Prevention Study. Antiherpetic
medication use and the risk of gastroschisis: findings from the National
Birth Defects Prevention Study, 1997–2007. Paediatr Perinat Epidemiol
2013;27:340–5. PMID:23772935 https://doi.org/10.1111/ppe.12064
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 153
US Department of Health and Human Services/Centers for Disease Control and Prevention
519. Stone KM, Reiff-Eldridge R, White AD, et al. Pregnancy outcomes
following systemic prenatal acyclovir exposure: conclusions from the
international acyclovir pregnancy registry, 1984–1999. Birth Defects
Res A Clin Mol Teratol 2004;70:201–7. PMID:15108247 https://doi.
org/10.1002/bdra.20013
520. Pasternak B, Hviid A. Use of acyclovir, valacyclovir, and famciclovir in
the first trimester of pregnancy and the risk of birth defects. JAMA
2010;304:85966. PMID:20736469 https://doi.org/10.1001/
jama.2010.1206
521. Sheffield JS, Sánchez PJ, Wendel GD Jr, et al. Placental histopathology
of congenital syphilis. Obstet Gynecol 2002;100:12633.
PMID:12100814
522. Watts DH, Brown ZA, Money D, et al. A double-blind, randomized,
placebo-controlled trial of acyclovir in late pregnancy for the reduction
of herpes simplex virus shedding and cesarean delivery. Am J Obstet
Gynecol 2003;188:83643. PMID:12634667 https://doi.org/10.1067/
mob.2003.185
523. Scott LL, Hollier LM, McIntire D, Sanchez PJ, Jackson GL, Wendel
GD Jr. Acyclovir suppression to prevent recurrent genital herpes at
delivery. Infect Dis Obstet Gynecol 2002;10:71–7. PMID:12530483
https://doi.org/10.1155/S1064744902000054
524. Pinninti SG, Angara R, Feja KN, et al. Neonatal herpes disease following
maternal antenatal antiviral suppressive therapy: a multicenter case
series. J Pediatr 2012;161:134–8.e1-3. PMID:22336576 https://doi.
org/10.1016/j.jpeds.2011.12.053
525. ACOG Committee on Practice Bulletins. ACOG Practice Bulletin.
Clinical management guidelines for obstetrician-gynecologists. No. 82
June 2007. Management of herpes in pregnancy. Obstet Gynecol
2007;109:148998. PMID:17569194 https://doi.org/10.1097/01.
AOG.0000263902.31953.3e
526. Winer RL, Hughes JP, Feng Q, et al. Early natural history of incident,
type-specific human papillomavirus infections in newly sexually active
young women. Cancer Epidemiol Biomarkers Prev 2011;20:699707.
PMID:21173170 https://doi.org/10.1158/1055-9965.EPI-10-1108
527. Ahmed N, Pillay A, Lawler M, Bobat R, Archary M. Donovanosis
causing lymphadenitis, mastoiditis, and meningitis in a child. Lancet
2015;385:2644. PMID:26122163 https://doi.org/10.1016/
S0140-6736(15)60992-8
528. Arora AK, Kumaran MS, Narang T, Saikia UN, Handa S. Donovanosis
and squamous cell carcinoma: the relationship conundrum! Int J STD
AIDS 2017;28:4114. PMID:27535727 https://doi.
org/10.1177/0956462416665996
529. Liverani CA, Lattuada D, Mangano S, et al. Hypertrophic donavanosis
in a young pregnant woman. J Pediatr Adolesc Gynecol 2012;25:e81–3.
PMID:22840941 https://doi.org/10.1016/j.jpag.2011.10.002
530. Magalhães BM, Veasey JV, Mayor SAS, Lellis RF. Donovanosis in a
child victim of sexual abuse: response to doxycycline treatment. An
Bras Dermatol 2018;93:5924. https://doi.org/10.1590/
abd1806-4841.20187948
531. Marfatia YS, Menon DS, Jose S, Patel BK. Nonhealing genital ulcer
in AIDS: a diagnostic dilemma! Indian J Sex Transm Dis AIDS
2016; 37: 197 200. PMID:27890958 https://doi.
org/10.4103/0253-7184.192130
532. Narang T, Kanwar AJ. Genital elephantiasis due to donovanosis:
forgotten but not gone yet. Int J STD AIDS 2012;23:8356.
PMID:23155109 https://doi.org/10.1258/ijsa.2012.012096
533. Pilani A, Vora R, Anjaneyan G. Granuloma inguinale
mimicking as squamous cell carcinoma of penis. Indian J Sex
Transm Dis AIDS 2014;35:56–8. PMID:24958990 https://doi.
org/10.4103/0253-7184.132433
534. Ramdial PK, Sing Y, Ramburan A, et al. Infantile donovanosis
presenting as external auditory canal polyps: a diagnostic trap. Am J
Dermatopathol 2012;34:81821. PMID:23169417 https://doi.
org/10.1097/DAD.0b013e3182540ccb
535. Wahal SP, Tuli D. Donovanosis: an incidental finding on Pap
test. J Cytol 2013;30:2178. PMID:24130421 https://doi.
org/10.4103/0970-9371.117638
536. Bowden FJ; National Donovanosis Eradication Advisory Committee.
Donovanosis in Australia: going, going. Sex Transm Infect
2005;81:365–6. PMID:16199732 https://doi.org/10.1136/
sti.2004.013227
537. Bright A. National Notifiable Diseases Surveillance System surveillance
report: sexually transmissible infections in Aboriginal and Torres Strait
Islander people. Commun Dis Intell Q Rep 2015;39:E5849.
PMID:26779731
538. O’Farrell N. Donovanosis. Sex Transm Infect 2002;78:4527.
PMID:12473810 https://doi.org/10.1136/sti.78.6.452
539. Mabey D, Peeling RW. Lymphogranuloma venereum. Sex Transm
Infect 2002;78:902. PMID:12081191 https://doi.org/10.1136/
sti.78.2.90
540. White JA. Manifestations and management of lymphogranuloma
venereum. Curr Opin Infect Dis 2009;22:57–66. PMID:19532081
https://doi.org/10.1097/QCO.0b013e328320a8ae
541. de Vries HJ, Zingoni A, White JA, Ross JD, Kreuter A. 2013
European Guideline on the management of proctitis, proctocolitis
and enteritis caused by sexually transmissible pathogens. Int
J STD AIDS 2014;25:46574. PMID:24352129 https://doi.
org/10.1177/0956462413516100
542. Ward H, Martin I, Macdonald N, et al. Lymphogranuloma venereum
in the United kingdom. Clin Infect Dis 2007;44:2632.
PMID:17143811 https://doi.org/10.1086/509922
543. Martin-Iguacel R, Llibre JM, Nielsen H, et al. Lymphogranuloma
venereum proctocolitis: a silent endemic disease in men who have sex
with men in industrialised countries. Eur J Clin Microbiol Infect Dis
2010;29:91725. PMID:20509036 https://doi.org/10.1007/
s10096-010-0959-2
544. de Voux A, Kent JB, Macomber K, et al. Notes from the field: cluster
of lymphogranuloma venereum cases among men who have sex with
men—Michigan, August 2015–April 2016. MMWR Morb Mortal
Wkly Rep 2016;65:9201. PMID:27583686 https://doi.org/10.15585/
mmwr.mm6534a6
545. Pallawela SN, Sullivan AK, Macdonald N, et al. Clinical predictors of
rectal lymphogranuloma venereum infection: results from a multicentre
case-control study in the U.K. Sex Transm Infect 2014;90:269–74.
PMID:24687130 https://doi.org/10.1136/sextrans-2013-051401
546. de Vrieze NH, de Vries HJ. Lymphogranuloma venereum among men
who have sex with men. An epidemiological and clinical review. Expert
Rev Anti Infect Ther 2014;12:697704. PMID:24655220 https://doi.
org/10.1586/14787210.2014.901169
547. Koper NE, van der Sande MA, Gotz HM, Koedijk FD; Dutch STI
Clinics. Lymphogranuloma venereum among men who have sex with
men in the Netherlands: regional differences in testing rates lead to
underestimation of the incidence, 2006–2012. Euro Surveill
2013;18:20561. PMID:23987831 https://doi.org/10.2807/1560-
7917.ES2013.18.34.20561
548. Haar K, Dudareva-Vizule S, Wisplinghoff H, et al. Lymphogranuloma
venereum in men screened for pharyngeal and rectal infection,
Germany. Emerg Infect Dis 2013;19:48892. PMID:23621949
https://doi.org/10.3201/eid1903.121028
549. Riera-Monroig J, Fuertes de Vega I. Lymphogranuloma venereum
presenting as an ulcer on the tongue. Sex Transm Infect 2019;95:169–70.
PMID:30554142 https://doi.org/10.1136/sextrans-2018-053787
550. Andrada MT, Dhar JK, Wilde H. Oral lymphogranuloma venereum
and cervical lymphadenopathy. Case report. Mil Med 1974;139:99–101.
PMID:4204816 https://doi.org/10.1093/milmed/139.2.99
551. Ilyas S, Richmond D, Burns G, et al. Orolabial lymphogranuloma
venereum, Michigan, USA. Emerg Infect Dis 2019;25:2112–4.
PMID:31625852 https://doi.org/10.3201/eid2511.190819
Recommendations and Reports
154 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
552. Kersh EN, Pillay A, de Voux A, Chen C. Laboratory processes for
confirmation of lymphogranuloma venereum infection during a 2015
investigation of a cluster of cases in the United States. Sex Transm Dis
2017;44:6914. PMID:28876314 https://doi.org/10.1097/
OLQ.0000000000000667
553. CDC. Recommendations for the laboratory-based detection of
Chlamydia trachomatis and Neisseria gonorrhoeae—2014. MMWR
Recomm Rep 2014;63(No. RR-2). PMID:24622331
554. Pathela P, Jamison K, Kornblum J, Quinlan T, Halse TA, Schillinger
JA. Lymphogranuloma venereum: an increasingly common anorectal
infection among men who have sex with men attending New York City
sexual health clinics. Sex Transm Dis 2019;46:e14–7. PMID:30278027
https://doi.org/10.1097/OLQ.0000000000000921
555. Cohen S, Brosnan H, Kohn R, et al. P494 Diagnosis and management
of lymphogranuloma venereum (LGV) in a municipal STD clinic, San
Francisco, 2016–18. Sex Transm Infect 2019;95(Suppl 1):A229.
556. Leeyaphan C, Ong JJ, Chow EP, et al. Systematic review and meta-
analysis of doxycycline efficacy for rectal lymphogranuloma venereum
in men who have sex with men. Emerg Infect Dis 2016;22:1778–84.
PMID:27513890 https://doi.org/10.3201/eid2210.160986
557. Cabello Úbeda A, Fernández Roblas R, García Delgado R, et al.
Anorectal lymphogranuloma venereum in Madrid: a persistent
emerging problem in men who have sex with men. Sex Transm Dis
2016;43:4149. PMID:27322040 https://doi.org/10.1097/
OLQ.0000000000000459
558. Simons R, Candfield S, French P, White JA. Observed treatment
responses to short-course doxycycline therapy for rectal
lymphogranuloma venereum in men who have sex with men. Sex
Transm Dis 2018;45:4068. PMID:29465660 https://doi.org/10.1097/
OLQ.0000000000000772
559. Vall-Mayans M, Isaksson J, Caballero E, Sallés B, Herrmann B. Bubonic
lymphogranuloma venereum with multidrug treatment failure. Int J
STD AIDS 2014;25:3068. PMID:24216037 https://doi.
org/10.1177/0956462413501158
560. Blanco JL, Fuertes I, Bosch J, et al. Effective treatment of
lymphogranuloma venereum (LGV) with 1g azithormycin administered
weekly for 3 weeks in HIV-infected population. Presented at the
Conference on Retroviruses and Opportunist Infections, Seattle, WA;
February 23–26, 2015.
561. Kong FY, Rupasinghe TW, Simpson JA, et al. Pharmacokinetics of a
single 1g dose of azithromycin in rectal tissue in men. PLoS One
2017;12:e0174372. PMID:28350806 https://doi.org/10.1371/journal.
pone.0174372
562. Elgalib A, Alexander S, Tong CY, White JA. Seven days of doxycycline
is an effective treatment for asymptomatic rectal Chlamydia trachomatis
infection. Int J STD AIDS 2011;22:474–7. PMID:21764781 https://
doi.org/10.1258/ijsa.2011.011134
563. Wormser GP, Wormser RP, Strle F, Myers R, Cunha BA. How safe is
doxycycline for young children or for pregnant or breastfeeding women?
Diagn Microbiol Infect Dis 2019;93:23842. PMID:30442509
https://doi.org/10.1016/j.diagmicrobio.2018.09.015
564. Towns JM, Leslie DE, Denham I, Azzato F, Fairley CK, Chen M.
Painful and multiple anogenital lesions are common in men with
Treponema pallidum PCR-positive primary syphilis without herpes
simplex virus coinfection: a cross-sectional clinic-based study. Sex
Transm Infect 2016;92:1105. PMID:26378262 https://doi.
org/10.1136/sextrans-2015-052219
565. Theel ES, Katz SS, Pillay A. Molecular and direct detection tests for
Treponema pallidum subspecies pallidum: a review of the literature,
1964–2017. Clin Infect Dis 2020;71(Suppl 1):S412. PMID:32578865
https://doi.org/10.1093/cid/ciaa176
566. Tuddenham S, Katz SS, Ghanem KG. Syphilis laboratory guidelines:
performance characteristics of nontreponemal antibody tests. Clin
Infect Dis 2020;71(Suppl 1):S21–42. PMID:32578862 https://doi.
org/10.1093/cid/ciaa306
567. Park IU, Tran A, Pereira L, Fakile Y. Sensitivity and specificity of
treponemal-specific tests for the diagnosis of syphilis. Clin Infect Dis
2020;71(Suppl 1):S1320. PMID:32578866 https://doi.org/10.1093/
cid/ciaa349
568. Bristow CC, Klausner JD, Tran A. Clinical test performance of a rapid
point-of-care syphilis treponemal antibody test: a systematic review
and meta-analysis. Clin Infect Dis 2020;71(Suppl 1):S527.
PMID:32578863 https://doi.org/10.1093/cid/ciaa350
569. Nandwani R, Evans DT. Are you sure its syphilis? A review of false
positive serology. Int J STD AIDS 1995;6:241–8. PMID:7548285
https://doi.org/10.1177/095646249500600404
570. Romanowski B, Sutherland R, Fick GH, Mooney D, Love
EJ. Serologic response to treatment of infectious syphilis. Ann
Intern Med 1991;114:10059. PMID:2029095 https://doi.
org/10.7326/0003-4819-114-12-1005
571. CDC. Syphilis testing algorithms using treponemal tests for initial
screening—four laboratories, New York City, 2005–2006. MMWR
Morb Mortal Wkly Rep 2008;57:872–5. PMID:18701877
572. CDC. Discordant results from reverse sequence syphilis screening—five
laboratories, United States, 2006–2010. MMWR Morb Mortal Wkly
Rep 2011;60:133–7. PMID:21307823
573. Ortiz-Lopez N, Diez M, Diaz O, Simon F, Diaz A. Epidemiological
surveillance of congenital syphilis in Spain, 2000–2010. Pediatr Infect
Dis J 2012;31:98890. PMID:22572752 https://doi.org/10.1097/
INF.0b013e31825d3152
574. Ortiz DA, Shukla MR, Loeffelholz MJ. The traditional or reverse
algorithm for diagnosis of syphilis: pros and cons. Clin Infect Dis
2020;71(Suppl 1):S4351. PMID:32578864 https://doi.org/10.1093/
cid/ciaa307
575. Berry GJ, Loeffelholz MJ. Use of treponemal screening assay strength
of signal to avoid unnecessary confirmatory testing. Sex Transm
Dis 2016;43:73740. PMID:27835625 https://doi.org/10.1097/
OLQ.0000000000000524
576. Park IU, Chow JM, Bolan G, Stanley M, Shieh J, Schapiro JM.
Screening for syphilis with the treponemal immunoassay: analysis of
discordant serology results and implications for clinical management.
J Infect Dis 2011;204:1297304. PMID:21930610 https://doi.
org/10.1093/infdis/jir524
577. Loeffelholz MJ, Wen T, Patel JA. Analysis of bioplex syphilis IgG
quantitative results in different patient populations. Clin Vaccine
Immunol 2011;18:2005–6. PMID:21880852 https://doi.org/10.1128/
CVI.05335-11
578. Fakile YF, Jost H, Hoover KW, et al. Correlation of treponemal
immunoassay signal strength values with reactivity of confirmatory
treponemal testing. J Clin Microbiol 2017;56:e01165-17.
PMID:29046410 https://doi.org/10.1128/JCM.01165-17
579. Wong EH, Klausner JD, Caguin-Grygiel G, et al. Evaluation of an IgM/
IgG sensitive enzyme immunoassay and the utility of index values for
the screening of syphilis infection in a high-risk population. Sex Transm
Dis 2011;38:52832. PMID:21233789 https://doi.org/10.1097/
OLQ.0b013e318205491a
580. Dai S, Chi P, Lin Y, et al. Improved reverse screening algorithm
for Treponema pallidum antibody using signal-to-cutoff ratios
from chemiluminescence microparticle immunoassay. Sex Transm
Dis 2014;41:2934. PMID:24326578 https://doi.org/10.1097/
OLQ.0000000000000066
581. Li Z, Feng Z, Liu P, Yan C. Screening for antibodies against Treponema
pallidum with chemiluminescent microparticle immunoassay: analysis
of discordant serology results and clinical characterization. Ann Clin
Biochem 2016;53:58892. PMID:26680646 https://doi.
org/10.1177/0004563215623806
582. Yen-Lieberman B, Daniel J, Means C, Waletzky J, Daly TM.
Identification of false-positive syphilis antibody results using a
semiquantitative algorithm. Clin Vaccine Immunol 2011;18:103840.
PMID:21508162 https://doi.org/10.1128/CVI.05066-11
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 155
US Department of Health and Human Services/Centers for Disease Control and Prevention
583. Yimtae K, Srirompotong S, Lertsukprasert K. Otosyphilis: a review of
85 cases. Otolaryngol Head Neck Surg 2007;136:6771.
PMID:17210336 https://doi.org/10.1016/j.otohns.2006.08.026
584. Gleich LL, Linstrom CJ, Kimmelman CP. Otosyphilis: a diagnostic and
therapeutic dilemma. Laryngoscope 1992;102:1255–9. PMID:1307698
https://doi.org/10.1288/00005537-199211000-00010
585. Lukehart SA, Hook EW 3rd, Baker-Zander SA, Collier AC, Critchlow
CW, Handsfield HH. Invasion of the central nervous system by
Treponema pallidum: implications for diagnosis and treatment. Ann
Intern Med 1988;109:85562. PMID:3056164 https://doi.
org/10.7326/0003-4819-109-11-855
586. Harding AS, Ghanem KG. The performance of cerebrospinal fluid
treponemal-specific antibody tests in neurosyphilis: a systematic
review. Sex Transm Dis 2012;39:291–7. PMID:22421696 https://doi.
org/10.1097/OLQ.0b013e31824c0e62
587. Jaffe HW, Larsen SA, Peters M, Jove DF, Lopez B, Schroeter
AL. Tests for treponemal antibody in CSF. Arch Intern Med
1978;138:2525. PMID:343742 https://doi.org/10.1001/
archinte.1978.03630260050016
588. Marra CM, Maxwell CL, Smith SL, et al. Cerebrospinal fluid
abnormalities in patients with syphilis: association with clinical and
laboratory features. J Infect Dis 2004;189:369–76. PMID:14745693
https://doi.org/10.1086/381227
589. CDC. Inadvertent use of Bicillin C-R to treat syphilis infection—Los
Angeles, California, 1999–2004. MMWR Morb Mortal Wkly Rep
2005;54:217–9. PMID:15758893
590. Butler T. The Jarisch-Herxheimer reaction after antibiotic treatment of
spirochetal infections: a review of recent cases and our understanding of
pathogenesis. Am J Trop Med Hyg 2017;96:46–52. PMID:28077740
https://doi.org/10.4269/ajtmh.16-0434
591. Rolfs RT, Joesoef MR, Hendershot EF, et al.; The Syphilis and HIV
Study Group. A randomized trial of enhanced therapy for early
syphilis in patients with and without human immunodeficiency virus
infection. N Engl J Med 1997;337:307–14. PMID:9235493 https://
doi.org/10.1056/NEJM199707313370504
592. Yang CJ, Lee NY, Chen TC, et al. One dose versus three weekly doses
of benzathine penicillin G for patients co-infected with HIV and early
syphilis: a multicenter, prospective observational study. PLoS One
2014;9:e109667. PMID:25286091 https://doi.org/10.1371/journal.
pone.0109667
593. Ganesan A, Mesner O, Okulicz JF, et al.; Infectious Disease Clinical
Research Program HIV/STI Working Group. A single dose of
benzathine penicillin G is as effective as multiple doses of benzathine
penicillin G for the treatment of HIV-infected persons with early
syphilis. Clin Infect Dis 2015;60:653–60. PMID:25389249 https://
doi.org/10.1093/cid/ciu888
594. Ghanem KG, Erbelding EJ, Wiener ZS, Rompalo AM. Serological
response to syphilis treatment in HIV-positive and HIV-negative
patients attending sexually transmitted diseases clinics. Sex Transm
Infect 2007;83:97101. PMID:16943224 https://doi.org/10.1136/
sti.2006.021402
595. Seña AC, Wolff M, Martin DH, et al. Predictors of serological cure
and serofast state after treatment in HIV-negative persons with early
syphilis. Clin Infect Dis 2011;53:1092–9. PMID:21998287 https://
doi.org/10.1093/cid/cir671
596. Zhang RL, Wang QQ, Zhang JP, Yang LJ. Molecular subtyping of
Treponema pallidum and associated factors of serofast status in early
syphilis patients: identified novel genotype and cytokine marker. PLoS
One 2017;12:e0175477. PMID:28410389 https://doi.org/10.1371/
journal.pone.0175477
597. Seña AC, Zhang XH, Li T, et al. A systematic review of syphilis
serological treatment outcomes in HIV-infected and HIV-uninfected
persons: rethinking the significance of serological non-responsiveness
and the serofast state after therapy. BMC Infect Dis 2015;15:479.
PMID:26511465 https://doi.org/10.1186/s12879-015-1209-0
598. Tong ML, Lin LR, Liu GL, et al. Factors associated with serological
cure and the serofast state of HIV-negative patients with primary,
secondary, latent, and tertiary syphilis. PLoS One 2013;8:e70102.
PMID:23894598 https://doi.org/10.1371/journal.pone.0070102
599. Seña AC, Wolff M, Behets F, et al. Response to therapy following
retreatment of serofast early syphilis patients with benzathine penicillin.
Clin Infect Dis 2013;56:4202. PMID:23118269 https://doi.
org/10.1093/cid/cis918
600. Ghanem KG, Erbelding EJ, Cheng WW, Rompalo AM. Doxycycline
compared with benzathine penicillin for the treatment of early
syphilis. Clin Infect Dis 2006;42:e45–9. PMID:16477545 https://
doi.org/10.1086/500406
601. Wong T, Singh AE, De P. Primary syphilis: serological treatment
response to doxycycline/tetracycline versus benzathine penicillin. Am
J Med 2008;121:903–8. PMID:18823862 https://doi.org/10.1016/j.
amjmed.2008.04.042
602. Hook EW 3rd, Martin DH, Stephens J, Smith BS, Smith K.
A randomized, comparative pilot study of azithromycin versus
benzathine penicillin G for treatment of early syphilis. Sex
Transm Dis 2002;29:48690. PMID:12172535 https://doi.
org/10.1097/00007435-200208000-00010
603. Cao Y, Su X, Wang Q, et al. A multicenter study evaluating ceftriaxone
and benzathine penicillin G as treatment agents for early syphilis in
Jiangsu, China. Clin Infect Dis 2017;65:1683–8. PMID:29020150
https://doi.org/10.1093/cid/cix611
604. Riedner G, Rusizoka M, Todd J, et al. Single-dose azithromycin versus
penicillin G benzathine for the treatment of early syphilis. N Engl J
Med 2005;353:1236–44. PMID:16177249 https://doi.org/10.1056/
NEJMoa044284
605. Hook EW 3rd, Behets F, Van Damme K, et al. A phase III equivalence
trial of azithromycin versus benzathine penicillin for treatment of early
syphilis. J Infect Dis 2010;201:172935. PMID:20402591 https://doi.
org/10.1086/652239
606. Lukehart SA, Godornes C, Molini BJ, et al. Macrolide resistance
in Treponema pallidum in the United States and Ireland. N Engl J
Med 2004;351:154–8. PMID:15247355 https://doi.org/10.1056/
NEJMoa040216
607. Mitchell SJ, Engelman J, Kent CK, Lukehart SA, Godornes C, Klausner
JD. Azithromycin-resistant syphilis infection: San Francisco, California,
2000–2004. Clin Infect Dis 2006;42:33745. PMID:16392078
https://doi.org/10.1086/498899
608. A2058G Prevalence Workgroup. Prevalence of the 23S rRNA A2058G
point mutation and molecular subtypes in Treponema pallidum in the
United States, 2007 to 2009. Sex Transm Dis 2012;39:7948.
PMID:23001267
609. Rolfs RT, Joesoef MR, Hendershot EF, et al.; The Syphilis and HIV
Study Group. A randomized trial of enhanced therapy for early syphilis
in patients with and without human immunodeficiency virus infection.
N Engl J Med 1997;337:30714. PMID:9235493 https://doi.
org/10.1056/NEJM199707313370504
610. Collart P, Poitevin M, Milovanovic A, Herlin A, Durel J. Kinetic study
of serum penicillin concentrations after single doses of benzathine and
benethamine penicillins in young and old people. Br J Vener Dis
1980;56:35562. PMID:7448577 https://doi.org/10.1136/
sti.56.6.355
611. Hagdrup HK, Lange Wantzin G, Secher L, Rosdahl VT. Penicillin
concentrations in serum following weekly injections of benzathine
penicillin G. Chemotherapy 1986;32:99101. PMID:3698728 https://
doi.org/10.1159/000238397
612. Frentz G, Nielsen PB, Espersen F, Czartoryski A, Aastrup H. Penicillin
concentrations in blood and spinal fluid after a single intramuscular
injection of penicillin G benzathine. Eur J Clin Microbiol
1984;3:147–9. PMID:6723638 https://doi.org/10.1007/BF02014334
Recommendations and Reports
156 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
613. Nathan L, Bawdon RE, Sidawi JE, Stettler RW, McIntire DM, Wendel
GD Jr. Penicillin levels following the administration of benzathine
penicillin G in pregnancy. Obstet Gynecol 1993;82:33842.
PMID:8355931
614. Marra CM, Maxwell CL, Tantalo LC, Sahi SK, Lukehart SA.
Normalization of serum rapid plasma reagin titer predicts normalization
of cerebrospinal fluid and clinical abnormalities after treatment of
neurosyphilis. Clin Infect Dis 2008;47:893–9. PMID:18715154
https://doi.org/10.1086/591534
615. Xiao Y, Tong ML, Lin LR, et al. Serological response predicts
normalization of cerebrospinal fluid abnormalities at six months after
treatment in HIV-negative neurosyphilis patients. Sci Rep 2017;7:9911.
PMID:28855625 https://doi.org/10.1038/s41598-017-10387-x
616. Hook EW 3rd, Baker-Zander SA, Moskovitz BL, Lukehart SA, Handsfield
HH. Ceftriaxone therapy for asymptomatic neurosyphilis. Case report
and Western blot analysis of serum and cerebrospinal fluid IgG response
to therapy. Sex Transm Dis 1986;13(Suppl):185–8. PMID:3764632
https://doi.org/10.1097/00007435-198607000-00018
617. Shann S, Wilson J. Treatment of neurosyphilis with ceftriaxone.
Sex Transm Infect 2003;79:4156. PMID:14573840 https://doi.
org/10.1136/sti.79.5.415
618. Ahmed KA, Fox SJ, Frigas E, Park MA. Clinical outcome in the use
of cephalosporins in pediatric patients with a history of penicillin
allergy. Int Arch Allergy Immunol 2012;158:40510. PMID:22487723
https://doi.org/10.1159/000333553
619. Park MA, Koch CA, Klemawesch P, Joshi A, Li JT. Increased adverse drug
reactions to cephalosporins in penicillin allergy patients with positive
penicillin skin test. Int Arch Allergy Immunol 2010;153:268–73.
PMID:20484925 https://doi.org/10.1159/000314367
620. Novalbos A, Sastre J, Cuesta J, et al. Lack of allergic cross-reactivity
to cephalosporins among patients allergic to penicillins. Clin
Exp Allergy 2001;31:43843. PMID:11260156 https://doi.
org/10.1046/j.1365-2222.2001.00992.x
621. Pichichero ME, Casey JR. Safe use of selected cephalosporins in
penicillin-allergic patients: a meta-analysis. Otolaryngol Head Neck
Surg 2007;136:340–7. PMID:17321857 https://doi.org/10.1016/j.
otohns.2006.10.007
622. Kingston AA, Vujevich J, Shapiro M, et al. Seronegative secondary
syphilis in 2 patients coinfected with human immunodeficiency
virus. Arch Dermatol 2005;141:431–3. PMID:15837859 https://doi.
org/10.1001/archderm.141.4.431
623. CDC. Symptomatic early neurosyphilis among HIV-positive men who
have sex with men—four cities, United States, January 2002–June 2004.
MMWR Morb Mortal Wkly Rep 2007;56:625–8. PMID:17597693
624. Ghanem KG, Moore RD, Rompalo AM, Erbelding EJ, Zenilman JM,
Gebo KA. Neurosyphilis in a clinical cohort of HIV-1-infected patients.
AIDS 2008;22:1145–51. PMID:18525260 https://doi.org/10.1097/
QAD.0b013e32830184df
625. Ghanem KG, Moore RD, Rompalo AM, Erbelding EJ, Zenilman JM,
Gebo KA. Antiretroviral therapy is associated with reduced serologic
failure rates for syphilis among HIV-infected patients. Clin Infect Dis
2008;47:258–65. PMID:18532887 https://doi.org/10.1086/589295
626. Tomkins A, Ahmad S, Cousins DE, Thng CM, Vilar FJ, Higgins
SP. Screening for asymptomatic neurosyphilis in HIV patients after
treatment of early syphilis: an observational study. Sex Transm
Infect 2018;94:337–9. PMID:28196838 https://doi.org/10.1136/
sextrans-2016-052938
627. Yang CJ, Chang SY, Hung CC. Sensitivity and specificity of lumbar
puncture in HIV-infected patients with syphilis and no neurologic
symptoms. Clin Infect Dis 2009;49:1623, author reply 162–3.
PMID:19500029 https://doi.org/10.1086/599616
628. Marra CM, Boutin P, McArthur JC, et al. A pilot study evaluating
ceftriaxone and penicillin G as treatment agents for neurosyphilis in
human immunodeficiency virus-infected individuals. Clin Infect Dis
2000;30:540–4. PMID:10722441 https://doi.org/10.1086/313725
629. Dowell ME, Ross PG, Musher DM, Cate TR, Baughn RE. Response of
latent syphilis or neurosyphilis to ceftriaxone therapy in persons infected
with human immunodeficiency virus. Am J Med 1992;93:481–8.
PMID:1442850 https://doi.org/10.1016/0002-9343(92)90574-U
630. Smith NH, Musher DM, Huang DB, et al. Response of HIV-infected
patients with asymptomatic syphilis to intensive intramuscular therapy
with ceftriaxone or procaine penicillin. Int J STD AIDS 2004;15:328–32.
PMID:15117503 https://doi.org/10.1177/095646240401500511
631. Ahmed KA, Fox SJ, Frigas E, Park MA. Clinical outcome in the use
of cephalosporins in pediatric patients with a history of penicillin
allergy. Int Arch Allergy Immunol 2012;158:40510. PMID:22487723
https://doi.org/10.1159/000333553
632. Trivedi S, Williams C, Torrone E, Kidd S. National trends and reported
risk factors among pregnant women with syphilis in the United States,
2012–2016. Obstet Gynecol 2019;133:2732. PMID:30531570
https://doi.org/10.1097/AOG.0000000000003000
633. Biswas HH, Chew Ng RA, Murray EL, et al. Characteristics associated
with delivery of an infant with congenital syphilis and missed
opportunities for prevention—California, 2012 to 2014. Sex Transm
Dis 2018;45:43541. PMID:29465666 https://doi.org/10.1097/
OLQ.0000000000000782
634. Slutsker JS, Hennessy RR, Schillinger JA. Factors contributing to
congenital syphilis cases—New York City, 2010–2016. MMWR Morb
Mortal Wkly Rep 2018;67:1088–93. PMID:30286056 https://doi.
org/10.15585/mmwr.mm6739a3
635. DiOrio D, Kroeger K, Ross A. Social vulnerability in congenital
syphilis case mothers: qualitative assessment of cases in Indiana, 2014
to 2016. Sex Transm Dis 2018;45:447–51. PMID:29465662 https://
doi.org/10.1097/OLQ.0000000000000783
636. Kimball A, Torrone E, Miele K, et al. Missed opportunities for
prevention of congenital syphilis—United States, 2018. MMWR
Morb Mortal Wkly Rep 2020;69:661–5. PMID:32497029 https://
doi.org/10.15585/mmwr.mm6922a1
637. Park IU, Chow JM, Bolan G, Stanley M, Shieh J, Schapiro JM.
Screening for syphilis with the treponemal immunoassay: analysis of
discordant serology results and implications for clinical management.
J Infect Dis 2011;204:1297304. PMID:21930610 https://doi.
org/10.1093/infdis/jir524
638. Mmeje O, Chow JM, Davidson L, Shieh J, Schapiro JM, Park
IU. Discordant syphilis immunoassays in pregnancy: perinatal
outcomes and implications for clinical management. Clin Infect Dis
2015;61:104953. PMID:26063719 https://doi.org/10.1093/cid/
civ445
639. Alexander JM, Sheffield JS, Sanchez PJ, Mayfield J, Wendel GD
Jr. Efficacy of treatment for syphilis in pregnancy. Obstet Gynecol
1999;93:5–8. PMID:9916946
640. Walker GJ. Antibiotics for syphilis diagnosed during pregnancy.
Cochrane Database Syst Rev 2001;(3):CD001143. PMID:11686978
641. Wendel GD Jr, Sheffield JS, Hollier LM, Hill JB, Ramsey PS, Sánchez
PJ. Treatment of syphilis in pregnancy and prevention of congenital
syphilis. Clin Infect Dis 2002;35(Suppl 2):S200–9. PMID:12353207
https://doi.org/10.1086/342108
642. Zhu L, Qin M, Du L, Xie RH, Wong T, Wen SW. Maternal and
congenital syphilis in Shanghai, China, 2002 to 2006. Int J Infect Dis
2010;14(Suppl 3):e45–8. PMID:20137991 https://doi.org/10.1016/j.
ijid.2009.09.009
643. Hawkes S, Matin N, Broutet N, Low N. Effectiveness of interventions
to improve screening for syphilis in pregnancy: a systematic review and
meta-analysis. Lancet Infect Dis 2011;11:684–91. PMID:21683653
https://doi.org/10.1016/S1473-3099(11)70104-9
644. Hollier LM, Harstad TW, Sanchez PJ, Twickler DM, Wendel GD Jr.
Fetal syphilis: clinical and laboratory characteristics. Obstet Gynecol
2001;97:947–53. PMID:11384701
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 157
US Department of Health and Human Services/Centers for Disease Control and Prevention
645. Rac MW, Bryant SN, McIntire DD, et al. Progression of ultrasound
findings of fetal syphilis after maternal treatment. Am J Obstet Gynecol
2014;211:426.e1–6. PMID:24907700 https://doi.org/10.1016/j.
ajog.2014.05.049
646. Zhou P, Gu Z, Xu J, Wang X, Liao K. A study evaluating ceftriaxone
as a treatment agent for primary and secondary syphilis in pregnancy.
