You are on page 1of 11

Annals of Internal Medicine Original Research

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
Connie Celum, MD, MPH; Rhoda A. Morrow, PhD; Deborah Donnell, PhD; Ting Hong, MD, PhD; Craig W. Hendrix, MD, PhD;
Katherine K. Thomas, MS; Kenneth H. Fife, MD, PhD; Edith Nakku-Joloba, MBChB, PhD; Andrew Mujugira, MBChB, MPH; and
Jared M. Baeten, MD, PhD, for the Partners PrEP Study Team*

Background: Daily oral preexposure prophylaxis (PrEP) using the per 100 person-years] and 52 of 481 assigned to placebo [HSV-2
antiretroviral tenofovir disoproxil fumarate (TDF) alone or in com- incidence, 7.7 per 100 person-years]). The hazard ratio (HR) for
bination with emtricitabine (FTC–TDF) reduces the risk for HIV-1 HSV-2 acquisition with daily oral PrEP was 0.70 (95% CI, 0.49 to
acquisition. Tenofovir has in vitro activity against herpes simplex 0.99; P ⫽ 0.047) compared with placebo, and the absolute risk
virus type 2 (HSV-2). reduction was 2.1 per 100 person-years. Among the 1044 partic-
ipants with HSV-2–infected partners, the HR for PrEP was 0.67 (CI,
Objective: To assess the efficacy of daily oral PrEP with tenofovir 0.46 to 0.98; P ⫽ 0.038) compared with placebo, and the absolute
and FTC–TDF in the prevention of HSV-2 acquisition. risk reduction was 3.1 per 100 person-years.
Design: Subgroup analysis of data from a randomized, placebo- Limitation: Randomization was not stratified by HSV-2 status, and
controlled trial with concealed allocation. (ClinicalTrials.gov: diagnostic tests to exclude participants with acute HSV-2 at base-
NCT00557245) line are not available.
Setting: Multiple sites in Kenya and Uganda. Conclusion: Daily oral tenofovir-based PrEP significantly reduced
Participants: Heterosexual men and women who were seronega- the risk for HSV-2 acquisition among heterosexual men and
tive for HIV-1 and HSV-2 and at high risk for HIV-1 acquisition due women. Modest protection against HSV-2 is an added benefit of
to having an HIV-1–infected partner. HIV-1 prevention with oral tenofovir-based PrEP.

Intervention: Once-daily oral tenofovir disoproxil fumarate (TDF), Primary Funding Source: Bill & Melinda Gates Foundation.
alone or combined with emtricitabine (FTC-TDF), compared with
placebo.

Measurements: HSV-2 seroconversion. Ann Intern Med. 2014;161:11-19. doi:10.7326/M13-2471 www.annals.org


For author affiliations, see end of text.
Results: A total of 131 participants seroconverted to HSV-2 (79 of * For a list of the members of the Partners PrEP Study Team, see the
1041 assigned to tenofovir or FTC–TDF PrEP [HSV-2 incidence, 5.6 Appendix (available at www.annals.org).

P reexposure prophylaxis (PrEP) with the oral antiretro-


viral tenofovir disoproxil fumarate (TDF) alone or in
combination with emtricitabine (FTC–TDF) has been
The efficacy of daily oral tenofovir-based PrEP for
HSV-2 protection has not been assessed in heterosexual
populations. In vitro data have shown that high concentra-
shown to significantly reduce the risk for HIV-1 acquisi- tions of tenofovir are required for HSV-2 inhibition, and
tion in 4 randomized, placebo-controlled clinical trials in high adherence to oral PrEP may be necessary to achieve
high-risk populations (1– 4). In addition to its potent ac- such levels. We performed a secondary analysis of HSV-2
tivity as a nucleotide inhibitor of HIV-1 reverse transcrip- acquisition among participants who were initially seroneg-
tase, tenofovir has anti– herpes simplex virus type 2 ative for HIV-1 and HSV-2 in a randomized trial of PrEP
(HSV-2) activity in vitro, although less than the related for HIV-1 prevention that showed high efficacy for HIV-1
compounds cidofovir and adefovir (5). In a randomized protection (2).
trial of pericoital dosing of intravaginal 1% tenofovir gel,
HSV-2 incidence was reduced by 51% compared with pla-
cebo (6 – 8) and vaginal tenofovir levels were associated METHODS
with protection against HIV-1 and HSV-2 (9). These re- Study Population and Procedures
sults were consistent with in vitro studies showing antiher- Between July 2008 and November 2010, a total of
petic activity against HSV-2 isolates at concentrations of 4747 heterosexual HIV-1–serodiscordant couples were en-
10 to 200 ␮g/mL (10), levels that are in the range of those rolled in a randomized trial of PrEP for HIV-1 prevention
achieved with vaginal dosing of tenofovir gel (11). How- (the Partners PrEP Study) and followed at 9 research sites
ever, whether tenofovir concentrations achieved through in Kenya and Uganda (2, 12). The primary aims of the
oral dosing are sufficient for HSV-2 protection in humans study were to determine the efficacy and safety of PrEP
is unknown. with tenofovir and FTC–TDF, each compared with pla-
© 2014 American College of Physicians 11
This article has been corrected. The specific correction appears on the last page of this document. The original version (PDF) is available at www.annals.org.
Downloaded From: http://annals.org/pdfaccess.ashx?url=/data/journals/aim/930448/ by a Suny Health Sciences Ctr User on 06/17/2017
Original Research HSV-2 Protection With Tenofovir-Based Preexposure Prophylaxis

