ArticlePDF Available

Antiretroviral Therapy and Pre-exposure Prophylaxis: Combined Impact on HIV Transmission and Drug Resistance in South Africa

Authors:
  • Avenir Health
  • The Public Health Company

Abstract and Figures

Background: The potential impact of antiretroviral therapy (ART) and pre-exposure prophylaxis (PrEP) with overlapping and nonoverlapping antiretrovirals (ARVs) on human immunodeficiency virus (HIV) transmission and drug resistance is unknown. Methods: A detailed mathematical model was used to simulate the epidemiological impact of ART alone, PrEP alone, and combined ART + PrEP in South Africa. Results: ART alone initiated at a CD4 lymphocyte cell count <200 cells/µL (80% coverage and 96% effectiveness) prevents 20% of HIV infections over 10 years but increases drug resistance prevalence to 6.6%. PrEP alone (30% coverage and 75% effectiveness) also prevents 21% of infections but with lower resistance prevalence of 0.5%. The ratio of cumulative infections prevented to prevalent drug-resistant cases after 10 years is 7-fold higher for PrEP than for ART. Combined ART + PrEP with overlapping ARVs prevents 35% of infections but increases resistance prevalence to 8.2%, whereas ART + PrEP with nonoverlapping ARVs prevents slightly more infections (37%) and reduces resistance prevalence to 7.2%. Conclusions: Combined ART + PrEP is likely to prevent more HIV infections than either strategy alone, but with higher prevalence of drug resistance. ART is predicted to contribute more to resistance than is PrEP. Optimizing both ART and PrEP effectiveness and delivery are the keys to preventing HIV transmission and drug resistance.
Content may be subject to copyright.
MAJOR ARTICLE
Antiretroviral Therapy and Pre-exposure
Prophylaxis: Combined Impact on HIV
Transmission and Drug Resistance in South
Africa
Ume L. Abbas,1Robert Glaubius,1Anuj Mubayi,1,a Gregory Hood,2and John W. Mellors3
1
Departments of Infectious Diseases and Quantitative Health Sciences, Cleveland Clinic, Ohio;
2
Pittsburgh Supercomputing Center and
3
Division of
Infectious Diseases, School of Medicine, University of Pittsburgh, Pennsylvania
(See the editorial commentary by Celum et al on pages 18991)
Background.The potential impact of antiretroviral therapy (ART) and pre-exposure prophylaxis (PrEP) with
overlapping and nonoverlapping antiretrovirals (ARVs) on human immunodeciency virus (HIV) transmission
and drug resistance is unknown.
Methods.A detailed mathematical model was used to simulate the epidemiological impact of ART alone, PrEP
alone, and combined ART + PrEP in South Africa.
Results.ART alone initiated at a CD4 lymphocyte cell count <200 cells/µL (80% coverage and 96% effective-
ness) prevents 20% of HIV infections over 10 years but increases drug resistance prevalence to 6.6%. PrEP alone
(30% coverage and 75% effectiveness) also prevents 21% of infections but with lower resistance prevalence of 0.5%.
The ratio of cumulative infections prevented to prevalent drug-resistant cases after 10 years is 7-fold higher for
PrEP than for ART. Combined ART + PrEP with overlapping ARVs prevents 35% of infections but increases resis-
tance prevalence to 8.2%, whereas ART + PrEP with nonoverlapping ARVs prevents slightly more infections (37%)
and reduces resistance prevalence to 7.2%.
Conclusions.Combined ART + PrEP is likely to prevent more HIV infections than either strategy alone, but
with higher prevalence of drug resistance. ART is predicted to contribute more to resistance than is PrEP. Optimiz-
ing both ART and PrEP effectiveness and delivery are the keys to preventing HIV transmission and drug resistance.
Keywords.antiretroviral therapy; ART; chemoprophylaxis; HIV drug resistance; HIV epidemic; HIV transmis-
sion; mathematical model; pre-exposure prophylaxis; PrEP; South Africa.
Oral antiretroviral (ARV) pre-exposure prophylaxis
(PrEP) is a new biomedical intervention against human
immunodeciency virus (HIV) transmission with
proven efcacy [13]. There is concern, however, about
the potential emergence and spread of HIV drug resis-
tance arising from the rollout of PrEP, particularly
in resource-constrained settings, where antiretroviral
therapy (ART) options are limited [4]. This concern is
amplied by the possibility that the same ARVs will
be used for both ART and PrEP. The combination of 2
nucleoside reverse-transcriptase inhibitors, tenofovir
(TDF) and lamivudine or emtricitabine (3TC or FTC,
respectively), with 1 nonnucleoside reverse-transcriptase
inhibitor (NNRTI), efavirenz or nevirapine, is the
World Health Organizationrecommended rst-line
ART regimen in several countries worldwide, including
South Africa [4], and TDF or TDF + FTC have shown
efcacy in HIV prevention trials [13]. Thus far, only 9
Received 22 October 2012; accepted 15 January 2013; electronically published 9
April 2013.