Sex Transm Dis 2005;32:4958. PMID:16041252 https://doi.
org/10.1097/01.olq.0000170443.70739.cd
647. Katanami Y, Hashimoto T, Takaya S, et al. Amoxicillin and ceftriaxone
as treatment alternatives to penicillin for maternal syphilis. Emerg
Infect Dis 2017;23:827–9. PMID:28418316 https://doi.org/10.3201/
eid2305.161936
648. Kestenbaum LA, Ebberson J, Zorc JJ, Hodinka RL, Shah SS. Defining
cerebrospinal fluid white blood cell count reference values in neonates
and young infants. Pediatrics 2010;125:25764. PMID:20064869
https://doi.org/10.1542/peds.2009-1181
649. Shah SS, Ebberson J, Kestenbaum LA, Hodinka RL, Zorc JJ. Age-
specific reference values for cerebrospinal fluid protein concentration
in neonates and young infants. J Hosp Med 2011;6:227.
PMID:20629018 https://doi.org/10.1002/jhm.711
650. Thomson J, Sucharew H, Cruz AT, et al.; Pediatric Emergency Medicine
Collaborative Research Committee (PEM CRC) HSV Study Group.
Cerebrospinal fluid reference values for young infants undergoing
lumbar puncture. Pediatrics 2018;141:e20173405. PMID:29437883
https://doi.org/10.1542/peds.2017-3405
651. Kimberlin DW, Brady MT, Jackson MA, Long SS, eds. Red book:
2018 report of the Committee on Infectious Diseases. 31st ed. Itasca,
IL: American Academy of Pediatrics; 2018.
652. Macy E, Contreras R. Adverse reactions associated with oral and
parenteral use of cephalosporins: a retrospective population-based
analysis. J Allergy Clin Immunol 2015;135:74552.e5.
PMID:25262461 https://doi.org/10.1016/j.jaci.2014.07.062
653. Macy E, Vyles D. Who needs penicillin allergy testing? Ann Allergy
Asthma Immunol 2018;121:523–9. PMID:30092265 https://doi.
org/10.1016/j.anai.2018.07.041
654. Annè S, Reisman RE. Risk of administering cephalosporin antibiotics
to patients with histories of penicillin allergy. Ann Allergy Asthma
Immunol 1995;74:167–70. PMID:7697478
655. Albin S, Agarwal S. Prevalence and characteristics of reported penicillin
allergy in an urban outpatient adult population. Allergy Asthma Proc
2014;35:48994. PMID:25584917 https://doi.org/10.2500/
aap.2014.35.3791
656. Blumenthal KG, Peter JG, Trubiano JA, Phillips EJ. Antibiotic allergy.
Lancet 2019;393:18398. PMID:30558872 https://doi.org/10.1016/
S0140-6736(18)32218-9
657. Macy E, Poon K-Y T. Self-reported antibiotic allergy incidence and
prevalence: age and sex effects. Am J Med 2009;122:778.e17.
PMID:19635279 https://doi.org/10.1016/j.amjmed.2009.01.034
658. Shenoy ES, Macy E, Rowe T, Blumenthal KG. Evaluation and
management of penicillin allergy: a review. JAMA 2019;321:188–99.
PMID:30644987 https://doi.org/10.1001/jama.2018.19283
659. Gadde J, Spence M, Wheeler B, Adkinson NF Jr. Clinical experience
with penicillin skin testing in a large inner-city STD clinic. JAMA
1993;270:245663. PMID:8230623 https://doi.org/10.1001/
jama.1993.03510200062033
660. Macy E, Ngor EW. Safely diagnosing clinically significant penicillin
allergy using only penicilloyl-poly-lysine, penicillin, and oral
amoxicillin. J Allergy Clin Immunol Pract 2013;1:25863.
PMID:24565482 https://doi.org/10.1016/j.jaip.2013.02.002
661. Jares EJ, Sánchez-Borges M, Cardona-Villa R, et al.; Latin America
Drug Allergy Interest Group. Multinational experience with
hypersensitivity drug reactions in Latin America. Ann Allergy Asthma
Immunol 2014;113:2829. PMID:25065979 https://doi.
org/10.1016/j.anai.2014.06.019
662. Macy E, Contreras R. Health care use and serious infection prevalence
associated with penicillin “allergy” in hospitalized patients: A cohort
study. J Allergy Clin Immunol 2014;133:790–6. PMID:24188976
https://doi.org/10.1016/j.jaci.2013.09.021
663. Blumenthal KG, Lu N, Zhang Y, Li Y, Walensky RP, Choi HK. Risk
of meticillin resistant Staphylococcus aureus and Clostridium difficile in
patients with a documented penicillin allergy: population based
matched cohort study. BMJ 2018;361:k2400. PMID:29950489
https://doi.org/10.1136/bmj.k2400
664. Blumenthal KG, Ryan EE, Li Y, Lee H, Kuhlen JL, Shenoy ES. The
impact of a reported penicillin allergy on surgical site infection risk.
Clin Infect Dis 2018;66:32936. PMID:29361015 https://doi.
org/10.1093/cid/cix794
665. Tucker MH, Lomas CM, Ramchandar N, Waldram JD. Amoxicillin
challenge without penicillin skin testing in evaluation of penicillin
allergy in a cohort of Marine recruits. J Allergy Clin Immunol Pract
2017;5:8135. PMID:28341170 https://doi.org/10.1016/j.
jaip.2017.01.023
666. Goldberg A, Confino-Cohen R. Skin testing and oral penicillin
challenge in patients with a history of remote penicillin allergy. Ann
Allergy Asthma Immunol 2008;100:3743. PMID:18254480 https://
doi.org/10.1016/S1081-1206(10)60402-4
667. Iammatteo M, Alvarez Arango S, Ferastraoaru D, et al. Safety and
outcomes of oral graded challenges to amoxicillin without prior skin
testing. J Allergy Clin Immunol Pract 2019;7:23643. PMID:29802906
https://doi.org/10.1016/j.jaip.2018.05.008
668. Cook DJ, Barbara DW, Singh KE, Dearani JA. Penicillin skin testing
in cardiac surgery. J Thorac Cardiovasc Surg 2014;147:19315.
PMID:24530197 https://doi.org/10.1016/j.jtcvs.2014.01.019
669. McDanel DL, Azar AE, Dowden AM, et al. Screening for beta-lactam
allergy in joint arthroplasty patients to improve surgical prophylaxis
practice. J Arthroplasty 2017;32(9s):S101–8. PMID:28236547 https://
doi.org/10.1016/j.arth.2017.01.012
670. Trubiano JA, Thursky KA, Stewardson AJ, et al. Impact of an integrated
antibiotic allergy testing program on antimicrobial stewardship: a
multicenter evaluation. Clin Infect Dis 2017;65:16674.
PMID:28520865 https://doi.org/10.1093/cid/cix244
671. Siew LQC, Li PH, Watts TJ, et al. Identifying low-risk beta-lactam
allergy patients in a UK tertiary centre. J Allergy Clin Immunol Pract
2019;7:21732181.e1. PMID:30922992 https://doi.org/10.1016/j.
jaip.2019.03.015
672. Chen JR, Tarver SA, Alvarez KS, Tran T, Khan DA. A proactive
approach to penicillin allergy testing in hospitalized patients. J Allergy
Clin Immunol Pract 2017;5:686–93. PMID:27888034 https://doi.
org/10.1016/j.jaip.2016.09.045
673. Leis JA, Palmay L, Ho G, et al. Point-of-care β-lactam allergy skin
testing by antimicrobial stewardship programs: a pragmatic multicenter
prospective evaluation. Clin Infect Dis 2017;65:105965.
PMID:28575226 https://doi.org/10.1093/cid/cix512
674. Banks TA, Tucker M, Macy E. Evaluating penicillin allergies without
skin testing. Curr Allergy Asthma Rep 2019;19:27. PMID:30903298
https://doi.org/10.1007/s11882-019-0854-6
675. Pham MN, Ho HE, Desai M. Penicillin desensitization: treatment of
syphilis in pregnancy in penicillin-allergic patients. Ann Allergy Asthma
Immunol 2017;118:53741. PMID:28477786 https://doi.
org/10.1016/j.anai.2017.03.013
676. Sogn DD, Evans R 3rd, Shepherd GM, et al. Results of the National
Institute of Allergy and Infectious Diseases Collaborative Clinical Trial
to test the predictive value of skin testing with major and minor
penicillin derivatives in hospitalized adults. Arch Intern Med
1992;152:102532. PMID:1580706 https://doi.org/10.1001/
archinte.1992.00400170105020
Recommendations and Reports
158 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
677. Solensky R, Jacobs J, Lester M, et al. Penicillin allergy evaluation: a
prospective, multicenter, open-label evaluation of a comprehensive
penicillin skin test kit. J Allergy Clin Immunol Pract 2019;7:187685.
e3. PMID:30878711 https://doi.org/10.1016/j.jaip.2019.02.040
678. Heil EL, Bork JT, Schmalzle SA, et al. Implementation of an infectious
disease fellow-managed penicillin allergy skin testing service. Open
Forum Infect Dis 2016;3:ofw155. PMID:27704011 https://doi.
org/10.1093/ofid/ofw155
679. du Plessis T, Walls G, Jordan A, Holland DJ. Implementation of a
pharmacist-led penicillin allergy de-labelling service in a public hospital.
J Antimicrob Chemother 2019;74:143846. PMID:30753497 https://
doi.org/10.1093/jac/dky575
680. Macy E, Blumenthal KG. Are cephalosporins safe for use in penicillin
allergy without prior allergy evaluation? J Allergy Clin Immunol Pract
2018;6:829. PMID:28958745 https://doi.org/10.1016/j.
jaip.2017.07.033
681. Zagursky RJ, Pichichero ME. Cross-reactivity in β-lactam allergy. J
Allergy Clin Immunol Pract 2018;6:7281.e1. PMID:29017833
https://doi.org/10.1016/j.jaip.2017.08.027
682. Blumenthal KG, Shenoy ES, Varughese CA, Hurwitz S, Hooper DC,
Banerji A. Impact of a clinical guideline for prescribing antibiotics to
inpatients reporting penicillin or cephalosporin allergy. Ann Allergy
Asthma Immunol 2015;115:294–300.e2. PMID:26070805 https://
doi.org/10.1016/j.anai.2015.05.011
683. Kuruvilla M, Wolf F, Sexton M, Wiley Z, Thomas J. Perioperative use
of cefazolin without preliminary skin testing in patients with reported
penicillin allergy. Surgery 2019;165:48696. PMID:30001827 https://
doi.org/10.1016/j.surg.2018.05.054
684. Lee P, Shanson D. Results of a UK survey of fatal anaphylaxis after oral
amoxicillin. J Antimicrob Chemother 2007;60:11723.
PMID:17761735 https://doi.org/10.1093/jac/dkm315
685. Blumenthal KG, Shenoy ES, Wolfson AR, et al. Addressing inpatient
beta-lactam allergies: a multihospital implementation. J Allergy Clin
Immunol Pract 2017;5:61625.e7. PMID:28483315 https://doi.
org/10.1016/j.jaip.2017.02.019
686. Mustafa SS, Conn K, Ramsey A. Comparing direct challenge to
penicillin skin testing for the outpatient evaluation of penicillin allergy:
a randomized controlled trial. J Allergy Clin Immunol Pract
2019;7:216370. PMID:31170542 https://doi.org/10.1016/j.
jaip.2019.05.037
687. Chastain DB, Hutzley VJ, Parekh J, Alegro JVG. Antimicrobial
desensitization: a review of published protocols. Pharmacy (Basel)
2019;7:112. PMID:31405062 https://doi.org/10.3390/
pharmacy7030112
688. Wendel GD Jr, Stark BJ, Jamison RB, Molina RD, Sullivan TJ.
Penicillin allergy and desensitization in serious infections during
pregnancy. N Engl J Med 1985;312:122932. PMID:3921835 https://
doi.org/10.1056/NEJM198505093121905
689. Borish L, Tamir R, Rosenwasser LJ. Intravenous desensitization to
beta-lactam antibiotics. J Allergy Clin Immunol 1987;80:3149.
PMID:3040836 https://doi.org/10.1016/0091-6749(87)90037-6
690. Legere HJ 3rd, Palis RI, Rodriguez Bouza T, Uluer AZ, Castells MC.
A safe protocol for rapid desensitization in patients with cystic fibrosis
and antibiotic hypersensitivity. J Cyst Fibros 2009;8:41824.
PMID:19740711 https://doi.org/10.1016/j.jcf.2009.08.002
691. Manhart LE, Holmes KK, Hughes JP, Houston LS, Totten PA.
Mycoplasma genitalium among young adults in the United States: an
emerging sexually transmitted infection. Am J Public Health
2007;97:111825. PMID:17463380 https://doi.org/10.2105/
AJPH.2005.074062
692. Ross JDC, Jensen JS. Mycoplasma genitalium as a sexually transmitted
infection: implications for screening, testing, and treatment. Sex Transm
Infect 2006;82:26971. PMID:16877571 https://doi.org/10.1136/
sti.2005.017368
693. Taylor-Robinson D, Gilroy CB, Thomas BJ, Hay PE.
Mycoplasma genitalium in chronic non-gonococcal urethritis.
Int J STD AIDS 2004;15:215. PMID:14769166 https://doi.
org/10.1258/095646204322637209
694. Dupin N, Bijaoui G, Schwarzinger M, et al. Detection and
quantification of Mycoplasma genitalium in male patients with
urethritis. Clin Infect Dis 2003;37:602–5. PMID:12905147 https://
doi.org/10.1086/376990
695. Krieger JN, Riley DE, Roberts MC, Berger RE. Prokaryotic DNA
sequences in patients with chronic idiopathic prostatitis. J Clin
Microbiol 1996;34:3120–8. PMID:8940458 https://doi.org/10.1128/
JCM.34.12.3120-3128.1996
696. le Roux MC, Hoosen AA. Quantitative real-time polymerase chain
reaction for the diagnosis of Mycoplasma genitalium infection in South
African men with and without symptoms of urethritis. Sex Transm
Dis 2017;44:1720. PMID:27898565 https://doi.org/10.1097/
OLQ.0000000000000540
697. Bachmann LH, Kirkcaldy RD, Geisler WM, et al.; the MAGNUM
Laboratory Working Group. Prevalence of Mycoplasma genitalium
infection, antimicrobial resistance mutations and symptom resolution
following treatment of urethritis. Clin Infect Dis 2020;71:e624–32.
PMID:32185385 https://doi.org/10.1093/cid/ciaa293
698. Nye MB, Schwebke JR, Body BA. Comparison of APTIMA
Trichomonas vaginalis transcription-mediated amplification to wet
mount microscopy, culture, and polymerase chain reaction for
diagnosis of trichomoniasis in men and women. Am J Obstet Gynecol
2009;200:188.e1–7. PMID:19185101 https://doi.org/10.1016/j.
ajog.2008.10.005
699. Bradshaw CS, Tabrizi SN, Read TR, et al. Etiologies of nongonococcal
urethritis: bacteria, viruses, and the association with orogenital
exposure. J Infect Dis 2006;193:336–45. PMID:16388480 https://
doi.org/10.1086/499434
700. Dombrowski JC, Harrington RD, Golden MR. Evidence for the long-
term stability of HIV transmission-associated sexual behavior after HIV
diagnosis. Sex Transm Dis 2013;40:41–5. PMID:23254116 https://
doi.org/10.1097/OLQ.0b013e3182753327
701. Rane VS, Fairley CK, Weerakoon A, et al. Characteristics of acute
nongonococcal urethritis in men differ by sexual preference.
J Clin Microbiol 2014;52:29716. PMID:24899041 https://doi.
org/10.1128/JCM.00899-14
702. Pond MJ, Nori AV, Witney AA, Lopeman RC, Butcher PD, Sadiq
ST. High prevalence of antibiotic-resistant Mycoplasma genitalium
in nongonococcal urethritis: the need for routine testing and
the inadequacy of current treatment options. Clin Infect Dis
2014;58:631–7. PMID:24280088 https://doi.org/10.1093/cid/cit752
703. Khatib N, Bradbury C, Chalker V, et al. Prevalence of Trichomonas
vaginalis, Mycoplasma genitalium and Ureaplasma urealyticum in
men with urethritis attending an urban sexual health clinic. Int
J STD AIDS 2015;26:38892. PMID:24925897 https://doi.
org/10.1177/0956462414539464
704. Cox C, McKenna JP, Watt AP, Coyle PV. Ureaplasma parvum and
Mycoplasma genitalium are found to be significantly associated with
microscopy-confirmed urethritis in a routine genitourinary medicine
setting. Int J STD AIDS 2016;27:861–7. PMID:26378187 https://
doi.org/10.1177/0956462415597620
705. Li Y, Su X, Le W, et al. Mycoplasma genitalium in symptomatic male
urethritis: macrolide use is associated with increased resistance.
Clin Infect Dis 2020;70:80510. PMID:30972419 https://doi.
org/10.1093/cid/ciz294
706. Ito S, Hanaoka N, Shimuta K, et al. Male non-gonococcal urethritis:
from microbiological etiologies to demographic and clinical features.
Int J Urol 2016;23:32531. PMID:26845624 https://doi.org/10.1111/
iju.13044
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 159
US Department of Health and Human Services/Centers for Disease Control and Prevention
707. Horner P, Donders G, Cusini M, Gomberg M, Jensen JS, Unemo M.
Should we be testing for urogenital Mycoplasma hominis, Ureaplasma
parvum and Ureaplasma urealyticum in men and women?—A position
statement from the European STI Guidelines Editorial Board. J Eur
Acad Dermatol Venereol 2018;32:184551. PMID:29924422 https://
doi.org/10.1111/jdv.15146
708. van der Veer C, van Rooijen MS, Himschoot M, de Vries HJ, Bruisten
SM. Trichomonas vaginalis and Mycoplasma genitalium: age-specific
prevalence and disease burden in men attending a sexually transmitted
infections clinic in Amsterdam, the Netherlands. Sex Transm Infect
2016;92:835. PMID:26283740 https://doi.org/10.1136/
sextrans-2015-052118
709. Seike K, Maeda S, Kubota Y, Tamaki M, Yasuda M, Deguchi T.
Prevalence and morbidity of urethral Trichomonas vaginalis in Japanese
men with or without urethritis. Sex Transm Infect 2013;89:528–30.
PMID:23349337 https://doi.org/10.1136/sextrans-2012-050702
710. Napierala M, Munson E, Wenten D, et al. Detection of Mycoplasma
genitalium from male primary urine specimens: an epidemiologic
dichotomy with Trichomonas vaginalis. Diagn Microbiol Infect Dis
2015;82:1948. PMID:25934156 https://doi.org/10.1016/j.
diagmicrobio.2015.03.016
711. Sviben M, Missoni EM, Meštrović T, Vojnović G, Galinović GM.
Epidemiology and laboratory characteristics of Trichomonas vaginalis
infection in Croatian men with and without urethritis syndrome: a
case-control study. Sex Transm Infect 2015;91:3604. PMID:25568091
https://doi.org/10.1136/sextrans-2014-051771
712. Rietmeijer CA, Mungati M, Machiha A, et al. The etiology of male
urethral discharge in Zimbabwe: results from the Zimbabwe STI
Etiology Study. Sex Transm Dis 2018;45:5660. PMID:29240635
https://doi.org/10.1097/OLQ.0000000000000696
713. Bazan JA, Peterson AS, Kirkcaldy RD, et al. Notes from the field:
increase in Neisseria meningitidis-associated urethritis among men at
two sentinel clinics—Columbus, Ohio, and Oakland County,
Michigan, 2015. MMWR Morb Mortal Wkly Rep 2016;65:550–2.
PMID:27254649 https://doi.org/10.15585/mmwr.mm6521a5
714. Jannic A, Mammeri H, Larcher L, et al. Orogenital transmission of
Neisseria meningitidis causing acute urethritis in men who have sex with
men. Emerg Infect Dis 2019;25:175–6. PMID:30561300 https://doi.
org/10.3201/eid2501.171102
715. Hayakawa K, Itoda I, Shimuta K, Takahashi H, Ohnishi M. Urethritis
caused by novel Neisseria meningitidis serogroup W in man who has
sex with men, Japan. Emerg Infect Dis 2014;20:15857.
PMID:25154021 https://doi.org/10.3201/eid2009.140349
716. Bazan JA, Tzeng YL, Stephens DS, et al. Repeat episodes of symptomatic
urethritis due to a uropathogenic meningococcal clade. Sex Transm
Dis 2020;47:e14. PMID:31651709 https://doi.org/10.1097/
OLQ.0000000000001079
717. Ong JJ, Morton AN, Henzell HR, et al. Clinical characteristics of
herpes simplex virus urethritis compared with chlamydial urethritis
among men. Sex Transm Dis 2017;44:121–5. PMID:28079748 https://
doi.org/10.1097/OLQ.0000000000000547
718. Avolio M, De Rosa R, Modolo ML, Stano P, Camporese A. When
should adenoviral non-gonococcal urethritis be suspected? Two case
reports. New Microbiol 2014;37:109–12. PMID:24531179
719. You C, Hamasuna R, Ogawa M, et al. The first report: an analysis of
bacterial flora of the first voided urine specimens of patients with male
urethritis using the 16S ribosomal RNA gene-based clone library
method. Microb Pathog 2016;95:95–100. PMID:27013259 https://
doi.org/10.1016/j.micpath.2016.02.022
720. Deguchi T, Ito S, Hatazaki K, et al. Antimicrobial susceptibility of
Haemophilus influenzae strains isolated from the urethra of men with
acute urethritis and/or epididymitis. J Infect Chemother 2017;23:804–7.
PMID:28619239 https://doi.org/10.1016/j.jiac.2017.05.009
721. Ito S, Hatazaki K, Shimuta K, et al. Haemophilus influenzae isolated
from men with acute urethritis: its pathogenic roles, responses to
antimicrobial chemotherapies, and antimicrobial susceptibilities. Sex
Transm Dis 2017;44:20510. PMID:28282645 https://doi.
org/10.1097/OLQ.0000000000000573
722. Horie K, Ito S, Hatazaki K, et al. Haemophilus quentini’ in the urethra
of men complaining of urethritis symptoms. J Infect Chemother
2018;24:714. PMID:28889986 https://doi.org/10.1016/j.
jiac.2017.08.007
723. Frølund M, Falk L, Ahrens P, Jensen JS. Detection of ureaplasmas and
bacterial vaginosis associated bacteria and their association with non-
gonococcal urethritis in men. PLoS One 2019;14:e0214425.
PMID:30946763 https://doi.org/10.1371/journal.pone.0214425
724. Deza G, Martin-Ezquerra G, Gómez J, Villar-García J, Supervia A,
Pujol RM. Isolation of Haemophilus influenzae and Haemophilus
parainfluenzae in urethral exudates from men with acute urethritis: a
descriptive study of 52 cases. Sex Transm Infect 2016;92:2931.
PMID:26139207 https://doi.org/10.1136/sextrans-2015-052135
725. Magdaleno-Tapial J, Valenzuela-Oñate C, Giacaman-von der Weth
MM, et al. Haemophilus species isolated in urethral exudates as a possible
causative agent in acute urethritis: a study of 38 cases. Actas
Dermosifiliogr 2019;110:3842. PMID:30390917 https://doi.
org/10.1016/j.adengl.2018.11.011
726. Abdolrasouli A, Roushan A. Corynebacterium propinquum associated
with acute, nongonococcal urethritis. Sex Transm Dis 2013;40:82931.
PMID:24275738 https://doi.org/10.1097/OLQ.0000000000000027
727. Ongrádi J, Stercz B, Kövesdi V, Nagy K, Chatlynne L. Isolation of
Kurthia gibsonii from non-gonorrheal urethritis: implications for the
pathomechanism upon surveying the literature. Acta Microbiol
Immunol Hung 2014;61:7987. PMID:24631755 https://doi.
org/10.1556/AMicr.61.2014.1.8
728. Gherardi G, Di Bonaventura G, Pompilio A, Savini V. Corynebacterium
glucuronolyticum causing genitourinary tract infection: case report and
review of the literature. IDCases 2015;2:56–8. PMID:26793456
https://doi.org/10.1016/j.idcr.2015.03.001
729. Meštrović T. A microbial game of whack-a-mole: clinical case series of
the urethral uncloaking phenomenon caused by Corynebacterium
glucuronolyticum in men treated for Chlamydia trachomatis urethritis.
Infection 2019;47:121–4. PMID:30168068 https://doi.org/10.1007/
s15010-018-1211-8
730. Frikh M, El Yaagoubi I, Lemnouer A, Elouennass M. Urethritis due
to corynebacterium striatum: an emerging germ. Tunis Med
2015;93:43–4. PMID:25955369
731. Babaeer AA, Nader C, Iacoviello V, Tomera K. Necrotizing urethritis
due to Aerococcus urinae. Case Rep Urol 2015;2015:136147.
PMID:26171271 https://doi.org/10.1155/2015/136147
732. Grandolfo M, Vestita M, Bonamonte D, Filoni A. Acute urethritis and
balonoposthitis associated to Neisseria elongata. Sex Transm Dis
2016;43:7789. PMID:27832027 https://doi.org/10.1097/
OLQ.0000000000000532
733. Frølund M, Lidbrink P, Wikström A, Cowan S, Ahrens P, Jensen JS.
Urethritis-associated pathogens in urine from men with non-gonococcal
urethritis: a case-control study. Acta Derm Venereol 2016;96:689–94.
PMID:26658669 https://doi.org/10.2340/00015555-2314
734. Chambers LC, Morgan JL, Lowens MS, et al. Cross-sectional study of
urethral exposures at last sexual episode associated with non-gonococcal
urethritis among STD clinic patients. Sex Transm Infect 2019;95:212–8.
PMID:30181326 https://doi.org/10.1136/sextrans-2018-053634
735. Manhart LE, Khosropour CM, Liu C, et al. Bacterial vaginosis-
associated bacteria in men: association of Leptotrichia/Sneathia spp.
with nongonococcal urethritis. Sex Transm Dis 2013;40:9449.
PMID:24220356 https://doi.org/10.1097/OLQ.0000000000000054
Recommendations and Reports
160 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
736. Ashraf J, Radford AR, Turner A, Subramaniam R. Preliminary
experience with instillation of triamcinolone acetonide into the urethra
for idiopathic urethritis: a prospective pilot study. J Laparoendosc Adv
Surg Tech A 2017;27:121721. PMID:29023188 https://doi.
org/10.1089/lap.2017.0064
737. Taylor SN, DiCarlo RP, Martin DH. Comparison of methylene blue/
gentian violet stain to Grams stain for the rapid diagnosis of gonococcal
urethritis in men. Sex Transm Dis 2011;38:995–6. PMID:21992973
https://doi.org/10.1097/OLQ.0b013e318225f7c2
738. Rietmeijer CA, Mettenbrink CJ. Recalibrating the Gram stain diagnosis
of male urethritis in the era of nucleic acid amplification testing. Sex
Transm Dis 2012;39:1820. PMID:22183839 https://doi.org/10.1097/
OLQ.0b013e3182354da3
739. Geisler WM, Yu S, Hook EW 3rd. Chlamydial and gonococcal infection
in men without polymorphonuclear leukocytes on Gram stain:
implications for diagnostic approach and management. Sex Transm
Dis 2005;32:630–4. PMID:16205305 https://doi.org/10.1097/01.
olq.0000175390.45315.a1
740. Gottesman T, Yossepowitch O, Samra Z, Rosenberg S, Dan M.
Prevalence of Mycoplasma genitalium in men with urethritis and in high
risk asymptomatic males in Tel Aviv: a prospective study. Int J STD
AIDS 2017;28:12732. PMID:26826161 https://doi.
org/10.1177/0956462416630675
741. Kim HJ, Park JK, Park SC, et al. The prevalence of causative organisms
of community-acquired urethritis in an age group at high risk for
sexually transmitted infections in Korean soldiers. J R Army Med Corps
2017;163:202. PMID:26607860 https://doi.org/10.1136/
jramc-2015-000488
742. Libois A, Hallin M, Crucitti T, Delforge M, De Wit S. Prevalence of
Mycoplasma genitalium in men with urethritis in a large public hospital
in Brussels, Belgium: an observational, cross-sectional study. PLoS One
2018;13:e0196217. PMID:29698421 https://doi.org/10.1371/journal.
pone.0196217
743. Bachmann LH, Manhart LE, Martin DH, et al. Advances in the
understanding and treatment of male urethritis. Clin Infect Dis
2015;61(Suppl 8):S763–9. PMID:26602615 https://doi.org/10.1093/
cid/civ755
744. Samaraweera GR, Garcia K, Druce J, et al. Characteristics of adenovirus
urethritis among heterosexual men and men who have sex with men:
a review of clinical cases. Sex Transm Infect 2016;92:1724.
PMID:26574571 https://doi.org/10.1136/sextrans-2015-052243
745. Horner P, Blee K, O’Mahony C, Muir P, Evans C, Radcliffe K; Clinical
Effectiveness Group of the British Association for Sexual Health and
HIV. 2015 UK National Guideline on the management of non-
gonococcal urethritis. Int J STD AIDS 2016;27:8596.
PMID:26002319 https://doi.org/10.1177/0956462415586675
746. Sarier M, Sepin N, Duman I, et al. Microscopy of Gram-stained urethral
smear in the diagnosis of urethritis: which threshold value should be
selected? Andrologia 2018;50:e13143. PMID:30238498 https://doi.
org/10.1111/and.13143
747. Sarier M, Kukul E. Classification of non-gonococcal urethritis: a review.
Int Urol Nephrol 2019;51:901–7. PMID:30953260 https://doi.
org/10.1007/s11255-019-02140-2
748. Kong FY, Tabrizi SN, Law M, et al. Azithromycin versus doxycycline
for the treatment of genital chlamydia infection: a meta-analysis of
randomized controlled trials. Clin Infect Dis 2014;59:193205.
PMID:24729507 https://doi.org/10.1093/cid/ciu220
749. Páez-Canro C, Alzate JP, González LM, Rubio-Romero JA, Lethaby
A, Gaitán HG. Antibiotics for treating urogenital Chlamydia trachomatis
infection in men and non-pregnant women. Cochrane Database Syst
Rev 2019;1:CD010871. PMID:30682211 https://doi.
org/10.1002/14651858.CD010871.pub2
750. McIver R, Jalocon D, McNulty A, et al. Men who have sex with men
with Mycoplasma genitalium-positive nongonococcal urethritis are more
likely to have macrolide resistant strains than men with only female
partners: a prospective study. Sex Transm Dis 2019;46:513–7.
PMID:31295218 https://doi.org/10.1097/OLQ.0000000000001009
751. Lau A, Bradshaw CS, Lewis D, et al. The efficacy of azithromycin for
the treatment of genital Mycoplasma genitalium: a systematic review
and meta-analysis. Clin Infect Dis 2015;61:138999. PMID:26240201
https://doi.org/10.1093/cid/civ644
752. Horner P. Mycoplasma genitalium nongonococcal urethritis is likely to
increase in men who have sex with men who practice unsafe sex: what
should we do? Sex Transm Dis 2019;46:518–20. PMID:31295219
https://doi.org/10.1097/OLQ.0000000000001030
753. Hosenfeld CB, Workowski KA, Berman S, et al. Repeat infection with
Chlamydia and gonorrhea among females: a systematic review of the
literature. Sex Transm Dis 2009;36:47889. PMID:19617871 https://
doi.org/10.1097/OLQ.0b013e3181a2a933
754. Fung M, Scott KC, Kent CK, Klausner JD. Chlamydial and gonococcal
reinfection among men: a systematic review of data to evaluate the
need for retesting. Sex Transm Infect 2007;83:304–9. PMID:17166889
https://doi.org/10.1136/sti.2006.024059
755. Kissinger PJ, White S, Manhart LE, et al. Azithromycin treatment
failure for Chlamydia trachomatis among heterosexual men with
nongonococcal urethritis. Sex Transm Dis 2016;43:599602.
PMID:27631353 https://doi.org/10.1097/OLQ.0000000000000489
756. Schwebke JR, Rompalo A, Taylor S, et al. Re-evaluating the treatment
of nongonococcal urethritis: emphasizing emerging pathogens—a
randomized clinical trial. Clin Infect Dis 2011;52:16370.
PMID:21288838 https://doi.org/10.1093/cid/ciq074
757. Manhart LE, Khosropour CM, Gillespie CW, Lowens MS, Golden
MR, Totten PA. O02.3 Treatment outcomes for persistent Mycoplasma
genitalium-associated NGU: evidence of moxifloxacin treatment
failures. Sex Transm Infect 2013;89(Suppl 1):A29. https://doi.
org/10.1136/sextrans-2013-051184.0091
758. Romano SS, Jensen JS, Lowens MS, et al. Long duration of
asymptomatic Mycoplasma genitalium infection after syndromic
treatment for nongonococcal urethritis. Clin Infect Dis 2019;69:113–20.
PMID:30281079 https://doi.org/10.1093/cid/ciy843
759. Read TRH, Fairley CK, Murray GL, et al. Outcomes of resistance-
guided sequential treatment of Mycoplasma genitalium infections: a
prospective evaluation. Clin Infect Dis 2019;68:55460.
PMID:29873691 https://doi.org/10.1093/cid/ciy477
760. Dowe G, Smikle M, King SD, Baum M, Chout R, Williams Y.
Symptomatic and asymptomatic chlamydial non-gonococcal urethritis
in Jamaica: the potential for HIV transmission. Int J STD AIDS
2000; 11: 187 90. PMID:10726944 https://doi.
org/10.1258/0956462001915507
761. Lusk MJ, Garden FL, Rawlinson WD, Naing ZW, Cumming RG,
Konecny P. Cervicitis aetiology and case definition: a study in Australian
women attending sexually transmitted infection clinics. Sex Transm
Infect 2016;92:17581. PMID:26586777 https://doi.org/10.1136/
sextrans-2015-052332
762. Lusk MJ, Konecny P. Cervicitis: a review. Curr Opin Infect Dis
2008;21:49–55. PMID:18192786
763. Marrazzo JM, Martin DH. Management of women with cervicitis.
Clin Infect Dis 2007;44(Suppl 3):S10210. PMID:17342663 https://
doi.org/10.1086/511423
764. Manavi K, Young H, Clutterbuck D. Sensitivity of microscopy for the
rapid diagnosis of gonorrhoea in men and women and the role of
gonorrhoea serovars. Int J STD AIDS 2003;14:390–4. PMID:12816666
https://doi.org/10.1258/095646203765371277
765. Lillis RA, Martin DH, Nsuami MJ. Mycoplasma genitalium infections
in women attending a sexually transmitted disease clinic in New
Orleans. Clin Infect Dis 2019;69:459–65. PMID:30351348 https://
doi.org/10.1093/cid/ciy922
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 161
US Department of Health and Human Services/Centers for Disease Control and Prevention
766. Lis R, Rowhani-Rahbar A, Manhart LE. Mycoplasma genitalium
infection and female reproductive tract disease: a meta-analysis. Clin
Infect Dis 2015;61:41826. PMID:25900174 https://doi.org/10.1093/
cid/civ312
767. Oliphant J, Azariah S. Cervicitis: limited clinical utility for the detection
of Mycoplasma genitalium in a cross-sectional study of women attending
a New Zealand sexual health clinic. Sex Health 2013;10:263–7.
PMID:23702105 https://doi.org/10.1071/SH12168
768. Sethi S, Rajkumari N, Dhaliwal L, Roy A. P3.294 Association of
Mycoplasma genitalium with cervicitis in North Indian women attending
gynecologic clinics. Sex Transm Infect 2013;89(Suppl 1):A240–1.
https://doi.org/10.1136/sextrans-2013-051184.0749
769. Taylor SN, Lensing S, Schwebke J, et al. Prevalence and treatment
outcome of cervicitis of unknown etiology. Sex Transm Dis
2013;40:37985. PMID:23588127 https://doi.org/10.1097/
OLQ.0b013e31828bfcb1
770. Mobley VL, Hobbs MM, Lau K, Weinbaum BS, Getman DK, Seña
AC. Mycoplasma genitalium infection in women attending a sexually
transmitted infection clinic: diagnostic specimen type, coinfections,
and predictors. Sex Transm Dis 2012;39:706–9. PMID:22902666
https://doi.org/10.1097/OLQ.0b013e318255de03
771. Marrazzo JM, Wiesenfeld HC, Murray PJ, et al. Risk factors for
cervicitis among women with bacterial vaginosis. J Infect Dis
2006;193:61724. PMID:16453256 https://doi.org/10.1086/500149
772. Gaydos C, Maldeis NE, Hardick A, Hardick J, Quinn TC. Mycoplasma
genitalium as a contributor to the multiple etiologies of cervicitis in
women attending sexually transmitted disease clinics. Sex Transm Dis
2009;36:598606. PMID:19704398 https://doi.org/10.1097/
OLQ.0b013e3181b01948
773. Clark LR, Atendido M. Group B streptococcal vaginitis in postpubertal
adolescent girls. J Adolesc Health 2005;36:437–40. PMID:15837348
https://doi.org/10.1016/j.jadohealth.2004.03.009
774. Hester EE, Middleman AB. A clinical conundrum: chronic cervicitis.
J Pediatr Adolesc Gynecol 2019;32:342–4. PMID:30582974 https://
doi.org/10.1016/j.jpag.2018.12.004
775. Liu L, Cao G, Zhao Z, Zhao F, Huang Y. High bacterial loads of
Ureaplasma may be associated with non-specific cervicitis. Scand J
Infect Dis 2014;46:637–41. PMID:25017795 https://doi.org/10.31
09/00365548.2014.922696
776. Leli C, Mencacci A, Latino MA, et al. Prevalence of cervical colonization
by Ureaplasma parvum, Ureaplasma urealyticum, Mycoplasma hominis
and Mycoplasma genitalium in childbearing age women by a
commercially available multiplex real-time PCR: an Italian observational
multicentre study. J Microbiol Immunol Infect 2018;51:2205.