Context
A secondary analysis of HSV-2 protection from oral
tenofovir-based PrEP compared with placebo was planned
Herpes simplex virus type 2 (HSV-2) infection is common
after July 2010, when a randomized trial of pericoital dos-
worldwide, can lead to complications, and has been asso-
ciated with an increased risk for HIV-1 infection.
ing of 1% tenofovir gel showed efficacy against HSV-2
acquisition (6 – 8). This finding was not anticipated in the
Contribution original design of trials of tenofovir-based PrEP but could
In a secondary analysis of a large randomized clinical trial be assessed among the subset of participants who were
testing the ability of tenofovir-based preexposure regimens HSV-2– uninfected at baseline. All HSV-2 testing and de-
to prevent HIV-1 infection, participants receiving tenofovir termination of end points was concluded before unblind-
were less likely to acquire HSV-2 than those receiving ing of HSV-2 outcomes. To correspond with the period
placebo. covered in the primary HIV-1 acquisition results from the
Caution trial, we included data through July 2011 in our analysis of
The trial was not originally designed to study HSV-2 HSV-2 acquisition. Because the objective of this secondary
acquisition. analysis of HSV-2 prevention was to determine the effect
of daily oral tenofovir on HSV-2 incidence, this report
Implication includes data only on participants who did not have
Protection against HSV-2 acquisition may be an added HSV-2 infection at baseline (Figure 1).
benefit of using tenofovir for HIV-1 preexposure The study protocol was approved by the University
prophylaxis. of Washington Human Subjects Review Committee and
ethics review committees at each of the study sites. All
—The Editors
participants provided written informed consent in English
or their local language.
cebo, for prevention of HIV-1 acquisition. Eligibility cri-
teria for HIV-1–seronegative partners included normal re- Laboratory Procedures
nal function and not being pregnant or breastfeeding. At HSV-2 Serologic Tests
enrollment, HIV-1–seronegative partners were assigned in For HIV-1– uninfected partners, we tested archived
a 1:1:1 ratio to once-daily oral tenofovir, FTC–TDF, or baseline sera for HSV-2 antibodies by using an HSV-2–
placebo; investigators and participants were blinded to specific enzyme immunoassay (EIA) (Focus Technologies,
randomization assignments. Randomization was by fixed- Cypress, California). On the basis of previous experience
size blocks and was stratified by site but not by HSV-2 with this EIA in East Africa (13), index value results less
serostatus. Participants who were HIV-1–seronegative had than 0.9 were considered to represent HSV-2– uninfected
monthly visits for up to 36 months that included HIV-1 status, values of 3.5 or greater signified HSV-2–infected
testing, dispensation of study medication, and standardized status, and values of 0.9 to 3.4 were considered indetermi-
assessment of sexual behavior and clinical and laboratory nate. We used HSV-2–specific Western blot to clarify in-
safety. Study medication was withheld from women who determinate results (14). For participants determined to be
became pregnant or began breastfeeding. All participants HSV-2–seronegative at baseline, serostatus at the final visit
received a comprehensive package of HIV-1 prevention through July 2011 was then assessed with an HSV-2–
services, including individual and couples risk-reduction specific EIA and Western blot using the same approach as
counseling, screening for and treatment of sexually trans- for baseline samples. Finally, individuals found to have
mitted infections, condoms, and referral for medical male seroconverted to HSV-2 between baseline and their final
circumcision and postexposure prophylaxis according to follow-up visit had sera from study months 1, 3, 6, 12, 18,
national policies. Partners infected with HIV-1 were not
24, 30, and 36 tested in batch by HSV-2–specific Western
eligible for antiretroviral therapy (ART) under the national
blot to determine the timing of seroconversion. An HSV-2
policies of Kenya and Uganda at the time of their enroll-
end point committee reviewed the serologic data in a
ment in the trial; they were followed in parallel with their
HIV-1– uninfected partners and referred for initiation of blinded manner to adjudicate end points.
ART when they became eligible. Participants with HIV-1 We tested HSV-2 serostatus at baseline for the HIV-1–
seroconversion were referred for care, but the standard of infected partners of HSV-2–seronegative participants. In-
care in Kenya and Uganda is to not treat acute HIV-1 dex EIA values less than 0.9 signified HSV-2– uninfected
infection, although clinical teams referred pregnant women status, values of 3.5 or greater signified HSV-2–infected
with acute HIV-1 infection for prompt ART initiation. At status, and HSV-2–specific Western blot was used to con-
an interim review in July 2011, the trial’s independent data firm the status of EIA values between 0.9 and 3.4. We did
and safety monitoring board recommended that the pla- not conduct longitudinal testing among HIV-1–infected
cebo group be discontinued early because of clear demon- partners who were HSV-2–seronegative because of the
stration of PrEP efficacy for HIV-1 prevention (2). high baseline prevalence of HSV-2.
12 1 July 2014 Annals of Internal Medicine Volume 161 • Number 1 www.annals.org

Downloaded From: http://annals.org/pdfaccess.ashx?url=/data/journals/aim/930448/ by a Suny Health Sciences Ctr User on 06/17/2017


HSV-2 Protection With Tenofovir-Based Preexposure Prophylaxis Original Research

Figure 1. Study flow diagram.

Eligible couples randomly assigned (n = 4747)

Randomly assigned to TDF Randomly assigned to FTC–TDF Randomly assigned to placebo


(n = 1584) (n = 1579) (n = 1584)

Excluded (n = 1071) Excluded (n = 1051) Excluded (n = 1103)


HSV-2–positive: 973 HSV-2–positive: 964 HSV-2–positive: 1012
Indeterminate HSV-2 status: 56 Indeterminate HSV-2 status: 56 Indeterminate HSV-2 status: 55
Missing result: 42 Missing result: 31 Missing result: 36

HSV-2–negative at enrollment HSV-2–negative at enrollment HSV-2–negative at enrollment


(n = 513) (n = 528) (n = 481)

Retention Retention Retention


6 mo: 495/511 (96.9%) 6 mo: 501/525 (95.4%) 6 mo: 463/479 (96.7%)
12 mo: 396/425 (93.2%) 12 mo: 420/453 (92.7%) 12 mo: 399/422 (94.6%)
24 mo: 199/219 (90.9%) 24 mo: 214/233 (91.9%) 24 mo: 207/224 (92.4%)
36 mo: 2/2 (100%) 36 mo: 3/3 (100%) 36 mo: 2/2 (100%)

No follow-up (n = 8)
No follow-up (n = 7) Indeterminate HSV-2 status at No follow-up (n = 8)
last visit (n = 1)

Analyzed (n = 506) Analyzed (n = 519) Analyzed (n = 473)


HSV-2–positive: 42 HSV-2–positive: 37 HSV-2–positive: 52 Analytic
HSV-2–negative: 464 HSV-2–negative: 482 HSV-2–negative: 421 sample

Retention of partners who were seronegative for HIV-1 and HSV-2 was calculated as cumulative retention, which was defined as remaining in follow-up
and having available HSV-2 serostatus information. A total of 131 participants seroconverted to HSV-2 (42 in the tenofovir group, 37 in the FTC–TDF
group, and 52 in the placebo group) and were included in the intention-to-treat analysis. FTC–TDF ⫽ emtricitabine in combination with tenofovir
disoproxil fumarate; HSV-2 ⫽ herpes simplex virus type 2; TDF ⫽ tenofovir disoproxil fumarate.

Plasma Tenofovir Levels Statistical Analysis


We used a case– cohort design to select a random sam- The primary outcome was HSV-2 seroconversion
ple of participants from the active PrEP groups for assess- among persons who were HSV-2–seronegative at baseline,
ment of plasma concentrations of tenofovir. Case partici- and the primary analysis was intention-to-treat. The only
pants were those with HSV-2 seroconversion in the active exception was that participants who acquired HIV-1 were
PrEP groups, and control participants were a random sam- censored after the first visit at which they were HIV-1–
ple (selected in a 2:1 ratio compared with case participants) seropositive because the study drug was withheld at that
who were assigned to active PrEP and did not acquire time. Thus, those with HIV-1 seroconversion contributed
HSV-2. We assessed plasma tenofovir levels in case and follow-up time for assessment of HSV-2 up to and includ-
control participants at 1, 3, 6, 12, 18, 24, 30, and 36 ing the visit when HSV-2 seroconversion or HIV-1 sero-
months after enrollment, thus including testing at the visits conversion was detected, whichever occurred first. We
at which HSV-2 seroconversion was detected for case par- compared both active PrEP groups (FTC–TDF and teno-
ticipants. Plasma tenofovir concentrations were determined fovir combined) with the placebo group given that teno-
by previously described liquid chromatography–mass spec- fovir was the common active moiety against HSV-2 in
trometry methods (15). both groups and emtricitabine is not known to have activ-
ity against HSV-2 (16). Assessment of HSV-2 was interval-
Plasma HIV-1 RNA Levels and CD4 Cell Counts in censored at the specified study visits, so discrete time pro-
HIV-1–Infected Partners portional hazards models were used to estimate hazard
We quantified CD4 cell counts by using standard flow ratios (HRs) for HSV-2 infection (17). We fit these models
cytometry and plasma HIV-1 RNA levels by using the using the PROC LOGISTIC statement in SAS, version
COBAS AmpliPrep/COBAS TaqMan real-time HIV-1 9.3 (SAS Institute, Cary, North Carolina), with a comple-
RNA assay, version 1.0 (Roche Diagnostics, Indianapolis, mentary log-log link, including visit interval as a class vari-
Indiana), with a lower limit of quantification of 240 able without an intercept term, in addition to the covariate
copies/mL. of interest (treatment group). The proportional hazards as-
www.annals.org 1 July 2014 Annals of Internal Medicine Volume 161 • Number 1 13