Presented in part: 18th Conference on Retroviruses and Opportunistic Infections,
Boston, Massachusetts, 27 February2 March 2011. Abstract 98LB. 6th IAS Confer-
ence on HIV Pathogenesis, Treatment and Prevention, Rome, Italy, 1720 July
2011. Abstract TUPE364.
a
Present afliation/address: Department of Mathematics, NEIU, 5500 North St
Louis Ave, Chicago, Illinois.
Correspondence: Ume L. Abbas, MD, MS, Departments of Infectious Diseases
and Quantitative Health Sciences, Cleveland Clinic Foundation, 9500 Euclid Ave/
G21, Cleveland, OH 44195 (abbasu@ccf.org).
The Journal of Infectious Diseases 2013;208:22434
© The Author 2013. Published by Oxford University Press on behalf of the Infectious
Diseases Society of America. All rights reserved. For Permissions, please e-mail:
journals.permissions@oup.com.
DOI: 10.1093/infdis/jit150
224 JID 2013:208 (15 July) Abbas et al
by guest on September 10, 2016http://jid.oxfordjournals.org/Downloaded from
drug-resistant cases have been observed among clinical trial
participants on PrEP, most of whom had unrecognized acute
infection at enrollment. However, clinical trials of PrEP are not
designed to address the population-level and/or long-term epi-
demiological impact of PrEP, including consequences of drug
resistance. We therefore used a mathematical model [5]to
examine the potential impact of orally administered overlap-
ping and nonoverlapping PrEP and ART on HIV transmission
and drug resistance in South Africa.
METHODS
Model Structure
We developed and analyzed a detailed mathematical model to
assess the impact of PrEP and ART implementation on the
adult population (aged 1549 years) of South Africa, using de-
terministic and stochastic modeling techniques and the pro-
gramming language C/C++. The model describes population
and epidemiological stratications based on gender (male;
female), sexual activity (high; medium; low; lowest), PrEP and
ART use status (on; not on), infection status (susceptible; in-
fected), stage of HIV infection (acute preseroconversion; acute
postseroconversion; early chronic; late chronic; AIDS), and
HIV drug susceptibility (drug-sensitive; drug-resistant). Model
parameter assignments are made using recent results from
PrEP trials [13,6,7] and data mainly from South/sub
Saharan Africa on HIV disease progression [8], infectivity [9],
sexual behavior [10], ART rollout [4,1118], and HIV drug re-
sistance [1933]. The model is calibrated to simulate the HIV
epidemic in South Africa with adult HIV prevalence (Supple-
mentary Figure S1) reaching 17% at the end of 2003, having a
female-to-male prevalence ratio of 1.6 and HIV incidence near
2.4% [34]. A simplied model structure is shown in Supple-
mentary Figure S2 and model input parameters are shown in
Tables 1 and 2and Supplementary S1. Model equations and
details are provided in the Supplementary Text S1.
Table 1. Model ART-Related Input Parameters
Parameter
Uncertainty
Symbol Base Case LHS Range Unit Reference
ART coverage
Start of ART rollout 2004 [17]
% of eligible individuals enrolled in ART at 2010 55% [18]
% of eligible individuals enrolled in ART at 2012 Θ80% 65%95% [11]
Coverage beyond 2012 80% 65%95% Per year
ART dropout
During first year of ART 1/η
H
0.10 0.050.15 Per year [13,16]
During subsequent years of ART 1/η
T
0.05 0.0250.075 Per year [13,16]
Infectivity relative to WT virus
On suppressive ART 4% 1%27% [36]
With acquired ART resistance 75% 37.5%100% [19,22,25,27]
With transmitted ART resistance 100% 50%100% [24,27]
Disease progression
Mortality in first year of suppressive ART ϖ
H
0.1 0.050.15 Per year [13,14,16]
Mortality in subsequent years of suppressive ART ϖ
T
0.05 0.0250.075 Per year [13,14,16]
Relative to WT disease progression with acquired
majority ART resistance, on or off ART
75% 37.5%100% [19,22,25]
Relative to WT disease progression while
nonadherent to ART
100% 100%
Virologic failure
WT virus failure rate during first year of ART
^
LHW 20% 10%30% Per year [19,30,32]
WT virus failure rate during subsequent years of ART
^
LTW 5% 2.5%7.5% Per year [19,30]
DR virus failure rate during first year of ART
^
LHV 50% 25%75% Per year [29]
DR virus failure rate during subsequent years of ART
^
LTV 15% 7.5%22.5% Per year [29]
% failing first year ART due to NA (no acquired DR) ^
a
H4% Per year [32]
% failing in subsequent years of ART due to NA (no
acquired DR)
^
a
T2% Per year [32]
Persistence time of transmitted ART resistance ψ
R1
3 1.54.5 Years [23,26,33]
Persistence time of acquired ART resistance ψ
R2
0.25 0.1250.375 Years [20,31]
Abbreviations: ART, antiretroviral therapy; DR, drug resistance; LHS, Latin hypercube sampling; NA, nonadherence; PrEP, pre-exposure prophylaxis; WT, wild-type.