PMID:28711440 https://doi.org/10.1016/j.jmii.2017.05.004
777. Manhart LE. Has the time come to systematically test for Mycoplasma
genitalium? Sex Transm Dis 2009;36:607–8. PMID:19734818 https://
doi.org/10.1097/OLQ.0b013e3181b9d825
778. Liu HL, Chen CM, Pai LW, Hwu YJ, Lee HM, Chung YC.
Comorbidity profiles among women with postcoital bleeding: a
nationwide health insurance database. Arch Gynecol Obstet
2017;295:93541. PMID:28246983 https://doi.org/10.1007/
s00404-017-4327-7
779. Coleman JS, Hitti J, Bukusi EA, et al. Infectious correlates of HIV-1
shedding in the female upper and lower genital tracts. AIDS
2007;21:7559. PMID:17413697 https://doi.org/10.1097/
QAD.0b013e328012b838
780. Johnson LF, Lewis DA. The effect of genital tract infections on HIV-1
shedding in the genital tract: a systematic review and meta-analysis.
Sex Transm Dis 2008;35:94659. PMID:18685546 https://doi.
org/10.1097/OLQ.0b013e3181812d15
781. McClelland RS, Wang CC, Mandaliya K, et al. Treatment
of cervicitis is associated with decreased cervical shedding of
HIV-1. AIDS 2001;15:10510. PMID:11192850 https://doi.
org/10.1097/00002030-200101050-00015
782. Gatski M, Martin DH, Theall K, et al. Mycoplasma genitalium infection
among HIV-positive women: prevalence, risk factors and association
with vaginal shedding. Int J STD AIDS 2011;22:1559.
PMID:21464453 https://doi.org/10.1258/ijsa.2010.010320
783. Gitau RW, Graham SM, Masese LN, et al. Effect of acquisition and
treatment of cervical infections on HIV-1 shedding in women on
antiretroviral therapy. AIDS 2010;24:27337. PMID:20871388
https://doi.org/10.1097/QAD.0b013e32833f9f43
784. Kreisel KM, Weston EJ, St Cyr SB, Spicknall IH. Estimates of the
prevalence and incidence of chlamydia and gonorrhea among US men
and women, 2018. Sex Transm Dis 2021;48:22231. PMID:33492094
https://doi.org/10.1097/OLQ.0000000000001382
785. Aghaizu A, Reid F, Kerry S, et al. Frequency and risk factors for incident
and redetected Chlamydia trachomatis infection in sexually active,
young, multi-ethnic women: a community based cohort study. Sex
Transm Infect 2014;90:5248. PMID:25100744 https://doi.
org/10.1136/sextrans-2014-051607
786. Scholes D, Satterwhite CL, Yu O, Fine D, Weinstock H, Berman S.
Long-term trends in Chlamydia trachomatis infections and related
outcomes in a U.S. managed care population. Sex Transm Dis
2012;39:818. PMID:22249294 https://doi.org/10.1097/
OLQ.0b013e31823e3009
787. Kamwendo F, Forslin L, Bodin L, Danielsson D. Decreasing incidences
of gonorrhea- and chlamydia-associated acute pelvic inflammatory
disease. A 25-year study from an urban area of central Sweden. Sex
Transm Dis 1996;23:38491. PMID:8885069 https://doi.
org/10.1097/00007435-199609000-00007
788. Rietmeijer CA, Hopkins E, Geisler WM, Orr DP, Kent CK. Chlamydia
trachomatis positivity rates among men tested in selected venues in the
United States: a review of the recent literature. Sex Transm Dis
2008;35(Suppl):S818. PMID:18449072 https://doi.org/10.1097/
OLQ.0b013e31816938ba
789. Gift TL, Blake DR, Gaydos CA, Marrazzo JM. The cost-effectiveness
of screening men for Chlamydia trachomatis: a review of the literature.
Sex Transm Dis 2008;35(Suppl):S51–60. PMID:18520977 https://
doi.org/10.1097/OLQ.0b013e3181723dba
790. Gift TL, Gaydos CA, Kent CK, et al. The program cost and cost-
effectiveness of screening men for Chlamydia to prevent pelvic
inflammatory disease in women. Sex Transm Dis 2008;35(Suppl):S6675.
PMID:18830137 https://doi.org/10.1097/OLQ.0b013e31818b64ac
791. Gopalappa C, Huang YL, Gift TL, Owusu-Edusei K, Taylor M, Gales
V. Cost-effectiveness of screening men in Maricopa County jails for
chlamydia and gonorrhea to avert infections in women. Sex Transm
Dis 2013;40:77683. PMID:24275727 https://doi.org/10.1097/
OLQ.0000000000000023
792. Masek BJ, Arora N, Quinn N, et al. Performance of three nucleic acid
amplification tests for detection of Chlamydia trachomatis and Neisseria
gonorrhoeae by use of self-collected vaginal swabs obtained via an
Internet-based screening program. J Clin Microbiol 2009;47:1663–7.
PMID:19386838 https://doi.org/10.1128/JCM.02387-08
793. Knox J, Tabrizi SN, Miller P, et al. Evaluation of self-collected samples
in contrast to practitioner-collected samples for detection of Chlamydia
trachomatis, Neisseria gonorrhoeae, and Trichomonas vaginalis by
polymerase chain reaction among women living in remote areas. Sex
Transm Dis 2002;29:64754. PMID:12438900 https://doi.
org/10.1097/00007435-200211000-00006
794. Schachter J, Chernesky MA, Willis DE, et al. Vaginal swabs are
the specimens of choice when screening for Chlamydia trachomatis
and Neisseria gonorrhoeae: results from a multicenter evaluation
of the APTIMA assays for both infections. Sex Transm Dis
2005;32:7258. PMID:16314767 https://doi.org/10.1097/01.
olq.0000190092.59482.96
795. Doshi JS, Power J, Allen E. Acceptability of chlamydia screening using
self-taken vaginal swabs. Int J STD AIDS 2008;19:5079.
PMID:18663033 https://doi.org/10.1258/ijsa.2008.008056
Recommendations and Reports
162 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
796. Chernesky MA, Jang D, Portillo E, et al. Self-collected swabs of the
urinary meatus diagnose more Chlamydia trachomatis and Neisseria
gonorrhoeae infections than first catch urine from men. Sex Transm
Infect 2013;89:102–4. PMID:23024224 https://doi.org/10.1136/
sextrans-2012-050573
797. Dize L, Barnes P Jr, Barnes M, et al. Performance of self-collected penile-
meatal swabs compared to clinician-collected urethral swabs for the
detection of Chlamydia trachomatis, Neisseria gonorrhoeae, Trichomonas
vaginalis, and Mycoplasma genitalium by nucleic acid amplification
assays. Diagn Microbiol Infect Dis 2016;86:131–5. PMID:27497595
https://doi.org/10.1016/j.diagmicrobio.2016.07.018
798. Berry L, Stanley B. Comparison of self-collected meatal swabs with
urine specimens for the diagnosis of Chlamydia trachomatis and
Neisseria gonorrhoeae in men. J Med Microbiol 2017;66:1346.
PMID:28068218 https://doi.org/10.1099/jmm.0.000428
799. Chernesky M, Freund GG, Hook E 3rd, Leone P, D’Ascoli P, Martens
M. Detection of Chlamydia trachomatis and Neisseria gonorrhoeae
infections in North American women by testing SurePath liquid-based
Pap specimens in APTIMA assays. J Clin Microbiol 2007;45:2434–8.
PMID:17581931 https://doi.org/10.1128/JCM.00013-07
800. Schachter J, Moncada J, Liska S, Shayevich C, Klausner JD. Nucleic
acid amplification tests in the diagnosis of chlamydial and gonococcal
infections of the oropharynx and rectum in men who have sex with
men. Sex Transm Dis 2008;35:637–42. PMID:18520976 https://doi.
org/10.1097/OLQ.0b013e31817bdd7e
801. Mimiaga MJ, Mayer KH, Reisner SL, et al. Asymptomatic gonorrhea
and chlamydial infections detected by nucleic acid amplification
tests among Boston area men who have sex with men. Sex Transm
Dis 2008;35:4958. PMID:18354345 https://doi.org/10.1097/
OLQ.0b013e31816471ae
802. Bachmann LH, Johnson RE, Cheng H, Markowitz LE, Papp JR, Hook
EW 3rd. Nucleic acid amplification tests for diagnosis of Neisseria
gonorrhoeae oropharyngeal infections. J Clin Microbiol 2009;47:902–7.
PMID:19193848 https://doi.org/10.1128/JCM.01581-08
803. Bachmann LH, Johnson RE, Cheng H, et al. Nucleic acid amplification
tests for diagnosis of Neisseria gonorrhoeae and Chlamydia trachomatis
rectal infections. J Clin Microbiol 2010;48:182732. PMID:20335410
https://doi.org/10.1128/JCM.02398-09
804. Cosentino LA, Danby CS, Rabe LK, et al. Use of nucleic acid
amplification testing for diagnosis of extragenital sexually transmitted
infections. J Clin Microbiol 2017;55:28017. PMID:28679521
https://doi.org/10.1128/JCM.00616-17
805. Food and Drug Administration. Microbiology Devices Panel of
the Medical Devices Advisory Committee meeting annoucement
[Internet]. Silver Spring, MD: US Department of Agriculture,
Food and Drug Administration; 2019. https://www.fda.gov/
advisory-committees/advisory-committee-calendar/march-8-2019-
microbiology-devices-panel-medical-devices-advisory-committee-
meeting-announcement#event-materials
806. Sexton ME, Baker JJ, Nakagawa K, et al. How reliable is self-testing for
gonorrhea and chlamydia among men who have sex with men? J Fam
Pract 2013;62:70–8. PMID:23405376
807. Herbst de Cortina S, Bristow CC, Joseph Davey D, Klausner JD. A
systematic review of point of care testing for Chlamydia trachomatis,
Neisseria gonorrhoeae, and Trichomonas vaginalis. Infect Dis Obstet
Gynecol 2016;2016:4386127. Epub May 26, 2016. PMID:27313440
https://doi.org/10.1155/2016/4386127
808. Rivard KR, Dumkow LE, Draper HM, Brandt KL, Whalen DW,
Egwuatu NE. Impact of rapid diagnostic testing for chlamydia
and gonorrhea on appropriate antimicrobial utilization in the
emergency department. Diagn Microbiol Infect Dis 2017;87:175–9.
PMID:27836225 https://doi.org/10.1016/j.diagmicrobio.2016.10.019
809. Wingrove I, McOwan A, Nwokolo N, Whitlock G. Diagnostics within
the clinic to test for gonorrhoea and chlamydia reduces the time to
treatment: a service evaluation. Sex Transm Infect 2014;90:474.
PMID:25118322 https://doi.org/10.1136/sextrans-2014-051580
810. Geisler WM, Wang C, Morrison SG, Black CM, Bandea CI, Hook EW
3rd. The natural history of untreated Chlamydia trachomatis infection in
the interval between screening and returning for treatment. Sex Transm
Dis 2008;35:11923. PMID:17898680 https://doi.org/10.1097/
OLQ.0b013e318151497d
811. Dukers-Muijrers NHTM, Wolffs PFG, De Vries H, et al. Treatment
effectiveness of azithromycin and doxycycline in uncomplicated rectal
and vaginal Chlamydia trachomatis infections in women: a multicentre
observational study (FemCure). Clin Infect Dis 2019;69:194654.
PMID:30689759 https://doi.org/10.1093/cid/ciz050
812. Dombrowski JC, Wierzbicki MR, Newman LM, et al. Doxycycline
versus azithromycin for the treatment of rectal chlamydia in men who
have sex with men: a randomized controlled trial. Clin Infect Dis
2021;ciab153. PMID:33606009 https://doi.org/10.1093/cid/ciab153
813. Dukers-Muijrers NH, Schachter J, van Liere GA, Wolffs PF, Hoebe CJ.
What is needed to guide testing for anorectal and pharyngeal Chlamydia
trachomatis and Neisseria gonorrhoeae in women and men? Evidence and
opinion. BMC Infect Dis 2015;15:533. PMID:26576538 https://doi.
org/10.1186/s12879-015-1280-6
814. Marcus JL, Kohn RP, Barry PM, Philip SS, Bernstein KT. Chlamydia
trachomatis and Neisseria gonorrhoeae transmission from the female
oropharynx to the male urethra. Sex Transm Dis 2011;38:372–3.
PMID:21183864 https://doi.org/10.1097/OLQ.0b013e3182029008
815. Manavi K, Hettiarachchi N, Hodson J. Comparison of doxycycline
with azithromycin in treatment of pharyngeal chlamydia infection.
Int J STD AIDS 2016;27:1303–8. PMID:26511655 https://doi.
org/10.1177/0956462415614723
816. Rank RG, Yeruva L. An alternative scenario to explain rectal positivity
in Chlamydia-infected individuals. Clin Infect Dis 2015;60:1585–6.
PMID:25648236 https://doi.org/10.1093/cid/civ079
817. Geisler WM, Koltun WD, Abdelsayed N, et al. Safety and efficacy
of WC2031 versus vibramycin for the treatment of uncomplicated
urogenital Chlamydia trachomatis infection: a randomized, double-blind,
double-dummy, active-controlled, multicenter trial. Clin Infect Dis
2012;55:82–8. PMID:22431798 https://doi.org/10.1093/cid/cis291
818. Renault CA, Israelski DM, Levy V, Fujikawa BK, Kellogg TA, Klausner
JD. Time to clearance of Chlamydia trachomatis ribosomal RNA in
women treated for chlamydial infection. Sex Health 2011;8:69–73.
PMID:21371385 https://doi.org/10.1071/SH10030
819. Lazenby GB, Korte JE, Tillman S, Brown FK, Soper DE. A
recommendation for timing of repeat Chlamydia trachomatis test
following infection and treatment in pregnant and nonpregnant
women. Int J STD AIDS 2017;28:902–9. PMID:27864473 https://
doi.org/10.1177/0956462416680438
820. Dunne EF, Chapin JB, Rietmeijer CA, et al. Rate and predictors of
repeat Chlamydia trachomatis infection among men. Sex Transm Dis
2008;35(Suppl):S40–4. PMID:18520978 https://doi.org/10.1097/
OLQ.0b013e31817247b2
821. Kjaer HO, Dimcevski G, Hoff G, Olesen F, Ostergaard L. Recurrence of
urogenital Chlamydia trachomatis infection evaluated by mailed samples
obtained at home: 24 weeks’ prospective follow up study. Sex Transm
Infect 2000;76:16972. PMID:10961191 https://doi.org/10.1136/
sti.76.3.169
822. Whittington WL, Kent C, Kissinger P, et al. Determinants
of persistent and recurrent Chlamydia trachomatis infection
in young women: results of a multicenter cohort study. Sex
Transm Dis 2001;28:11723. PMID:11234786 https://doi.
org/10.1097/00007435-200102000-00011
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 163
US Department of Health and Human Services/Centers for Disease Control and Prevention
823. Kapil R, Press CG, Hwang ML, Brown L, Geisler WM. Investigating
the epidemiology of repeat Chlamydia trachomatis detection after
treatment by using C. trachomatis OmpA genotyping. J Clin Microbiol
2015;53:5469. PMID:25472488 https://doi.org/10.1128/
JCM.02483-14
824. Jacobson GF, Autry AM, Kirby RS, Liverman EM, Motley RU. A
randomized controlled trial comparing amoxicillin and azithromycin
for the treatment of Chlamydia trachomatis in pregnancy. Am J Obstet
Gynecol 2001;184:1352–6. PMID:11408852 https://doi.org/10.1067/
mob.2001.115050
825. Kacmar J, Cheh E, Montagno A, Peipert JF. A randomized trial of
azithromycin versus amoxicillin for the treatment of Chlamydia
trachomatis in pregnancy. Infect Dis Obstet Gynecol 2001;9:197202.
PMID:11916175 https://doi.org/10.1155/S1064744901000321
826. Rahangdale L, Guerry S, Bauer HM, et al. An observational cohort
study of Chlamydia trachomatis treatment in pregnancy. Sex Transm
Dis 2006;33:106–10. PMID:16432482 https://doi.org/10.1097/01.
olq.0000187226.32145.ea
827. Aggarwal A, Spitzer RF, Caccia N, Stephens D, Johnstone J, Allen L.
Repeat screening for sexually transmitted infection in adolescent
obstetric patients. J Obstet Gynaecol Can 2010;32:95661.
PMID:21176304 https://doi.org/10.1016/S1701-2163(16)34683-7
828. Phillips Campbell R, Kintner J, Whittimore J, Schoborg RV. Chlamydia
muridarum enters a viable but non-infectious state in amoxicillin-
treated BALB/c mice. Microbes Infect 2012;14:117785.
PMID:22943883 https://doi.org/10.1016/j.micinf.2012.07.017
829. Wyrick PB. Chlamydia trachomatis persistence in vitro: an overview.
J Infect Dis 2010;201(Suppl 2):S8895. PMID:20470046 https://doi.
org/10.1086/652394
830. Fan H, Li L, Wijlaars L, Gilbert RE. Associations between use of
macrolide antibiotics during pregnancy and adverse child outcomes: a
systematic review and meta-analysis. PLoS One 2019;14:e0212212.
PMID:30779772 https://doi.org/10.1371/journal.pone.0212212
831. Fan H, Gilbert R, O’Callaghan F, Li L. Associations between macrolide
antibiotics prescribing during pregnancy and adverse child outcomes
in the UK: population based cohort study. BMJ 2020;368:m331.
Erratum in: BMJ 2020;368:m766. PMID:32075790 https://doi.
org/10.1136/bmj.m331
832. Mallah N, Tohidinik HR, Etminan M, Figueiras A, Takkouche B.
Prenatal exposure to macrolides and risk of congenital malformations:
a meta-analysis. Drug Saf 2020;43:211–21. PMID:31721138 https://
doi.org/10.1007/s40264-019-00884-5
833. Hammerschlag MR, Cummings C, Roblin PM, Williams TH, Delke
I. Efficacy of neonatal ocular prophylaxis for the prevention of
chlamydial and gonococcal conjunctivitis. N Engl J Med
1989;320:769–72. PMID:2922026 https://doi.org/10.1056/
NEJM198903233201204
834. Zikic A, Schünemann H, Wi T, Lincetto O, Broutet N, Santesso N.
Treatment of neonatal chlamydial conjunctivitis: a systematic review
and meta-analysis. J Pediatric Infect Dis Soc 2018;7:e10715.
PMID:30007329 https://doi.org/10.1093/jpids/piy060
835. Hammerschlag MR, Chandler JW, Alexander ER, English M, Koutsky
L. Longitudinal studies on chlamydial infections in the first year of
life. Pediatr Infect Dis 1982;1:395–401. PMID:7163029 https://doi.
org/10.1097/00006454-198211000-00007
836. Beem MO, Saxon E, Tipple MA. Treatment of chlamydial pneumonia
of infancy. Pediatrics 1979;63:198–203. PMID:440807
837. Brownell AD, Shapiro RA, Hammerschlag MR. Caution is required
when using non-Food and Drug Administration-cleared assays to
diagnose sexually transmitted infections in children. J Pediatr
2019;206:2802. PMID:30466791 https://doi.org/10.1016/j.
jpeds.2018.10.038
838. Kreisel KM, Spicknall IH, Gargano JW, et al. Sexually transmitted
infections among US women and men: prevalence and incidence
estimates, 2018. Sex Transm Dis 2021;48:208–14. PMID:33492089
https://doi.org/10.1097/OLQ.0000000000001355
839. Lunny C, Taylor D, Hoang L, et al. Self-collected versus clinician-
collected sampling for chlamydia and gonorrhea screening: a systemic
review and meta-analysis. PLoS One 2015;10:e0132776.
PMID:26168051 https://doi.org/10.1371/journal.pone.0132776
840. Schick V, Van Der Pol B, Dodge B, Baldwin A, Fortenberry JD. A
mixed methods approach to assess the likelihood of testing for STI
using self-collected samples among behaviourally bisexual women. Sex
Transm Infect 2015;91:32933. PMID:25637328 https://doi.
org/10.1136/sextrans-2014-051842
841. Mustanski B, Feinstein BA, Madkins K, Sullivan P, Swann G. Prevalence
and risk factors for rectal and urethral sexually transmitted infections
from self-collected samples among young men who have sex with men
participating in the Keep It Up! 2.0 randomized controlled trial. Sex
Transm Dis 2017;44:4838. PMID:28703727 https://doi.org/10.1097/
OLQ.0000000000000636
842. Salow KR, Cohen AC, Bristow CC, McGrath MR, Klausner JD.
Comparing mail-in self-collected specimens sent via United States
Postal Service versus clinic-collected specimens for the detection
of Chlamydia trachomatis and Neisseria gonorrhoeae in extra-genital
sites. PLoS One 2017;12:e0189515. PMID:29240781 https://doi.
org/10.1371/journal.pone.0189515
843. Drake C, Barenfanger J, Lawhorn J, Verhulst S. Comparison of
Easy-Flow Copan Liquid Stuart’s and Starplex Swab transport
systems for recovery of fastidious aerobic bacteria. J Clin Microbiol
2005;43:1301–3. PMID:15750099 https://doi.org/10.1128/
JCM.43.3.1301-1303.2005
844. Wade JJ, Graver MA. Survival of six auxotypes of Neisseria gonorrhoeae in
transport media. J Clin Microbiol 2003;41:17201. PMID:12682168
https://doi.org/10.1128/JCM.41.4.1720-1721.2003
845. Arbique JC, Forward KR, LeBlanc J. Evaluation of four commercial
transport media for the survival of Neisseria gonorrhoeae. Diagn
Microbiol Infect Dis 2000;36:163–8. PMID:10729658 https://doi.
org/10.1016/S0732-8893(99)00134-0
846. Hook EW 3rd, Kirkcaldy RD. A brief history of evolving diagnostics and
therapy for gonorrhea: lessons learned. Clin Infect Dis 2018;67:1294–9.
PMID:29659749 https://doi.org/10.1093/cid/ciy271
847. Unemo M, Shafer WM. Future treatment of gonorrhea—novel
emerging drugs are essential and in progress? Expert Opin Emerg Drugs
2015;20:357–60. PMID:25907334 https://doi.org/10.1517/147282
14.2015.1039981
848. Sánchez-Busó L, Golparian D, Corander J, et al. The impact of
antimicrobials on gonococcal evolution. Nat Microbiol 2019;4:1941–50.
PMID:31358980 https://doi.org/10.1038/s41564-019-0501-y
849. Schwarcz SK, Zenilman JM, Schnell D, et al. National surveillance
of antimicrobial resistance in Neisseria gonorrhoeae. The Gonococcal
Isolate Surveillance Project. JAMA 1990;264:1413–7. PMID:2144026
https://doi.org/10.1001/jama.1990.03450110059027
850. CDC. Update to CDC’s sexually transmitted diseases treatment
guidelines, 2006: fluoroquinolones no longer recommended for
treatment of gonococcal infections. MMWR Morb Mortal Wkly Rep
2007;56:332–6. PMID:17431378
851. CDC. Sexually transmitted disease surveillance 2013. Atlanta, GA: US
Department of Health and Human Services, CDC; 2014. https://www.
cdc.gov/std/stats/archive/Surv2013-Print.pdf
852. Muratani T, Akasaka S, Kobayashi T, et al. Outbreak of
cefozopran (penicillin, oral cephems, and aztreonam)-resistant
Neisseria gonorrhoeae in Japan. Antimicrob Agents Chemother
2001;45:36036. PMID:11709349 https://doi.org/10.1128/
AAC.45.12.3603-3606.2001
853. Yokoi S, Deguchi T, Ozawa T, et al. Threat to cefixime treatment for
gonorrhea. Emerg Infect Dis 2007;13:1275–7. PMID:17953118
Recommendations and Reports
164 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
854. Lo JY, Ho KM, Leung AO, et al. Ceftibuten resistance and treatment
failure of Neisseria gonorrhoeae infection. Antimicrob Agents Chemother
2008;52:35647. PMID:18663018 https://doi.org/10.1128/
AAC.00198-08
855. Deguchi T, Yasuda M, Yokoi S, et al. Treatment of uncomplicated
gonococcal urethritis by double-dosing of 200 mg cefixime at a 6-h
interval. J Infect Chemother 2003;9:35–9. PMID:12673405 https://
doi.org/10.1007/s10156-002-0204-8
856. Unemo M, Golparian D, Hestner A. Ceftriaxone treatment failure of
pharyngeal gonorrhoea verified by international recommendations,
Sweden, July 2010. Euro Surveill 2011;16:19792. PMID:21329645
857. Unemo M, Golparian D, Syversen G, Vestrheim DF, Moi H. Two cases
of verified clinical failures using internationally recommended first-line
cefixime for gonorrhoea treatment, Norway, 2010. Euro Surveill
2010;15:19721. PMID:21144442 https://doi.org/10.2807/
ese.15.47.19721-en
858. Unemo M, Golparian D, Potočnik M, Jeverica S. Treatment failure of
pharyngeal gonorrhoea with internationally recommended first-line
ceftriaxone verified in Slovenia, September 2011. Euro Surveill
2012;17:20200. PMID:22748003
859. Ison CA, Hussey J, Sankar KN, Evans J, Alexander S. Gonorrhoea
treatment failures to cefixime and azithromycin in England, 2010.
Euro Surveill 2011;16:19833. PMID:21492528
860. Forsyth S, Penney P, Rooney G. Cefixime-resistant Neisseria gonorrhoeae
in the UK: a time to reflect on practice and recommendations. Int J
STD AIDS 2011;22:2967. PMID:21571983 https://doi.org/10.1258/
ijsa.2009.009191
861. Lewis DA, Sriruttan C, Müller EE, et al. Phenotypic and genetic
characterization of the first two cases of extended-spectrum-
cephalosporin-resistant Neisseria gonorrhoeae infection in South Africa
and association with cefixime treatment failure. J Antimicrob
Chemother 2013;68:126770. PMID:23416957 https://doi.
org/10.1093/jac/dkt034
862. Ota KV, Fisman DN, Tamari IE, et al. Incidence and treatment
outcomes of pharyngeal Neisseria gonorrhoeae and Chlamydia
trachomatis infections in men who have sex with men: a 13-year
retrospective cohort study. Clin Infect Dis 2009;48:123743.
PMID:19323630 https://doi.org/10.1086/597586
863. Allen VG, Mitterni L, Seah C, et al. Neisseria gonorrhoeae treatment
failure and susceptibility to cefixime in Toronto, Canada. JAMA
2013;309:16370. PMID:23299608 https://doi.org/10.1001/
jama.2012.176575
864. Chen MY, Stevens K, Tideman R, et al. Failure of 500 mg of ceftriaxone
to eradicate pharyngeal gonorrhoea, Australia. J Antimicrob Chemother
2013;68:14457. PMID:23390207 https://doi.org/10.1093/jac/
dkt017
865. Tapsall J, Read P, Carmody C, et al. Two cases of failed ceftriaxone
treatment in pharyngeal gonorrhoea verified by molecular
microbiological methods. J Med Microbiol 2009;58:6837.
PMID:19369534 https://doi.org/10.1099/jmm.0.007641-0
866. Ohnishi M, Saika T, Hoshina S, et al. Ceftriaxone-resistant Neisseria
gonorrhoeae, Japan. Emerg Infect Dis 2011;17:1489. PMID:21192886
https://doi.org/10.3201/eid1701.100397
867. Unemo M, Golparian D, Nicholas R, Ohnishi M, Gallay A, Sednaoui
P. High-level cefixime- and ceftriaxone-resistant Neisseria gonorrhoeae
in France: novel penA mosaic allele in a successful international clone
causes treatment failure. Antimicrob Agents Chemother 2012;56:1273
80. PMID:22155830 https://doi.org/10.1128/AAC.05760-11
868. CDC. Update to CDC’s sexually transmitted diseases treatment
guidelines, 2010: oral cephalosporins no longer a recommended
treatment for gonococcal infections. MMWR Morb Mortal Wkly Rep
2012;61:590–4. PMID:22874837
869. Wind CM, de Vries E, Schim van der Loeff MF, et al. Decreased
azithromycin susceptibility of Neisseria gonorrhoeae isolates in patients
recently treated with azithromycin. Clin Infect Dis 2017;65:37–45.
PMID:28510723 https://doi.org/10.1093/cid/cix249
870. Kong FYS, Horner P, Unemo M, Hocking JS. Pharmacokinetic
considerations regarding the treatment of bacterial sexually transmitted
infections with azithromycin: a review. J Antimicrob Chemother
2019;74:115766. PMID:30649333 https://doi.org/10.1093/jac/
dky548
871. CDC. Antibiotic resistance threats in the United States, 2019. Atlanta,
GA: US Department of Health and Human Services, CDC; 2019.
https://www.cdc.gov/drugresistance/pdf/threats-report/2019-ar-
threats-report-508.pdf
872. St Cyr S, Barbee L, Workowski KA, et al. Update to CDC’s treatment
guidelines for gonococcal infection, 2020. MMWR Morb Mortal Wkly
Rep 2020;69:1911–6. PMID:33332296 https://doi.org/10.15585/
mmwr.mm6950a6
873. Clinical and Laboratory Standards Institute. Performance standards for
antimicrobial susceptibility testing: twentieth informational supplement.
Clinical and Laboratory Standards Institute document M100-S20.
Wayne, PA: Clinical and Laboratory Standards Institute; 2010.
874. CDC. Cephalosporin-resistant Neisseria gonorrhoeae public health
response plan. Atlanta, GA: US Department of Health and Human
Services, CDC; 2012. https://www.cdc.gov/std/treatment/Ceph-R-
ResponsePlanJuly30-2012.pdf
875. Poncin T, Merimeche M, Braille A, et al. Two cases of multidrug-
resistant Neisseria gonorrhoeae related to travel in south-eastern Asia,
France, June 2019. Euro Surveill 2019;24:1900528. PMID:31507264
https://doi.org/10.2807/1560-7917.ES.2019.24.36.1900528
876. Carnicer-Pont D, Smithson A, Fina-Homar E, Bastida MT; Gonococcus
Antimicrobial Resistance Surveillance Working Group. First cases of
Neisseria gonorrhoeae resistant to ceftriaxone in Catalonia, Spain, May
2011. Enferm Infecc Microbiol Clin 2012;30:218–9. PMID:22244992
https://doi.org/10.1016/j.eimc.2011.11.010
877. Cámara J, Serra J, Ayats J, et al. Molecular characterization of two
high-level ceftriaxone-resistant Neisseria gonorrhoeae isolates detected
in Catalonia, Spain. J Antimicrob Chemother 2012;67:185860.
PMID:22566592 https://doi.org/10.1093/jac/dks162
878. Eyre DW, Sanderson ND, Lord E, et al. Gonorrhoea treatment failure
caused by a Neisseria gonorrhoeae strain with combined ceftriaxone and
high-level azithromycin resistance, England, February 2018. Euro
Surveill 2018;23:1800323. PMID:29991383 https://doi.
org/10.2807/1560-7917.ES.2018.23.27.1800323
879. Fifer H, Hughes G, Whiley D, Lahra MM. Lessons learnt from
ceftriaxone-resistant gonorrhoea in the UK and Australia. Lancet Infect
Dis 2020;20:2768. PMID:32112753 https://doi.org/10.1016/
S1473-3099(20)30055-4
880. Chisholm SA, Mouton JW, Lewis DA, Nichols T, Ison CA, Livermore
DM. Cephalosporin MIC creep among gonococci: time for a
pharmacodynamic rethink? J Antimicrob Chemother 2010;65:2141–8.
PMID:20693173 https://doi.org/10.1093/jac/dkq289
881. Connolly KL, Eakin AE, Gomez C, Osborn BL, Unemo M, Jerse AE.
Pharmacokinetic data are predictive of in vivo efficacy for cefixime and
ceftriaxone against susceptible and resistant Neisseria gonorrhoeae strains
in the gonorrhea mouse model. Antimicrob Agents Chemother
2019;63:e01644-18. PMID:30642924 https://doi.org/10.1128/
AAC.01644-18
882. Blondeau JM, Hansen G, Metzler K, Hedlin P. The role of PK/PD
parameters to avoid selection and increase of resistance: mutant
prevention concentration. J Chemother 2004;16(Suppl 3):1–19.
PMID:15334827 https://doi.org/10.1080/1120009X.2004.11782371
883. Moran JS, Levine WC. Drugs of choice for the treatment of
uncomplicated gonococcal infections. Clin Infect Dis 1995;20(Suppl
1):S4765. PMID:7795109 https://doi.org/10.1093/clinids/20.
Supplement_1.S47
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 165
US Department of Health and Human Services/Centers for Disease Control and Prevention
884. Unemo M, Golparian D, Eyre DW. Antimicrobial resistance
in Neisseria gonorrhoeae and treatment of gonorrhea. Methods
Mol Biol 2019;1997:3758. PMID:31119616 https://doi.
org/10.1007/978-1-4939-9496-0_3
885. Kirkcaldy RD, Weinstock HS, Moore PC, et al. The efficacy and
safety of gentamicin plus azithromycin and gemifloxacin plus
azithromycin as treatment of uncomplicated gonorrhea. Clin Infect
Dis 2014;59:1083–91. PMID:25031289 https://doi.org/10.1093/
cid/ciu521
886. Ross JDC, Brittain C, Cole M, et al.; G-ToG trial team. Gentamicin
compared with ceftriaxone for the treatment of gonorrhoea (G-ToG):
a randomised non-inferiority trial. Lancet 2019;393:251120.
PMID:31056291 https://doi.org/10.1016/S0140-6736(18)32817-4
887. Singh V, Bala M, Bhargava A, Kakran M, Bhatnagar R. In vitro efficacy
of 21 dual antimicrobial combinations comprising novel and currently
recommended combinations for treatment of drug resistant gonorrhoea
in future era. PLoS One 2018;13:e0193678. PMID:29509792 https://
doi.org/10.1371/journal.pone.0193678
888. Mayer KH, Klausner JD, Handsfield HH. Intersecting
epidemics and educable moments: sexually transmitted disease
risk assessment and screening in men who have sex with men.
Sex Transm Dis 2001;28:4647. PMID:11473219 https://doi.
org/10.1097/00007435-200108000-00008
889. Linhart Y, Shohat T, Amitai Z, et al. Sexually transmitted infections
among brothel-based sex workers in Tel-Aviv area, Israel: high
prevalence of pharyngeal gonorrhoea. Int J STD AIDS 2008;19:656–9.
PMID:18824615 https://doi.org/10.1258/ijsa.2008.008127
890. Johnson Jones ML, Chapin-Bardales J, Bizune D, et al.; National HIV
Behavioral Surveillance Sexually Transmitted Infection Study Group.
Extragenital chlamydia and gonorrhea among community venue-
attending men who have sex with men—five cities, United States, 2017.
MMWR Morb Mortal Wkly Rep 2019;68:321–5. PMID:30973847
https://doi.org/10.15585/mmwr.mm6814a1
891. Chow EP, Williamson DA, Fortune R, et al. Prevalence of genital and
oropharyngeal chlamydia and gonorrhoea among female sex workers
in Melbourne, Australia, 2015–2017: need for oropharyngeal testing.