Downloaded From: http://annals.org/pdfaccess.ashx?url=/data/journals/aim/930448/ by a Suny Health Sciences Ctr User on 06/17/2017


Original Research HSV-2 Protection With Tenofovir-Based Preexposure Prophylaxis

sumption was tested by including interactions between donated the study medication but had no role in data col-
visit and treatment group. In a sensitivity analysis, we also lection or analysis.
stratified the proportional hazards model by site. Prespeci-
fied subgroup analyses were performed to explore the uni-
formity of treatment effects found in the overall analysis. RESULTS
These included baseline covariates of sex of HIV-1– Of the 4747 HIV-1– uninfected partners enrolled in
uninfected partners; circumcision status of HIV-1– the trial, 4638 (97.7%) had baseline samples available for
uninfected male participants; and plasma HIV-1 RNA HSV-2 testing; of these, 1522 (32.8%) were HSV-2–
level (ⱖ50 000 vs. ⬍50 000 copies/mL), CD4 count seronegative (Figure 1), of whom 1498 (98.4%) had a final
(⬍0.350 vs. ⱖ0.350 ⫻ 109 cells/L), and HSV-2 serostatus study visit sample for HSV-2 testing. The median age of
of HIV-1–infected partners. the initially HSV-2–seronegative participants was 31 years,
In addition to the primary proportional hazards anal- and 80% were men, 40% of whom were uncircumcised. At
ysis, we conducted sensitivity analyses to examine the po- enrollment, 96% were married to their HIV-1–infected
tential effect of confounding and bias that could have re- partner, 8% reported an outside partner, the median num-
sulted from using the subset of the HSV-2– uninfected ber of sex acts in the prior month was 4, and 27% reported
cohort from the randomized trial, loss to follow-up, and unprotected sex (Table 1). A total of 237 HIV-1–infected
censoring of participants with HIV-1 seroconversion. A partners (15.6%) initiated ART during follow-up; 25
marginal structural model approach was used, adjusted for (1.6% of all HIV-1–infected partners) reported receiving a
age, sex, unprotected sex at baseline, and weighting that tenofovir-containing regimen.
accounted for time-dependent unprotected sex during Follow-up and Adherence
follow-up. Logistic regression was used to estimate stabi- Retention was greater than 90% for all groups during
lized inverse weights (18), which were used to conduct an the study period (Figure 1), with a median follow-up of 18
adjusted, weighted discrete proportional hazards analysis months (interquartile range, 12 to 24 months; range, 0 to
using the procedure described earlier. In a separate sensi- 36 months). Study medication was dispensed at 96% of
tivity analysis to assess the potential effects of early censor- attended visits. The most common reason for withholding
ing of participants who acquired HIV-1, we conducted study medication was pregnancy, which did not differ sig-
simulation modeling to estimate the HR if these partici- nificantly by study group and accounted for 1.5% of
pants had continued in follow-up at the observed HSV-2 follow-up time. Study medication interruptions due to
risk and at higher risk. safety-related reasons accounted for 0.5% of follow-up
We performed a case– cohort analysis of plasma teno- time.
fovir concentrations to assess the risk for HSV-2 acquisi- The primary measure of adherence was monthly pill
tion associated with detection (⬎0.3 ng/mL) versus non- counts of returned study tablets. Ninety-eight percent of
detection, using the same approach as in the case– cohort dispensed study bottles were returned, and pill counts in-
analysis of plasma tenofovir concentrations and HIV-1 dicated that 96% of dispensed tablets were taken. When
protection in the Partners PrEP Study (2). The case– missed visits, other reasons for nondispensation of study
cohort analysis was performed using drug concentrations medication, and nonadherence to dispensed study pills
in all selected participants, regardless of drug holds in the were taken into account, 90.9% of follow-up time for this
prior interval (for example, for pregnancy or clinical analysis was covered by study medication.
events). We used case– cohort methods in a Cox propor-
tional hazards model to estimate the HR of infection for HSV-2 Incidence by Group
detectable versus undetectable tenofovir in plasma (19). In A total of 131 participants had incident HSV-2 infec-
this nonrandomized comparison, the model was adjusted tion (79 in the active PrEP groups [incidence, 5.6 per 100
for factors that might be associated with adherence to the person-years] and 52 in the placebo group [incidence, 7.7
study drug, including age, polygamous marriage, and un- per 100 person-years]) (Table 2). The HR for HSV-2 ac-
protected sex as a time-dependent variable (20). This anal- quisition with PrEP compared with placebo was 0.70
ysis used the 2-phase design approach to the case– cohort (95% CI, 0.49 to 0.99; P ⫽ 0.047) (Figure 2), and the
analysis (21). absolute risk reduction was 2.1 per 100 person-years.
All analyses were conducted using SAS, version 9.3, When the 2 PrEP groups were considered separately, 42
with the exception of the case– cohort analysis, for which HSV-2 infections were observed among participants as-
we used R, version 3.0.0 (survey package 3.28-2 and signed tenofovir (incidence, 6.1 per 100 person-years) and
Hmisc package 3.10-1.1, R Foundation for Statistical 37 were seen among those assigned FTC–TDF (incidence,
Computing, Vienna, Austria). 5.1 per 100 person-years), indicating an HR for HSV-2
acquisition of 0.76 (CI, 0.51 to 1.14; P ⫽ 0.186) for teno-
Role of the Funding Source fovir and 0.64 (CI, 0.42 to 0.98; P ⫽ 0.041) for FTC–
The Bill & Melinda Gates Foundation funded the TDF, each compared with placebo. The causal effect of
study but did not oversee the protocol. Gilead Sciences PrEP, which we estimated using marginal structural mod-
14 1 July 2014 Annals of Internal Medicine Volume 161 • Number 1 www.annals.org

Downloaded From: http://annals.org/pdfaccess.ashx?url=/data/journals/aim/930448/ by a Suny Health Sciences Ctr User on 06/17/2017


HSV-2 Protection With Tenofovir-Based Preexposure Prophylaxis Original Research

Table 1. Baseline Characteristics of Participants Who Were Dually Seronegative for HIV-1 and HSV-2

Variable Tenofovir FTC–TDF Placebo Total


(n ⴝ 513) (n ⴝ 528) (n ⴝ 481) (n ⴝ 1522)
Demographic characteristics
Men n (%) 408 (80) 422 (80) 389 (81) 1219 (80)
Median age (IQR), y 30 (26–36) 30 (26–36) 31 (26–36) 31 (26–36)
Median education (IQR), y 8 (5–12) 8 (5–11) 8 (5–11) 8 (5–11)
Any monthly income, n (%) 435 (85) 433 (82) 398 (83) 1266 (83)