Overlapping and Non-Overlapping ARVs JID 2013:208 (15 July) 225
by guest on September 10, 2016http://jid.oxfordjournals.org/Downloaded from
HIV Drug Resistance
We stratify HIV-infected individuals based on their ARV
status, HIV drug susceptibility, type of drug resistance, and
virus population dynamics of drug-resistant HIV, including
persistence and reversion of resistance [35]. The model tracks
individuals infected with different viral variants over time,
either untreated, on PrEP, or on ART. We do not explicitly rep-
resent different drug-resistant mutants but assume the emer-
gence and transmission of 184V with PrEP use [1,2,6]; and
although several different mutations may arise with ART use
(such as 103N, 106M, 181C, 184V, 65R), 184V is the most
common. Transmitted resistancemay occur from a donor
either on PrEP, not on PrEP, on ART, or not on ART, having a
majority population of drug-resistant virus, to a recipient either
on or not on PrEP. Acquired resistancemay occur due to de
novo selection on PrEP or ART in persons with wild-type in-
fection, reemerge from archived drug-resistant variants on
PrEP or ART, or persist/accumulate on ART. Upon removal of
drug pressure, either by discontinuation of ART or PrEP or
transmission to a recipient not on PrEP or ART, the drug-
Table 2. Model PrEP-Related Input Parameters
Parameter
Uncertainty
Symbol Base Case LHS Range Unit Reference
PrEP Program
% of individuals enrolled in PrEP (coverage)
w
30% 15%45% . . .
% of inappropriate PrEP use among
individuals with established infection
~
w
2.5% . . . . . .
Initial year of PrEP deployment 2012 . . .
Time to reach target coverage 5 2.57.5 Years . . .
HIV testing frequency in PrEP program 1=
f
P639 Months . . .
HIV testing frequency in general population 1=
f
~
P1 . . . Years . . .
Average duration of PrEP use 1/σ5 2.57.5 Years . . .
Effects of PrEP
Efficacy of PrEP for WT or reverted virus ξ
W
90% 70%99% [1,2]
Adherence when highly/poorly adherent θ95%/1% 80%99%/179% [1]
Proportion highly/poorly adherent 88%/12% 10%90%/9010% [1]
Efficacy of PrEP against resistant virus ξ
R
,ξ
Q
0.25 ξ
W
0.125 ξ
W
0.375ξ
W
Relative infectivity while on PrEP with WT or
reverted virus
100% 50%100% [2]
Relative infectivity of acquired PrEP-resistant
virus, on or off PrEP
75% 50%100% [2]
Relative infectivity of transmitted PrEP-
resistant virus, on or off PrEP
100% 50%100% [2,21,27]
Time to acquisition of PrEP resistance with
WT virus in entire cohort
t
1
0.5 0.250.75 Years . . .
Time to acquisition of PrEP resistance with
reverted resistant virus in entire cohort
t
2
0.5t
1
0.25t
1
0.75t
1
Years . . .
Rate of PrEP resistance acquisition with WT
virus
π
W
ln (10.99θ)/t
1
ln (10.99θ)/t
1
Per year . . .
Rate of PrEP resistance acquisition with
reverted resistant virus
π
r
1
,π
q
1
ln (10.99θ)/t
2
ln (10.99θ)/t
2
Per year . . .