Sex Transm Infect 2019;95:398–401. PMID:31113904 https://doi.
org/10.1136/sextrans-2018-053957
892. Cornelisse VJ, Williamson D, Zhang L, et al. Evidence for a new
paradigm of gonorrhoea transmission: cross-sectional analysis of
Neisseria gonorrhoeae infections by anatomical site in both partners in
60 male couples. Sex Transm Infect 2019;95:43742. PMID:30996106
https://doi.org/10.1136/sextrans-2018-053803
893. Kissinger PJ, Reilly K, Taylor SN, Leichliter JS, Rosenthal S, Martin
DH. Early repeat Chlamydia trachomatis and Neisseria gonorrhoeae
infections among heterosexual men. Sex Transm Dis 2009;36:498500.
PMID:19617870 https://doi.org/10.1097/OLQ.0b013e3181a4d147
894. Berenger BM, Demczuk W, Gratrix J, Pabbaraju K, Smyczek P,
Martin I. Genetic characterization and enhanced surveillance of
ceftriaxone-resistant Neisseria gonorrhoeae strain, Alberta, Canada,
2018. Emerg Infect Dis 2019;25:1660–7. PMID:31407661 https://
doi.org/10.3201/eid2509.190407
895. Rob F, Klubalová B, Nyčová E, Hercogová J, Unemo M. Gentamicin
240 mg plus azithromycin 2 g vs. ceftriaxone 500 mg plus
azithromycin 2 g for treatment of rectal and pharyngeal gonorrhoea: a
randomized controlled trial. Clin Microbiol Infect 2020;26:207–12.
PMID:31419483 https://doi.org/10.1016/j.cmi.2019.08.004
896. Romano A, Gaeta F, Valluzzi RL, Caruso C, Rumi G, Bousquet PJ.
IgE-mediated hypersensitivity to cephalosporins: cross-reactivity and
tolerability of penicillins, monobactams, and carbapenems. J Allergy
Clin Immunol 2010;126:9949. PMID:20888035 https://doi.
org/10.1016/j.jaci.2010.06.052
897. American College of Obstetricians and Gynecologists’ Committee
on Obstetric Practice. Committee opinion No. 717: sulfonamides,
nitrofurantoin, and risk of birth defects. Obstet Gynecol
2017;130:e1502. PMID:28832488 https://doi.org/10.1097/
AOG.0000000000002300
898. Haimovici R, Roussel TJ. Treatment of gonococcal
conjunctivitis with single-dose intramuscular ceftriaxone. Am
J Ophthalmol 1989;107:511–4. PMID:2496606 https://doi.
org/10.1016/0002-9394(89)90495-9
899. Bleich AT, Sheffield JS, Wendel GD Jr, Sigman A, Cunningham
FG. Disseminated gonococcal infection in women. Obstet Gynecol
2012;119:597602. PMID:22353959 https://doi.org/10.1097/
AOG.0b013e318244eda9
900. Belkacem A, Caumes E, Ouanich J, et al.; Working Group FRA-DGI.
Changing patterns of disseminated gonococcal infection in France:
cross-sectional data 2009–2011. Sex Transm Infect 2013;89:613–5.
PMID:23920397 https://doi.org/10.1136/sextrans-2013-051119
901. Birrell JM, Gunathilake M, Singleton S, Williams S, Krause V.
Characteristics and impact of disseminated gonococcal infection in
the “Top End” of Australia. Am J Trop Med Hyg 2019;101:753–60.
PMID:31392956 https://doi.org/10.4269/ajtmh.19-0288
902. Crew PE, Abara WE, McCulley L, et al. Disseminated gonococcal
infections in patients receiving eculizumab: a case series. Clin Infect
Dis 2019;69:596600. PMID:30418536 https://doi.org/10.1093/cid/
ciy958
903. Curry SJ, Krist AH, Owens DK, et al.; US Preventive Services Task
Force. Ocular prophylaxis for gonococcal ophthalmia neonatorum:
US Preventive Services Task Force reaffirmation recommendation
statement. JAMA 2019;321:3948. PMID:30694327 https://doi.
org/10.1001/jama.2018.21367
904. Kreisel K, Weston E, Braxton J, Llata E, Torrone E. Keeping an eye
on chlamydia and gonorrhea conjunctivitis in infants in the United
States, 2010–2015. Sex Transm Dis 2017;44:356–8. PMID:28499285
https://doi.org/10.1097/OLQ.0000000000000613
905. Scott WJ, Eck CD. Povidone-iodine and ophthalmia neonatorum.
Ophthalmology 2012;119:6534. PMID:22385492 https://doi.
org/10.1016/j.ophtha.2011.11.037
906. David M, Rumelt S, Weintraub Z. Efficacy comparison between
povidone iodine 2.5% and tetracycline 1% in prevention of ophthalmia
neonatorum. Ophthalmology 2011;118:1454–8. PMID:21439642
https://doi.org/10.1016/j.ophtha.2010.12.003
907. Binenbaum G, Bruno CJ, Forbes BJ, et al. Periocular ulcerative
dermatitis associated with gentamicin ointment prophylaxis in
newborns. J Pediatr 2010;156:320–1. PMID:20105641 https://doi.
org/10.1016/j.jpeds.2009.11.060
908. Nathawad R, Mendez H, Ahmad A, et al. Severe ocular reactions after
neonatal ocular prophylaxis with gentamicin ophthalmic ointment.
Pediatr Infect Dis J 2011;30:175–6. PMID:20885334 https://doi.
org/10.1097/INF.0b013e3181f6c2e5
909. Taylor-Robinson D, Jensen JS. Mycoplasma genitalium: from chrysalis
to multicolored butterfly. Clin Microbiol Rev 2011;24:498514.
PMID:21734246 https://doi.org/10.1128/CMR.00006-11
910. Seña AC, Lensing S, Rompalo A, et al. Chlamydia trachomatis,
Mycoplasma genitalium, and Trichomonas vaginalis infections in
men with nongonococcal urethritis: predictors and persistence after
therapy. J Infect Dis 2012;206:357–65. PMID:22615318 https://doi.
org/10.1093/infdis/jis356
911. Huppert JS, Mortensen JE, Reed JL, Kahn JA, Rich KD, Hobbs MM.
Mycoplasma genitalium detected by transcription-mediated amplification
is associated with Chlamydia trachomatis in adolescent women. Sex
Transm Dis 2008;35:2504. PMID:18490867 https://doi.org/10.1097/
OLQ.0b013e31815abac6
Recommendations and Reports
166 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
912. Mena L, Wang X, Mroczkowski TF, Martin DH. Mycoplasma genitalium
infections in asymptomatic men and men with urethritis attending a
sexually transmitted diseases clinic in New Orleans. Clin Infect Dis
2002;35:116773. PMID:12410476 https://doi.org/10.1086/343829
913. Falk L. The overall agreement of proposed definitions of mucopurulent
cervicitis in women at high risk of Chlamydia infection. Acta Derm
Venereol 2010;90:50611. PMID:20814628 https://doi.
org/10.2340/00015555-0924
914. Anagrius C, Loré B, Jensen JS. Mycoplasma genitalium: prevalence,
clinical significance, and transmission. Sex Transm Infect
2005;81:458–62. PMID:16326846 https://doi.org/10.1136/
sti.2004.012062
915. Manhart LE, Critchlow CW, Holmes KK, et al. Mucopurulent cervicitis
and Mycoplasma genitalium. J Infect Dis 2003;187:6507.
PMID:12599082 https://doi.org/10.1086/367992
916. Lusk MJ, Konecny P, Naing ZW, Garden FL, Cumming RG, Rawlinson
WD. Mycoplasma genitalium is associated with cervicitis and HIV
infection in an urban Australian STI clinic population. Sex Transm
Infect 2011;87:107–9. PMID:21071566 https://doi.org/10.1136/
sti.2010.045138
917. Dehon PM, McGowin CL. The immunopathogenesis of Mycoplasma
genitalium infections in women: a narrative review. Sex Transm Dis
2017;44:42832. PMID:28608793 https://doi.org/10.1097/
OLQ.0000000000000621
918. Bjartling C, Osser S, Persson K. The association between Mycoplasma
genitalium and pelvic inflammatory disease after termination of
pregnancy. BJOG 2010;117:3614. PMID:20015303 https://doi.
org/10.1111/j.1471-0528.2009.02455.x
919. Bjartling C, Osser S, Persson K. Mycoplasma genitalium in cervicitis
and pelvic inflammatory disease among women at a gynecologic
outpatient service. Am J Obstet Gynecol 2012;206:476.e18.
PMID:22483084 https://doi.org/10.1016/j.ajog.2012.02.036
920. Taylor BD, Zheng X, O’Connell CM, Wiesenfeld HC, Hillier SL,
Darville T. Risk factors for Mycoplasma genitalium endometritis and
incident infection: a secondary data analysis of the T cell Response
Against Chlamydia (TRAC) Study. Sex Transm Infect 2018;94:41420.
PMID:29563165 https://doi.org/10.1136/sextrans-2017-053376
921. Cohen CR, Mugo NR, Astete SG, et al. Detection of Mycoplasma
genitalium in women with laparoscopically diagnosed acute salpingitis.
Sex Transm Infect 2005;81:4636. PMID:16326847 https://doi.
org/10.1136/sti.2005.015701
922. Haggerty CL, Totten PA, Astete SG, Ness RB. Mycoplasma genitalium
among women with nongonococcal, nonchlamydial pelvic inflammatory
disease. Infect Dis Obstet Gynecol 2006;2006:30184. PMID:17485798
https://doi.org/10.1155/IDOG/2006/30184
923. Short VL, Totten PA, Ness RB, Astete SG, Kelsey SF, Haggerty CL.
Clinical presentation of Mycoplasma genitalium infection versus Neisseria
gonorrhoeae infection among women with pelvic inflammatory disease.
Clin Infect Dis 2009;48:417. PMID:19025498 https://doi.
org/10.1086/594123
924. Simms I, Eastick K, Mallinson H, et al. Associations between
Mycoplasma genitalium, Chlamydia trachomatis, and pelvic inflammatory
disease. Sex Transm Infect 2003;79:154–6. PMID:12690141 https://
doi.org/10.1136/sti.79.2.154
925. Oakeshott P, Aghaizu A, Hay P, et al. Is Mycoplasma genitalium in
women the “New Chlamydia?” A community-based prospective cohort
study. Clin Infect Dis 2010;51:1160–6. PMID:20942656 https://doi.
org/10.1086/656739
926. Wiesenfeld HC, Hillier SL, Meyn L, et al. Mycoplasma genitalium—is
it a pathogen in acute pelvic inflammatory disease (PID)? Sex Transm
Infect 2013;89(Suppl 1):A34. https://doi.org/10.1136/
sextrans-2013-051184.0106
927. Møller BR, Taylor-Robinson D, Furr PM, Freundt EA. Acute upper
genital-tract disease in female monkeys provoked experimentally by
Mycoplasma genitalium. Br J Exp Pathol 1985;66:41726.
PMID:4027175
928. Wiesenfeld HC, Manhart LE. Mycoplasma genitalium in women:
current knowledge and research priorities for this recently emerged
pathogen. J Infect Dis 2017;216(suppl_2):S389–95.
929. Clausen HF, Fedder J, Drasbek M, et al. Serological investigation of
Mycoplasma genitalium in infertile women. Hum Reprod
2001;16:1866–74. PMID:11527890 https://doi.org/10.1093/
humrep/16.9.1866
930. Svenstrup HF, Fedder J, Kristoffersen SE, Trolle B, Birkelund S,
Christiansen G. Mycoplasma genitalium, Chlamydia trachomatis, and
tubal factor infertility—a prospective study. Fertil Steril 2008;90:513–20.
PMID:17548070 https://doi.org/10.1016/j.fertnstert.2006.12.056
931. Idahl A, Jurstrand M, Olofsson JI, Fredlund H. Mycoplasma genitalium
serum antibodies in infertile couples and fertile women. Sex Transm
Infect 2015;91:58991. PMID:25921018 https://doi.org/10.1136/
sextrans-2015-052011
932. Edwards RK, Ferguson RJ, Reyes L, Brown M, Theriaque DW, Duff
P. Assessing the relationship between preterm delivery and various
microorganisms recovered from the lower genital tract. J Matern Fetal
Neonatal Med 2006;19:35763. PMID:16801313 https://doi.
org/10.1080/00207170600712071
933. Vandepitte J, Bukenya J, Hughes P, et al. Clinical characteristics
associated with Mycoplasma genitalium infection among women at high
risk of HIV and other STI in Uganda. Sex Transm Dis 2012;39:48791.
PMID:22592838 https://doi.org/10.1097/OLQ.0b013e31824b1cf3
934. Rowlands S, Danielewski JA, Tabrizi SN, Walker SP, Garland SM.
Microbial invasion of the amniotic cavity in midtrimester pregnancies
using molecular microbiology. Am J Obstet Gynecol 2017;217:71.
e1–5. PMID:28268197 https://doi.org/10.1016/j.ajog.2017.02.051
935. Jurstrand M, Jensen JS, Magnuson A, Kamwendo F, Fredlund H. A
serological study of the role of Mycoplasma genitalium in pelvic
inflammatory disease and ectopic pregnancy. Sex Transm Infect
2007;83:31923. PMID:17475688 https://doi.org/10.1136/
sti.2007.024752
936. Ashshi AM, Batwa SA, Kutbi SY, Malibary FA, Batwa M, Refaat B.
Prevalence of 7 sexually transmitted organisms by multiplex real-time
PCR in Fallopian tube specimens collected from Saudi women with
and without ectopic pregnancy. BMC Infect Dis 2015;15:569.
PMID:26666587 https://doi.org/10.1186/s12879-015-1313-1
937. Bissessor M, Tabrizi SN, Bradshaw CS, et al. The contribution of
Mycoplasma genitalium to the aetiology of sexually acquired infectious
proctitis in men who have sex with men. Clin Microbiol Infect
2016;22:2605. PMID:26686807 https://doi.org/10.1016/j.
cmi.2015.11.016
938. Ong JJ, Aung E, Read TRH, et al. Clinical characteristics of anorectal
Mycoplasma genitalium infection and microbial cure in men who have
sex with men. Sex Transm Dis 2018;45:522–6. PMID:29465653
https://doi.org/10.1097/OLQ.0000000000000793
939. Read TRH, Murray GL, Danielewski JA, et al. symptoms, sites, and
significance of Mycoplasma genitalium in men who have sex with men.
Emerg Infect Dis 2019;25:71927. PMID:30882306 https://doi.
org/10.3201/eid2504.181258
940. Cina M, Baumann L, Egli-Gany D, et al. Mycoplasma genitalium
incidence, persistence, concordance between partners and progression:
systematic review and meta-analysis. Sex Transm Infect 2019;95:328–35.
PMID:31055469 https://doi.org/10.1136/sextrans-2018-053823
941. Baumann L, Cina M, Egli-Gany D, et al. Prevalence of Mycoplasma
genitalium in different population groups: systematic review andmeta-
analysis. Sex Transm Infect 2018;94:25562. PMID:29440466 https://
doi.org/10.1136/sextrans-2017-053384
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 167
US Department of Health and Human Services/Centers for Disease Control and Prevention
942. Vandepitte J, Weiss HA, Bukenya J, et al. Association between
Mycoplasma genitalium infection and HIV acquisition among female
sex workers in Uganda: evidence from a nested case-control study. Sex
Transm Infect 2014;90:5459. PMID:24687129 https://doi.
org/10.1136/sextrans-2013-051467
943. Ferré VM, Ekouevi DK, Gbeasor-Komlanvi FA, et al. Prevalence of
human papillomavirus, human immunodeficiency virus and other
sexually transmitted infections among female sex workers in Togo: a
national cross-sectional survey. Clin Microbiol Infect 2019;25:1560.
e1–7. PMID:31051265 https://doi.org/10.1016/j.cmi.2019.04.015
944. Mavedzenge SN, Van Der Pol B, Weiss HA, et al. The association
between Mycoplasma genitalium and HIV-1 acquisition in African
women. AIDS 2012;26:61724. PMID:22210630 https://doi.
org/10.1097/QAD.0b013e32834ff690
945. Salado-Rasmussen K, Jensen JS. Mycoplasma genitalium testing pattern
and macrolide resistance: a Danish nationwide retrospective survey.
Clin Infect Dis 2014;59:2430. PMID:24729494 https://doi.
org/10.1093/cid/ciu217
946. Wold C, Sorthe J, Hartgill U, Olsen AO, Moghaddam A, Reinton N.
Identification of macrolide-resistant Mycoplasma genitalium using real-
time PCR. J Eur Acad Dermatol Venereol 2015;29:161620.
PMID:25622510 https://doi.org/10.1111/jdv.12963
947. Gesink D, Racey CS, Seah C, et al. Mycoplasma genitalium in Toronto,
Ont: estimates of prevalence and macrolide resistance. Can Fam
Physician 2016;62:e96–101. PMID:27331225
948. Kristiansen GQ, Lisby JG, Schønning K. 5’ nuclease genotyping assay
for identification of macrolide-resistant Mycoplasma genitalium in
clinical specimens. J Clin Microbiol 2016;54:1593–7. PMID:27053672
https://doi.org/10.1128/JCM.00012-16
949. Braam JF, Slotboom B, Van Marm S, et al. High prevalence of the
A2058T macrolide resistance-associated mutation in Mycoplasma
genitalium strains from the Netherlands. J Antimicrob Chemother
2017;72:152930. PMID:28158595 https://doi.org/10.1093/jac/
dkw584
950. Murray GL, Bradshaw CS, Bissessor M, et al. Increasing macrolide and
fluoroquinolone resistance in Mycoplasma genitalium. Emerg Infect Dis
2017;23:80912. PMID:28418319 https://doi.org/10.3201/
eid2305.161745
951. Chernesky MA, Jang D, Martin I, et al.; Canadian MG Study Group.
Mycoplasma genitalium antibiotic resistance-mediating mutations in
Canadian women with or without Chlamydia trachomatis infection.
Sex Transm Dis 2017;44:4335. PMID:28608794 https://doi.
org/10.1097/OLQ.0000000000000617
952. Barberá MJ, Fernández-Huerta M, Jensen JS, Caballero E, Andreu A.
Mycoplasma genitalium macrolide and fluoroquinolone resistance:
prevalence and risk factors among a 2013–2014 cohort of patients in
Barcelona, Spain. Sex Transm Dis 2017;44:45762. PMID:28703723
https://doi.org/10.1097/OLQ.0000000000000631
953. Sweeney EL, Trembizki E, Bletchly C, et al. Levels of Mycoplasma
genitalium antimicrobial resistance differ by both region and gender in
the state of Queensland, Australia: implications for treatment
guidelines. J Clin Microbiol 2019;57:e01555-18. PMID:30602443
https://doi.org/10.1128/JCM.01555-18
954. Bissessor M, Tabrizi SN, Tw in J, et al. Macrolide resistance and
azithromycin failure in a Mycoplasma genitalium-infected cohort and
response of azithromycin failures to alternative antibiotic regimens.
Clin Infect Dis 2015;60:122836. PMID:25537875 https://doi.
org/10.1093/cid/ciu1162
955. Piñeiro L, Idigoras P, de la Caba I, López-Olaizola M, Cilla G. Guided
antibiotic therapy for Mycoplasma genitalium infections: analysis of
mutations associated with resistance to macrolides and fluoroquinolones
[Spanish]. Enferm Infecc Microbiol Clin 2019;37:3947.
PMID:30396750
956. Dionne-Odom J, Geisler WM, Aaron KJ, et al. High prevalence of
multidrug-resistant Mycoplasma genitalium in human immunodeficiency
virus-infected men who have sex with men in Alabama. Clin Infect Dis
2018;66:796–8. PMID:29028993 https://doi.org/10.1093/cid/cix853
957. Pitt R, Fifer H, Woodford N, Alexander S. Detection of markers
predictive of macrolide and fluoroquinolone resistance in Mycoplasma
genitalium from patients attending sexual health services in England.
Sex Transm Infect 2018;94:913. PMID:28717051 https://doi.
org/10.1136/sextrans-2017-053164
958. Unemo M, Salado-Rasmussen K, Hansen M, et al. Clinical and
analytical evaluation of the new Aptima Mycoplasma genitalium assay,
with data on M.genitalium prevalence and antimicrobial resistance in
M.genitalium in Denmark, Norway and Sweden in 2016. Clin
Microbiol Infect 2018;24:5339. PMID:28923377 https://doi.
org/10.1016/j.cmi.2017.09.006
959. Anderson T, Coughlan E, Werno A. Mycoplasma genitalium macrolide
and fluoroquinolone resistance detection and clinical implications in
a selected cohort in New Zealand. J Clin Microbiol 2017;55:3242–8.
PMID:28878004 https://doi.org/10.1128/JCM.01087-17
960. Shimada Y, Deguchi T, Nakane K, et al. Emergence of clinical strains
of Mycoplasma genitalium harbouring alterations in ParC associated
with fluoroquinolone resistance. Int J Antimicrob Agents
2010;36:255–8. PMID:20580532 https://doi.org/10.1016/j.
ijantimicag.2010.05.011
961. Muller EE, Mahlangu MP, Lewis DA, Kularatne RS. Macrolide and
fluoroquinolone resistance-associated mutations in Mycoplasma
genitalium in Johannesburg, South Africa, 2007–2014. BMC Infect
Dis 2019;19:148. PMID:30760230 https://doi.org/10.1186/
s12879-019-3797-6
962. Chambers LC, Jensen JS, Morgan JL, et al. Lack of association between
the S83I ParC mutation in Mycoplasma genitalium and treatment
outcomes among men who have sex with men with nongonococcal
urethritis. Sex Transm Dis 2019;46:805–9. PMID:31259853 https://
doi.org/10.1097/OLQ.0000000000001035
963. Durukan D, Read TRH, Murray G, et al. Resistance-guided
antimicrobial therapy using doxycycline-moxifloxacin and doxycycline-
2.5g azithromycin for the treatment of Mycoplasma genitalium infection:
efficacy and tolerability. Clin Infect Dis 2020;71:14618.
PMID:31629365 https://doi.org/10.1093/cid/ciz1031
964. Li Y, Le WJ, Li S, Cao YP, Su XH. Meta-analysis of the efficacy of
moxifloxacin in treating Mycoplasma genitalium infection. Int J STD
AIDS 2017;28:110614. PMID:28118803 https://doi.
org/10.1177/0956462416688562
965. Mondeja BA, Couri J, Rodríguez NM, Blanco O, Fernández C, Jensen
JS. Macrolide-resistant Mycoplasma genitalium infections in Cuban
patients: an underestimated health problem. BMC Infect Dis
2018;18:601. PMID:30486786 https://doi.org/10.1186/
s12879-018-3523-9
966. Glaser AM, Geisler WM, Ratliff AE, Xiao L, Waites KB, Gaisa M. Tw o
cases of multidrug-resistant genitourinary Mycoplasma genitalium
infection successfully eradicated with minocycline. Int J STD AIDS
2019; 30: 512 4 . PMID:30999836 https://doi.
org/10.1177/0956462418816757
967. Xiao L, Waites KB, Van Der Pol B, Aaron KJ, Hook EW 3rd, Geisler
WM. Mycoplasma genitalium infections with macrolide and
fluoroquinolone resistance-associated mutations in heterosexual African
American couples in Alabama. Sex Transm Dis 2019;46:1824.
PMID:29979336 https://doi.org/10.1097/OLQ.0000000000000891
968. Slifirski JB, Vodstrcil LA, Fairley CK, et al. Mycoplasma genitalium
infection in adults reporting sexual contact with infected partners,
Australia, 2008–2016. Emerg Infect Dis 2017;23:182633.
PMID:29047422 https://doi.org/10.3201/eid2311.170998
969. Anderson MR, Klink K, Cohrssen A. Evaluation of vaginal complaints.
JAMA 2004;291:136879. PMID:15026404 https://doi.org/10.1001/
jama.291.11.1368
Recommendations and Reports
168 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
970. Swidsinski A, Mendling W, Loening-Baucke V, et al. Adherent biofilms
in bacterial vaginosis. Obstet Gynecol 2005;106:101323.
PMID:16260520 https://doi.org/10.1097/01.
AOG.0000183594.45524.d2
971. Brotman RM, Klebanoff MA, Nansel TR, et al. Bacterial vaginosis
assessed by Gram stain and diminished colonization resistance to
incident gonococcal, chlamydial, and trichomonal genital infection.
J Infect Dis 2010;202:190715. PMID:21067371 https://doi.
org/10.1086/657320
972. Peebles K, Velloza J, Balkus JE, McClelland RS, Barnabas RV. High
global burden and costs of bacterial vaginosis: a systematic review and
meta-analysis. Sex Transm Dis 2019;46:30411. PMID:30624309
https://doi.org/10.1097/OLQ.0000000000000972
973. Kenyon CR, Buyze J, Klebanoff M, Brotman RM. Association between
bacterial vaginosis and partner concurrency: a longitudinal study. Sex
Transm Infect 2018;94:757. PMID:27645157 https://doi.
org/10.1136/sextrans-2016-052652
974. Sanchez S, Garcia PJ, Thomas KK, Catlin M, Holmes KK. Intravaginal
metronidazole gel versus metronidazole plus nystatin ovules for bacterial
vaginosis: a randomized controlled trial. Am J Obstet Gynecol
2004;191:1898906. PMID:15592270 https://doi.org/10.1016/j.
ajog.2004.06.089
975. Ness RB, Soper DE, Holley RL, et al.; PID Evaluation and Clinical
Health (PEACH) Study Investigators. Douching and endometritis:
results from the PID evaluation and clinical health (PEACH) study.
Sex Transm Dis 2001;28:2405. PMID:11318257 https://doi.
org/10.1097/00007435-200104000-00010
976. Gondwe T, Ness R, Totten PA, et al. Novel bacterial vaginosis-associated
organisms mediate the relationship between vaginal douching and
pelvic inflammatory disease. Sex Transm Infect 2020;96:43944.
PMID:31810995 https://doi.org/10.1136/sextrans-2019-054191
977. Abbai NS, Reddy T, Ramjee G. Prevalent bacterial vaginosis
infection—a risk factor for incident sexually transmitted infections in
women in Durban, South Africa. Int J STD AIDS 2016;27:1283–8.
PMID:26538552 https://doi.org/10.1177/0956462415616038
978. Morris BJ, Hankins CA, Banerjee J, et al. Does male circumcision
reduce womens risk of sexually transmitted infections, cervical cancer,
and associated conditions? Front Public Health 2019;7:4.
PMID:30766863 https://doi.org/10.3389/fpubh.2019.00004
979. Srinivasan S, Liu C, Mitchell CM, et al. Temporal variability of human
vaginal bacteria and relationship with bacterial vaginosis. PLoS One
2010;5:e10197. PMID:20419168 https://doi.org/10.1371/journal.
pone.0010197
980. Gajer P, Brotman RM, Bai G, et al. Temporal dynamics of the human
vaginal microbiota. Sci Transl Med 2012;4:132ra52. PMID:22553250
https://doi.org/10.1126/scitranslmed.3003605
981. Fethers KA, Fairley CK, Morton A, et al. Early sexual experiences and
risk factors for bacterial vaginosis. J Infect Dis 2009;200:1662–70.
PMID:19863439 https://doi.org/10.1086/648092
982. Achilles SL, Austin MN, Meyn LA, Mhlanga F, Chirenje ZM, Hillier
SL. Impact of contraceptive initiation on vaginal microbiota. Am
J Obstet Gynecol 2018;218:622.e110. PMID:29505773 https://doi.
org/10.1016/j.ajog.2018.02.017
983. Vodstrcil LA, Plummer ME, Fairley CK, et al. Combined oral
contraceptive pill-exposure alone does not reduce the risk of bacterial
vaginosis recurrence in a pilot randomised controlled trial. Sci Rep
2019;9:3555. PMID:30837554 https://doi.org/10.1038/
s41598-019-39879-8
984. Brooks JP, Edwards DJ, Blithe DL, et al. Effects of combined oral
contraceptives, depot medroxyprogesterone acetate and the
levonorgestrel-releasing intrauterine system on the vaginal microbiome.
Contraception 2017;95:40513. PMID:27913230 https://doi.
org/10.1016/j.contraception.2016.11.006
985. Moore KR, Harmon QE, Baird DD. Serum 25-hydroxyvitamin D and
risk of self-reported bacterial vaginosis in a prospective cohort study
of young African American women. J Womens Health (Larchmt)
2018;27:127884. PMID:29897832 https://doi.org/10.1089/
jwh.2017.6804
986. Lokken EM, Balkus JE, Kiarie J, et al. Association of recent bacterial
vaginosis with acquisition of Mycoplasma genitalium. Am J Epidemiol
2017;186:194201. PMID:28472225 https://doi.org/10.1093/aje/
kwx043
987. Brusselaers N, Shrestha S, van de Wijgert J, Verstraelen H. Vaginal
dysbiosis and the risk of human papillomavirus and cervical cancer:
systematic review and meta-analysis. Am J Obstet Gynecol
2019;221:9–18.e8. PMID:30550767 https://doi.org/10.1016/j.
ajog.2018.12.011
988. Abbai NS, Nyirenda M, Naidoo S, Ramjee G. Prevalent herpes simplex
virus-2 increases the risk of incident bacterial vaginosis in women
from South Africa. AIDS Behav 2018;22:217280. PMID:28956191
https://doi.org/10.1007/s10461-017-1924-1
989. Laxmi U, Agrawal S, Raghunandan C, Randhawa VS, Saili A.
Association of bacterial vaginosis with adverse fetomaternal outcome
in women with spontaneous preterm labor: a prospective cohort study.
J Matern Fetal Neonatal Med 2012;25:64–7. PMID:21557693
https://doi.org/10.3109/14767058.2011.565390
990. Cherpes TL, Wiesenfeld HC, Melan MA, et al. The associations
between pelvic inflammatory disease, Trichomonas vaginalis infection,
and positive herpes simplex virus type 2 serology. Sex Transm Dis
2006;33:74752. PMID:16691155 https://doi.org/10.1097/01.
olq.0000218869.52753.c7
991. Nelson DB, Hanlon A, Hassan S, et al. Preterm labor and bacterial
vaginosis-associated bacteria among urban women. J Perinat Med
2009;37:1304. PMID:18999913 https://doi.org/10.1515/
JPM.2009.026
992. Atashili J, Poole C, Ndumbe PM, Adimora AA, Smith JS. Bacterial
vaginosis and HIV acquisition: a meta-analysis of published studies.
AIDS 2008;22:1493501. PMID:18614873 https://doi.org/10.1097/
QAD.0b013e3283021a37
993. Gosmann C, Anahtar MN, Handley SA, et al. Lactobacillus-deficient
cervicovaginal bacterial communities are associated with increased
HIV acquisition in young South African women. Immunity
2017;46:2937. PMID:28087240 https://doi.org/10.1016/j.
immuni.2016.12.013
994. McClelland RS, Lingappa JR, Srinivasan S, et al. Evaluation of the
association between the concentrations of key vaginal bacteria and
the increased risk of HIV acquisition in African women from five
cohorts: a nested case-control study. Lancet Infect Dis 2018;18:554–
64. PMID:29396006 https://doi.org/10.1016/
S1473-3099(18)30058-6
995. Johnston C, Magaret A, Srinivasan S, et al. P239 Genital HSV-2
suppression is not associated with alterations in the vaginal
microbiome: a one-way, cross-over study. Sex Transm Infect
2019;95(Suppl 1):A148.
996. Zozaya M, Ferris MJ, Siren JD, et al. Bacterial communities in penile
skin, male urethra, and vaginas of heterosexual couples with and
without bacterial vaginosis. Microbiome 2016;4:16. PMID:27090518
https://doi.org/10.1186/s40168-016-0161-6
997. Liu CM, Hungate BA, Tobian AA, et al. Penile microbiota and female
partner bacterial vaginosis in Rakai, Uganda. MBio 2015;6:e00589.
PMID:26081632 https://doi.org/10.1128/mBio.00589-15
998. Mehta SD. Systematic review of randomized trials of treatment of
male sexual partners for improved bacteria vaginosis outcomes in
women. Sex Transm Dis 2012;39:82230. PMID:23007709 https://
doi.org/10.1097/OLQ.0b013e3182631d89
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 169
US Department of Health and Human Services/Centers for Disease Control and Prevention
999. Amsel R, Totten PA, Spiegel CA, Chen KC, Eschenbach D, Holmes
KK. Nonspecific vaginitis. Diagnostic criteria and microbial and
epidemiologic associations. Am J Med 1983;74:1422.
PMID:6600371 https://doi.org/10.1016/0002-9343(83)91112-9
1000. Nugent RP, Krohn MA, Hillier SL. Reliability of diagnosing bacterial
vaginosis is improved by a standardized method of Gram stain
interpretation. J Clin Microbiol 1991;29:297–301. PMID:1706728
https://doi.org/10.1128/JCM.29.2.297-301.1991
1001. Schwebke JR, Hillier SL, Sobel JD, McGregor JA, Sweet RL. Validity
of the vaginal Gram stain for the diagnosis of bacterial vaginosis.
Obstet Gynecol 1996;88:573–6. PMID:8841221 https://doi.
org/10.1016/0029-7844(96)00233-5
1002. Coleman JS, Gaydos CA. Molecular diagnosis of bacterial vaginosis:
an update. J Clin Microbiol 2018;56:e00342-18. PMID:29769280
https://doi.org/10.1128/JCM.00342-18
1003. Myziuk L, Romanowski B, Johnson SC. BVBlue test for diagnosis of
bacterial vaginosis. J Clin Microbiol 2003;41:1925–8. PMID:12734228
https://doi.org/10.1128/JCM.41.5.1925-1928.2003
1004. Bradshaw CS, Morton AN, Garland SM, Horvath LB, Kuzevska
I, Fairley CK. Evaluation of a point-of-care test, BVBlue, and
clinical and laboratory criteria for diagnosis of bacterial vaginosis.
J Clin Microbiol 2005;43:1304–8. PMID:15750100 https://doi.
org/10.1128/JCM.43.3.1304-1308.2005
1005. West B, Morison L, Schim van der Loeff M, et al. Evaluation
of a new rapid diagnostic kit (FemExam) for bacterial vaginosis
in patients with vaginal discharge syndrome in The Gambia.
Sex Transm Dis 2003;30:483–9. PMID:12782948 https://doi.
org/10.1097/00007435-200306000-00003
1006. Fredricks DN, Fiedler TL, Thomas KK, Oakley BB, Marrazzo JM.
Targeted PCR for detection of vaginal bacteria associated with bacterial
vaginosis. J Clin Microbiol 2007;45:3270–6. PMID:17687006
https://doi.org/10.1128/JCM.01272-07
1007. Gaydos CA, Beqaj S, Schwebke JR, et al. Clinical validation of a
test for the diagnosis of vaginitis. Obstet Gynecol 2017;130:181–9.
PMID:28594779 https://doi.org/10.1097/AOG.0000000000002090
1008. Cartwright CP, Lembke BD, Ramachandran K, et al. Development
and validation of a semiquantitative, multitarget PCR assay for
diagnosis of bacterial vaginosis. J Clin Microbiol 2012;50:2321–9.
PMID:22535982 https://doi.org/10.1128/JCM.00506-12
1009. Hilbert DW, Smith WL, Chadwick SG, et al. Development and
validation of a highly accurate quantitative real-time PCR assay for
diagnosis of bacterial vaginosis. J Clin Microbiol 2016;54:1017–24.
Erratum in: J Clin Microbiol 2016;54:1930. PMID:26818677
https://doi.org/10.1128/JCM.03104-15
1010. Schwebke JR, Desmond R. A randomized trial of metronidazole
in asymptomatic bacterial vaginosis to prevent the acquisition of
sexually transmitted diseases. Am J Obstet Gynecol 2007;196:517.
e1–6. PMID:17547876 https://doi.org/10.1016/j.ajog.2007.02.048
1011. Fjeld H, Raknes G. Is combining metronidazole and alcohol really
hazardous? [Norwegian]. Tidsskr Nor Laegeforen 2014;134:1661–3.
PMID:25223673 https://doi.org/10.4045/tidsskr.14.0081
1012. Hillier SL, Nyirjesy P, Waldbaum AS, et al. Secnidazole treatment
of bacterial vaginosis: a randomized controlled trial. Obstet Gynecol
2017;130:37986. PMID:28697102 https://doi.org/10.1097/
AOG.0000000000002135
1013. Schwebke JR, Morgan FG Jr, Koltun W, Nyirjesy P. A phase-3,
double-blind, placebo-controlled study of the effectiveness and safety
of single oral doses of secnidazole 2 g for the treatment of women
with bacterial vaginosis. Am J Obstet Gynecol 2017;217:678.e1–9.