Couple characteristics
Married to study partner, n (%) 489 (95) 506 (96) 466 (97) 1461 (96)
Median time living with study partner (IQR), y 4.7 (1.5–10.0) 5.0 (2.0–11.0) 4.5 (1.6–10.0) 5.0 (1.6–10.0)

Sexual risk behavior


Median sex acts in prior month (IQR), n 4 (3–8) 4 (3–8) 4 (3–9) 4 (3–8)
Any unprotected sex acts in prior month, n (%) 150 (29) 145 (27) 113 (23) 408 (27)
Any sex with outside partner in prior month, n (%) 48 (9) 39 (7) 36 (7) 123 (8)

Clinical characteristics of HIV-1–seronegative partner


Median CD4 count (IQR), ⫻ 109 cells/L 0.481 (0.366–0.680) 0.493 (0.387–0.680) 0.519 (0.395–0.677) 0.496 (0.382–0.680)
Median plasma HIV-1 RNA level (IQR), log10 copies/mL* 3.8 (3.1–4.3) 3.7 (3.0–4.4) 3.9 (3.2–4.5) 3.8 (3.1–4.4)

Clinical characteristics of HIV-1/HSV-2–seronegative participant, n (%)


Uncircumcised (men only) 162 (40) 177 (42) 150 (39) 489 (40)
Curable sexually transmitted infection† 26 (5) 19 (4) 35 (7) 80 (5)

FTC–TDF ⫽ emtricitabine in combination with tenofovir disoproxil fumarate; HSV-2 ⫽ herpes simplex virus type 2; IQR ⫽ interquartile range.
* Data were not available for 16 HIV-1–infected partners.
† Neisseria gonorrhoeae, Chlamydia trachomatis, or Trichomonas vaginalis detected by nucleic acid amplification testing. Data were not available for 14 participants.

eling, was essentially the same as the result from the pro- censoring of participants with HIV-1 seroconversion that
portional hazards model (HR, 0.69 [CI, 0.49 to 0.99]; assumed increased risk for HSV-2 (1.5 times the placebo
P ⫽ 0.041). The magnitude of reduction in incident rate) and no protective effect (because the study drug was
HSV-2 in the PrEP groups compared with placebo was withheld at detection of HIV-1 seroconversion) produced
similar in subgroup categories, including men and women, a narrow range of HRs over 1000 simulations (median,
circumcised and uncircumcised HIV-1–infected men, and 0.697 [CI, 0.496 to 0.998]) (Table 2), indicating little
individuals whose HIV-1–infected partners had higher or sensitivity to censoring.
lower plasma HIV-1 RNA levels. However, PrEP was more Tenofovir Exposure and HSV-2 Protection
effective in reducing HSV-2 acquisition among individuals We compared plasma tenofovir levels among the 79
whose HIV-1–infected partner had a baseline CD4 count case participants with and 160 control participants without
of at least 0.350 ⫻ 109 cells/L than among those whose HSV-2 seroconversion randomly selected from the active
partner had a CD4 count below this level. PrEP groups. Overall, 76.8% of control participants had
Baseline HSV-2 serostatus was available for 1493 detectable tenofovir (⬎0.3 ng/mL) compared with 75.9%
HIV-1–infected partners, of whom 1044 (69.9%) were of case participants at the visit where HSV-2 infection was
HSV-2–seropositive. For HIV-1– uninfected persons with first detected, and detection of tenofovir was not associated
a partner co-infected with HIV-1 and HSV-2, incidence of with HSV-2 protection (adjusted HR, 1.72 [CI, 0.86 to
HSV-2 was 10.1 per 100 person-years in the placebo group 3.44]; P ⫽ 0.123).
and 7.0 per 100 person-years in the PrEP groups (HR,
0.67 [CI, 0.46 to 0.98]; P ⫽ 0.038). Participants whose
HIV-1–infected partners were HSV-2–seronegative could DISCUSSION
have acquired HSV-2 from outside partners; among the 13 In this secondary analysis within a randomized,
such participants with incident HSV-2 infection, none re- placebo-controlled trial of PrEP for HIV-1 prevention among
ported outside partners at study enrollment, but 6 reported 4747 HIV-1–serodiscordant couples, oral tenofovir-based
an outside partner during follow-up (5 of the HIV-1– PrEP reduced HSV-2 acquisition by 30% compared with
uninfected partners and 1 of the HIV-1–infected partners). placebo among initially HSV-2–seronegative persons. In
Twenty-three participants seroconverted to HIV-1; of the subgroup analysis of participants with known exposure
these, 5 seroconverted to HSV-2 before HIV-1 and 18 (11 to HSV-2 due to having a partner co-infected with HIV-1
in the placebo group and 7 in the PrEP groups) were HSV-2– and HSV-2, oral tenofovir-based PrEP reduced HSV-2
seronegative at the last visit before HIV-1 seroconversion. seroincidence by 33%. This is, to our knowledge, the first
A sensitivity analysis of the potential effect of informative evidence of efficacy of daily oral tenofovir-based PrEP in
www.annals.org 1 July 2014 Annals of Internal Medicine Volume 161 • Number 1 15

Downloaded From: http://annals.org/pdfaccess.ashx?url=/data/journals/aim/930448/ by a Suny Health Sciences Ctr User on 06/17/2017


Original Research HSV-2 Protection With Tenofovir-Based Preexposure Prophylaxis

Table 2. HSV-2 Seroincidence, by Study Group

Variable Combined Treatment Group Placebo


(Tenofovir or FTC–TDF)

Participants, n Events, n Person-Years Incidence, Participants, n Events, n Person-Years Incidence,


events per 100 events per 100
person-years person-years
Overall 1041 79 1422.2 5.55 481 52 672.0 7.74

Sex
Male 830 60 1143.9 5.25 389 42 556.9 7.54
Female 211 19 278.3 6.83 92 10 115.1 8.69

Circumcision among HIV-1/HSV-2–


seronegative men
Circumcised 491 35 649.0 5.39 239 28 337.8 8.29
Uncircumcised 339 25 494.9 5.05 150 14 219.1 6.39

Baseline plasma HIV-1 RNA level of


HIV-1–infected partner
⬍50 000 copies/mL 888 64 1226.2 5.22 392 41 548.9 7.47
ⱖ50 000 copies/mL 141 15 186.5 8.04 85 8 121.9 6.56

Baseline HSV-2 status of


HIV-1–infected partner
Positive 722 69 987.5 6.99 322 45 445.5 10.10
Negative 297 7 402.1 1.74 152 6 219.4 2.73

Baseline CD4 count of


HIV-1–infected partner
⬍0.350 ⫻ 109 cells/L 193 22 266.0 8.27 81 4 128.9 3.10
ⱖ0.350 ⫻ 109 cells/L 848 57 1156.2 4.93 400 48 543.1 8.84