Persistence time of transmitted PrEP
resistance
ψ
Q
1
213 Years [2,26,27,33]
Persistence time of acquired PrEP resistance ψ
Q
2
0.125 0.06250.1875 Years [2]
Relative disease progression rates
While on PrEP with WT infection 100% 50%100% [2]
With acquired resistance to PrEP, on or off
PrEP
75% 50%100% [2]
While on PrEP with transmitted or reverted
resistant infection
100% 50%100% [2,21,27]
With transmitted resistant virus and no ARV
pressure
100% 50%100% [2,21,24,27]
Abbreviations: ART, antiretroviral therapy; ARV, antiretroviral; HIV, human immunodeficiency virus; LHS, Latin hypercube sampling, PrEP, pre-exposure prophylaxis;
WT, wild-type.
226 JID 2013:208 (15 July) Abbas et al
by guest on September 10, 2016http://jid.oxfordjournals.org/Downloaded from
resistant virus may revert to drug-sensitive virus after a period
of persistence. Prior to reversion, drug-resistant variants com-
prise the majority population, whereas following reversion,
they become a minority population [35].
ARV Interventions, Base-Case Scenarios, and Model Analyses
We simulate 3 different rollout strategies for ARV-mediated HIV
preventionART alone, PrEP alone (a hypothetical illustration),
and ART + PrEPand compare the epidemiological outcomes
with an ARV-naive epidemic. For each strategy, we rst construct
and analyze a reference-case (base-case) scenario using a dened
set of input parameters, including estimates of the effectiveness
of ART and PrEP for prevention of HIV from the HPTN 052
clinical trial [36] and the Partners PrEP study [1], respectively;
followed by uncertainty and sensitivity analyses [37].
Base-Case Analyses
ART Rollout and Effectiveness. In our model, individuals
become treatment eligible at CD4 lymphocyte cell counts <200
cells/µL [11]. Treatment scale-up starts at the end of 2003 [17]
and the proportion of eligible persons on ART (ie, coverage)
reaches 55% by the end of 2009 [18] and 80% by the end of
2011 [11]. Coverage is then maintained at 80% throughout the
simulation [11]. To represent the current situation in South
Africa, we simulate 2 additional scenarios of expanded ART
rollout in which treatment eligibility threshold changes at the
end of 2009 to include individuals with CD4 counts between
200 and 350 cells/µL [4], reaching 66% coverage at CD4 count
threshold <350 cells/µL by the end of 2011 [15]. Coverage is
then: (1) maintained at the 66% level (termed status-quo cover-
age) or (2) increased to reach 80% at the end of 2016 [12] and
maintained thereafter (termed optimized coverage). We model
only rst-line ART with conservative coverage to focus on the
interplay between rst-line ART and PrEP, assuming that
access to second-line regimens [38] and drug-resistance testing
[39] is limited. In base-case analyses, we assume ART reduces
HIV transmission by 96% [36]. Our model represents virologic
suppression and failure (with/without drug resistance),
dropout, survival, and HIV transmission during the rst and
subsequent years of ART.
PrEP Rollout and Effectiveness. The effectiveness of PrEP
against HIV acquisition is a composite of efcacy and adher-
ence [40]. The Partners PrEP study showed the effectiveness of
oral TDF + FTC PrEP to be 75% (95% condence interval [CI],
5587); with 90% efcacy of PrEP in those with near-perfect
adherence, and only 12% of subjects having less than 80% ad-
herence [1].
We therefore stratify individuals into 2 groups based on their
level of adherence to PrEP: high or low. For base-case analyses,
we assume that close to 90% of individuals have 95% adherence
and about 10% have low (near zero) adherence. However, given
the conicting results from different PrEP trials (TDF + FTC
was ineffective in the Fem-PrEP trial [6], and oral TDF was in-
effective in the VOICE trial [7]), for uncertainty and sensitivity
analyses we use a wide range of input estimates for PrEP efca-
cy and adherence and the proportion of individuals in the 2
(high/low) adherence groups.
PrEP (TDF + FTC) scale-up starts in 2012 and achieves 30%
coverage over a 5-year period that is then maintained. We
assume that PrEP is about 90% efcacious against wild-type
virus [1,2] and that the average duration of PrEP use is 5 years
in susceptible individuals with HIV testing every 6 months
(and PrEP discontinuation if HIV infection occurs). For the
ART + PrEP strategy, in addition to our base-case scenario with
overlapping drugs (ie, cross-resistance) between PrEP (TDF +
FTC) and ART (TDF + FTC + NNRTI), we simulate an alter-
nate scenario with identical model input and structural as-
sumptions except for there being no overlap/cross-resistance
between ART and PrEP.