Erratum in: Am J Obstet Gynecol 2018;219;110. PMID:28867602
https://doi.org/10.1016/j.ajog.2017.08.017
1014. Chavoustie SE, Gersten JK, Samuel MJ, Schwebke JR. A phase 3,
multicenter, prospective, open-label study to evaluate the safety of
a single dose of secnidazole 2 g for the treatment of women and
postmenarchal adolescent girls with bacterial vaginosis. J Womens
Health (Larchmt) 2018;27:492–7. PMID:29323627 https://doi.
org/10.1089/jwh.2017.6500
1015. Livengood CH 3rd, Ferris DG, Wiesenfeld HC, et al. Effectiveness of
two tinidazole regimens in treatment of bacterial vaginosis: a randomized
controlled trial. Obstet Gynecol 2007;110:302–9. PMID:17666604
https://doi.org/10.1097/01.AOG.0000275282.60506.3d
1016. Sobel JD, Nyirjesy P, Brown W. Tinidazole therapy for metronidazole-
resistant vaginal trichomoniasis. Clin Infect Dis 2001;33:1341–6.
PMID:11565074 https://doi.org/10.1086/323034
1017. Chavoustie SE, Jacobs M, Reisman HA, et al. Metronidazole vaginal
gel 1.3% in the treatment of bacterial vaginosis: a dose-ranging study.
J Low Genit Tract Dis 2015;19:129–34. PMID:24983350 https://
doi.org/10.1097/LGT.0000000000000062
1018. Schwebke JR, Marrazzo J, Beelen AP, Sobel JD. A phase 3,
multicenter, randomized, double-blind, vehicle-controlled study
evaluating the safety and efficacy of metronidazole vaginal gel
1.3% in the treatment of bacterial vaginosis. Sex Transm Dis
2015;42:37681. PMID:26222750 https://doi.org/10.1097/
OLQ.0000000000000300
1019. Faro S, Skokos CK; Clindesse Investigators Group. The efficacy and
safety of a single dose of Clindesse vaginal cream versus a seven-dose
regimen of Cleocin vaginal cream in patients with bacterial vaginosis.
Infect Dis Obstet Gynecol 2005;13:15560. PMID:16240515
https://doi.org/10.1080/10647440500148321
1020. Marrazzo JM, Dombrowski JC, Wierzbicki MR, et al. Safety and
efficacy of a novel vaginal anti-infective, TOL-463, in the treatment
of bacterial vaginosis and vulvovaginal candidiasis: a randomized,
single-blind, phase 2, controlled trial. Clin Infect Dis 2019;68:803–9.
PMID:30184181 https://doi.org/10.1093/cid/ciy554
1021. Antonio MA, Meyn LA, Murray PJ, Busse B, Hillier SL.
Vaginal colonization by probiotic Lactobacillus crispatus CTV-
05 is decreased by sexual activity and endogenous Lactobacilli.
J Infect Dis 2009;199:150613. PMID:19331578 https://doi.
org/10.1086/598686
1022. Senok AC, Verstraelen H, Temmerman M, Botta GA. Probiotics
for the treatment of bacterial vaginosis. Cochrane Database
Syst Rev 2009;(4):CD006289. PMID:19821358 https://doi.
org/10.1002/14651858.CD006289.pub2
1023. Abad CL, Safdar N. The role of Lactobacillus probiotics in the
treatment or prevention of urogenital infections—a systematic
review. J Chemother 2009;21:24352. PMID:19567343 https://
doi.org/10.1179/joc.2009.21.3.243
1024. Mastromarino P, Macchia S, Meggiorini L, et al. Effectiveness
of Lactobacillus-containing vaginal tablets in the treatment
of symptomatic bacterial vaginosis. Clin Microbiol
Infect 2009;15:6774. PMID:19046169 https://doi.
org/10.1111/j.1469-0691.2008.02112.x
1025. Hemmerling A, Harrison W, Schroeder A, et al. Phase 2a study
assessing colonization efficiency, safety, and acceptability of
Lactobacillus crispatus CTV-05 in women with bacterial vaginosis.
Sex Transm Dis 2010;37:74550. PMID:20644497 https://doi.
org/10.1097/OLQ.0b013e3181e50026
1026. Bunge KE, Beigi RH, Meyn LA, Hillier SL. The efficacy of retreatment
with the same medication for early treatment failure of bacterial
vaginosis. Sex Transm Dis 2009;36:711–3. PMID:19652628 https://
doi.org/10.1097/OLQ.0b013e3181af6cfd
1027. Aguin T, Akins RA, Sobel JD. High-dose vaginal maintenance
metronidazole for recurrent bacterial vaginosis: a pilot study.
Sex Transm Dis 2014;41:290–1. PMID:24722380 https://doi.
org/10.1097/OLQ.0000000000000123
Recommendations and Reports
170 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
1028. Sobel JD, Ferris D, Schwebke J, et al. Suppressive antibacterial therapy
with 0.75% metronidazole vaginal gel to prevent recurrent bacterial
vaginosis. Am J Obstet Gynecol 2006;194:1283–9. PMID:16647911
https://doi.org/10.1016/j.ajog.2005.11.041
1029. Reichman O, Akins R, Sobel JD. Boric acid addition to suppressive
antimicrobial therapy for recurrent bacterial vaginosis. Sex Transm
Dis 2009;36:7324. PMID:19704395 https://doi.org/10.1097/
OLQ.0b013e3181b08456
1030. McClelland RS, Richardson BA, Hassan WM, et al. Improvement
of vaginal health for Kenyan women at risk for acquisition of
human immunodeficiency virus type 1: results of a randomized
trial. J Infect Dis 2008;197:1361–8. PMID:18444793 https://doi.
org/10.1086/587490
1031. Schwebke J, Carter B, Waldbaum A, et al. Results of a phase 3,
randomized, double-blind, placebo-controlled study to evaluate
the efficacy and safety of astodrimer gel for prevention of recurrent
bacterial vaginosis. Am J Obstet Gynecol 2019;221:672–3. https://
doi.org/10.1016/j.ajog.2019.10.087
1032. Cohen CR, Wierzbicki MR, French AL, et al. Randomized trial of
Lactin-V to prevent recurrence of bacterial vaginosis. N Engl J Med
2020;382:190615. PMID:32402161 https://doi.org/10.1056/
NEJMoa1915254
1033. Turner AN, Carr Reese P, Fields KS, et al. A blinded, randomized
controlled trial of high-dose vitamin D supplementation to reduce
recurrence of bacterial vaginosis. Am J Obstet Gynecol 2014;211:479.
e113. PMID:24949544 https://doi.org/10.1016/j.ajog.2014.06.023
1034. Plummer EL, Vodstrcil LA, Danielewski JA, et al. Combined oral
and topical antimicrobial therapy for male partners of women with
bacterial vaginosis: acceptability, tolerability and impact on the genital
microbiota of couples—a pilot study. PLoS One 2018;13:e0190199.
PMID:29293559 https://doi.org/10.1371/journal.pone.0190199
1035. Schwebke JR, Lensing SY, Lee J, et al. Treatment of male sexual
partners of women with bacterial vaginosis (BV): a randomized,
double-blind, placebo-controlled trial. Clin Infect Dis 2020; Epub
December 31, 2020. PMID: 33383580
1036. Koumans EH, Kendrick JS; CDC Bacterial Vaginosis
Working Group. Preventing adverse sequelae of bacterial
vaginosis: a public health program and research agenda. Sex
Transm Dis 2001;28:292–7. PMID:11354269 https://doi.
org/10.1097/00007435-200105000-00011
1037. Hauth JC, Goldenberg RL, Andrews WW, DuBard MB, Copper
RL. Reduced incidence of preterm delivery with metronidazole
and erythromycin in women with bacterial vaginosis. N Engl
J Med 1995;333:1732–6. PMID:7491136 https://doi.org/10.1056/
NEJM199512283332603
1038. Morales WJ, Schorr S, Albritton J. Effect of metronidazole in
patients with preterm birth in preceding pregnancy and bacterial
vaginosis: a placebo-controlled, double-blind study. Am J Obstet
Gynecol 1994;171:345–9. PMID:8059811 https://doi.org/10.1016/
S0002-9378(94)70033-8
1039. Yudin MH, Landers DV, Meyn L, Hillier SL. Clinical and cervical
cytokine response to treatment with oral or vaginal metronidazole
for bacterial vaginosis during pregnancy: a randomized trial. Obstet
Gynecol 2003;102:527–34. PMID:12962937
1040. Ugwumadu A, Reid F, Hay P, Manyonda I. Natural history of bacterial
vaginosis and intermediate flora in pregnancy and effect of oral
clindamycin. Obstet Gynecol 2004;104:1149. PMID:15229009
https://doi.org/10.1097/01.AOG.0000130068.21566.4e
1041. Burtin P, Taddio A, Ariburnu O, Einarson TR, Koren G.
Safety of metronidazole in pregnancy: a meta-analysis. Am
J Obstet Gynecol 1995;172:5259. PMID:7856680 https://doi.
org/10.1016/0002-9378(95)90567-7
1042. Piper JM, Mitchel EF, Ray WA. Prenatal use of metronidazole and
birth defects: no association. Obstet Gynecol 1993;82:34852.
PMID:8355932
1043. Sheehy O, Santos F, Ferreira E, Berard A. The use of metronidazole
during pregnancy: a review of evidence. Curr Drug Saf 2015;10:170–9.
PMID:25986038 https://doi.org/10.2174/1574886310021505151
24548
1044. Lamont RF, Nhan-Chang CL, Sobel JD, Workowski K, Conde-
Agudelo A, Romero R. Treatment of abnormal vaginal flora in early
pregnancy with clindamycin for the prevention of spontaneous
preterm birth: a systematic review and metaanalysis. Am J Obstet
Gynecol 2011;205:17790. PMID:22071048 https://doi.
org/10.1016/j.ajog.2011.03.047
1045. Odendaal HJ, Popov I, Schoeman J, Smith M, Grové D. Preterm
labour—is bacterial vaginosis involved? S Afr Med J 2002;92:231–4.
PMID:12040953
1046. Carey JC, Klebanoff MA, Hauth JC, et al.; National Institute of
Child Health and Human Development Network of Maternal-
Fetal Medicine Units. Metronidazole to prevent preterm delivery in
pregnant women with asymptomatic bacterial vaginosis. N Engl J Med
2000;342:53440. PMID:10684911 https://doi.org/10.1056/
NEJM200002243420802
1047. Vermeulen GM, Bruinse HW. Prophylactic administration
of clindamycin 2% vaginal cream to reduce the incidence of
spontaneous preterm birth in women with an increased recurrence
risk: a randomised placebo-controlled double-blind trial. Br J
Obstet Gynaecol 1999;106:652–7. PMID:10428520 https://doi.
org/10.1111/j.1471-0528.1999.tb08363.x
1048. McDonald HM, O’Loughlin JA, Vigneswaran R, et al. Impact
of metronidazole therapy on preterm birth in women with
bacterial vaginosis flora (Gardnerella vaginalis): a randomised,
placebo controlled trial. Br J Obstet Gynaecol 1997;104:1391–7.
PMID:9422018 https://doi.org/10.1111/j.1471-0528.1997.
tb11009.x
1049. Ugwumadu A, Manyonda I, Reid F, Hay P. Effect of early
oral clindamycin on late miscarriage and preterm delivery in
asymptomatic women with abnormal vaginal flora and bacterial
vaginosis: a randomised controlled trial. Lancet 2003;361:983–8.
PMID:12660054 https://doi.org/10.1016/S0140-6736(03)12823-1
1050. Subtil D, Brabant G, Tilloy E, et al. Early clindamycin for bacterial
vaginosis in pregnancy (PREMEVA): a multicentre, double-
blind, randomised controlled trial. Lancet 2018;392:21719.
PMID:30322724 https://doi.org/10.1016/S0140-6736(18)31617-9
1051. Erickson SH, Oppenheim GL, Smith GH. Metronidazole in breast
milk. Obstet Gynecol 1981;57:48–50. PMID:7454176
1052. Passmore CM, McElnay JC, Rainey EA, D’Arcy PF. Metronidazole
excretion in human milk and its effect on the suckling neonate. Br
J Clin Pharmacol 1988;26:4551. PMID:3203060 https://doi.
org/10.1111/j.1365-2125.1988.tb03362.x
1053. United Kingdom National Health Service. Medicines Q&A:
metronidazole—is it safe to use with breastfeeding? [Internet].
London, England: United Kingdom National Health Service, UK
Medicines Information; 2012. https://studylib.net/doc/7341270/
metronidazole-in-breastfeeding-mothers
1054. Jamieson DJ, Duerr A, Klein RS, et al. Longitudinal analysis of
bacterial vaginosis: findings from the HIV epidemiology research
study. Obstet Gynecol 2001;98:656–63. PMID:11576584 https://
doi.org/10.1097/00006250-200110000-00023
1055. Rowley J, Vander Hoorn S, Korenromp E, et al. Chlamydia,
gonorrhoea, trichomoniasis and syphilis: global prevalence
and incidence estimates, 2016. Bull World Health Organ
2019;97:548–562P. PMID:31384073 https://doi.org/10.2471/
BLT.18.228486
1056. Hoots BE, Peterman TA, Torrone EA, Weinstock H, Meites E, Bolan
GA. A Trich-y question: should Trichomonas vaginalis infection be
reportable? Sex Transm Dis 2013;40:113–6. PMID:23321992 https://
doi.org/10.1097/OLQ.0b013e31827c08c3
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 171
US Department of Health and Human Services/Centers for Disease Control and Prevention
1057. Flagg EW, Meites E, Phillips C, Papp J, Torrone EA. Prevalence
of Trichomonas vaginalis among civilian, noninstitutionalized male
and female population aged 14 to 59 years: United States, 2013 to
2016. Sex Transm Dis 2019;46:e93–6. PMID:31517807 https://doi.
org/10.1097/OLQ.0000000000001013
1058. Daugherty M, Glynn K, Byler T. Prevalence of Trichomonas vaginalis
infection among US males, 2013–2016. Clin Infect Dis 2019;68:460–5.
PMID:29893808 https://doi.org/10.1093/cid/ciy499c
1059. Alcaide ML, Feaster DJ, Duan R, et al. The incidence of Trichomonas
vaginalis infection in women attending nine sexually transmitted
diseases clinics in the USA. Sex Transm Infect 2016;92:5862.
PMID:26071390 https://doi.org/10.1136/sextrans-2015-052010
1060. Muzny CA, Blackburn RJ, Sinsky RJ, Austin EL, Schwebke JR.
Added benefit of nucleic acid amplification testing for the diagnosis
of Trichomonas vaginalis among men and women attending a
sexually transmitted diseases clinic. Clin Infect Dis 2014;59:83441.
PMID:24928292 https://doi.org/10.1093/cid/ciu446
1061. Meites E, Llata E, Braxton J, et al. Trichomonas vaginalis in selected
U.S. sexually transmitted disease clinics: testing, screening, and
prevalence. Sex Transm Dis 2013;40:8659. PMID:24113409
https://doi.org/10.1097/OLQ.0000000000000038
1062. Ginocchio CC, Chapin K, Smith JS, et al. Prevalence of
Trichomonas vaginalis and coinfection with Chlamydia trachomatis
and Neisseria gonorrhoeae in the United States as determined by
the Aptima Trichomonas vaginalis nucleic acid amplification assay.
J Clin Microbiol 2012;50:2601–8. PMID:22622447 https://doi.
org/10.1128/JCM.00748-12
1063. Shuter J, Bell D, Graham D, Holbrook KA, Bellin EY. Rates of and
risk factors for trichomoniasis among pregnant inmates in New York
City. Sex Transm Dis 1998;25:303–7. PMID:9662764 https://doi.
org/10.1097/00007435-199807000-00006
1064. Sosman JM, MacGowan RJ, Margolis AD, et al.; Project START
Study Group. Screening for sexually transmitted diseases and hepatitis
in 18–29-year-old men recently released from prison: feasibility and
acceptability. Int J STD AIDS 2005;16:117–22. PMID:15825246
https://doi.org/10.1258/0956462053057594
1065. Rogers SM, Turner CF, Hobbs M, et al. Epidemiology of undiagnosed
trichomoniasis in a probability sample of urban young adults. PLoS
One 2014;9:e90548. PMID:24626058 https://doi.org/10.1371/
journal.pone.0090548
1066. Mayer KH, Bush T, Henry K, et al.; SUN Investigators. Ongoing
sexually transmitted disease acquisition and risk-taking behavior
among US HIV-infected patients in primary care: implications
for prevention interventions. Sex Transm Dis 2012;39:17.
PMID:22183836 https://doi.org/10.1097/OLQ.0b013e31823b1922
1067. Seña AC, Miller WC, Hobbs MM, et al. Trichomonas vaginalis
infection in male sexual partners: implications for diagnosis, treatment,
and prevention. Clin Infect Dis 2007;44:13–22. PMID:17143809
https://doi.org/10.1086/511144
1068. Kelley CF, Rosenberg ES, O’Hara BM, Sanchez T, del Rio C,
Sullivan PS. Prevalence of urethral Trichomonas vaginalis in black and
white men who have sex with men. Sex Transm Dis 2012;39:739.
PMID:22902674 https://doi.org/10.1097/OLQ.0b013e318264248b
1069. Sutton M, Sternberg M, Koumans EH, McQuillan G, Berman S,
Markowitz L. The prevalence of Trichomonas vaginalis infection
among reproductive-age women in the United States, 2001–2004.
Clin Infect Dis 2007;45:131926. PMID:17968828 https://doi.
org/10.1086/522532
1070. Peterman TA, Tian LH, Metcalf CA, Malotte CK, Paul SM,
Douglas JM Jr; RESPECT-2 Study Group. Persistent, undetected
Trichomonas vaginalis infections? Clin Infect Dis 2009;48:259–60.
PMID:19113985 https://doi.org/10.1086/595706
1071. Wølner-Hanssen P, Krieger JN, Stevens CE, et al. Clinical manifestations
of vaginal trichomoniasis. JAMA 1989;261:571–6. PMID:2783346
https://doi.org/10.1001/jama.1989.03420040109029
1072. Gray RH, Kigozi G, Serwadda D, et al. The effects of male
circumcision on female partners’ genital tract symptoms and
vaginal infections in a randomized trial in Rakai, Uganda. Am J
Obstet Gynecol 2009;200:42.e1–7. PMID:18976733 https://doi.
org/10.1016/j.ajog.2008.07.069
1073. Sobngwi-Tambekou J, Taljaard D, Nieuwoudt M, Lissouba P,
Puren A, Auvert B. Male circumcision and Neisseria gonorrhoeae,
Chlamydia trachomatis and Trichomonas vaginalis: observations
after a randomised controlled trial for HIV prevention. Sex Transm
Infect 2009;85:116–20. PMID:19074928 https://doi.org/10.1136/
sti.2008.032334
1074. Tsai CS, Shepherd BE, Vermund SH. Does douching increase
risk for sexually transmitted infections? A prospective study in
high-risk adolescents. Am J Obstet Gynecol 2009;200:38.e18.
PMID:18667177 https://doi.org/10.1016/j.ajog.2008.06.026
1075. Silver BJ, Guy RJ, Kaldor JM, Jamil MS, Rumbold AR. Trichomonas
vaginalis as a cause of perinatal morbidity: a systematic review and
meta-analysis. Sex Transm Dis 2014;41:369–76. PMID:24825333
https://doi.org/10.1097/OLQ.0000000000000134
1076. Yang S, Zhao W, Wang H, Wang Y, Li J, Wu X. Trichomonas vaginalis
infection-associated risk of cervical cancer: a meta-analysis. Eur J
Obstet Gynecol Reprod Biol 2018;228:166–73. PMID:29980111
https://doi.org/10.1016/j.ejogrb.2018.06.031
1077. Najafi A, Chaechi Nosrati MR, Ghasemi E, et al. Is there association
between Trichomonas vaginalis infection and prostate cancer
risk?: A systematic review and meta-analysis. Microb Pathog
2019;137:103752. PMID:31539586 https://doi.org/10.1016/j.
micpath.2019.103752
1078. Wang CC, McClelland RS, Reilly M, et al. The effect of treatment
of vaginal infections on shedding of human immunodeficiency virus
type 1. J Infect Dis 2001;183:1017–22. PMID:11237825 https://
doi.org/10.1086/319287
1079. Kissinger P, Amedee A, Clark RA, et al. Trichomonas vaginalis treatment
reduces vaginal HIV-1 shedding. Sex Transm Dis 2009;36:11–6.
PMID:19008776 https://doi.org/10.1097/OLQ.0b013e318186decf
1080. Minkoff H, Grunebaum AN, Schwarz RH, et al. Risk factors
for prematurity and premature rupture of membranes: a
prospective study of the vaginal flora in pregnancy. Am J
Obstet Gynecol 1984;150:965–72. PMID:6391179 https://doi.
org/10.1016/0002-9378(84)90392-2
1081. Cotch MF, Pastorek JG 2nd, Nugent RP, et al.; The Vaginal
Infections and Prematurity Study Group. Trichomonas vaginalis
associated with low birth weight and preterm delivery. Sex
Transm Dis 1997;24:35360. PMID:9243743 https://doi.
org/10.1097/00007435-199707000-00008
1082. Moodley P, Wilkinson D, Connolly C, Moodley J, Sturm AW.
Trichomonas vaginalis is associated with pelvic inflammatory disease in
women infected with human immunodeficiency virus. Clin Infect Dis
2002;34:51922. PMID:11797180 https://doi.org/10.1086/338399
1083. Francis SC, Kent CK, Klausner JD, et al. Prevalence of rectal
Trichomonas vaginalis and Mycoplasma genitalium in male patients
at the San Francisco STD clinic, 2005–2006. Sex Transm Dis
2008;35:797800. PMID:18607317 https://doi.org/10.1097/
OLQ.0b013e318177ec39
1084. Hollman D, Coupey SM, Fox AS, Herold BC. Screening for
Trichomonas vaginalis in high-risk adolescent females with a new
transcription-mediated nucleic acid amplification test (NAAT):
associations with ethnicity, symptoms, and prior and current STIs.
J Pediatr Adolesc Gynecol 2010;23:312–6. PMID:20493735 https://
doi.org/10.1016/j.jpag.2010.03.004
1085. Roth AM, Williams JA, Ly R, et al. Changing sexually transmitted
infection screening protocol will result in improved case finding
for Trichomonas vaginalis among high-risk female populations.
Sex Transm Dis 2011;38:398400. PMID:21217417 https://doi.
org/10.1097/OLQ.0b013e318203e3ce
Recommendations and Reports
172 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
1086. Hobbs MM, Seña AC. Modern diagnosis of Trichomonas vaginalis
infection. Sex Transm Infect 2013;89:434–8. PMID:23633669
https://doi.org/10.1136/sextrans-2013-051057
1087. Kingston MA, Bansal D, Carlin EM. ‘Shelf life’ of Trichomonas
vaginalis. Int J STD AIDS 2003;14:28–9. PMID:12590789 https://
doi.org/10.1258/095646203321043228
1088. Schwebke JR, Hobbs MM, Taylor SN, et al. Molecular testing for
Trichomonas vaginalis in women: results from a prospective U.S.
clinical trial. J Clin Microbiol 2011;49:4106–11. PMID:21940475
https://doi.org/10.1128/JCM.01291-11
1089. Huppert JS, Mortensen JE, Reed JL, et al. Rapid antigen testing
compares favorably with transcription-mediated amplification
assay for the detection of Trichomonas vaginalis in young women.
Clin Infect Dis 2007;45:194–8. PMID:17578778 https://doi.
org/10.1086/518851
1090. Van Der Pol B, Williams JA, Taylor SN, et al. Detection of Trichomonas
vaginalis DNA by use of self-obtained vaginal swabs with the BD
ProbeTec Qx assay on the BD Viper system. J Clin Microbiol
2014;52:8859. PMID:24391200 https://doi.org/10.1128/
JCM.02966-13
1091. Van Der Pol B, Williams JA, Fuller D, Taylor SN, Hook EW 3rd.
Combined testing for chlamydia, gonorrhea, and trichomonas by
use of the BD Max CT/GC/TV assay with genitourinary specimen
types. J Clin Microbiol 2016;55:155–64. PMID:27795343 https://
doi.org/10.1128/JCM.01766-16
1092. Schwebke JR, Gaydos CA, Davis T, et al. Clinical evaluation of the
Cepheid Xpert TV Assay for detection of Trichomonas vaginalis with
prospectively collected specimens from men and women. J Clin
Microbiol 2018;56:e01091-17. PMID:29167292 https://doi.
org/10.1128/JCM.01091-17
1093. Campbell L, Woods V, Lloyd T, Elsayed S, Church DL. Evaluation
of the OSOM Trichomonas rapid test versus wet preparation
examination for detection of Trichomonas vaginalis vaginitis
in specimens from women with a low prevalence of infection.
J Clin Microbiol 2008;46:3467–9. PMID:18685008 https://doi.
org/10.1128/JCM.00671-08
1094. Huppert JS, Hesse E, Kim G, et al. Adolescent women can perform
a point-of-care test for trichomoniasis as accurately as clinicians.
Sex Transm Infect 2010;86:514–9. PMID:20595142 https://doi.
org/10.1136/sti.2009.042168
1095. Sheele JM, Crandall CJ, Arko BL, et al. The OSOM® Trichomonas
Test is unable to accurately diagnose Trichomonas vaginalis from urine
in men. Am J Emerg Med 2019;37:1002–3. PMID:30361151 https://
doi.org/10.1016/j.ajem.2018.10.022
1096. Gaydos CA, Schwebke J, Dombrowski J, et al. Clinical performance
of the Solana® Point-of-Care Trichomonas Assay from clinician-
collected vaginal swabs and urine specimens from symptomatic
and asymptomatic women. Expert Rev Mol Diagn 2017;17:303–6.
PMID:28092466 https://doi.org/10.1080/14737159.2017.1282823
1097. Gaydos CA, Hobbs M, Marrazzo J, et al. Rapid diagnosis of Trichomonas
vaginalis by testing vaginal swabs in an isothermal helicase-dependent
AmpliVue Assay. Sex Transm Dis 2016;43:36973. PMID:27196258
https://doi.org/10.1097/OLQ.0000000000000447
1098. Patil MJ, Nagamoti JM, Metgud SC. Diagnosis of Trichomonas
vaginalis from vaginal specimens by wet mount microscopy, In
Pouch TV culture system, and PCR. J Glob Infect Dis 2012;4:22–5.
PMID:22529623 https://doi.org/10.4103/0974-777X.93756
1099. Lawing LF, Hedges SR, Schwebke JR. Detection of trichomonosis
in vaginal and urine specimens from women by culture and PCR.
J Clin Microbiol 2000;38:3585–8. PMID:11015368 https://doi.
org/10.1128/JCM.38.10.3585-3588.2000
1100. Mohamed OA, Cohen CR, Kungu D, et al. Urine proves a poor
specimen for culture of Trichomonas vaginalis in women. Sex Transm
Infect 2001;77:78–9. PMID:11158705 https://doi.org/10.1136/
sti.77.1.78
1101. Rivers CA, Muzny CA, Schwebke JR. Diagnostic rates differ on
the basis of the number of read days with the use of the InPouch
culture system for Trichomonas vaginalis screening. J Clin Microbiol
2013;51:38756. PMID:24006006 https://doi.org/10.1128/
JCM.02006-13
1103. Audisio T, Pigini T, de Riutort SV, et al. Validity of the Papanicolaou
smear in the diagnosis of Candida spp., Trichomonas vaginalis,
and bacterial vaginosis. J Low Genit Tract Dis 2001;5:2235.
PMID:17050980
1104. Loo SK, Tang WY, Lo KK. Clinical significance of Trichomonas
vaginalis detected in Papanicolaou smear: a survey in female Social
Hygiene Clinic. Hong Kong Med J 2009;15:90–3. PMID:19342733
1105. Howe K, Kissinger PJ. Single-dose compared with multidose
metronidazole for the treatment of trichomoniasis in women: a meta-
analysis. Sex Transm Dis 2017;44:29–34. PMID:27898571 https://
doi.org/10.1097/OLQ.0000000000000537
1105. Kissinger P, Mena L, Levison J, et al. A randomized treatment
trial: single versus 7-day dose of metronidazole for the treatment
of Trichomonas vaginalis among HIV-infected women. J Acquir
Immune Defic Syndr 2010;55:565–71. PMID:21423852 https://
doi.org/10.1097/QAI.0b013e3181eda955
1106. Wood BA, Monro AM. Pharmacokinetics of tinidazole and
metronidazole in women after single large oral doses. Br J Vener Dis
1975;51:51–3. PMID:1092424 https://doi.org/10.1136/sti.51.1.51
1107. Viitanen J, Haataja H, Männistö PT. Concentrations of metronidazole
and tinidazole in male genital tissues. Antimicrob Agents Chemother
1985;28:8124. PMID:4083864 https://doi.org/10.1128/
AAC.28.6.812
1108. Gabriel G, Robertson E, Thin RN. Single dose treatment of
trichomoniasis. J Int Med Res 1982;10:12930. PMID:7067925
https://doi.org/10.1177/030006058201000212
1109. Mati JK, Wallace RJ. The treatment of trichomonal vaginitis using a
single dose of tinidazole by mouth. East Afr Med J 1974;51:883–8.
PMID:4616829
1110. Anjaeyulu R, Gupte SA, Desai DB. Single-dose treatment of
trichomonal vaginitis: a comparison of tinidazole and metronidazole.
J Int Med Res 1977;5:438–41. PMID:590601
1111. Apte VV, Packard RS. Tinidazole in the treatment of trichomoniasis,
giardiasis and amoebiasis. Report of a multicentre study.
Drugs 1978;15(Suppl 1):438. PMID:657995 https://doi.
org/10.2165/00003495-197800151-00009
1112. O-Prasertsawat P, Jetsawangsri T. Split-dose metronidazole or
single-dose tinidazole for the treatment of vaginal trichomoniasis.
Sex Transm Dis 1992;19:2957. PMID:1411848 https://doi.
org/10.1097/00007435-199209000-00011
1113. Kawamura N. Metronidazole and tinidazole in a single large dose for
treating urogenital infections with Trichomonas vaginalis in men. Br
J Vener Dis 1978;54:81–3. PMID:305809 https://doi.org/10.1136/
sti.54.2.81
1114. Forna F, Gülmezoglu AM. Interventions for treating trichomoniasis
in women. Cochrane Database Syst Rev 2003;(2):CD000218.
PMID:12804391
1115. Cu-Uvin S, Ko H, Jamieson DJ, et al.; HIV Epidemiology Research
Study (HERS) Group. Prevalence, incidence, and persistence or
recurrence of trichomoniasis among human immunodeficiency
virus (HIV)-positive women and among HIV-negative women at
high risk for HIV infection. Clin Infect Dis 2002;34:140611.
PMID:11981738 https://doi.org/10.1086/340264
1116. Schwebke JR, Barrientes FJ. Prevalence of Trichomonas vaginalis
isolates with resistance to metronidazole and tinidazole. Antimicrob
Agents Chemother 2006;50:420910. PMID:17000740 https://doi.
org/10.1128/AAC.00814-06
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 173
US Department of Health and Human Services/Centers for Disease Control and Prevention
1117. Van Der Pol B, Williams JA, Orr DP, Batteiger BE, Fortenberry
JD. Prevalence, incidence, natural history, and response to
treatment of Trichomonas vaginalis infection among adolescent
women. J Infect Dis 2005;192:2039–44. PMID:16288365 https://
doi.org/10.1086/498217
1118. Kirkcaldy RD, Augostini P, Asbel LE, et al. Trichomonas vaginalis
antimicrobial drug resistance in 6 US cities, STD Surveillance
Network, 2009–2010. Emerg Infect Dis 2012;18:93943.
PMID:22608054 https://doi.org/10.3201/eid1806.111590
1119. Crowell AL, Sanders-Lewis KA, Secor WE. In vitro metronidazole
and tinidazole activities against metronidazole-resistant strains
of Trichomonas vaginalis. Antimicrob Agents Chemother
2003;47:14079. PMID:12654679 https://doi.org/10.1128/
AAC.47.4.1407-1409.2003
1120. Muzny CA, Mena L, Lillis RA, et al. A comparison of 2 g single-dose
versus 7-day 500 mg twice daily metronidazole for the treatment
trichomoniasis in women by selected clinical factors. Am J Obstet
Gynecol 2019;221:669. https://doi.org/10.1016/j.ajog.2019.10.079
1121. Sobel JD, Nyirjesy P, Brown W. Tinidazole therapy for metronidazole-
resistant vaginal trichomoniasis. Clin Infect Dis 2001;33:1341–6.
PMID:11565074 https://doi.org/10.1086/323034
1122. Nyirjesy P, Gilbert J, Mulcahy LJ. Resistant trichomoniasis: successful
treatment with combination therapy. Sex Transm Dis 2011;38:962–3.
PMID:21934573 https://doi.org/10.1097/OLQ.0b013e31822037e4
1123. Muzny C, Barnes A, Mena L. Symptomatic Trichomonas
vaginalis infection in the setting of severe nitroimidazole allergy:
successful treatment with boric acid. Sex Health 2012;9:38991.
PMID:22877600 https://doi.org/10.1071/SH11114
1124. Aggarwal A, Shier RM. Recalcitrant Trichomonas vaginalis infections
successfully treated with vaginal acidification. J Obstet Gynaecol
Can 2008;30:55–8. PMID:18198069 https://doi.org/10.1016/
S1701-2163(16)32714-1
1125. Dan M, Sobel JD. Failure of nitazoxanide to cure trichomoniasis in
three women. Sex Transm Dis 2007;34:813–4. PMID:17551415
https://doi.org/10.1097/NMD.0b013e31802f5d9a
1126. Seña AC, Bachmann LH, Hobbs MM. Persistent and recurrent
Trichomonas vaginalis infections: epidemiology, treatment and
management considerations. Expert Rev Anti Infect Ther
2014;12:67385. PMID:24555561 https://doi.org/10.1586/1478
7210.2014.887440
1127. Helms DJ, Mosure DJ, Secor WE, Workowski KA. Management
of Trichomonas vaginalis in women with suspected metronidazole
hypersensitivity. Am J Obstet Gynecol 2008;198:370.e17.
PMID:18221927 https://doi.org/10.1016/j.ajog.2007.10.795
1128. Gendelman SR, Pien LC, Gutta RC, Abouhassan SR. Modified oral
metronidazole desensitization protocol. Allergy Rhinol (Providence)
2014;5:669. PMID:24612959 https://doi.org/10.2500/
ar.2014.5.0080
1129. Nyirjesy P, Sobel JD, Weitz MV, Leaman DJ, Gelone SP. Difficult-to-
treat trichomoniasis: results with paromomycin cream. Clin Infect Dis
1998;26:986–8. PMID:9564487 https://doi.org/10.1086/513951
1130. Klebanoff MA, Carey JC, Hauth JC, et al.; National Institute of
Child Health and Human Development Network of Maternal-Fetal
Medicine Units. Failure of metronidazole to prevent preterm delivery
among pregnant women with asymptomatic Trichomonas vaginalis
infection. N Engl J Med 2001;345:48793. PMID:11519502 https://
doi.org/10.1056/NEJMoa003329
1131. Stringer E, Read JS, Hoffman I, Valentine M, Aboud S, Goldenberg
RL. Treatment of trichomoniasis in pregnancy in sub-Saharan Africa
does not appear to be associated with low birth weight or preterm
birth. S Afr Med J 2010;100:58–64. PMID:20429491
1132. Caro-Patón T, Carvajal A, Martin de Diego I, Martin-Arias
LH, Alvarez Requejo A, Rodríguez Pinilla E. Is metronidazole
teratogenic? A meta-analysis. Br J Clin Pharmacol 1997;44:179–82.