FTC–TDF ⫽ emtricitabine in combination with tenofovir disoproxil fumarate; HR ⫽ hazard ratio; HSV-2 ⫽ herpes simplex virus type 2.
* The HR was 0.69 (95% CI, 0.49 – 0.99; P ⫽ 0.041) in the Cox proportional hazards model and 0.70 (CI, 0.49 –1.00; P ⫽ 0.053) in the marginal structural model when
each model was stratified by site. To account for potential informative censoring of participants with HIV-1 seroconversion (n ⫽ 18 censored at the time of seroconversion,
with no additional HSV-2 testing after that point), we conducted simulation modeling to assess the effect of following participants after seroconversion. We simulated a
conservative scenario 1000 times under the assumption that the risk for HSV-2 acquisition after HIV-1 seroconversion was elevated (1.5 times the HSV-2 seroconversion rate
in the placebo group among those who were HIV-1– uninfected) and the same in the preexposure prophylaxis and placebo groups (HR, 1). In this scenario, the overall
median HR was 0.697 (CI, 0.496 – 0.998)— essentially the same as the HRs from the proportional hazards and marginal structural models—suggesting that censoring
participants at the time of HIV-1 seroconversion had little effect on the overall findings.
† Values in bold are statistically significant.
‡ Test for effect modification.

providing protection against HSV-2 in a study population months (in contrast to HIV-1 serostatus, which was as-
with high adherence to daily oral PrEP. sessed monthly). In addition, 80% of the HSV-2–suscep-
The efficacy of tenofovir to reduce the risk for HSV-2 tible participants in this study were men; the biology of
acquisition found in this study is consistent with the heterosexual HSV-2 transmission with respect to HSV-2
HSV-2 protective effect of 51% observed with pericoital target cells and the bioavailability of oral tenofovir in pe-
use of topical 1% tenofovir gel (6, 8). Topical use of teno- nile epithelium are not well-characterized.
fovir or high adherence to daily dosing of oral tenofovir Protection from HSV-2 with daily oral tenofovir-
may be needed to achieve sufficient levels to provide pro- based PrEP is biologically plausible given recent in vitro
tection against HSV-2 given that the 90% effective con- data showing that tenofovir inhibits HSV-2 replication in
centration (EC90) of tenofovir for HSV-2 is high (10, 11). epithelial raft cultures, a mouse model, and ex vivo human
We did not find a relationship between detection of teno- lymphoid and cervicovaginal tissue from persons monoin-
fovir in plasma and protection against HSV-2, although fected with HSV-2 and those co-infected with HIV-1 and
similar analyses for HIV-1 protection among the HIV-1– HSV-2 (10). Steady-state tenofovir concentrations in the
uninfected participants in the Partners PrEP Study showed genital tract after daily oral FTC–TDF are approximately
an association (2). The lack of relationship between detec- 100 ng/mL (22), and in vitro studies indicate HSV sup-
tion of tenofovir in plasma and protection against HSV-2 pression at tenofovir concentrations between 10 and 200
may indicate either less ability to discern a concentration– ␮g/mL (10), which would only be achieved in the context
response relationship between tenofovir exposure and of high adherence to daily oral dosing and accumulation of
HSV-2 protection if the actual protective effect is modest active metabolites in the genital tract. The lack of HSV-2
or less precise timing of drug exposure and HSV-2 sero- protection with oral FTC–TDF among men who have sex
conversion due to assessing HSV-2 serostatus up to every 6 with men in another PrEP trial was potentially due to
16 1 July 2014 Annals of Internal Medicine Volume 161 • Number 1 www.annals.org

Downloaded From: http://annals.org/pdfaccess.ashx?url=/data/journals/aim/930448/ by a Suny Health Sciences Ctr User on 06/17/2017


HSV-2 Protection With Tenofovir-Based Preexposure Prophylaxis Original Research

lower adherence in that study (23); only about half of


Table 2—Continued
participants had objective evidence of PrEP adherence (1)
compared with more than 80% in our study (2). Given the
Proportional Hazards Model* Marginal Structural Model*
strong association between adherence and HIV-1 protec-
tion with daily oral dosing of PrEP, new formulations for
HR vs. Placebo P Value† HR vs. Placebo P Value†
(95% CI) (95% CI)
sustained delivery of PrEP products, such as an intravaginal
ring containing tenofovir, may provide a less user-
0.70 (0.49–0.99) 0.047 0.69 (0.49–0.99) 0.041 dependent formulation with sustained release of tenofovir
in the genital tract (24).
0.67‡
Our results and those of others indicate that tenofovir-
0.67 (0.45–1.00) 0.048 0.66 (0.45–0.99) 0.042
0.81 (0.38–1.74) 0.59 0.77 (0.36–1.67) 0.51 based PrEP may have prophylactic activity against other
viruses in addition to HIV-1. Tenofovir-containing ART
0.58‡
regimens have been shown to be associated with reduced
0.62 (0.38–1.02) 0.059 0.61 (0.37–1.00) 0.051 incidence of hepatitis B infection in HIV-1–infected per-
0.78 (0.41–1.50) 0.46 0.76 (0.40–1.47) 0.42 sons (25), which is consistent with in vitro and in vivo
0.24‡
activity of tenofovir (and emtricitabine) in persons with
chronic hepatitis B infection (26, 27). The efficacy of
0.68 (0.46–1.01) 0.057 0.67 (0.45–0.99) 0.049 tenofovir-containing ART regimens has not been evaluated
1.19 (0.51–2.82) 0.69 1.19 (0.52–2.73) 0.68
for HSV-2 protection in HIV-1–infected persons who are
0.50‡ HSV-2– uninfected. Ongoing studies are evaluating the
therapeutic efficacy of tenofovir in reducing HSV-2 shed-
0.67 (0.46–0.98) 0.038 0.65 (0.44–0.95) 0.024
0.62 (0.21–1.85) 0.39 0.63 (0.21–1.90) 0.41
ding in persons with chronic HSV-2 infection with and
without HIV-1 infection. One small study in persons co-
0.003‡ infected with HIV-1 and HSV-2 found that tenofovir-
2.50 (0.86–7.26) 0.092 2.24 (0.78–6.48) 0.140
containing ART was not associated with reduced rates of
0.55 (0.37–0.81) 0.002 0.55 (0.37–0.80) 0.002 HSV-2 shedding, but the study had limited power to de-
tect a difference (28).
Interventions to reduce the risk for HSV-2 acquisition
are a high priority. Incidence of HSV-2 is high in popula-

Figure 2. Cumulative probability of HSV-2 seroconversion in the active PrEP (tenofovir and FTC–TDF) versus placebo groups.

0.20
Cumulative Probability of HSV-2 Seroconversion

PrEP
0.15
Placebo

0.10

0.05

0.00

0 3 6 9 12 15 18 21 24 27 30

Months Since Start


At risk, n
PrEP 1025 973 948 774 576 386 130
Placebo 473 438 428 371 279 189 62

FTC–TDF ⫽ emtricitabine in combination with tenofovir disoproxil fumarate; HSV-2 ⫽ herpes simplex virus type 2; PrEP ⫽ preexposure prophylaxis.

www.annals.org 1 July 2014 Annals of Internal Medicine Volume 161 • Number 1 17

Downloaded From: http://annals.org/pdfaccess.ashx?url=/data/journals/aim/930448/ by a Suny Health Sciences Ctr User on 06/17/2017