Uncertainty Analyses
We perform uncertainty analyses to estimate the extent of varia-
tion in our projections across a broad range of input parameter
estimates that include the following assumptions (Tables 1and
2): ART effectiveness is 73%99%; PrEP efcacy against wild-
type virus is 70%99%; PrEP adherence among individuals
highly adherent is 80%99% and among poorly adherent is 1%
79%; the proportion of individuals highly adherent is 10%90%;
PrEP coverage is 15%45%; average duration of PrEP use is 2.5
7.5 years; the frequency of HIV testing under the PrEP program
is 39 months; and the time by which about 100% of wild-type
virus recipients acquire PrEP resistance from inappropriate PrEP
use with perfect adherence is 39 months with the median time
to acquired resistance of about 1 month [41]. We perform
50 000 simulations using Latin hypercube sampling (LHS) for
each ARV-based strategy, and compute the epidemiological out-
comes (median and interquartile range [IQR]) in comparison
with an ARV-naive baseline epidemic. We also calculate the out-
comes for the overlapping and nonoverlapping ART + PrEP
strategies in comparison with ART alone as baseline.
Sensitivity Analyses
We conduct sensitivity analyses to identify those parameters
that exert the greatest inuence on the predicted model out-
comes for each strategy. For these time-dependent multivariate
analyses, we use the input and output data from our uncertain-
ty analyses to derive standardized regression coefcients. In ad-
dition, we examine the sensitivity of the models predictions to
the modeling technique by comparative analyses of our sto-
chastic and deterministic model simulations.
Inappropriate PrEP Use
We simulate 2 contexts of inappropriate PrEP initiation and
use by previously infected individuals by extending our
Overlapping and Non-Overlapping ARVs JID 2013:208 (15 July) 227
by guest on September 10, 2016http://jid.oxfordjournals.org/Downloaded from
PrEP-alone and ART + PrEP base-case scenarios. In the rst,
individuals in the preseroconversion phase of acute HIV infec-
tion are started on PrEP (window use). In the second, indi-
viduals with undiagnosed established HIV infection start PrEP
inappropriately at a rate of 2.5% per year (general use). The
duration of inappropriate PrEP use following seroconversion is
determined by the HIV testing interval assumed for the PrEP
program (6 months for base-case; LHS range: 39 months). For
general use, the duration is determined by the frequency of
population surveillance (1 year for base-case).
RESULTS
Prevention of HIV Transmission
Base-Case Scenarios
Figure 1Ashows the impact of different ARV-based strategies
on HIV prevention after 10 years compared with an ARV-naive
epidemic. ART alone is projected to prevent 20% of HIV
infections (0.92 million). Similarly, PrEP alone prevents 21%
(0.96 million) of HIV infections. The combined strategy of
ART + PrEP is predicted to be most effective, reducing infec-
tions by 35% (>1.6 million) with overlapping regimens and
37% (>1.7 million) with nonoverlapping ARV regimens.
Expanded ART Rollout
The scenarios, which expand treatment rollout to include cov-
erage at a CD4 count <350 cells/µL, result in modest increase in
infections prevented when measured against the base-case sce-
narios of ART alone and overlapping ART + PrEP (Figure 1B).
Coverage at 66% (status-quo coverage) respectively prevents
23% and 38% of infections, while 80% coverage (optimized cov-
erage) prevents 28% and 41% of infections versus 20% and 35%
for the base-case ART-alone and ART + PrEP scenarios.
Prediction Uncertainty of HIV Prevention
Figure 2Ashows the results of uncertainty analyses for the 3
ARV-based strategies. The median decrease in HIV infections with
ART alone after 10 years is 15% (IQR, 12%19%), PrEP alone is
14% (IQR, 10%18%), overlapping ART + PrEP is 27% (IQR,
22%31%) and non-overlapping PrEP is 28% (IQR: 23%33%).
Overlapping ART + PrEP (Figure 2C) prevents a median of
12.7% (IQR, 9.1%17.2%) more infections than ART alone.
Results are similar for nonoverlapping ART + PrEP (median,
14%; IQR, 10%18.9%).