PMID:9278206 https://doi.org/10.1046/j.1365-2125.1997.00660.x
1133. Gülmezoglu AM. Interventions for trichomoniasis in pregnancy.
Cochrane Database Syst Rev 2002;(3):CD000220. PMID:12137609
1134. Goldenberg RL, Mwatha A, Read JS, et al.; Hptn024 Team.
The HPTN 024 Study: the efficacy of antibiotics to prevent
chorioamnionitis and preterm birth. Am J Obstet Gynecol
2006;194:65061. PMID:16522393 https://doi.org/10.1016/j.
ajog.2006.01.004
1135. Mann JR, McDermott S, Zhou L, Barnes TL, Hardin J. Treatment
of trichomoniasis in pregnancy and preterm birth: an observational
study. J Womens Health (Larchmt) 2009;18:493–7. PMID:19361316
https://doi.org/10.1089/jwh.2008.0964
1136. Carter JE, Whithaus KC. Neonatal respiratory tract involvement
by Trichomonas vaginalis: a case report and review of the literature.
Am J Trop Med Hyg 2008;78:17–9. PMID:18187779 https://doi.
org/10.4269/ajtmh.2008.78.17
1137. Trintis J, Epie N, Boss R, Riedel S. Neonatal Trichomonas vaginalis
infection: a case report and review of literature. Int J STD AIDS
2010;21:6067. PMID:20975098 https://doi.org/10.1258/
ijsa.2010.010174
1138. Miller M, Liao Y, Wagner M, Korves C. HIV, the clustering of sexually
transmitted infections, and sex risk among African American women
who use drugs. Sex Transm Dis 2008;35:696702. PMID:18418289
https://doi.org/10.1097/OLQ.0b013e31816b1fb8
1139. Anderson BL, Firnhaber C, Liu T, et al. Effect of trichomoniasis
therapy on genital HIV viral burden among African women.
Sex Transm Dis 2012;39:63842. PMID:22797689 https://doi.
org/10.1097/OLQ.0b013e31825725ad
1140. Masese LN, Graham SM, Gitau R, et al. A prospective study of vaginal
trichomoniasis and HIV-1 shedding in women on antiretroviral
therapy. BMC Infect Dis 2011;11:307. PMID:22047086 https://
doi.org/10.1186/1471-2334-11-307
1141. Balkus JE, Richardson BA, Mochache V, et al. A prospective
cohort study comparing the effect of single-dose 2 g metronidazole
on Trichomonas vaginalis infection in HIV-seropositive versus
HIV-seronegative women. Sex Transm Dis 2013;40:499505.
PMID:23677023 https://doi.org/10.1097/OLQ.0b013e31828fce34
1142. Gumbo FZ, Duri K, Kandawasvika GQ, et al. Risk factors of HIV
vertical transmission in a cohort of women under a PMTCT program
at three peri-urban clinics in a resource-poor setting. J Perinatol
2010;30:71723. PMID:20336078 https://doi.org/10.1038/
jp.2010.31
1143. Brüggemann RJ, Alffenaar JW, Blijlevens NM, et al. Clinical relevance
of the pharmacokinetic interactions of azole antifungal drugs with
other coadministered agents. Clin Infect Dis 2009;48:144158.
PMID:19361301 https://doi.org/10.1086/598327
1144. Shahid Z, Sobel JD. Reduced fluconazole susceptibility of Candida
albicans isolates in women with recurrent vulvovaginal candidiasis:
effects of long-term fluconazole therapy. Diagn Microbiol Infect
Dis 2009;64:354–6. PMID:19501794 https://doi.org/10.1016/j.
diagmicrobio.2009.03.021
1145. Marchaim D, Lemanek L, Bheemreddy S, Kaye KS, Sobel JD.
Fluconazole-resistant Candida albicans vulvovaginitis. Obstet Gynecol
2012;120:140714. PMID:23168767 https://doi.org/10.1097/
AOG.0b013e31827307b2
1146. Denning DW, Kneale M, Sobel JD, Rautemaa-Richardson R. Global
burden of recurrent vulvovaginal candidiasis: a systematic review.
Lancet Infect Dis 2018;18:e339–47. PMID:30078662 https://doi.
org/10.1016/S1473-3099(18)30103-8
1147. Crouss T, Sobel JD, Smith K, Nyirjesy P. Long-term outcomes
of women with recurrent vulvovaginal candidiasis after a course
of maintenance antifungal therapy. J Low Genit Tract Dis
2018;22:3826. PMID:29975334 https://doi.org/10.1097/
LGT.0000000000000413
Recommendations and Reports
174 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
1148. Kennedy MA, Sobel JD. Vulvovaginal candidiasis caused by
non-albicans Candida species: new insights. Curr Infect Dis Rep
2010;12:46570. PMID:21308556 https://doi.org/10.1007/
s11908-010-0137-9
1149. Sobel JD, Chaim W, Nagappan V, Leaman D. Treatment of vaginitis
caused by Candida glabrata: use of topical boric acid and flucytosine.
Am J Obstet Gynecol 2003;189:1297300. PMID:14634557 https://
doi.org/10.1067/S0002-9378(03)00726-9
1150. Mølgaard-Nielsen D, Svanström H, Melbye M, Hviid A, Pasternak
B. Association between use of oral fluconazole during pregnancy and
risk of spontaneous abortion and stillbirth. JAMA 2016;315:58–67.
PMID:26746458 https://doi.org/10.1001/jama.2015.17844
1151. Bérard A, Sheehy O, Zhao JP, et al. Associations between low- and
high-dose oral fluconazole and pregnancy outcomes: 3 nested case-
control studies. CMAJ 2019;191:E17987. PMID:30782643 https://
doi.org/10.1503/cmaj.180963
1152. Ohmit SE, Sobel JD, Schuman P, et al.; HIV Epidemiology Research
Study (HERS) Group. Longitudinal study of mucosal Candida species
colonization and candidiasis among human immunodeficiency
virus (HIV)-seropositive and at-risk HIV-seronegative women.
J Infect Dis 2003;188:11827. PMID:12825180 https://doi.
org/10.1086/375746
1153. Duerr A, Heilig CM, Meikle SF, et al.; HER Study Group.
Incident and persistent vulvovaginal candidiasis among human
immunodeficiency virus-infected women: risk factors and severity.
Obstet Gynecol 2003;101:54856. PMID:12636961 https://doi.
org/10.1097/00006250-200303000-00022
1154. Vazquez JA, Peng G, Sobel JD, et al. Evolution of antifungal
susceptibility among Candida species isolates recovered from human
immunodeficiency virus-infected women receiving fluconazole
prophylaxis. Clin Infect Dis 2001;33:106975. PMID:11528582
https://doi.org/10.1086/322641
1155. Darville T; Pelvic Inflammatory Disease Workshop Proceedings
Committee. Pelvic inflammatory disease: identifying research gaps—
proceedings of a workshop sponsored by Department of Health and
Human Services/National Institutes of Health/National Institute of
Allergy and Infectious Diseases, November 3–4, 2011. Sex Transm
Dis 2013;40:7617. PMID:24275724 https://doi.org/10.1097/
OLQ.0000000000000028
1156. Wiesenfeld HC, Sweet RL, Ness RB, Krohn MA, Amortegui AJ,
Hillier SL. Comparison of acute and subclinical pelvic inflammatory
disease. Sex Transm Dis 2005;32:400–5. PMID:15976596 https://
doi.org/10.1097/01.olq.0000154508.26532.6a
1157. Wiesenfeld HC, Hillier SL, Meyn LA, Amortegui AJ, Sweet RL.
Subclinical pelvic inflammatory disease and infertility. Obstet Gynecol
2012;120:3743. PMID:22678036 https://doi.org/10.1097/
AOG.0b013e31825a6bc9
1158. Ness RB, Soper DE, Holley RL, et al. Effectiveness of inpatient and
outpatient treatment strategies for women with pelvic inflammatory
disease: results from the Pelvic Inflammatory Disease Evaluation and
Clinical Health (PEACH) Randomized Trial. Am J Obstet Gynecol
2002;186:92937. PMID:12015517 https://doi.org/10.1067/
mob.2002.121625
1159. Burnett AM, Anderson CP, Zwank MD. Laboratory-confirmed
gonorrhea and/or chlamydia rates in clinically diagnosed pelvic
inflammatory disease and cervicitis. Am J Emerg Med 2012;30:1114–7.
PMID:22030186 https://doi.org/10.1016/j.ajem.2011.07.014
1160. Wiesenfeld HC, Meyn LA, Darville T, Macio IS, Hillier SL. A
randomized controlled trial of ceftriaxone and doxycycline, with or
without metronidazole, for the treatment of acute pelvic inflammatory
disease. Clin Infect Dis 2021;72:1181–9. PMID:32052831 https://
doi.org/10.1093/cid/ciaa101
1161. Ness RB, Kip KE, Hillier SL, et al. A cluster analysis of bacterial
vaginosis-associated microflora and pelvic inflammatory disease.
Am J Epidemiol 2005;162:585–90. PMID:16093289 https://doi.
org/10.1093/aje/kwi243
1162. Scholes D, Stergachis A, Heidrich FE, Andrilla H, Holmes KK,
Stamm WE. Prevention of pelvic inflammatory disease by screening
for cervical chlamydial infection. N Engl J Med 1996;334:1362–6.
PMID:8614421 https://doi.org/10.1056/NEJM199605233342103
1163. Oakeshott P, Kerry S, Aghaizu A, et al. Randomised controlled trial of
screening for Chlamydia trachomatis to prevent pelvic inflammatory
disease: the POPI (Prevention of Pelvic Infection) trial. BMJ
2010;340:c1642. PMID:20378636 https://doi.org/10.1136/bmj.
c1642
1164. Peipert JF, Ness RB, Blume J, et al.; Pelvic Inflammatory Disease
Evaluation and Clinical Health Study Investigators. Clinical
predictors of endometritis in women with symptoms and signs of
pelvic inflammatory disease. Am J Obstet Gynecol 2001;184:85664.
PMID:11303192 https://doi.org/10.1067/mob.2001.113847
1165. Gaitán H, Angel E, Diaz R, Parada A, Sanchez L, Vargas C. Accuracy
of five different diagnostic techniques in mild-to-moderate pelvic
inflammatory disease. Infect Dis Obstet Gynecol 2002;10:171–80.
PMID:12648310 https://doi.org/10.1155/S1064744902000194
1166. Vicetti Miguel RD, Chivukula M, Krishnamurti U, et al.
Limitations of the criteria used to diagnose histologic endometritis
in epidemiologic pelvic inflammatory disease research. Pathol Res
Pract 2011;207:680–5. PMID:21996319 https://doi.org/10.1016/j.
prp.2011.08.007
1167. Jacobson L, Weström L. Objectivized diagnosis of acute pelvic
inflammatory disease. Diagnostic and prognostic value of
routine laparoscopy. Am J Obstet Gynecol 1969;105:108898.
PMID:4242830 https://doi.org/10.1016/0002-9378(69)90132-X
1168. Sellors J, Mahony J, Goldsmith C, et al. The accuracy of clinical
findings and laparoscopy in pelvic inflammatory disease. Am J
Obstet Gynecol 1991;164:11320. PMID:1824740 https://doi.
org/10.1016/0002-9378(91)90639-9
1169. Bevan CD, Johal BJ, Mumtaz G, Ridgway GL, Siddle NC. Clinical,
laparoscopic and microbiological findings in acute salpingitis: report
on a United Kingdom cohort. Br J Obstet Gynaecol 1995;102:407–14.
PMID:7612536 https://doi.org/10.1111/j.1471-0528.1995.
tb11294.x
1170. Jaiyeoba O, Soper DE. A practical approach to the diagnosis of pelvic
inflammatory disease. Infect Dis Obstet Gynecol 2011;2011:753037.
PMID:21822367 https://doi.org/10.1155/2011/753037
1171. Sweet RL. Treatment of acute pelvic inflammatory disease. Infect Dis
Obstet Gynecol 2011;2011:561909. PMID:22228985 https://doi.
org/10.1155/2011/561909
1172. Smith KJ, Ness RB, Wiesenfeld HC, Roberts MS. Cost-effectiveness
of alternative outpatient pelvic inflammatory disease treatment
strategies. Sex Transm Dis 2007;34:960–6. PMID:18077847 https://
doi.org/10.1097/01.olq.0000225321.61049.13
1173. Petrina MAB, Cosentino LA, Wiesenfeld HC, Darville T, Hillier SL.
Susceptibility of endometrial isolates recovered from women with
clinical pelvic inflammatory disease or histological endometritis to
antimicrobial agents. Anaerobe 2019;56:615. PMID:30753898
https://doi.org/10.1016/j.anaerobe.2019.02.005
1174. Haggerty CL, Ness RB, Amortegui A, et al. Endometritis does not
predict reproductive morbidity after pelvic inflammatory disease.
Am J Obstet Gynecol 2003;188:141–8. PMID:12548208 https://
doi.org/10.1067/mob.2003.87
1175. Haggerty CL, Totten PA, Tang G, et al. Identification of novel
microbes associated with pelvic inflammatory disease and infertility.
Sex Transm Infect 2016;92:441–6. PMID:26825087 https://doi.
org/10.1136/sextrans-2015-052285
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 175
US Department of Health and Human Services/Centers for Disease Control and Prevention
1176. Ness RB, Randall H, Richter HE, et al.; Pelvic Inflammatory Disease
Evaluation and Clinical Health Study Investigators. Condom use and
the risk of recurrent pelvic inflammatory disease, chronic pelvic pain,
or infertility following an episode of pelvic inflammatory disease.
Am J Public Health 2004;94:1327–9. PMID:15284036 https://doi.
org/10.2105/AJPH.94.8.1327
1177. McGregor JA, Crombleholme WR, Newton E, Sweet RL, Tuomala
R, Gibbs RS. Randomized comparison of ampicillin-sulbactam
to cefoxitin and doxycycline or clindamycin and gentamicin in
the treatment of pelvic inflammatory disease or endometritis.
Obstet Gynecol 1994;83:9981004. PMID:8190448 https://doi.
org/10.1097/00006250-199406000-00020
1178. Bevan CD, Ridgway GL, Rothermel CD. Efficacy and safety of
azithromycin as monotherapy or combined with metronidazole
compared with two standard multidrug regimens for the treatment
of acute pelvic inflammatory disease. J Int Med Res 2003;31:45–54.
PMID:12635534 https://doi.org/10.1177/147323000303100108
1179. Heystek M, Ross JD; PID Study Group. A randomized double-
blind comparison of moxifloxacin and doxycycline/metronidazole/
ciprofloxacin in the treatment of acute, uncomplicated pelvic
inflammatory disease. Int J STD AIDS 2009;20:6905.
PMID:19815913 https://doi.org/10.1258/ijsa.2008.008495
1180. Boothby M, Page J, Pryor R, Ross JD. A comparison of treatment
outcomes for moxifloxacin versus ofloxacin/metronidazole for first-
line treatment of uncomplicated non-gonococcal pelvic inflammatory
disease. Int J STD AIDS 2010;21:195–7. PMID:20215625 https://
doi.org/10.1258/ijsa.2009.009374
1181. Judlin P, Liao Q, Liu Z, Reimnitz P, Hampel B, Arvis P. Efficacy and
safety of moxifloxacin in uncomplicated pelvic inflammatory disease:
the MONALISA study. BJOG 2010;117:147584. PMID:20716255
https://doi.org/10.1111/j.1471-0528.2010.02687.x
1182. Korn AP. Pelvic inflammatory disease in women infected with HIV.
AIDS Patient Care STDS 1998;12:431–4. PMID:11361990 https://
doi.org/10.1089/apc.1998.12.431
1183. Irwin KL, Moorman AC, O’Sullivan MJ, et al. Influence of human
immunodeficiency virus infection on pelvic inflammatory disease.
Obstet Gynecol 2000;95:525–34. PMID:10725484
1184. Bukusi EA, Cohen CR, Stevens CE, et al. Effects of human
immunodeficiency virus 1 infection on microbial origins of pelvic
inflammatory disease and on efficacy of ambulatory oral therapy. Am
J Obstet Gynecol 1999;181:1374–81. PMID:10601915 https://doi.
org/10.1016/S0002-9378(99)70378-9
1185. Mugo NR, Kiehlbauch JA, Nguti R, et al. Effect of human
immunodeficiency virus-1 infection on treatment outcome of acute
salpingitis. Obstet Gynecol 2006;107:80712. PMID:16582116
https://doi.org/10.1097/01.AOG.0000207597.70524.e8
1186. Grimes DA. Intrauterine device and upper-genital-tract infection.
Lancet 2000;356:1013–9. PMID:11041414 https://doi.org/10.1016/
S0140-6736(00)02699-4
1187. Viberga I, Odlind V, Lazdane G, Kroica J, Berglund L, Olofsson S.
Microbiology profile in women with pelvic inflammatory disease in
relation to IUD use. Infect Dis Obstet Gynecol 2005;13:183–90.
PMID:16338777 https://doi.org/10.1155/2005/376830
1188. Jatlaoui TC, Riley HEM, Curtis KM. The safety of intrauterine
devices among young women: a systematic review. Contraception
2017;95:1739. PMID:27771475 https://doi.org/10.1016/j.
contraception.2016.10.006
1189. Chen MJ, Kim CR, Whitehouse KC, Berry-Bibee E, Gaffield ME.
Development, updates, and future directions of the World Health
Organization Selected Practice Recommendations for Contraceptive
Use. Int J Gynaecol Obstet 2017;136:1139. PMID:28099730
https://doi.org/10.1002/ijgo.12064
1190. Tepper NK, Steenland MW, Gaffield ME, Marchbanks PA, Curtis
KM. Retention of intrauterine devices in women who acquire
pelvic inflammatory disease: a systematic review. Contraception
2013;87:65560. PMID:23040135 https://doi.org/10.1016/j.
contraception.2012.08.011
1191. Louette A, Krahn J, Caine V, Ha S, Lau TTY, Singh AE. Treatment
of acute epididymitis: a systematic review and discussion of the
implications for treatment based on etiology. Sex Transm Dis
2018;45:e1048. PMID:30044339 https://doi.org/10.1097/
OLQ.0000000000000901
1192. Pilatz A, Hossain H, Kaiser R, et al. Acute epididymitis revisited: impact
of molecular diagnostics on etiology and contemporary guideline
recommendations. Eur Urol 2015;68:42835. PMID:25542628
https://doi.org/10.1016/j.eururo.2014.12.005
1193. Hongo H, Kikuchi E, Matsumoto K, et al. Novel algorithm for
management of acute epididymitis. Int J Urol 2017;24:827.
PMID:27714879 https://doi.org/10.1111/iju.13236
1194. de Villiers EM, Fauquet C, Broker TR, Bernard HU, zur Hausen
H. Classification of papillomaviruses. Virology 2004;324:1727.
PMID:15183049 https://doi.org/10.1016/j.virol.2004.03.033
1195. Myers ER, McCrory DC, Nanda K, Bastian L, Matchar
DB. Mathematical model for the natural history of human
papillomavirus infection and cervical carcinogenesis. Am J Epidemiol
2000;151:115871. PMID:10905528 https://doi.org/10.1093/
oxfordjournals.aje.a010166
1196. Chesson HW, Dunne EF, Hariri S, Markowitz LE. The estimated
lifetime probability of acquiring human papillomavirus in the United
States. Sex Transm Dis 2014;41:660–4. PMID:25299412 https://doi.
org/10.1097/OLQ.0000000000000193
1197. Cogliano V, Baan R, Straif K, Grosse Y, Secretan B, El Ghissassi F;
WHO International Agency for Research on Cancer. Carcinogenicity of
human papillomaviruses. Lancet Oncol 2005;6:204. PMID:15830458
https://doi.org/10.1016/S1470-2045(05)70086-3
1198. Senkomago V, Henley SJ, Thomas CC, Mix JM, Markowitz LE,
Saraiya M. Human papillomavirus-attributable cancers—United
States, 2012–2016. MMWR Morb Mortal Wkly Rep 2019;68:724–8.
PMID:31437140 https://doi.org/10.15585/mmwr.mm6833a3
1199. Chesson HW, Ekwueme DU, Saraiya M, Watson M, Lowy DR,
Markowitz LE. Estimates of the annual direct medical costs of
the prevention and treatment of disease associated with human
papillomavirus in the United States. Vaccine 2012;30:60169.
PMID:22867718 https://doi.org/10.1016/j.vaccine.2012.07.056
1200. Markowitz LE, Hariri S, Lin C, et al. Reduction in human
papillomavirus (HPV) prevalence among young women following
HPV vaccine introduction in the United States, National Health
and Nutrition Examination Surveys, 2003–2010. J Infect Dis
2013;208:38593. PMID:23785124 https://doi.org/10.1093/infdis/
jit192
1201. Flagg EW, Schwartz R, Weinstock H. Prevalence of anogenital warts
among participants in private health plans in the United States,
2003–2010: potential impact of human papillomavirus vaccination.
Am J Public Health 2013;103:1428–35. PMID:23763409 https://
doi.org/10.2105/AJPH.2012.301182
1202. McClung NM, Lewis RM, Gargano JW, Querec T, Unger ER,
Markowitz LE. Declines in vaccine-type human papillomavirus
prevalence in females across racial/ethnic groups: data from a national
survey. J Adolesc Health 2019;65:71522. PMID:31515134 https://
doi.org/10.1016/j.jadohealth.2019.07.003
1203. Drolet M, Bénard É, rez N, et al.; HPV Vaccination Impact
Study Group. Population-level impact and herd effects
following the introduction of human papillomavirus vaccination
programmes: updated systematic review and meta-analysis. Lancet
2019;394:497509. PMID:31255301 https://doi.org/10.1016/
S0140-6736(19)30298-3
Recommendations and Reports
176 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
1204. Mayhew A, Mullins TL, Ding L, et al. Risk perceptions and subsequent
sexual behaviors after HPV vaccination in adolescents. Pediatrics
2014;133:40411. PMID:24488747 https://doi.org/10.1542/
peds.2013-2822
1205. Brouwer AF, Delinger RL, Eisenberg MC, et al. HPV vaccination has
not increased sexual activity or accelerated sexual debut in a college-
aged cohort of men and women. BMC Public Health 2019;19:821.
PMID:31238911 https://doi.org/10.1186/s12889-019-7134-1
1206. Garland SM, Steben M, Sings HL, et al. Natural history of genital
warts: analysis of the placebo arm of 2 randomized phase III trials
of a quadrivalent human papillomavirus (types 6, 11, 16, and 18)
vaccine. J Infect Dis 2009;199:805–14. PMID:19199546 https://
doi.org/10.1086/597071
1207. Flagg EW, Torrone EA. Declines in anogenital warts among age groups
most likely to be impacted by human papillomavirus vaccination,
United States, 2006–2014. Am J Public Health 2018;108:112–9.
PMID:29161070 https://doi.org/10.2105/AJPH.2017.304119
1208. Hariri S, Schuler MS, Naleway AL, et al. Human papillomavirus
vaccine effectiveness against incident genital warts among
female health-plan enrollees, United States. Am J Epidemiol
2018;187:298–305. PMID:28641366 https://doi.org/10.1093/aje/
kwx253
1209. Wangu Z, Hsu KK. Impact of HPV vaccination on anogenital
warts and respiratory papillomatosis. Hum Vaccin Immunother
2016;12:135762. PMID:27217191 https://doi.org/10.1080/216
45515.2016.1172754
1210. Swedish KA, Goldstone SE. Prevention of anal condyloma with
quadrivalent human papillomavirus vaccination of older men who
have sex with men. PLoS One 2014;9:e93393. PMID:24714693
https://doi.org/10.1371/journal.pone.0093393
1211. Sandø N, Kofoed K, Zachariae C, Fouchard J. A reduced national
incidence of anogenital warts in young Danish men and women
after introduction of a national quadrivalent human papillomavirus
vaccination programme for young women—an ecological study.
Acta Derm Venereol 2014;94:28892. PMID:24150529 https://doi.
org/10.2340/00015555-1721
1212. Herweijer E, Ploner A, Sparén P. Substantially reduced incidence
of genital warts in women and men six years after HPV vaccine
availability in Sweden. Vaccine 2018;36:191720. PMID:29523448
https://doi.org/10.1016/j.vaccine.2018.02.097
1213. Harrison C, Britt H, Garland S, et al. Decreased management of
genital warts in young women in Australian general practice post
introduction of national HPV vaccination program: results from a
nationally representative cross-sectional general practice study. PLoS
One 2014;9:e105967. PMID:25180698 https://doi.org/10.1371/
journal.pone.0105967
1214. Canvin M, Sinka K, Hughes G, Mesher D. Decline in genital
warts diagnoses among young women and young men since the
introduction of the bivalent HPV (16/18) vaccination programme
in England: an ecological analysis. Sex Transm Infect 2017;93:125–8.
PMID:27365492 https://doi.org/10.1136/sextrans-2016-052626
1215. Chow EP, Read TR, Wigan R, et al. Ongoing decline in genital
warts among young heterosexuals 7 years after the Australian
human papillomavirus (HPV) vaccination programme. Sex Transm
Infect 2015;91:214–9. PMID:25305210 https://doi.org/10.1136/
sextrans-2014-051813
1216. Petrosky EY, Liu G, Hariri S, Markowitz LE. Human papillomavirus
vaccination and age at first sexual activity, National Health and
Nutrition Examination Survey. Clin Pediatr (Phila) 2017;56:36370.
PMID:27609513 https://doi.org/10.1177/0009922816660541
1217. Gotovtseva EP, Kapadia AS, Smolensky MH, Lairson DR. Optimal
frequency of imiquimod (aldara) 5% cream for the treatment of
external genital warts in immunocompetent adults: a meta-analysis.
Sex Transm Dis 2008;35:34651. PMID:18360317 https://doi.
org/10.1097/OLQ.0b013e31815ea8d1
1218. Baker DA, Ferris DG, Martens MG, et al. Imiquimod 3.75%
cream applied daily to treat anogenital warts: combined results from
women in two randomized, placebo-controlled studies. Infect Dis
Obstet Gynecol 2011;2011:806105. PMID:21876641 https://doi.
org/10.1155/2011/806105
1219. Mashiah J, Brenner S. Possible mechanisms in the induction of vitiligo-
like hypopigmentation by topical imiquimod. Clin Exp Dermatol
2008;33:74–6. PMID:17979992
1220. Domingues E, Chaney KC, Scharf MJ, Wiss K. Imiquimod reactivation
of lichen planus. Cutis 2012;89:276–7, 283. PMID:22838091
1221. Patel U, Mark NM, Machler BC, Levine VJ. Imiquimod 5% cream
induced psoriasis: a case report, summary of the literature and
mechanism. Br J Dermatol 2011;164:6702. PMID:21062268
https://doi.org/10.1111/j.1365-2133.2010.10124.x
1222. Kumar B, Narang T. Local and systemic adverse effects to topical
imiquimod due to systemic immune stimulation. Sex Transm
Infect 2011;87:432. PMID:21606474 https://doi.org/10.1136/
sextrans-2011-050025
1223. Stockfleth E, Beti H, Orasan R, et al. Topical Polyphenon E in
the treatment of external genital and perianal warts: a randomized
controlled trial. Br J Dermatol 2008;158:132938. PMID:18363746
https://doi.org/10.1111/j.1365-2133.2008.08520.x
1224. Gross G, Meyer KG, Pres H, Thielert C, Tawfik H, Mescheder A. A
randomized, double-blind, four-arm parallel-group, placebo-controlled
Phase II/III study to investigate the clinical efficacy of two galenic
formulations of Polyphenon E in the treatment of external genital warts.
J Eur Acad Dermatol Venereol 2007;21:1404–12. PMID:17958849
https://doi.org/10.1111/j.1468-3083.2007.02441.x
1225. Tatti S, Swinehart JM, Thielert C, Tawfik H, Mescheder A, Beutner
KR. Sinecatechins, a defined green tea extract, in the treatment
of external anogenital warts: a randomized controlled trial.
Obstet Gynecol 2008;111:1371–9. PMID:18515521 https://doi.
org/10.1097/AOG.0b013e3181719b60
1226. National Institute for Occupational Safety and Health. Control of
smoke from laser/electric surgical procedures. Washington, DC:
US Department of Health and Human Services, CDC, National
Institute for Occupational Safety and Health; 1996. https://www.
cdc.gov/niosh/docs/hazardcontrol/pdfs/hc11.pdf?id=10.26616/
NIOSHPUB96128
1227. Filley CM, Graff-Richard NR, Lacy JR, Heitner MA, Earnest MP.
Neurologic manifestations of podophyllin toxicity. Neurology
1982;32:30811. PMID:7199647 https://doi.org/10.1212/
WNL.32.3.308
1228. Conard PF, Hanna N, Rosenblum M, Gross JB. Delayed recognition
of podophyllum toxicity in a patient receiving epidural morphine.
Anesth Analg 1990;71:1913. PMID:2375521 https://doi.
org/10.1213/00000539-199008000-00013
1229. Karol MD, Conner CS, Watanabe AS, Murphrey KJ.
Podophyllum: suspected teratogenicity from topical application.
Clin Toxicol 1980;16:2836. PMID:7398215 https://doi.
org/10.3109/15563658008989950
1230. Silverberg MJ, Thorsen P, Lindeberg H, Grant LA, Shah KV.
Condyloma in pregnancy is strongly predictive of juvenile-
onset recurrent respiratory papillomatosis. Obstet Gynecol
2003;101:645–52. PMID:12681865
1231. Dolev JC, Maurer T, Springer G, et al. Incidence and risk factors
for verrucae in women. AIDS 2008;22:1213–9. PMID:18525267
https://doi.org/10.1097/QAD.0b013e3283021aa3
1232. Silverberg MJ, Ahdieh L, Munoz A, et al. The impact of HIV infection and
immunodeficiency on human papillomavirus type 6 or 11 infection and
on genital warts. Sex Transm Dis 2002;29:42735. PMID:12172526
https://doi.org/10.1097/00007435-200208000-00001
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 177
US Department of Health and Human Services/Centers for Disease Control and Prevention
1233. De Panfilis G, Melzani G, Mori G, Ghidini A, Graifemberghi
S. Relapses after treatment of external genital warts are more
frequent in HIV-positive patients than in HIV-negative controls.
Sex Transm Dis 2002;29:121–5. PMID:11875372 https://doi.
org/10.1097/00007435-200203000-00001
1234. Conley LJ, Ellerbrock TV, Bush TJ, Chiasson MA, Sawo D,
Wright TC. HIV-1 infection and risk of vulvovaginal and perianal
condylomata acuminata and intraepithelial neoplasia: a prospective
cohort study. Lancet 2002;359:108–13. PMID:11809252 https://
doi.org/10.1016/S0140-6736(02)07368-3
1235. Schlecht HP, Fugelso DK, Murphy RK, et al. Frequency of occult high-
grade squamous intraepithelial neoplasia and invasive cancer within
anal condylomata in men who have sex with men. Clin Infect Dis
2010;51:10710. PMID:20482370 https://doi.org/10.1086/653426
1236. Maniar KP, Ronnett BM, Vang R, Yemelyanova A. Coexisting
high-grade vulvar intraepithelial neoplasia (VIN) and condyloma
acuminatum: independent lesions due to different HPV types
occurring in immunocompromised patients. Am J Surg Pathol
2013;37:5360. PMID:23026935 https://doi.org/10.1097/
PAS.0b013e318263cda6
1237. Massad LS, Xie X, Darragh T, et al.; Womens Interagency HIV Study
Collaborative Study Group. Genital warts and vulvar intraepithelial
neoplasia: natural history and effects of treatment and human
immunodeficiency virus infection. Obstet Gynecol 2011;118:831–9.
PMID:21934446 https://doi.org/10.1097/AOG.0b013e31821a0f4d
1238. Forman D, de Martel C, Lacey CJ, et al. Global burden of human
papillomavirus and related diseases. Vaccine 2012;30(Suppl 5):F12–23.
PMID:23199955 https://doi.org/10.1016/j.vaccine.2012.07.055
1239. Darragh TM, Colgan TJ, Cox JT, et al.; Members of LAST Project
Work Groups. The lower anogenital squamous terminology
standardization project for HPV-associated lesions: background
and consensus recommendations from the College of American
Pathologists and the American Society for Colposcopy and
Cervical Pathology. Arch Pathol Lab Med 2012;136:126697.
PMID:22742517 https://doi.org/10.5858/arpa.LGT200570
1240. Committee on Practice Bulletins—Gynecology. Practice Bulletin
No. 168 Summary: Cervical cancer screening and prevention.
Obstet Gynecol 2016;128:923–5. PMID:27661643 https://doi.
org/10.1097/AOG.0000000000001699
1241. Perkins RB, Guido RL, Saraiya M, et al. Summary of current
guidelines for cervical cancer screening and management of abnormal
test results: 2016–2020. J Womens Health (Larchmt) 2021;30:5–13.
PMID:33464997 https://doi.org/10.1089/jwh.2020.8918
1242. Kim JJ, Burger EA, Regan C, Sy S. Screening for cervical cancer
in primary care: a decision analysis for the US Preventive Services
Task Force. JAMA 2018;320:706–14. PMID:30140882 https://doi.
org/10.1001/jama.2017.19872
1243. Sawaya GF, Sanstead E, Alarid-Escudero F, et al. Estimated quality
of life and economic outcomes associated with 12 cervical cancer
screening strategies: a cost-effectiveness analysis. JAMA Intern Med
2019;179:86778. PMID:31081851 https://doi.org/10.1001/
jamainternmed.2019.0299
1244. Saslow D, Solomon D, Lawson HW, et al.; ACS-ASCCP-ASCP
Cervical Cancer Guideline Committee. American Cancer Society,
American Society for Colposcopy and Cervical Pathology, and
American Society for Clinical Pathology screening guidelines for
the prevention and early detection of cervical cancer. CA Cancer
J Clin 2012;62:147–72. PMID:22422631 https://doi.org/10.3322/
caac.21139
1245. Committee on Practice Bulletins—Gynecology. ACOG Practice
Bulletin No. 131: Screening for cervical cancer. Obstet Gynecol
2012;120:122238. PMID:23090560 https://doi.org/10.1097/
AOG.0b013e318277c92a
1246. Meyerson BE, Sayegh MA, Davis A, et al. Cervical cancer screening in a
sexually transmitted disease clinic: screening adoption experiences from
a midwestern clinic. Am J Public Health 2015;105(Suppl 2):e8–14.
PMID:25689199 https://doi.org/10.2105/AJPH.2014.302272
1247. Perkins RB, Guido RS, Castle PE, et al.; 2019 ASCCP Risk-Based
Management Consensus Guidelines Committee. 2019 ASCCP
risk-based management consensus guidelines for abnormal cervical
cancer screening tests and cancer precursors. J Low Genit Tract
Dis 2020;24:10231. PMID:32243307 https://doi.org/10.1097/
LGT.0000000000000525
1248. Saraiya M, Lee NC, Blackman D, Smith MJ, Morrow B, McKenna
MA. Self-reported Papanicolaou smears and hysterectomies among
women in the United States. Obstet Gynecol 2001;98:26978.
PMID:11506844
1249. Sirovich BE, Welch HG. Cervical cancer screening among women
without a cervix. JAMA 2004;291:2990–3. PMID:15213211 https://
doi.org/10.1001/jama.291.24.2990
1250. Stokes-Lampard H, Wilson S, Waddell C, Ryan A, Holder R,
Kehoe S. Vaginal vault smears after hysterectomy for reasons
other than malignancy: a systematic review of the literature.
BJOG 2006;113:135465. PMID:17081187 https://doi.
org/10.1111/j.1471-0528.2006.01099.x
1251. Arbyn M, Herbert A, Schenck U, et al. European guidelines for
quality assurance in cervical cancer screening: recommendations
for collecting samples for conventional and liquid-based cytology.
Cytopathology 2007;18:1339. PMID:17573762 https://doi.
org/10.1111/j.1365-2303.2007.00464.x
1252. Daley E, Perrin K, Vamos C, et al. Confusion about Pap smears: lack
of knowledge among high-risk women. J Womens Health (Larchmt)
2013;22:6774. PMID:23215902 https://doi.org/10.1089/
jwh.2012.3667
1253. Drolet M, Bénard É, Boily MC, et al. Population-level impact
and herd effects following human papillomavirus vaccination
programmes: a systematic review and meta-analysis. Lancet Infect
Dis 2015;15:56580. PMID:25744474 https://doi.org/10.1016/
S1473-3099(14)71073-4
1254. Ogbechie OA, Hacker MR, Dodge LE, Patil MM, Ricciotti HA.
Confusion regarding cervical cancer screening and chlamydia
screening among sexually active young women. Sex Transm
Infect 2012;88:35–7. PMID:22123163 https://doi.org/10.1136/
sextrans-2011-050289
1255. Dunne EF, Friedman A, Datta SD, Markowitz LE, Workowski
KA. Updates on human papillomavirus and genital warts and
counseling messages from the 2010 Sexually Transmitted Diseases
Treatment Guidelines. Clin Infect Dis 2011;53(Suppl 3):S143–52.