Original Research HSV-2 Protection With Tenofovir-Based Preexposure Prophylaxis

tions worldwide; annualized incidence was 20% among that the HRs for HSV-2 acquisition were relatively stable
young South African women (6) and 15% among HIV-1– despite this potentially informative censoring.
serodiscordant couples in Kenya who reported a high rate In summary, tenofovir-based PrEP provides modest
of condom use (29). In addition, observational data indi- protection against HSV-2. Incidence was 7.7 per 100
cate a 3-fold increased risk for HIV-1 acquisition due to person-years in the placebo group of this study, nearly
prevalent HSV-2 infection and up to a 6-fold increase with 4-fold greater than the HIV-1 incidence (2.0 per 100
incident HSV-2 infection (30). Interventions to reduce the person-years). Given the high prevalence of HSV-2 in
risk for HSV-2 transmission and acquisition are limited. HIV-1–infected persons, and given that HSV-2 is a signif-
Condoms are estimated to reduce HSV-2 acquisition by icant risk factor for HIV-1 acquisition, identifying effective
30% (31), which is substantially lower than the 80% effi- primary prevention strategies for HSV-2 is a public health
cacy of condoms in reducing the per-contact risk for priority. The HSV-2 protective effects of oral tenofovir-
HIV-1 acquisition (32), and medical circumcision pro- based PrEP add benefit to the high protection against
vided modest protection against HSV-2 acquisition in HIV-1 in populations who are often at risk for HIV-1 and
HIV-1– uninfected men in 2 of 3 trials (33–35). Candidate HSV-2.
preventive HSV-2 vaccines to date have not shown efficacy
in preventing HSV-2 infection (36, 37). Widely used From University of Washington and Fred Hutchinson Cancer Research
agents for treatment of HSV-2 infection have shown mixed Center, Seattle, Washington; Johns Hopkins University, Baltimore,
Maryland; Indiana University, Indianapolis, Indiana; and Makerere Uni-
results when used for prevention of HSV-2 infection, with versity, Kampala, Uganda.
1 study finding that valacyclovir, 500 mg once daily, re-
duced HSV-2 transmission by 50% to partners of HSV-2– Note: All authors vouch for the completeness and accuracy of the data
infected persons who were not infected with HIV-1 (38), presented.
but other studies found that acyclovir, 400 mg twice daily,
did not reduce HSV-2 transmission to partners of persons Acknowledgment: The authors thank the study participants and the
co-infected with HIV-1 and HSV-2 (39) or HSV-2 acqui- study team.
sition in HIV-1– uninfected persons (40, 41). No studies
are available of valacyclovir or acyclovir for primary pro- Grant Support: By the Bill & Melinda Gates Foundation (OOP47674).
phylaxis against HSV-2 acquisition.
Strengths of this study include the use of a blinded Disclosures: Disclosures can be viewed at www.acponline.org/authors
end point determination process based on the best avail- /icmje/ConflictOfInterestForms.do?msNum⫽M13-2471.
able HSV-2 serologic testing algorithm and sufficient
power with 131 end points to determine modest effective- Reproducible Research Statement: Study protocol and statistical code:
ness. Subgroup analysis based on known exposure to an Available from Dr. Celum (e-mail, ccelum@uw.edu). Data set: Portions
HSV-2–infected partner was feasible because of the couples of the analytic data set are available to approved individuals through
written agreement with Dr. Celum (e-mail, ccelum@uw.edu).
design. The consistency of results between the proportional
hazards and marginal structural models indicates that
Requests for Single Reprints: Connie Celum, MD, MPH, University
intention-to-treat assumptions were appropriate and that
of Washington, 325 Ninth Avenue, UW Box 359927, Seattle, WA
significant confounding was not present in the subsample 98104.
of initially HSV-2–seronegative persons and was not in-
duced by censoring at HIV-1 seroconversion or loss to Current author addresses and author contributions are available at
follow-up. www.annals.org.
Limitations of this study include the inability to detect
and exclude individuals with acute HSV-2 infection at
baseline due to a lack of laboratory methods. Ten percent References
of incident HSV-2 infections occurred in partnerships 1. Grant RM, Lama JR, Anderson PL, McMahan V, Liu AY, Vargas L, et al;
where the HIV-1–infected partner was HSV-2– iPrEx Study Team. Preexposure chemoprophylaxis for HIV prevention in men
seronegative at baseline, but laboratory methods are not who have sex with men. N Engl J Med. 2010;363:2587-99. [PMID: 21091279]
2. Baeten JM, Donnell D, Ndase P, Mugo NR, Campbell JD, Wangisi J, et al;
available for linkage of HSV-2 strains. Because of availabil- Partners PrEP Study Team. Antiretroviral prophylaxis for HIV prevention in
ity of testing and samples, we were unable to assess the heterosexual men and women. N Engl J Med. 2012;367:399-410. [PMID:
timing of HSV-2 acquisition with greater precision than 22784037]
3. Choopanya K, Martin M, Suntharasamai P, Sangkum U, Mock PA, Leet-
according to the schedule of follow-up testing we per-
hochawalit M, et al; Bangkok Tenofovir Study Group. Antiretroviral prophy-
formed, which may have limited our ability to demonstrate laxis for HIV infection in injecting drug users in Bangkok, Thailand (the Bang-
an association between HSV-2 protection and plasma kok Tenofovir Study): a randomised, double-blind, placebo-controlled phase 3
tenofovir detection. Finally, we censored participants after trial. Lancet. 2013;381:2083-90. [PMID: 23769234]
4. Thigpen MC, Kebaabetswe PM, Paxton LA, Smith DK, Rose CE, Segolodi
HIV-1 seroconversion because the study drug was discon- TM, et al; TDF2 Study Group. Antiretroviral preexposure prophylaxis for het-
tinued and we could not determine HSV-2 seroconversion erosexual HIV transmission in Botswana. N Engl J Med. 2012;367:423-34.
in these participants; however, sensitivity analyses indicated [PMID: 22784038]

18 1 July 2014 Annals of Internal Medicine Volume 161 • Number 1 www.annals.org

Downloaded From: http://annals.org/pdfaccess.ashx?url=/data/journals/aim/930448/ by a Suny Health Sciences Ctr User on 06/17/2017