PMID:22080267 https://doi.org/10.1093/cid/cir703
1256. Fry AM, Ferries-Rowe EA, Learman LA, Haas DM. Pap smear versus
speculum examination: can we teach providers to educate patients?
J Womens Health (Larchmt) 2010;19:1715–9. PMID:20662627
https://doi.org/10.1089/jwh.2009.1862
1257. Adab P, Marshall T, Rouse A, Randhawa B, Sangha H, Bhangoo
N. Randomised controlled trial of the effect of evidence based
information on womens willingness to participate in cervical cancer
screening. J Epidemiol Community Health 2003;57:58993.
PMID:12883063 https://doi.org/10.1136/jech.57.8.589
1258. Drolet M, Brisson M, Maunsell E, et al. The psychosocial impact of
an abnormal cervical smear result. Psychooncology 2012;21:107181.
PMID:21695747 https://doi.org/10.1002/pon.2003
1259. McCaffery KJ, Irwig L, Turner R, et al. Psychosocial outcomes of three
triage methods for the management of borderline abnormal cervical
smears: an open randomised trial. BMJ 2010;340(feb23 1):b4491.
PMID:20179125 https://doi.org/10.1136/bmj.b4491
Recommendations and Reports
178 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
1260. Daley EM, Perrin KM, McDermott RJ, et al. The psychosocial
burden of HPV: a mixed-method study of knowledge, attitudes and
behaviors among HPV+ women. J Health Psychol 2010;15:279–90.
PMID:20207671 https://doi.org/10.1177/1359105309351249
1261. Pirotta M, Ung L, Stein A, et al. The psychosocial burden of human
papillomavirus related disease and screening interventions. Sex Transm
Infect 2009;85:508–13. PMID:19703844 https://doi.org/10.1136/
sti.2009.037028
1262. Lin L, Benard VB, Greek A, Roland KB, Hawkins NA, Saraiya M.
Communication practices about HPV testing among providers in
Federally Qualified Health Centers. Prev Med Rep 2015;2:436–9.
PMID:26213683 https://doi.org/10.1016/j.pmedr.2015.05.006
1263. Kapeu AS, Luostarinen T, Jellum E, et al. Is smoking an independent
risk factor for invasive cervical cancer? A nested case-control
study within Nordic biobanks. Am J Epidemiol 2009;169:480–8.
PMID:19074773 https://doi.org/10.1093/aje/kwn354
1264. Plummer M, Herrero R, Franceschi S, et al.; IARC Multi-centre
Cervical Cancer Study Group. Smoking and cervical cancer: pooled
analysis of the IARC multi-centric case-control study. Cancer
Causes Control 2003;14:805–14. PMID:14682438 https://doi.
org/10.1023/B:CACO.0000003811.98261.3e
1265. de Sanjosé S, Brotons M, Pavón MA. The natural history of human
papillomavirus infection. Best Pract Res Clin Obstet Gynaecol
2018;47:213. PMID:28964706 https://doi.org/10.1016/j.
bpobgyn.2017.08.015
1266. Louie KS, Castellsague X, de Sanjose S, et al.; International Agency
for Research on Cancer Multicenter Cervical Cancer Study Group.
Smoking and passive smoking in cervical cancer risk: pooled analysis
of couples from the IARC multicentric case-control studies. Cancer
Epidemiol Biomarkers Prev 2011;20:137990. PMID:21610224
https://doi.org/10.1158/1055-9965.EPI-11-0284
1267. Nelson HD, Cantor A, Wagner J, et al. Effectiveness of patient
navigation to increase cancer screening in populations adversely
affected by health disparities: a meta-analysis. J Gen Intern Med
2020;35:302635. PMID:32700218 https://doi.org/10.1007/
s11606-020-06020-9
1268. Stillson T, Knight AL, Elswick RK Jr. The effectiveness and safety
of two cervical cytologic techniques during pregnancy. J Fam Pract
1997;45:159–63. PMID:9267375
1269. Foster JC, Smith HL. Use of the Cytobrush for Papanicolaou smear
screens in pregnant women. J Nurse Midwifery 1996;41:211–7.
PMID:8708804 https://doi.org/10.1016/0091-2182(96)00013-4
1270. Paraiso MF, Brady K, Helmchen R, Roat TW. Evaluation of the
endocervical Cytobrush and Cervex-Brush in pregnant women.
Obstet Gynecol 1994;84:539–43. PMID:8090390
1271. Silverberg MJ, Leyden WA, Chi A, et al. Human immunodeficiency
virus (HIV)- and non-HIV-associated immunosuppression and risk of
cervical neoplasia. Obstet Gynecol 2018;131:4755. PMID:29215531
https://doi.org/10.1097/AOG.0000000000002371
1272. Videla S, Tarrats A, Ornelas A, et al. Incidence of cervical high-
grade squamous intraepithelial lesions in HIV-1-infected women
with no history of cervical pathology: up to 17 years of follow-up.
Int J STD AIDS 2019;30:5663. PMID:30170532 https://doi.
org/10.1177/0956462418792653
1273. Liu G, Sharma M, Tan N, Barnabas RV. HIV-positive women have
higher risk of human papilloma virus infection, precancerous lesions,
and cervical cancer. AIDS 2018;32:795808. PMID:29369827
https://doi.org/10.1097/QAD.0000000000001765
1274. Sawaya GF, Lamar R, Perkins RB. Managing minimally abnormal
cervical cancer screening test results. JAMA 2020;324:1557.
PMID:32975557 https://doi.org/10.1001/jama.2020.12488
1275. Silverberg MJ, Lau B, Justice AC, et al.; North American AIDS Cohort
Collaboration on Research and Design (NA-ACCORD) of IeDEA.
Risk of anal cancer in HIV-infected and HIV-uninfected individuals in
North America. Clin Infect Dis 2012;54:102634. PMID:22291097
https://doi.org/10.1093/cid/cir1012
1276. Tomassi MJ, Abbas MA, Klaristenfeld DD. Expectant management
surveillance for patients at risk for invasive squamous cell carcinoma
of the anus: a large US healthcare system experience. Int J Colorectal
Dis 2019;34:4754. PMID:30244347 https://doi.org/10.1007/
s00384-018-3167-7
1277. Colón-López V, Shiels MS, Machin M, et al. Anal cancer risk
among people with HIV infection in the United States. J Clin
Oncol 2018;36:68–75. PMID:29140774 https://doi.org/10.1200/
JCO.2017.74.9291
1278. Machalek DA, Grulich AE, Jin F, Templeton DJ, Poynten IM. The
epidemiology and natural history of anal human papillomavirus
infection in men who have sex with men. Sex Health 2012;9:52737.
PMID:23380235 https://doi.org/10.1071/SH12043
1279. Deshmukh AA, Suk R, Shiels MS, et al. Recent trends in squamous cell
carcinoma of the anus incidence and mortality in the United States,
2001–2015. J Natl Cancer Inst 2020;112:82938. PMID:31742639
https://doi.org/10.1093/jnci/djz219
1280. Edgren G, Sparén P. Risk of anogenital cancer after diagnosis of cervical
intraepithelial neoplasia: a prospective population-based study. Lancet
Oncol 2007;8:3116. PMID:17395104 https://doi.org/10.1016/
S1470-2045(07)70043-8
1281. Chaturvedi AK, Engels EA, Gilbert ES, et al. Second cancers among
104,760 survivors of cervical cancer: evaluation of long-term risk.
J Natl Cancer Inst 2007;99:1634–43. PMID:17971527 https://doi.
org/10.1093/jnci/djm201
1282. Suk R, Mahale P, Sonawane K, et al. Trends in risks for second primary
cancers associated with index human papillomavirus-associated
cancers. JAMA Netw Open 2018;1:e181999. PMID:30646145
https://doi.org/10.1001/jamanetworkopen.2018.1999
1283. Hillman RJ, Berry-Lawhorn JM, Ong JJ, et al.; International Anal
Neoplasia Society. International Anal Neoplasia Society guidelines
for the practice of digital anal rectal examination. J Low Genit Tract
Dis 2019;23:13846. PMID:30907777 https://doi.org/10.1097/
LGT.0000000000000458
1284. Ong JJ, Grulich A, Walker S, et al. Baseline findings from the
Anal Cancer Examination (ACE) study: screening using digital
ano-rectal examination in HIV-positive men who have sex with
men. J Med Screen 2016;23:70–6. PMID:26462726 https://doi.
org/10.1177/0969141315604658
1285. Read TR, Vodstrcil L, Grulich AE, et al. Acceptability of digital anal
cancer screening examinations in HIV-positive homosexual men. HIV
Med 2013;14:491–6. PMID:23590621 https://doi.org/10.1111/
hiv.12035
1286. Jin F, Grulich AE, Poynten IM, et al.; SPANC Study Team. The
performance of anal cytology as a screening test for anal HSILs
in homosexual men. Cancer Cytopathol 2016;124:41524.
PMID:26915346 https://doi.org/10.1002/cncy.21702
1287. Silva M, Peixoto A, Sarmento JA, Coelho R, Macedo G. Anal cytology,
histopathology and anoscopy in an anal dysplasia screening program:
is anal cytology enough? Rev Esp Enferm Dig 2018;110:109–14.
PMID:29168646 https://doi.org/10.17235/reed.2018.5678/2018
1288. Iribarren Díaz M, Ocampo Hermida A, González-Carreró Fojón
J, et al. Preliminary results of a screening program for anal cancer
and its precursors for HIV-infected men who have sex with men
in Vigo-Spain [Spanish]. Rev Esp Enferm Dig 2017;109:242–9.
PMID:28229612 https://doi.org/10.17235/reed.2017.4274/2016
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 179
US Department of Health and Human Services/Centers for Disease Control and Prevention
1289. Burgos J, Hernández-Losa J, Landolfi S, et al. The role of oncogenic
human papillomavirus determination for diagnosis of high-grade
anal intraepithelial neoplasia in HIV-infected MSM. AIDS
2017;31:222733. PMID:28723712 https://doi.org/10.1097/
QAD.0000000000001605
1290. Cheng SH, Wang CC, Chang SL, Chu FY, Hsueh YM. Oncogenic
human papillomavirus is not helpful for cytology screening of the
precursor lesions of anal cancers in Taiwanese men who are infected
with human immunodeficiency virus. Int J Clin Oncol 2015;20:943
51. PMID:25712159 https://doi.org/10.1007/s10147-015-0804-9
1291. Hidalgo-Tenorio C, Rivero-Rodriguez M, Gil-Anguita C, et al. The
role of polymerase chain reaction of high-risk human papilloma virus
in the screening of high-grade squamous intraepithelial lesions in the
anal mucosa of human immunodeficiency virus-positive males having
sex with males. PLoS One 2015;10:e0123590. PMID:25849412
https://doi.org/10.1371/journal.pone.0123590
1292. Richel O, de Vries HJ, van Noesel CJ, Dijkgraaf MG, Prins JM.
Comparison of imiquimod, topical fluorouracil, and electrocautery
for the treatment of anal intraepithelial neoplasia in HIV-positive
men who have sex with men: an open-label, randomised controlled
trial. Lancet Oncol 2013;14:346–53. PMID:23499546 https://doi.
org/10.1016/S1470-2045(13)70067-6
1293. Goldstone SE, Johnstone AA, Moshier EL. Long-term outcome
of ablation of anal high-grade squamous intraepithelial lesions:
recurrence and incidence of cancer. Dis Colon Rectum
2014;57:31623. PMID:24509453 https://doi.org/10.1097/
DCR.0000000000000058
1294. Tong WW, Shepherd K, Garland S, et al.; Study of the Prevention
of Anal Cancer (SPANC) team. Human papillomavirus 16-specific
T-cell responses and spontaneous regression of anal high-grade
squamous intraepithelial lesions. J Infect Dis 2015;211:40515.
PMID:25139018 https://doi.org/10.1093/infdis/jiu461
1295. Tong WW, Jin F, McHugh LC, et al. Progression to and spontaneous
regression of high-grade anal squamous intraepithelial lesions
in HIV-infected and uninfected men. AIDS 2013;27:223343.
PMID:24157904 https://doi.org/10.1097/QAD.0b013e3283633111
1296. Shin EC, Jeong SH. Natural history, clinical manifestations, and
pathogenesis of hepatitis A. Cold Spring Harb Perspect Med
2018;8:a031708. PMID:29440324 https://doi.org/10.1101/
cshperspect.a031708
1297. Nelson NP, Weng MK, Hofmeister MG, et al. Prevention of hepatitis
A virus infection in the United States: recommendations of the
Advisory Committee on Immunization Practices, 2020. MMWR
Recomm Rep 2020;69(No. RR-5). PMID:32614811 https://doi.
org/10.15585/mmwr.rr6905a1
1298. Foster MA, Hofmeister MG, Kupronis BA, et al. Increase in hepatitis
A virus infections—United States, 2013–2018. MMWR Morb
Mortal Wkly Rep 2019;68:413–5. PMID:31071072 https://doi.
org/10.15585/mmwr.mm6818a2
1299. Bower WA, Nainan OV, Han X, Margolis HS. Duration of
viremia in hepatitis A virus infection. J Infect Dis 2000;182:12–7.
PMID:10882576 https://doi.org/10.1086/315701
1300. Clemens R, Safary A, Hepburn A, Roche C, Stanbury WJ, André FE.
Clinical experience with an inactivated hepatitis A vaccine. J Infect Dis
1995;171(Suppl 1):S44–9. PMID:7876648 https://doi.org/10.1093/
infdis/171.Supplement_1.S44
1301. Sharapov UM, Bulkow LR, Negus SE, et al. Persistence of hepatitis
A vaccine induced seropositivity in infants and young children
by maternal antibody status: 10-year follow-up. Hepatology
2012;56:51622. PMID:22371069 https://doi.org/10.1002/
hep.25687
1302. Mosites E, Gounder P, Snowball M, et al. Hepatitis A vaccine immune
response 22 years after vaccination. J Med Virol 2018;90:1418–22.
PMID:29663458 https://doi.org/10.1002/jmv.25197
1303. Theeten H, Van Herck K, Van Der Meeren O, Crasta P, Van
Damme P, Hens N. Long-term antibody persistence after vaccination
with a 2-dose Havrix (inactivated hepatitis A vaccine): 20 years of
observed data, and long-term model-based predictions. Vaccine
2015;33:5723–7. PMID:26190091 https://doi.org/10.1016/j.
vaccine.2015.07.008
1304. Hens N, Habteab Ghebretinsae A, Hardt K, Van Damme P,
Van Herck K. Model based estimates of long-term persistence of
inactivated hepatitis A vaccine-induced antibodies in adults. Vaccine
2014;32:150713. PMID:24508042 https://doi.org/10.1016/j.
vaccine.2013.10.088
1305. Mosites E, Seeman S, Negus S, et al. Immunogenicity of the hepatitis A
vaccine 20years after infant immunization. Vaccine 2020;38:4940–3.
PMID:32535018 https://doi.org/10.1016/j.vaccine.2020.05.069
1306. Yin S, Barker L, Ly KN, et al. Susceptibility to hepatitis A virus
infection in the United States, 2007–2016. Clin Infect Dis
2020;71:e5719. PMID:32193542 https://doi.org/10.1093/cid/
ciaa298
1307. Moro PL, Arana J, Marquez PL, et al. Is there any harm in
administering extra-doses of vaccine to a person? Excess doses of
vaccine reported to the Vaccine Adverse Event Reporting System
(VAERS), 2007–2017. Vaccine 2019;37:3730–4. PMID:31155414
https://doi.org/10.1016/j.vaccine.2019.04.088
1308. Winokur PL, Stapleton JT. Immunoglobulin prophylaxis for hepatitis
A. Clin Infect Dis 1992;14:580–6. PMID:1554845 https://doi.
org/10.1093/clinids/14.2.580
1309. Alter HJ, Purcell RH, Gerin JL, et al. Transmission of hepatitis B
to chimpanzees by hepatitis B surface antigen-positive saliva and
semen. Infect Immun 1977;16:928–33. PMID:892901 https://doi.
org/10.1128/IAI.16.3.928-933.1977
1310. Villarejos VM, Visoná KA, Gutiérrez A, Rodríguez A. Role of saliva,
urine and feces in the transmission of type B hepatitis. N Engl J
Med 1974;291:1375–8. PMID:4427641 https://doi.org/10.1056/
NEJM197412262912602
1311. Busch K, Thimme R. Natural history of chronic hepatitis B
virus infection. Med Microbiol Immunol (Berl) 2015;204:510.
PMID:25540037 https://doi.org/10.1007/s00430-014-0369-7
1312. Hyams KC. Risks of chronicity following acute hepatitis B
virus infection: a review. Clin Infect Dis 1995;20:9921000.
PMID:7795104 https://doi.org/10.1093/clinids/20.4.992
1313. Goldstein ST, Zhou F, Hadler SC, Bell BP, Mast EE, Margolis
HS. A mathematical model to estimate global hepatitis B disease
burden and vaccination impact. Int J Epidemiol 2005;34:1329–39.
PMID:16249217 https://doi.org/10.1093/ije/dyi206
1314. Thompson ND, Perz JF, Moorman AC, Holmberg SD. Nonhospital
health care-associated hepatitis B and C virus transmission: United
States, 1998–2008. Ann Intern Med 2009;150:339. PMID:19124818
https://doi.org/10.7326/0003-4819-150-1-200901060-00007
1315. Davis LG, Weber DJ, Lemon SM. Horizontal transmission of
hepatitis B virus. Lancet 1989;1:889–93. PMID:2564960 https://
doi.org/10.1016/S0140-6736(89)92876-6
1316. Martinson FE, Weigle KA, Royce RA, Weber DJ, Suchindran CM,
Lemon SM. Risk factors for horizontal transmission of hepatitis B
virus in a rural district in Ghana. Am J Epidemiol 1998;147:47887.
PMID:9525535 https://doi.org/10.1093/oxfordjournals.aje.a009474
1317. CDC. Healthcare-associated hepatitis B and C outbreaks (≥2 cases)
reported to the CDC 2008–2019. Atlanta, GA: US Department of
Health and Human Services, CDC; 2019. https://www.cdc.gov/
hepatitis/outbreaks/pdfs/HealthcareInvestigationTable.pdf
1318. Schillie S, Harris A, Link-Gelles R, Romero J, Ward J, Nelson N.
Recommendations of the Advisory Committee on Immunization
Practices for use of a hepatitis B vaccine with a novel adjuvant.
MMWR Morb Mortal Wkly Rep 2018;67:455–8. PMID:29672472
https://doi.org/10.15585/mmwr.mm6715a5
Recommendations and Reports
180 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
1319. Lu PJ, Byrd KK, Murphy TV, Weinbaum C. Hepatitis B vaccination
coverage among high-risk adults 18–49 years, U.S., 2009. Vaccine
2011;29:704957. PMID:21782873 https://doi.org/10.1016/j.
vaccine.2011.07.030
1320. Williams WW, Lu PJ, O’Halloran A, et al. Surveillance of vaccination
coverage among adult populations—United States, 2015. MMWR
Surveill Summ 2017;66(No. SS-11). PMID:28472027 https://doi.
org/10.15585/mmwr.ss6611a1
1321. CDC. Hepatitis B vaccination coverage among adults—United
States, 2004. MMWR Morb Mortal Wkly Rep 2006;55:509–11.
PMID:16691181
1322. MacKellar DA, Valleroy LA, Secura GM, et al.; Young Mens
Survey Study Group. Two decades after vaccine license: hepatitis B
immunization and infection among young men who have sex with
men. Am J Public Health 2001;91:96571. PMID:11392942 https://
doi.org/10.2105/AJPH.91.6.965
1323. Terrault NA, Lok ASF, McMahon BJ, et al. Update on prevention,
diagnosis, and treatment of chronic hepatitis B: AASLD 2018 hepatitis
B guidance. Hepatology 2018;67:156099. PMID:29405329 https://
doi.org/10.1002/hep.29800
1324. Ezeanolue E, Harriman K, Hunter P, Kroger A, Pellegrini C. General
best practice guidelines for immunization: best practices guidance
of the Advisory Committee on Immunization Practices (ACIP)
[Internet]. Atlanta, GA: US Department of Health and Human
Services, CDC, Advisory Committee on Immunization Practices;
2020. https://www.cdc.gov/vaccines/hcp/acip-recs/general-recs/
downloads/general-recs.pdf
1325. Bruce MG, Bruden D, Hurlburt D, et al. Antibody levels and
protection after hepatitis B vaccine: results of a 30-year follow-up
study and response to a booster dose. J Infect Dis 2016;214:16–22.
PMID:26802139 https://doi.org/10.1093/infdis/jiv748
1326. McMahon BJ, Bulkow LR, Singleton RJ, et al. Elimination of
hepatocellular carcinoma and acute hepatitis B in children 25 years
after a hepatitis B newborn and catch-up immunization program.
Hepatology 2011;54:8017. PMID:21618565 https://doi.
org/10.1002/hep.24442
1327. Simons BC, Spradling PR, Bruden DJ, et al. A longitudinal hepatitis
B vaccine cohort demonstrates long-lasting hepatitis B virus (HBV)
cellular immunity despite loss of antibody against HBV surface
antigen. J Infect Dis 2016;214:273–80. PMID:27056956 https://
doi.org/10.1093/infdis/jiw142
1328. Bohlke K, Davis RL, Marcy SM, et al.; Vaccine Safety Datalink Team.
Risk of anaphylaxis after vaccination of children and adolescents.
Pediatrics 2003;112:81520. PMID:14523172 https://doi.
org/10.1542/peds.112.4.815
1329. André FE. Summary of safety and efficacy data on a yeast-derived
hepatitis B vaccine. Am J Med 1989;87(3A):14S20. PMID:2528292
https://doi.org/10.1016/0002-9343(89)90525-1
1330. Abara WE, Qaseem A, Schillie S, McMahon BJ, Harris AM; High
Value Care Task Force of the American College of Physicians and the
Centers for Disease Control and Prevention. Hepatitis B vaccination,
screening, and linkage to care: best practice advice from the American
College of Physicians and the Centers for Disease Control and
Prevention. Ann Intern Med 2017;167:794–804. PMID:29159414
https://doi.org/10.7326/M17-1106
1331. Minuk GY, Bohme CE, Bowen TJ, et al. Efficacy of commercial
condoms in the prevention of hepatitis B virus infection.
Gastroenterology 1987;93:7104. PMID:3040512 https://doi.
org/10.1016/0016-5085(87)90431-8
1332. Hofmeister MG, Rosenthal EM, Barker LK, et al. Estimating
prevalence of hepatitis C virus infection in the United States, 2013–
2016. Hepatology 2019;69:1020–31. PMID:30398671 https://doi.
org/10.1002/hep.30297
1333. Lockart I, Matthews GV, Danta M. Sexually transmitted hepatitis C
infection: the evolving epidemic in HIV-positive and HIV-negative
MSM. Curr Opin Infect Dis 2019;32:31–7. PMID:30531370 https://
doi.org/10.1097/QCO.0000000000000515
1334. Terrault NA, Dodge JL, Murphy EL, et al. Sexual transmission of
hepatitis C virus among monogamous heterosexual couples: the HCV
partners study. Hepatology 2013;57:881–9. PMID:23175457 https://
doi.org/10.1002/hep.26164
1335. Price JC, McKinney JE, Crouch PC, et al. Sexually acquired hepatitis
C infection in HIV-uninfected men who have sex with men using
preexposure prophylaxis against HIV. J Infect Dis 2019;219:1373–6.
PMID:30462305 https://doi.org/10.1093/infdis/jiy670
1336. Tohme RA, Holmberg SD. Transmission of hepatitis C virus infection
through tattooing and piercing: a critical review. Clin Infect Dis
2012;54:116778. PMID:22291098 https://doi.org/10.1093/cid/
cir991
1337. Brettler DB, Mannucci PM, Gringeri A, et al. The low risk of
hepatitis C virus transmission among sexual partners of hepatitis
C-infected hemophilic males: an international, multicenter study.
Blood 1992;80:540–3. PMID:1627805 https://doi.org/10.1182/
blood.V80.2.540.540
1338. Kao JH, Hwang YT, Chen PJ, et al. Transmission of hepatitis C
virus between spouses: the important role of exposure duration. Am
J Gastroenterol 1996;91:2087–90. PMID:8855726
1339. Fierer DS, Mullen MP, Dieterich DT, Isabel Fiel M, Branch AD. Early-
onset liver fibrosis due to primary hepatitis C virus infection is higher
over time in HIV-infected men. Clin Infect Dis 2012;55:887–8,
author reply 888–9. PMID:22677713 https://doi.org/10.1093/cid/
cis538
1340. van de Laar TJ, van der Bij AK, Prins M, et al. Increase in HCV
incidence among men who have sex with men in Amsterdam most
likely caused by sexual transmission. J Infect Dis 2007;196:230–8.
PMID:17570110 https://doi.org/10.1086/518796
1341. Nijmeijer BM, Koopsen J, Schinkel J, Prins M, Geijtenbeek TB.
Sexually transmitted hepatitis C virus infections: current trends, and
recent advances in understanding the spread in men who have sex with
men. J Int AIDS Soc 2019;22(Suppl 6):e25348. PMID:31468692
https://doi.org/10.1002/jia2.25348
1342. Todesco E, Day N, Amiel C, et al. High clustering of acute HCV
infections and high rate of associated STIs among Parisian HIV-
positive male patients. Int J Antimicrob Agents 2019;53:678–81.
PMID:30742957 https://doi.org/10.1016/j.ijantimicag.2019.02.002
1343. Jin F, Matthews GV, Grulich AE. Sexual transmission of hepatitis C
virus among gay and bisexual men: a systematic review. Sex Health
2017;14:2841. PMID:27712618 https://doi.org/10.1071/SH16141
1344. Hegazi A, Lee MJ, Whittaker W, et al. Chemsex and the city: sexualised
substance use in gay bisexual and other men who have sex with men
attending sexual health clinics. Int J STD AIDS 2017;28:362–6.
Erratum in: Int J STD AIDS 2017;28:423. PMID:27178067 https://
doi.org/10.1177/0956462416651229
1345. Page EE, Nelson M. Hepatitis C and sex. Clin Med (Lond)
2016;16:18992. PMID:27037392 https://doi.org/10.7861/
clinmedicine.16-2-189
1346. Apers L, Vanden Berghe W, De Wit S, et al. Risk factors for HCV
acquisition among HIV-positive MSM in Belgium. J Acquir
Immune Defic Syndr 2015;68:585–93. PMID:25763786 https://
doi.org/10.1097/QAI.0000000000000528
1347. Daskalopoulou M, Rodger AJ, Phillips AN, et al.; ASTRA Study
Group. Condomless sex in HIV-diagnosed men who have sex with
men in the UK: prevalence, correlates, and implications for HIV
transmission. Sex Transm Infect 2017;93:590–8. PMID:28679630
https://doi.org/10.1136/sextrans-2016-053029
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 181
US Department of Health and Human Services/Centers for Disease Control and Prevention
1348. Vanhommerig JW, Lambers FA, Schinkel J, et al.; MOSAIC (MSM
Observational Study of Acute Infection With Hepatitis C) Study
Group. Risk factors for sexual transmission of hepatitis C virus among
human immunodeficiency virus-infected men who have sex with
men: a case-control study. Open Forum Infect Dis 2015;2:ofv115.
PMID:26634219 https://doi.org/10.1093/ofid/ofv115
1349. Turner SS, Gianella S, Yip MJ, et al. Shedding of hepatitis C virus in
semen of human immunodeficiency virus-infected men. Open Forum
Infect Dis 2016;3:ofw057.
1350. Foster AL, Gaisa MM, Hijdra RM, et al. Shedding of hepatitis C
virus into the rectum of HIV-infected men who have sex with men.
Clin Infect Dis 2017;64:284–8. PMID:28013267 https://doi.
org/10.1093/cid/ciw740
1351. Hammer GP, Kellogg TA, McFarland WC, et al. Low incidence and
prevalence of hepatitis C virus infection among sexually active non-
intravenous drug-using adults, San Francisco, 1997–2000. Sex Transm
Dis 2003;30:91924. PMID:14646642 https://doi.org/10.1097/01.
OLQ.0000091152.31366.E6
1352. Roy KM, Goldberg DJ, Hutchinson S, Cameron SO, Wilson K,
MacDonald L. Hepatitis C virus among self declared non-injecting
sexual partners of injecting drug users. J Med Virol 2004;74:62–6.
PMID:15258969 https://doi.org/10.1002/jmv.20146
1353. Mele A, Stroffolini T, Tosti ME, et al. Heterosexual
transmission of hepatitis C in Italy. J Med Virol
1999;57:1113. PMID:9892393 https://doi.org/10.1002/
(SICI)1096-9071(199902)57:2<111::AID-JMV4>3.0.CO;2-C
1354. Rauch A, Rickenbach M, Weber R, et al.; Swiss HIV Cohort Study.
Unsafe sex and increased incidence of hepatitis C virus infection
among HIV-infected men who have sex with men: the Swiss HIV
Cohort Study. Clin Infect Dis 2005;41:395–402. PMID:16007539
https://doi.org/10.1086/431486
1355. Browne R, Asboe D, Gilleece Y, et al. Increased numbers of acute
hepatitis C infections in HIV positive homosexual men; is sexual
transmission feeding the increase? Sex Transm Infect 2004;80:326–7.
PMID:15295139 https://doi.org/10.1136/sti.2003.008532
1356. Danta M, Brown D, Bhagani S, et al.; HIV and Acute HCV (HAAC)
group. Recent epidemic of acute hepatitis C virus in HIV-positive
men who have sex with men linked to high-risk sexual behaviours.
AIDS 2007;21:983–91. PMID:17457092 https://doi.org/10.1097/
QAD.0b013e3281053a0c
1357. Ghosn J, Pierre-François S, Thibault V, et al. Acute
hepatitis C in HIV-infected men who have sex with men.
HIV Med 2004;5:3036. PMID:15236621 https://doi.
org/10.1111/j.1468-1293.2004.00225.x
1358. van de Laar T, Pybus O, Bruisten S, et al. Evidence of a large,
international network of HCV transmission in HIV-positive men
who have sex with men. Gastroenterology 2009;136:160917.
PMID:19422083 https://doi.org/10.1053/j.gastro.2009.02.006
1359. Hoornenborg E, Achterbergh RCA, Schim van der Loeff MF, et al.;
Amsterdam PrEP Project team in the HIV Transmission Elimination
AMsterdam Initiative, MOSAIC study group. MSM starting
preexposure prophylaxis are at risk of hepatitis C virus infection.
AIDS 2017;31:160310. PMID:28657964 https://doi.org/10.1097/
QAD.0000000000001522
1360. Gras J, Mahjoub N, Charreau I, et al.; IPERGAY Study Group. Early
diagnosis and risk factors of acute hepatitis C in high-risk MSM on
preexposure prophylaxis. AIDS 2020;34:47–52. PMID:31789889
https://doi.org/10.1097/QAD.0000000000002364
1361. Hoofnagle JH. Hepatitis C: the clinical spectrum of disease.
Hepatology 1997;26(Suppl 1):15S20S. PMID:9305658 https://
doi.org/10.1002/hep.510260703
1362. Orland JR, Wright TL, Cooper S. Acute hepatitis C. Hepatology
2001;33:3217. PMID:11172332 https://doi.org/10.1053/
jhep.2001.22112
1363. Alter MJ, Margolis HS, Krawczynski K, et al. The natural history of
community-acquired hepatitis C in the United States. The Sentinel
Counties Chronic non-A, non-B Hepatitis Study Team. N Engl J
Med 1992;327:1899905. PMID:1280771 https://doi.org/10.1056/
NEJM199212313272702
1364. Farci P, Alter HJ, Wong D, et al. A long-term study of hepatitis
C virus replication in non-A, non-B hepatitis. N Engl J Med
1991;325:98104. PMID:1646962 https://doi.org/10.1056/
NEJM199107113250205
1365. Liang TJ, Rehermann B, Seeff LB, Hoofnagle JH. Pathogenesis,
natural history, treatment, and prevention of hepatitis C. Ann
Intern Med 2000;132:296305. PMID:10681285 https://doi.
org/10.7326/0003-4819-132-4-200002150-00008
1366. Thomas DL, Seeff LB. Natural history of hepatitis C. Clin Liver Dis
2005;9:38398, vi. PMID:16023972 https://doi.org/10.1016/j.
cld.2005.05.003
1367. Westbrook RH, Dusheiko G. Natural history of hepatitis C. J
Hepatol 2014;61(Suppl):S5868. PMID:25443346 https://doi.
org/10.1016/j.jhep.2014.07.012
1368. Zou S, Stramer SL, Dodd RY. Donor testing and risk: current
prevalence, incidence, and residual risk of transfusion-transmissible
agents in US allogeneic donations. Transfus Med Rev 2012;26:119–28.
PMID:21871776 https://doi.org/10.1016/j.tmrv.2011.07.007
1369. Bixler D, Annambholta P, Abara WE, et al. Hepatitis B and C virus
infections transmitted through organ transplantation investigated by
CDC, United States, 2014–2017. Am J Transplant 2019;19:257082.
PMID:30861300 https://doi.org/10.1111/ajt.15352
1370. CDC. Testing for HCV infection: an update of guidance for clinicians
and laboratorians. MMWR Morb Mortal Wkly Rep 2013;62:362–5.
PMID:23657112
1371. Marincovich B, Castilla J, del Romero J, et al. Absence of hepatitis
C virus transmission in a prospective cohort of heterosexual
serodiscordant couples. Sex Transm Infect 2003;79:1602.
PMID:12690143 https://doi.org/10.1136/sti.79.2.160
1372. Tahan V, Karaca C, Yildirim B, et al. Sexual transmission of HCV between
spouses. Am J Gastroenterol 2005;100:8214. PMID:15784025
https://doi.org/10.1111/j.1572-0241.2005.40879.x
1373. Vandelli C, Renzo F, Romanò L, et al. Lack of evidence
of sexual transmission of hepatitis C among monogamous
couples: results of a 10-year prospective follow-up study. Am
J Gastroenterol 2004;99:8559. PMID:15128350 https://doi.
org/10.1111/j.1572-0241.2004.04150.x
1374. Fierer DS, Uriel AJ, Carriero DC, et al. Liver fibrosis during an
outbreak of acute hepatitis C virus infection in HIV-infected
men: a prospective cohort study. J Infect Dis 2008;198:683–6.
PMID:18627270 https://doi.org/10.1086/590430
1375. Cottrell EB, Chou R, Wasson N, Rahman B, Guise JM. Reducing
risk for mother-to-infant transmission of hepatitis C virus: a
systematic review for the U.S. Preventive Services Task Force. Ann
Intern Med 2013;158:10913. PMID:23437438 https://doi.
org/10.7326/0003-4819-158-2-201301150-00575
1376. Mast EE, Hwang LY, Seto DS, et al. Risk factors for perinatal
transmission of hepatitis C virus (HCV) and the natural history of
HCV infection acquired in infancy. J Infect Dis 2005;192:1880–9.