HSV-2 Protection With Tenofovir-Based Preexposure Prophylaxis Original Research
5. Balzarini J, Holy A, Jindrich J, Naesens L, Snoeck R, Schols D, 23. Lama J, Mayer K, Schechter M, Kallas E, Bekker LG, Chariyalertsak S,
et al. Differential antiherpesvirus and antiretrovirus effects of the (S) and (R) et al. Oral TDF and its impact in HSV-2 acquisition and clinical expression
enantiomers of acyclic nucleoside phosphonates: potent and selective in vitro and [Abstract]. Presented at the 18th Conference on Retroviruses and Opportunistic
in vivo antiretrovirus activities of (R)-9-(2-phosphonomethoxypropyl)-2,6- Infections, Boston, Massachusetts, 27 February–2 March 2011. Abstract no.
diaminopurine. Antimicrob Agents Chemother. 1993;37:332-8. [PMID: 1002.
8452366] 24. Mesquita PM, Rastogi R, Segarra TJ, Teller RS, Torres NM, Huber AM,
6. Abdool Karim Q, Abdool Karim SS, Frohlich JA, Grobler AC, Baxter C, et al. Intravaginal ring delivery of tenofovir disoproxil fumarate for prevention of
Mansoor LE, et al; CAPRISA 004 Trial Group. Effectiveness and safety of HIV and herpes simplex virus infection. J Antimicrob Chemother. 2012;67:
tenofovir gel, an antiretroviral microbicide, for the prevention of HIV infection in 1730-8. [PMID: 22467632]
women. Science. 2010;329:1168-74. [PMID: 20643915] 25. Gatanaga H, Hayashida T, Tanuma J, Oka S. Prophylactic effect of anti-
7. Tan D. Potential role of tenofovir vaginal gel for reduction of risk of herpes retroviral therapy on hepatitis B virus infection. Clin Infect Dis. 2013;56:1812-9.
simplex virus in females. Int J Womens Health. 2012;4:341-50. [PMID: [PMID: 23487374]
22927765] 26. Price H, Dunn D, Pillay D, Bani-Sadr F, de Vries-Sluijs T, Jain MK, et al.
8. Centre for the AIDS Programme of Research in South Africa. Effectiveness Suppression of HBV by tenofovir in HBV/HIV coinfected patients: a systematic
& safety of vaginal microbicide 1% tenofovir gel for prevention of HIV infection review and meta-analysis. PLoS One. 2013;8:e68152. [PMID: 23874527]
in women. Congella, South Africa: Centre for the AIDS Programme of Research 27. Soriano V, Poveda E, Vispo E, Barreiro P. Hepatitis B in HIV-infected
in South Africa; 2010. Accessed at www.conrad.org/media/news/92 patients. Clin Liver Dis. 2013;17:489-501. [PMID: 23905818]
_CAPRISA%20004%20Results.pdf on 7 May 2014. 28. Tan DH, Kaul R, Raboud JM, Walmsley SL. No impact of oral tenofovir
9. Karim SS, Kashuba AD, Werner L, Karim QA. Drug concentrations after disoproxil fumarate on herpes simplex virus shedding in HIV-infected adults.
topical and oral antiretroviral pre-exposure prophylaxis: implications for HIV AIDS. 2011;25:207-10. [PMID: 21150556]
prevention in women. Lancet. 2011;378:279-81. [PMID: 21763939] 29. Muiru AN, Guthrie BL, Bosire R, Merkel M, Liu AY, Choi RY, et al.
10. Andrei G, Lisco A, Vanpouille C, Introini A, Balestra E, van den Oord J, Incident HSV-2 infections are common among HIV-1-discordant couples. J In-
et al. Topical tenofovir, a microbicide effective against HIV, inhibits herpes sim- fect Dis. 2013;208:1093-101. [PMID: 23840044]
plex virus-2 replication. Cell Host Microbe. 2011;10:379-89. [PMID: 30. Freeman EE, Weiss HA, Glynn JR, Cross PL, Whitworth JA, Hayes RJ.
22018238] Herpes simplex virus 2 infection increases HIV acquisition in men and women:
11. Schwartz JL, Rountree W, Kashuba AD, Brache V, Creinin MD, Poin- systematic review and meta-analysis of longitudinal studies. AIDS. 2006;20:73-
dexter A, et al. A multi-compartment, single and multiple dose pharmacokinetic 83. [PMID: 16327322]
study of the vaginal candidate microbicide 1% tenofovir gel. PLoS One. 2011;6: 31. Martin ET, Krantz E, Gottlieb SL, Magaret AS, Langenberg A, Stanberry
e25974. [PMID: 22039430] L, et al. A pooled analysis of the effect of condoms in preventing HSV-2 acqui-
12. Mujugira A, Baeten JM, Donnell D, Ndase P, Mugo NR, Barnes L, et al; sition. Arch Intern Med. 2009;169:1233-40. [PMID: 19597073]
Partners PrEP Study Team. Characteristics of HIV-1 serodiscordant couples 32. Hughes JP, Baeten JM, Lingappa JR, Magaret AS, Wald A, de Bruyn G,
enrolled in a clinical trial of antiretroviral pre-exposure prophylaxis for HIV-1 et al; Partners in Prevention HSV/HIV Transmission Study Team. Determi-
prevention. PLoS One. 2011;6:e25828. [PMID: 21998703] nants of per-coital-act HIV-1 infectivity among African HIV-1-serodiscordant
13. Lingappa J, Nakku-Joloba E, Magaret A, Friedrich D, Dragavon J, Kam- couples. J Infect Dis. 2012;205:358-65. [PMID: 22241800]
bugu F, et al. Sensitivity and specificity of herpes simplex virus-2 serological 33. Mahiane SG, Legeai C, Taljaard D, Latouche A, Puren A, Peillon A, et al.
assays among HIV-infected and uninfected urban Ugandans. Int J STD AIDS. Transmission probabilities of HIV and herpes simplex virus type 2, effect of male
2010;21:611-6. [PMID: 21097732] circumcision and interaction: a longitudinal study in a township of South Africa.
14. Ashley RL, Militoni J, Lee F, Nahmias A, Corey L. Comparison of Western AIDS. 2009;23:377-383. [PMID: 19198042]
blot (immunoblot) and glycoprotein G-specific immunodot enzyme assay for 34. Tobian AA, Serwadda D, Quinn TC, Kigozi G, Gravitt PE, Laeyendecker
detecting antibodies to herpes simplex virus types 1 and 2 in human sera. J Clin O, et al. Male circumcision for the prevention of HSV-2 and HPV infections and
Microbiol. 1988;26:662-7. [PMID: 2835389] syphilis. N Engl J Med. 2009;360:1298-309. [PMID: 19321868]
15. Keller MJ, Madan RP, Torres NM, Fazzari MJ, Cho S, Kalyoussef S, et al. 35. Mehta SD, Moses S, Parker CB, Agot K, Maclean I, Bailey RC. Circum-
A randomized trial to assess anti-HIV activity in female genital tract secretions cision status and incident herpes simplex virus type 2 infection, genital ulcer
and soluble mucosal immunity following application of 1% tenofovir gel. PLoS disease, and HIV infection. AIDS. 2012;26:1141-9. [PMID: 22382150]
One. 2011;6:e16475. [PMID: 21283552] 36. Belshe RB, Leone PA, Bernstein DI, Wald A, Levin MJ, Stapleton JT, et al;
16. Dropulic LK, Cohen JI. Update on new antivirals under development for the Herpevac Trial for Women. Efficacy results of a trial of a herpes simplex vaccine.
treatment of double-stranded DNA virus infections. Clin Pharmacol Ther. 2010; N Engl J Med. 2012;366:34-43. [PMID: 22216840]
88:610-9. [PMID: 20881959] 37. Johnston C, Koelle DM, Wald A. Current status and prospects for develop-
17. Prentice RL, Gloeckler LA. Regression analysis of grouped survival data with ment of an HSV vaccine. Vaccine. 2014;32:1553-60. [PMID: 24016811]
application to breast cancer data. Biometrics. 1978;34:57-67. [PMID: 630037] 38. Corey L, Wald A, Patel R, Sacks SL, Tyring SK, Warren T, et al; Valacy-
18. Hernán MA, Brumback B, Robins JM. Marginal structural models to esti- clovir HSV Transmission Study Group. Once-daily valacyclovir to reduce the
mate the causal effect of zidovudine on the survival of HIV-positive men. Epide- risk of transmission of genital herpes. N Engl J Med. 2004;350:11-20. [PMID:
miology. 2000;11:561-70. [PMID: 10955409] 14702423]
19. Breslow NE, Lumley T, Ballantyne CM, Chambless LE, Kulich M. Im- 39. Mujugira A, Magaret AS, Celum C, Baeten JM, Lingappa JR, Morrow RA,
proved Horvitz-Thompson estimation of model parameters from two-phase strat- et al; Partners in Prevention HSV/HIV Transmission Study Team. Daily acy-
ified samples: applications in epidemiology. Stat Biosci. 2009;1:32. [PMID: clovir to decrease herpes simplex virus type 2 (HSV-2) transmission from HSV-
20174455] 2/HIV-1 coinfected persons: a randomized controlled trial. J Infect Dis. 2013;
20. Haberer JE, Baeten JM, Campbell J, Wangisi J, Katabira E, Ronald A, et al. 208:1366-74. [PMID: 23901094]
Adherence to antiretroviral prophylaxis for HIV prevention: a substudy cohort 40. Celum C, Wald A, Hughes J, Sanchez J, Reid S, Delany-Moretlwe S, et al;
within a clinical trial of serodiscordant couples in East Africa. PLoS Med. 2013; HPTN 039 Protocol Team. Effect of aciclovir on HIV-1 acquisition in herpes
10:e1001511. [PMID: 24058300] simplex virus 2 seropositive women and men who have sex with men: a ran-
21. Lumley T. Complex Surveys: A Guide to Analysis Using R. Hoboken, NJ: domised, double-blind, placebo-controlled trial. Lancet. 2008;371:2109-19.
J Wiley; 2010. [PMID: 18572080]
22. Dumond JB, Yeh RF, Patterson KB, Corbett AH, Jung BH, Rezk NL, 41. Watson-Jones D, Weiss HA, Rusizoka M, Changalucha J, Baisley K, Mug-
et al. Antiretroviral drug exposure in the female genital tract: implications for oral eye K, et al; HSV trial team. Effect of herpes simplex suppression on incidence of
pre- and post-exposure prophylaxis. AIDS. 2007;21:1899-907. [PMID: HIV among women in Tanzania. N Engl J Med. 2008;358:1560-71. [PMID:
17721097] 18337596]