PMID:16267758 https://doi.org/10.1086/497701
1377. Barbosa C, Fraser H, Hoerger TJ, et al. Cost-effectiveness of scaling-up
HCV prevention and treatment in the United States for people who
inject drugs. Addiction 2019;114:226778. PMID:31307116 https://
doi.org/10.1111/add.14731
1378. Lambers FA, Prins M, Thomas X, et al.; MOSAIC (MSM
Observational Study of Acute Infection with hepatitis C) study
group. Alarming incidence of hepatitis C virus re-infection after
treatment of sexually acquired acute hepatitis C virus infection
in HIV-infected MSM. AIDS 2011;25:F217. PMID:21857492
https://doi.org/10.1097/QAD.0b013e32834bac44
Recommendations and Reports
182 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
1379. Martin TC, Singh GJ, McClure M, Nelson M. HCV reinfection
among HIV-positive men who have sex with men: a pragmatic
approach. Hepatology 2015;61:1437. PMID:25147047 https://doi.
org/10.1002/hep.27391
1380. Ingiliz P, Martin TC, Rodger A, et al.; NEAT study group. HCV
reinfection incidence and spontaneous clearance rates in HIV-
positive men who have sex with men in Western Europe. J Hepatol
2017;66:2827. PMID:27650285 https://doi.org/10.1016/j.
jhep.2016.09.004
1381. Briat A, Dulioust E, Galimand J, et al. Hepatitis C virus in the
semen of men coinfected with HIV-1: prevalence and origin. AIDS
2005;19:182735. PMID:16227790 https://doi.org/10.1097/01.
aids.0000189847.98569.2d
1382. Bissessor M, Fairley CK, Read T, Denham I, Bradshaw C,
Chen M. The etiology of infectious proctitis in men who have
sex with men differs according to HIV status. Sex Transm Dis
2013;40:76870. PMID:24275725 https://doi.org/10.1097/
OLQ.0000000000000022
1383. Gutierrez-Fernandez J, Medina V, Hidalgo-Tenorio C, Abad R. Two
cases of Neisseria meningitidis proctitis in HIV-positive men who have
sex with men. Emerg Infect Dis 2017;23:542–3. PMID:28221124
https://doi.org/10.3201/eid2303.161039
1384. Levy I, Gefen-Halevi S, Nissan I, et al. Delayed diagnosis of colorectal
sexually transmitted diseases due to their resemblance to inflammatory
bowel diseases. Int J Infect Dis 2018;75:34–8. PMID:30125691
https://doi.org/10.1016/j.ijid.2018.08.004
1385. Lebari D. Syphilis presenting as colorectal cancer [Abstract 34216].
Sex Transm Dis 2014;41(Suppl 1):S4.
1386. Hines JZ, Pinsent T, Rees K, et al. Notes from the field: shigellosis
outbreak among men who have sex with men and homeless
persons—Oregon, 2015–2016. MMWR Morb Mortal Wkly Rep
2016;65:812–3. PMID:27513523 https://doi.org/10.15585/mmwr.
mm6531a5
1387. Marchand-Senécal X, Bekal S, Pilon PA, Sylvestre JL, Gaudreau
C. Campylobacter fetus cluster among men who have sex with
men, Montreal, Quebec, Canada, 2014–2016. Clin Infect Dis
2017;65:17513. PMID:29020280 https://doi.org/10.1093/cid/
cix610
1388. Klausner JD, Kohn R, Kent C. Etiology of clinical proctitis among
men who have sex with men. Clin Infect Dis 2004;38:3002.
PMID:14699467 https://doi.org/10.1086/380838
1389. Stoner BP, Cohen SE. Lymphogranuloma venereum 2015: clinical
presentation, diagnosis, and treatment. Clin Infect Dis 2015;61(Suppl
8):S865–73. PMID:26602624 https://doi.org/10.1093/cid/civ756
1390. Mohrmann G, Noah C, Sabranski M, Sahly H, Stellbrink HJ.
Ongoing epidemic of lymphogranuloma venereum in HIV-positive
men who have sex with men: how symptoms should guide treatment.
J Int AIDS Soc 2014;17(Suppl 3):19657. PMID:25394161 https://
doi.org/10.7448/IAS.17.4.19657
1391. Hoffmann C, Sahly H, Jessen A, et al. High rates of quinolone-
resistant strains of Shigella sonnei in HIV-infected MSM. Infection
2013;41:9991003. PMID:23852945 https://doi.org/10.1007/
s15010-013-0501-4
1392. Heiman KE, Karlsson M, Grass J, et al.; CDC. Notes from the field:
Shigella with decreased susceptibility to azithromycin among men
who have sex with men—United States, 2002–2013. MMWR Morb
Mortal Wkly Rep 2014;63:132–3. PMID:24522098
1393. Galiczynski EM Jr, Elston DM. Whats eating you? Pubic lice (Pthirus
pubis). Cutis 2008;81:109–14. PMID:18441761
1394. Meinking TL, Serrano L, Hard B, et al. Comparative in vitro
pediculicidal efficacy of treatments in a resistant head lice
population in the United States. Arch Dermatol 2002;138:220–4.
PMID:11843643 https://doi.org/10.1001/archderm.138.2.220
1395. Yoon KS, Gao JR, Lee SH, Clark JM, Brown L, Taplin D. Permethrin-
resistant human head lice, Pediculus capitis, and their treatment.
Arch Dermatol 2003;139:994–1000. PMID:12925385 https://doi.
org/10.1001/archderm.139.8.994
1396. Burkhart CG, Burkhart CN. Oral ivermectin for Phthirus pubis.
J Am Acad Dermatol 2004;51:1037–8. PMID:15583618 https://
doi.org/10.1016/j.jaad.2004.04.041
1397. Scott GR, Chosidow O; IUSTI/WHO. European guideline for
the management of pediculosis pubis, 2010. Int J STD AIDS
2011;22:3045. PMID:21680662 https://doi.org/10.1258/
ijsa.2011.011114
1398. Goldust M, Rezaee E, Raghifar R, Hemayat S. Comparing the efficacy
of oral ivermectin vs malathion 0.5% lotion for the treatment of
scabies. Skinmed 2014;12:284–7. PMID:25632646
1399. Veraldi S, Schianchi R, Ramoni S, Nazzaro G. Pubic hair removal
and Phthirus pubis infestation. Int J STD AIDS 2018;29:103–4.
PMID:29130406 https://doi.org/10.1177/0956462417740292
1400. Leung AKC, Lam JM, Leong KF. Scabies: a neglected global disease.
Curr Pediatr Rev 2020;16:33–42. PMID:31544694 https://doi.org
/10.2174/1573396315666190717114131
1401. Engelman D, Cantey PT, Marks M, et al. The public health control
of scabies: priorities for research and action. Lancet 2019;394:8192.
PMID:31178154 https://doi.org/10.1016/S0140-6736(19)31136-5
1402. Shimose L, Munoz-Price LS. Diagnosis, prevention, and treatment
of scabies. Curr Infect Dis Rep 2013;15:426–31. PMID:23904181
https://doi.org/10.1007/s11908-013-0354-0
1403. Walter B, Heukelbach J, Fengler G, Worth C, Hengge U, Feldmeier H.
Comparison of dermoscopy, skin scraping, and the adhesive tape test
for the diagnosis of scabies in a resource-poor setting. Arch Dermatol
2011;147:46873. PMID:21482897 https://doi.org/10.1001/
archdermatol.2011.51
1404. Micali G, Lacarrubba F, Verzì AE, Chosidow O, Schwartz RA.
Scabies: advances in noninvasive diagnosis. PLoS Negl Trop Dis
2016;10:e0004691. PMID:27311065 https://doi.org/10.1371/
journal.pntd.0004691
1405. Strong M, Johnstone P. Interventions for treating scabies. Cochrane
Database Syst Rev 2007;(3):CD000320. PMID:17636630
1406. Abdel-Raheem TA, Méabed EM, Nasef GA, Abdel Wahed WY,
Rohaim RM. Efficacy, acceptability and cost effectiveness of four
therapeutic agents for treatment of scabies. J Dermatolog Treat
2016;27:473–9. PMID:27027929 https://doi.org/10.3109/09546
634.2016.1151855
1407. Alipour H, Goldust M. The efficacy of oral ivermectin vs. sulfur 10%
ointment for the treatment of scabies. Ann Parasitol 2015;61:79–84.
PMID:26342502
1408. Al Jaff DAA, Amin MHM. Comparison of the effectiveness of sulphur
ointment, permethrin and oral ivermectin in treatment of scabies. Res
J Pharm Biol Chem Sci 2018;9:670–6.
1409. Goldust M, Rezaee E, Raghifar R, Hemayat S. Treatment of scabies:
the topical ivermectin vs. permethrin 2.5% cream. Ann Parasitol
2013;59:79–84. PMID:24171301
1410. Ahmad HM, Abdel-Azim ES, Abdel-Aziz RT. Clinical efficacy and
safety of topical versus oral ivermectin in treatment of uncomplicated
scabies. Dermatol Ther (Heidelb) 2016;29:5863. PMID:26555785
https://doi.org/10.1111/dth.12310
1411. Currie BJ, McCarthy JS. Permethrin and ivermectin for scabies.
N Engl J Med 2010;362:71725. PMID:20181973 https://doi.
org/10.1056/NEJMct0910329
1412. Chiu S, Argaez C. Ivermectin for parasitic skin infections of scabies:
a review of comparative clinical effectiveness, cost-effectiveness,
and guidelines. Ottawa, ON: Canadian Agency for Drugs and
Technologies in Health; 2019. https://www.ncbi.nlm.nih.gov/books/
NBK545083/
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 183
US Department of Health and Human Services/Centers for Disease Control and Prevention
1413. Nolan K, Kamrath J, Levitt J. Lindane toxicity: a
comprehensive review of the medical literature. Pediatr
Dermatol 2012;29:1416. PMID:21995612 https://doi.
org/10.1111/j.1525-1470.2011.01519.x
1414. Mounsey KE, Holt DC, McCarthy J, Currie BJ, Walton SF.
Scabies: molecular perspectives and therapeutic implications in the
face of emerging drug resistance. Future Microbiol 2008;3:57–66.
PMID:18230034 https://doi.org/10.2217/17460913.3.1.57
1415. Mounsey KE, Holt DC, McCarthy JS, Currie BJ, Walton SF.
Longitudinal evidence of increasing in vitro tolerance of scabies
mites to ivermectin in scabies-endemic communities. Arch Dermatol
2009;145:8401. PMID:19620572 https://doi.org/10.1001/
archdermatol.2009.125
1416. Mounsey KE, McCarthy JS, Walton SF. Scratching the itch: new tools
to advance understanding of scabies. Trends Parasitol 2013;29:3542.
PMID:23088958 https://doi.org/10.1016/j.pt.2012.09.006
1417. van der Linden N, van Gool K, Gardner K, et al. A systematic review of
scabies transmission models and data to evaluate the cost-effectiveness
of scabies interventions. PLoS Negl Trop Dis 2019;13:e0007182.
PMID:30849124 https://doi.org/10.1371/journal.pntd.0007182
1418. Roberts LJ, Huffam SE, Walton SF, Currie BJ. Crusted scabies: clinical
and immunological findings in seventy-eight patients and a review
of the literature. J Infect 2005;50:375–81. PMID:15907543 https://
doi.org/10.1016/j.jinf.2004.08.033
1419. Ortega-Loayza AG, McCall CO, Nunley JR. Crusted scabies and
multiple dosages of ivermectin. J Drugs Dermatol 2013;12:584–5.
PMID:23652958
1420. Bouvresse S, Chosidow O. Scabies in healthcare settings. Curr
Opin Infect Dis 2010;23:1118. PMID:20075729 https://doi.
org/10.1097/QCO.0b013e328336821b
1421. Marotta M, Toni F, Dallolio L, Toni G, Leoni E. Management of a
family outbreak of scabies with high risk of spread to other community
and hospital facilities. Am J Infect Control 2018;46:80813.
PMID:29397231 https://doi.org/10.1016/j.ajic.2017.12.004
1422. Romani L, Whitfeld MJ, Koroivueta J, et al. Mass drug administration
for scabies control in a population with endemic disease. N Engl J Med
2015;373:230513. PMID:26650152 https://doi.org/10.1056/
NEJMoa1500987
1423. Ackerman DR, Sugar NF, Fine DN, Eckert LO. Sexual assault
victims: factors associated with follow-up care. Am J Obstet Gynecol
2006;194:16539. PMID:16635464 https://doi.org/10.1016/j.
ajog.2006.03.014
1424. Parekh V, Beaumont Brown C. Follow up of patients who have
been recently sexually assaulted. Sex Transm Infect 2003;79:349.
PMID:12902602 https://doi.org/10.1136/sti.79.4.349-a
1425. Vrees RA. Evaluation and management of female victims of sexual
assault. Obstet Gynecol Surv 2017;72:3953. PMID:28134394
https://doi.org/10.1097/OGX.0000000000000390
1426. Unger ER, Fajman NN, Maloney EM, et al. Anogenital human
papillomavirus in sexually abused and nonabused children: a
multicenter study. Pediatrics 2011;128:e658–65. PMID:21844060
https://doi.org/10.1542/peds.2010-2247
1427. Kreimer AR, Rodriguez AC, Hildesheim A, et al.; CVT Vaccine
Group. Proof-of-principle evaluation of the efficacy of fewer than
three doses of a bivalent HPV16/18 vaccine. J Natl Cancer Inst
2011;103:144451. PMID:21908768 https://doi.org/10.1093/jnci/
djr319
1428. Claydon E, Murphy S, Osborne EM, Kitchen V, Smith JR, Harris
JR. Rape and HIV. Int J STD AIDS 1991;2:200–1. PMID:1863649
https://doi.org/10.1177/095646249100200310
1429. Murphy S, Kitchen V, Harris JR, Forster SM. Rape and subsequent
seroconversion to HIV. BMJ 1989;299:718. PMID:2508885 https://
doi.org/10.1136/bmj.299.6701.718
1430. Cardo DM, Culver DH, Ciesielski CA, et al.; CDC Needlestick
Surveillance Group. A case-control study of HIV seroconversion
in health care workers after percutaneous exposure. N Engl J Med
1997;337:148590. PMID:9366579 https://doi.org/10.1056/
NEJM199711203372101
1431. Kuhar DT, Henderson DK, Struble KA, et al.; US Public Health
Service Working Group. Updated US Public Health Service
guidelines for the management of occupational exposures to human
immunodeficiency virus and recommendations for postexposure
prophylaxis. Infect Control Hosp Epidemiol 2013;34:87592.
PMID:23917901 https://doi.org/10.1086/672271
1432. Du Mont J, Myhr TL, Husson H, Macdonald S, Rachlis A, Loutfy
MR. HIV postexposure prophylaxis use among Ontario female
adolescent sexual assault victims: a prospective analysis. Sex Transm
Dis 2008;35:973–8. PMID:18836390
1433. Neu N, Heffernan-Vacca S, Millery M, Stimell M, Brown J.
Postexposure prophylaxis for HIV in children and adolescents after
sexual assault: a prospective observational study in an urban medical
center. Sex Transm Dis 2007;34:65–8. PMID:16794560 https://doi.
org/10.1097/01.olq.0000225329.07765.d8
1434. Loutfy MR, Macdonald S, Myhr T, et al. Prospective cohort study of
HIV post-exposure prophylaxis for sexual assault survivors. Antivir
Ther 2008;13:87–95. PMID:18389902
1435. Inciarte A, Leal L, Masfarre L, et al.; Sexual Assault Victims Study
Group. Post-exposure prophylaxis for HIV infection in sexual assault
victims. HIV Med 2020;21:4352. PMID:31603619 https://doi.
org/10.1111/hiv.12797
1436. Announcement: Updated guidelines for antiretroviral postexposure
prophylaxis after sexual, injection-drug use, or other nonoccupational
exposure to HIV—United States, 2016. MMWR Morb Mortal Wkly
Rep 2016;65:458. PMID:27149423 https://doi.org/10.15585/
mmwr.mm6517a5
1437. Ford N, Venter F, Irvine C, Beanland RL, Shubber Z. Starter packs
versus full prescription of antiretroviral drugs for postexposure
prophylaxis: a systematic review. Clin Infect Dis 2015;60(Suppl
3):S182–6. PMID:25972501 https://doi.org/10.1093/cid/civ093
1438. Jenny C, Crawford-Jakubiak JE; Committee on Child Abuse and
Neglect; American Academy of Pediatrics. The evaluation of children
in the primary care setting when sexual abuse is suspected. Pediatrics
2013;132:e55867. PMID:23897912 https://doi.org/10.1542/
peds.2013-1741
1439. Girardet RG, Lahoti S, Howard LA, et al. Epidemiology of
sexually transmitted infections in suspected child victims of sexual
assault. Pediatrics 2009;124:7986. PMID:19564286 https://doi.
org/10.1542/peds.2008-2947
1440. Black CM, Driebe EM, Howard LA, et al. Multicenter study of nucleic
acid amplification tests for detection of Chlamydia trachomatis and
Neisseria gonorrhoeae in children being evaluated for sexual abuse.
Pediatr Infect Dis J 2009;28:608–13. PMID:19451856 https://doi.
org/10.1097/INF.0b013e31819b592e
1441. Trotman GE, Young-Anderson C, Deye KP. Acute sexual assault in
the pediatric and adolescent population. J Pediatr Adolesc Gynecol
2016;29:51826. PMID:26702774 https://doi.org/10.1016/j.
jpag.2015.05.001
1442. Schwandt A, Williams C, Beigi RH. Perinatal transmission of
Trichomonas vaginalis: a case report. J Reprod Med 2008;53:59–61.
PMID:18251366
1443. Bell TA, Stamm WE, Wang SP, Kuo CC, Holmes KK, Grayston
JT. Chronic Chlamydia trachomatis infections in infants. JAMA
1992;267:4002. PMID:1727964 https://doi.org/10.1001/
jama.1992.03480030078041
Recommendations and Reports
184 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
1444. Adachi K, Nielsen-Saines K, Klausner JD. Chlamydia trachomatis
infection in pregnancy: the global challenge of preventing adverse
pregnancy and infant outcomes in sub-Saharan Africa and Asia.
BioMed Res Int 2016;2016:9315757. PMID:27144177 https://doi.
org/10.1155/2016/9315757
1445. Schachter J, Grossman M, Sweet RL, Holt J, Jordan C, Bishop E.
Prospective study of perinatal transmission of Chlamydia trachomatis.
JAMA 1986;255:3374–7. PMID:3712696 https://doi.org/10.1001/
jama.1986.03370240044034
1446. Smith EM, Swarnavel S, Ritchie JM, Wang D, Haugen TH, Turek
LP. Prevalence of human papillomavirus in the oral cavity/oropharynx
in a large population of children and adolescents. Pediatr Infect
Dis J 2007;26:836–40. PMID:17721381 https://doi.org/10.1097/
INF.0b013e318124a4ae
1447. Sabeena S, Bhat P, Kamath V, Arunkumar G. Possible non-sexual
modes of transmission of human papilloma virus. J Obstet Gynaecol
Res 2017;43:42935. PMID:28165175 https://doi.org/10.1111/
jog.13248
1448. Adams JA, Farst KJ, Kellogg ND. Interpretation of medical findings
in suspected child sexual abuse: an update for 2018. J Pediatr
Adolesc Gynecol 2018;31:22531. PMID:29294380 https://doi.
org/10.1016/j.jpag.2017.12.011
1449. Kellogg ND, Melville JD, Lukefahr JL, Nienow SM, Russell EL.
Genital and extragenital gonorrhea and chlamydia in children
and adolescents evaluated for sexual abuse. Pediatr Emerg Care
2018;34:7616. PMID:28072668 https://doi.org/10.1097/
PEC.0000000000001014
1450. Gavril AR, Kellogg ND, Nair P. Value of follow-up examinations
of children and adolescents evaluated for sexual abuse and
assault. Pediatrics 2012;129:282–9. PMID:22291113 https://doi.
org/10.1542/peds.2011-0804
1451. Bandea CI, Joseph K, Secor EW, et al. Development of PCR assays
for detection of Trichomonas vaginalis in urine specimens. J Clin
Microbiol 2013;51:1298300. PMID:23390274 https://doi.
org/10.1128/JCM.03101-12
1452. Gallion HR, Dupree LJ, Scott TA, Arnold DH. Diagnosis of
Trichomonas vaginalis in female children and adolescents evaluated
for possible sexual abuse: a comparison of the InPouch TV culture
method and wet mount microscopy. J Pediatr Adolesc Gynecol
2009;22:300–5. PMID:19576816 https://doi.org/10.1016/j.
jpag.2008.12.006
1453. Lalor K, McElvaney R. Child sexual abuse, links to later
sexual exploitation/high-risk sexual behavior, and prevention/
treatment programs. Trauma Violence Abuse 2010;11:15977.
PMID:20679329 https://doi.org/10.1177/1524838010378299
1454. Girardet RG, Lemme S, Biason TA, Bolton K, Lahoti S. HIV
post-exposure prophylaxis in children and adolescents presenting
for reported sexual assault. Child Abuse Negl 2009;33:173–8.
PMID:19324415 https://doi.org/10.1016/j.chiabu.2008.05.010
1455. Panel on Antiretroviral Therapy and Medical Management of HIV-
Infected Children. Guidelines for the use of antiretroviral agents in
pediatric HIV infection. Washington, DC: US Department of Health
and Human Services, National Institutes of Health, AIDSinfo; 2020.
https://aidsinfo.nih.gov/contentfiles/lvguidelines/pediatricguidelines.pdf
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 185
US Department of Health and Human Services/Centers for Disease Control and Prevention
STI Treatment Guidelines, 2021, Work Group Members
Chairperson: Kimberly A. Workowski, MD, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, CDC, and Emory University,
Atlanta, Georgia.
Work Group Members: Elizabeth Alderman, MD, The Childrens Hospital at Montefiore, The Pediatric Hospital for Albert Einstein College of Medicine,
Bronx, New York; Lindley Barbee, MD, University of Washington and Public Health Seattle and King County STD Clinic, Seattle, Washington; Luis
Barroso, MD, Wake Forest Baptist Health, Winston-Salem, North Carolina; Gale Burstein, MD, Erie County Department of Health, Buffalo, New York;
Virginia A. Caine, MD, Marion County Public Health Department, Indiana University, Indianapolis, Indiana; Carla Chibwesha, MD, University of North
Carolina at Chapel Hill, Chapel Hill, North Carolina; Stephanie Cohen, MD, San Francisco Department of Public Health, San Francisco, California; Susan
Cu-Uvin, MD, Brown University, Providence, Rhode Island; Jodie Dionne-Odom, MD, University of Alabama at Birmingham, Birmingham, Alabama; Julia
Dombrowski, MD, Public Health Seattle and King County, University of Washington, Seattle, Washington; Carolyn Gardella, MD, Veterans Affairs Puget
Sound University of Washington, Seattle, Washington; William M. Geisler, MD, University of Alabama at Birmingham, Birmingham, Alabama; Khalil G.
Ghanem, MD, PhD, Johns Hopkins University, Baltimore, Maryland; Matthew Golden, MD, Seattle and King County STD/HIV Program University of
Washington, Seattle, Washington; Margaret R. Hammerschlag, MD, SUNY Downstate Medical Center, Brooklyn, New York; H. Hunter Handsfield, MD,
University of Washington Center for AIDS and STD, Seattle, Washington; King K. Holmes, MD, PhD, University of Washington, Seattle, Washington;
Edward W. Hook, III, MD, University of Alabama at Birmingham, Birmingham, Alabama; Katherine Hsu, MD, Ratelle STD/HIV Prevention Training
Center, Massachusetts Department of Public Health, Boston, Massachusetts; Christine Johnston, MD, University of Washington, Seattle, Washington; Patricia
Kissinger, PhD, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana; Jeffrey D. Klausner, MD, University of California
Los Angeles, Los Angeles, California; Lisa E. Manhart, PhD, University of Washington, Seattle, Washington; Jeanne Marrazzo, MD, University of Alabama at
Birmingham, Birmingham, Alabama; David H. Martin, MD, Tulane University, Louisiana State University Health Sciences Center, New Orleans, Louisiana;
Leslie Massad, MD, Washington University, St. Louis, Missouri; Kenneth H. Mayer, MD, Fenway Health, The Fenway Institute, Harvard Medical School,
Beth Israel Deaconess Hospital, Boston, Massachusetts; Candice McNeil, MD, Wake Forest Baptist Health, Alabama/North Carolina HIV/STD Prevention
Training Center, Forsyth County Department of Public Health, Winston-Salem, North Carolina; Leandro Mena, MD, University of Mississippi Medical
Center, Jackson, Mississippi; Caroline Mitchell, MD, Vincent Center for Reproductive Biology, Boston, Massachusetts; Okeoma Mmeje, MD, University
of Michigan, Ann Arbor, Michigan; Anna-Barbara Moscicki, MD, University of California Los Angeles, Los Angeles, California; Christina A. Muzny, MD,
University of Alabama at Birmingham, Birmingham, Alabama; Natalie Neu, MD, Columbia University, New York, New York; Paul Nyirjesy, MD, Sidney
Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania; Jeff Peipert, MD, PhD, Indiana University, Indianapolis, Indiana;
Rebecca Perkins, MD, Boston University, Boston, Massachusetts; Susan Philip, MD, San Francisco Department of Public Health, San Francisco, California;
Asa Radix, MD, PhD, Callen-Lorde Community Health Center, New York, New York; Anne Rompalo, MD, Johns Hopkins University, Baltimore, Maryland;
Sarah Rudman, MD, County of Santa Clara Public Health Department, San Jose, California; Pablo J. Sanchez, MD, The Ohio State University Nationwide
Childrens Hospital, Columbus, Ohio; George Sawaya, MD, University of San Francisco, San Francisco, California; Arlene Seña, MD, University of North
Carolina at Chapel Hill and Durham County Department of Public Health, Durham, North Carolina; Jeanne Sheffield, MD, Johns Hopkins University,
Baltimore, Maryland; Jack Sobel, MD, Wayne State University, Detroit, Michigan; David Soper, MD, Medical University of South Carolina, Charleston, South
Carolina; Anne Spaulding, MD, Emory University, Atlanta, Georgia; Bradley Stoner, MD, PhD, Washington University, St. Louis, Missouri; Stephanie Taylor,
MD, Louisiana State University Crescent Care Sexual Health Center, New Orleans, Louisiana; Susan Tuddenham, MD, Johns Hopkins University School
of Medicine, Baltimore, Maryland; Karen Wendel, MD, Denver Public Health, Denver, Colorado; Harold C. Wiesenfeld, MD, University of Pittsburgh,
Pittsburgh, Pennsylvania; Jonathan Zenilman, MD, Johns Hopkins University, Baltimore, Maryland.
Moderators: Philip Chan, MD, Brown University, Providence, Rhode Island; Edward W. Hook, III, MD, University of Alabama at Birmingham, Birmingham,
Alabama; Christine Johnston, MD, University of Washington, Seattle, Washington; David H. Martin, MD, Tulane University, Louisiana State University
Health Sciences Center, New Orleans, Louisiana; Jeanne Marrazzo, MD, University of Alabama at Birmingham, Birmingham, Alabama; Christina A. Muzny,
MD, University of Alabama at Birmingham, Birmingham, Alabama; Ina Park, MD, University of California San Francisco, San Francisco, California; Hilary
Reno, MD, PhD, Washington University, St. Louis, Missouri; Jonathan Zenilman, MD, Johns Hopkins University, Baltimore, Maryland.
Rapporteurs: Philip Chan, MD, Brown University, Providence, Rhode Island; Hilary Reno, MD, PhD, Washington University, St. Louis, Missouri; Christine
Johnston, MD, University of Washington, Seattle, Washington; Christina A. Muzny, MD, University of Alabama at Birmingham, Birmingham, Alabama;
Ina Park, MD, University of California San Francisco, San Francisco, California; Jonathan Zenilman, MD, Johns Hopkins University, Baltimore, Maryland
Liaison Participants: Jean Anderson, MD, CDC/HRSA Advisory Committee on HIV, Viral Hepatitis and STD Prevention and Treatment, Johns Hopkins
University, Baltimore, Maryland; Kevin Ault, MD, American College of Obstetricians and Gynecologists, University of Kansas, Lawrence, Kansas; Himani
Bisht, PhD, Food and Drug Administration, Silver Spring, Maryland; J. Quentin Clemens, MD, American Urological Association, University of Michigan, Ann
Arbor, Michigan; Gina Coleman, MD, Public Health Agency of Canada, Ottawa, Ontario; Carolyn Deal, PhD, National Institute of Allergy and Infectious
Diseases, Bethesda, Maryland; Maeve B. Mello, PhD, Pan American Health Organization/World Health Organization, Washington, DC; Diane Foley, MD,
U.S. Department of Health and Human Services, Washington, DC; Michael Ganio, PharmD, American Society of Health-System Pharmacists, Bethesda,
Maryland; Eric Garges, MD, U.S. Department of Defense, Arlington, Virginia, and Uniformed Services University, Bethesda, Maryland; Brent Gibson, MD,
National Commission on Correctional Health Care, Chicago, Illinois; William A. Glover, II, PhD, Association of Public Health Laboratories, Silver Spring,
Maryland; W. David Hardy, MD, HIV Medical Association, Johns Hopkins University, Baltimore, Maryland; David Harvey, National Coalition of STD
Directors, Washington, DC; Letha Healey, MD, Health Resources and Services Administration (HIV/AIDS Bureau), Rockville, Maryland; Michael Huey,
MD, American College Health Association, Silver Spring, Maryland, and Emory University, Atlanta, Georgia; Gal Mayer, MD, GLMA: Health Professionals
Advancing LGBTQ Equality, Washington, DC; Sumathi Nambiar, MD, Food and Drug Administration, Silver Spring, Maryland; Seema Nayak, MD, National
Institutes of Health, Rockville, Maryland; Lori Newman, MD, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland; Samantha Ritter,
MPH, National Association of County and City Health Officials, Washington, DC; Elizabeth Ruebush, Association of State and Territorial Health Officials,
Arlington, Virginia; Bisan Salhi, MD, PhD, American College of Emergency Physicians, Irving, Texas, Emory University, Atlanta, Georgia; Natasha Travis,
MD, National Medical Association, Silver Spring, Maryland, and Johns Hopkins University, Baltimore, Maryland; Maria Trent, MD, Society for Adolescent
Health and Medicine, Chicago, Illinois, and Johns Hopkins University, Baltimore, Maryland; Julie Vaishampayan, MD, Infectious Disease Society of America,
Arlington, Virginia; Barbara Van Der Pol, PhD, American Sexually Transmitted Diseases Association, Research Triangle Park, North Carolina, and University
of Alabama at Birmingham, Birmingham, Alabama; Teodora Elvira Wi, MD, Department of Global HIV, Hepatitis and STI Programmes, World Health
Organization, Geneva, Switzerland.
Recommendations and Reports
186 MMWR / July 23, 2021 / Vol. 70 / No. 4
US Department of Health and Human Services/Centers for Disease Control and Prevention
CDC and Outside CDC Consultants: Sevgi O. Aral, PhD; Laura H. Bachmann, MD; Kyle Bernstein, PhD; Gail Bolan, MD; John Brooks, MD; Philip Chan,
MD, Brown University, Providence, Rhode Island; Aaron Harris, MD; Matthew Hogben, PhD; Christine Johnston, MD, University of Washington, Seattle,
Washington; Sarah Kidd, MD; Kristen Kreisel, PhD; Lauri Markowitz, MD; Christina Muzny, MD, University of Alabama at Birmingham, Birmingham,
Alabama; Ina Park, MD, University of California San Francisco, San Francisco, California; Allan Pillay, PhD; Laura Quilter, MD; Hilary Reno, MD, PhD,
Washington University, St. Louis, Missouri; Sancta St. Cyr, MD; Mona Saraiya, MD; Julie Schillinger, MD; Virginia Senkomago, PhD; Philip Spradling,
MD; Phoebe Thorpe, MD; Mark Weng, MD; Jonathan Zenilman, MD, Johns Hopkins University, Baltimore, Maryland.
CDC, Division of Sexually Transmitted Disease Prevention Treatment Guidelines, 2021, Project Coordinator: Kimberly A. Workowski, MD, National
Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, CDC, and Emory University, Atlanta, Georgia.
CDC Support Staff: Alexandra Coor, MPH; Catherine Carter; Quinn Haaga; Amber Herald; Patricia Jackson; Brenda Kelly.
Recommendations and Reports
MMWR / July 23, 2021 / Vol. 70 / No. 4 187
US Department of Health and Human Services/Centers for Disease Control and Prevention
Disclosure of Relationships
All STI Treatment Guidelines, 2021, Work Group members have completed and submitted the International Committee of Medical Journal Editors form for
disclosure of potential conflicts of interest. Lindley Barbee received research support from Hologic and SpeeDx and consults for Nabriva. Christine Johnston
received research funding from Sanofi-Pasteur; royalties from UpToDate; and personal fees from MedPace, Gilead, AbbVie, and UpToDate. Jeffrey Klausner
received research supplies from Hologic, Cepheid, and SpeeDx; advisory fees from Shield Diagnostics, Click Diagnostics, and SpeeDx; and research funding
from AbbVie and Gilead Sciences. Lisa Manhart received research supplies, honoraria, and research contracts from Hologic and served as a consultant to
diagnostic test manufacturers (SpeeDx, BioFire Diagnostics, Roche Diagnostics, and Click Diagnostics). Jeanne Marrazzo serves on expert advisory boards for
Becton Dickinson, BioFire Diagnostics, and Gilead Sciences and has research and grant support and supplies from Merck & Co. and Toltec. David Martin
served as a consultant in 2019 to Hologic, which recently gained Food and Drug Administration clearance for a Mycoplasma genitalium diagnostic test and
markets diagnostic tests for gonorrhea, chlamydia, and trichomonas. Kenneth Mayer serves on the scientific advisory boards for Gilead Sciences and Merck
& Co. Pharmaceuticals and receives grant funds from Gilead Sciences. Leandro Mena received research funding through his institution from Atlas Genetics,
Merck & Co., Becton Dickinson, Hologic, Biolytical, and Roche and received consulting honoraria from Roche Molecular, Merck & Co., Gilead Sciences, and
ViiV Healthcare and from speaker bureaus for Gilead Sciences and ViiV Healthcare. Caroline Mitchell served as a consultant for Lupin Pharmaceuticals and
Scynexis and received grant funding from Merck & Co. Christina A. Muzny served as a consultant and received research support from Lupin Pharmaceuticals
for a randomized controlled trial of secnidazole versus placebo for treatment of trichomoniasis and grants from the National Institutes of Health/National
Institute of Allergy and Infectious Diseases; was a speaker for Abbott Molecular, Cepheid, and Roche Diagnostics on topics related to STIs; received personal
fees from Lupin Pharmaceuticals, PhagoMed, Cepheid, and Beckton Dickinson; and served as a consultant for BioFire Diagnostics. Paul Nyirjesy received
research support from Mycovia Pharmaceuticals, Curatek Pharmaceuticals, Scynexis, and Hologic and served as a consultant for Mycovia Pharmaceuticals,
Hologic, Scynexis, Daré Bioscience, and Becton Dickinson. Jeffery Pepest serves on advisory boards for Cooper Surgical and Bayer and provides research support
to Merck & Co. Susan Philip is an unpaid public health advisor to GlaxoSmithKline. Anne Rompalo serves on the BioFire Diagnostics advisory board and
has financial ties to UpToDate. Hilary Reno is a site principle investigator on a project evaluating the prevalence of M. genitalium funded by Hologic (funds
are allocated via her institution). Arlene Seña serves on a speakers bureau and scientific advisory board for M. genitalium at Hologic, works with the Gilead
Focus grant for hepatitis C testing and linkage to care, and mentored Sancta St. Cyr, MD, at the University of North Carolina Division of Infectious Diseases
before employment at CDC. Anne Spaulding received consulting fees or honoraria (either directly or through third parties) from Gilead Sciences, Merck &
Co., and AbbVie and grants through her institution from Gilead Sciences and ViiV. Susan Tuddenham served as a consultant for Biofire Diagnostics and Roche
Molecular Diagnostics and received a speaker honorarium from Roche Molecular Diagnostics. Karen Wendel has stock ownership in Pfizer.
ISSN: 1057-5987 (Print)
The Morbidity and Mortality Weekly Report (MMWR) Series is prepared by the Centers for Disease Control and Prevention (CDC) and is available free
of charge in electronic format. To receive an electronic copy each week, visit MMWR at https://www.cdc.gov/mmwr/index.html.
Readers who have difficulty accessing this PDF file may access the HTML file at https://www.cdc.gov/mmwr/volumes/70/rr/rr7004a1.htm?s_
cid=rr7004a1_w. Address all inquiries about the MMWR Series to Editor-in-Chief, MMWR Series, Mailstop V25-5, CDC, 1600 Clifton Rd., N.E.,
Atlanta, GA 30329-4027 or to [email protected]v.
All material in the MMWR Series is in the public domain and may be used and reprinted without permission; citation as to source, however, is appreciated.
MMWR and Morbidity and Mortality Weekly Report are service marks of the U.S. Department of Health and Human Services.
Use of trade names and commercial sources is for identification only and does not imply endorsement by the U.S. Department of Health and Human Services.
References to non-CDC sites on the Internet are provided as a service to MMWR readers and do not constitute or imply endorsement of these organizations
or their programs by CDC or the U.S. Department of Health and Human Services. CDC is not responsible for the content of these sites. URL addresses
listed in MMWR were current as of the date of publication.