www.annals.org 1 July 2014 Annals of Internal Medicine Volume 161 • Number 1 19

Downloaded From: http://annals.org/pdfaccess.ashx?url=/data/journals/aim/930448/ by a Suny Health Sciences Ctr User on 06/17/2017


Current Author Addresses: Drs. Celum, Donnell, Hong, and Baeten; APPENDIX: PARTNERS PREP STUDY TEAM
Ms. Thomas; and Mr. Mujugira: University of Washington, 325 Ninth University of Washington Coordinating Center and
Avenue, UW Box 359927, Seattle, WA 98104. Central Laboratories, Seattle, Washington
Dr. Morrow: Fred Hutchinson Cancer Research Center, 1100 Fairview Connie Celum (principal investigator and protocol co-
Avenue North, LE-500, Seattle, WA 98109.
chair), Jared M. Baeten (medical director and protocol co-chair),
Dr. Hendrix: Department of Medicine, Johns Hopkins University, 600
Deborah Donnell (protocol statistician), Robert W. Coombs
North Wolfe Street, Harvey 502, Baltimore, MD 21287-5554.
Dr. Fife: Department of Medicine, Indiana University, Emerson (University of Washington Retrovirology Laboratory), Anne
H/Room 435, 545 Barnhill Drive, Indianapolis, IN 46202. Cent (University of Washington Virology Laboratory), and Jai-
Dr. Nakku-Joloba: School of Public Health, Makerere University, PO ram Lingappa (University of Washington International Clinical
Box 22418, Plot 106, Block 213, Bukoto, Kampala, Uganda. Research Center Biorepository).
Study Sites and Principal Investigators
Author Contributions: Conception and design: C. Celum, D. Donnell, Eldoret, Kenya (Moi University and Indiana University):
J.M. Baeten. Kenneth Fife and Edwin Were; Kabwohe, Uganda (Kabwohe
Analysis and interpretation of the data: C. Celum, R.A. Morrow, Clinical Research Center): Elioda Tumwesigye; Jinja, Uganda
D. Donnell, T. Hong, C.W. Hendrix, K.K. Thomas, K.H. Fife, J.M. (Makerere University and University of Washington): Patrick
Baeten. Ndase and Elly Katabira; Kampala, Uganda (Makerere Univer-
Drafting of the article: C. Celum, D. Donnell, J.M. Baeten. sity): Elly Katabira and Allan Ronald; Kisumu, Kenya (Kenya
Critical revision of the article for important intellectual content: C.
Medical Research Institute and University of California, San
Celum, D. Donnell, K.H. Fife, E. Nakku-Joloba, J.M. Baeten.
Francisco): Elizabeth Bukusi and Craig Cohen; Mbale, Uganda
Final approval of the article: C. Celum, R.A. Morrow, D. Donnell, C.W.
Hendrix, K.H. Fife, E. Nakku-Joloba, A. Mujugira, J.M. Baeten. (The AIDS Support Organization and Centers for Disease
Provision of study materials or patients: K.H. Fife, A. Mujugira, J.M. Control and Prevention–Uganda): Jonathan Wangisi, James
Baeten. Campbell, and Jordan Tappero; Nairobi, Kenya (University of
Statistical expertise: D. Donnell, T. Hong, K.K. Thomas. Nairobi and University of Washington): James Kiarie, Carey Far-
Obtaining of funding: C. Celum, J.M. Baeten. quhar, and Grace John-Stewart; Thika, Kenya (University of
Administrative, technical, or logistic support: E. Nakku-Joloba, A. Nairobi and University of Washington): Nelly Rwamba Mugo;
Mujugira, J.M. Baeten. Tororo, Uganda (Centers for Disease Control and Prevention–
Collection and assembly of data: C. Celum, R.A. Morrow, D. Donnell, Uganda and The AIDS Support Organization): James Campbell,
C.W. Hendrix, K.H. Fife, E. Nakku-Joloba, A. Mujugira, J.M. Baeten. Jordan Tappero, and Jonathan Wangisi.
Data management was provided by DF/Net Research (Seat-
tle, Washington), and site laboratory oversight was provided by
Contract Laboratory Services (University of the Witwatersrand,
Johannesburg, South Africa).

www.annals.org 1 July 2014 Annals of Internal Medicine Volume 161 • Number 1

Downloaded From: http://annals.org/pdfaccess.ashx?url=/data/journals/aim/930448/ by a Suny Health Sciences Ctr User on 06/17/2017


CORRECTION: TENOFOVIR AND EMTRICITABINE–TENOFOVIR
PREEXPOSURE PROPHYLAXIS FOR HERPES SIMPLEX VIRUS
TYPE 2
An article (1) was missing the Intervention section of its ab-
stract. The section should read as follows:
Once-daily oral tenofovir disoproxil fumarate (TDF), alone or
combined with emtricitabine (FTC-TDF), compared with placebo.
This has been corrected in the online version.

Reference
1. Celum C, Morrow RA, Donnell D, Hong T, Hendrix CW, Thomas KK, et al.
Daily oral tenofovir and emtricitabine–tenofovir preexposure prophylaxis reduces her-
pes simplex virus type 2 acquisition among heterosexual HIV-1–uninfected men and
women: a subgroup analysis of a randomized trial. Ann Intern Med. 2014;161:11-9.
[PMID: 24979446] doi: 10.7326/M13-2471

1 July 2014 Annals of Internal Medicine Volume 161 • Number 1 www.annals.org

Downloaded From: http://annals.org/pdfaccess.ashx?url=/data/journals/aim/930448/ by a Suny Health Sciences Ctr User on 06/17/2017

You might also like