HIV Drug Resistance
Base-Case Scenarios
Figure 3Ashows the impact of different ARV-based strategies
on HIV drug resistance prevalence compared with an ARV-
naive epidemic. After 10 years of PrEP alone, the prevalence of
overall resistance is low at 0.5% (20 090 cases). Drug resistance
prevalence is higher from the ART-alone strategy at 6.6%
overall (307 254 cases) with 4.2% acquired (195 758 cases) and
2.4% transmitted resistance (111 497 cases). The prevalence of
resistance increases further from overlapping ART + PrEP to
8.2% (339 895 cases) with the prevalence of acquired and trans-
mitted ART resistance increasing to 4.6% and 3.3%, respective-
ly. With nonoverlapping ART + PrEP, drug resistance
prevalence falls modestly to 7.2% due to a lower prevalence of
transmitted ART resistance (2.2%). In terms of the number of
prevalent cases of drug resistance (data not shown), acquired
ART resistance falls modestly from both overlapping and non-
overlapping ART + PrEP, when measured against ART alone;
however, transmitted ART resistance rises with overlapping but
falls with nonoverlapping ART + PrEP. Both acquired and
transmitted cases of PrEP resistance fall from ART + PrEP
when measured against PrEP alone.
Expanded ART Rollout
The scenarios of expanded ART rollout result in a modest in-
crease in drug resistance prevalence when measured against the
Figure 1. A, Cumulative new HIV infections prevented after 10 years
(20122022) compared to a naive epidemic, assuming base-case scenari-
os. B, Cumulative new HIV infections prevented after 10 years (2012
2022) compared to a naive epidemic, assuming scenarios with different
treatment eligibility thresholds and levels of coverage. Abbreviations: ART,
antiretroviral therapy; HIV, human immunodeciency virus; PrEP, pre-expo-
sure prophylaxis.
228 JID 2013:208 (15 July) Abbas et al
by guest on September 10, 2016http://jid.oxfordjournals.org/Downloaded from
base-case scenarios of ART alone and overlapping ART + PrEP
strategies (Figure 3B). Drug resistance prevalence increases to
8.3% and 10.1% in status-quo coverage scenarios and 11.4%
and 13.4% in optimized coverage scenarios, versus 6.6% and
8.2% in the base-case scenarios of ART alone and ART + PrEP,
respectively.
Ratio of Cumulative Infections Prevented to Prevalent and
Incident Drug-Resistant Cases
To compare the resistance consequences of different ARV-based
strategies, we calculated ratios of cumulative infections prevented
to resistance over 10 years, either dened as prevalent cases (pre-
vailing cases with majority drug-resistant variants; Figure 4A)or
incident (new cases of transmitted or acquired drug resistance;
Figure 4B). PrEP alone prevents about 48 infections for each
prevalent drug-resistant case and more than 5 infections for each
incident drug-resistant case. Inappropriate window-use in the
PrEP-alone strategy decreases these ratios modestly to 46 and
4.8, respectively. By contrast, inappropriate general-use PrEP
markedly reduces the ratios to 10 and 1, respectively. ART alone
prevents about 7 infections for each prevalent drug-resistant case
and about 1 infection for each incident drug-resistant case,
which is 6- to 7-fold lower than for PrEP. The prevention-
resistance ratios for prevalent and incident cases are 9.8 and 1.4,
respectively, for overlapping ART + PrEP, and 14.7 and 1.7,
respectively, for nonoverlapping ART + PrEP.
Figure 3. A, Prevalence of drug resistance after 10 years (20122022),
assuming base-case scenarios. B, Prevalence of drug resistance after 10
years (20122022), assuming scenarios with different treatment eligibility
thresholds and levels of coverage. Columns of different colors represent
the prevalence of overall drug resistance and acquired and transmitted
drug resistance from ART and PrEP. Abbreviations: ART, antiretroviral
therapy; PrEP, pre-exposure prophylaxis.
Figure 2. Uncertainty analyses. Results of 50 000 simulations are
shown as columns representing the median values and bars representing
the interquartile range. A, Cumulative new HIV infections prevented after
10 years (20122022), compared to a naive epidemic. B, Prevalence of
drug resistance after 10 years (20122022). Panel C: Cumulative new HIV
infections prevented and prevalence of drug resistance from ART + PrEP
after 10 years (20122022), compared to an epidemic with ART alone. Ab-
breviations: ART, antiretroviral therapy; HIV, human immunodeciency
virus; PrEP, pre-exposure prophylaxis.
Overlapping and Non-Overlapping ARVs JID 2013:208 (15 July) 229
by guest on September 10, 2016http://jid.oxfordjournals.org/Downloaded from
Prediction Uncertainty of HIV Drug Resistance
Figure 2Bshows the results of uncertainty analyses for HIV
drug resistance outcomes from different ARV-based strategies.
After 10 years, the median overall prevalence of drug resistance
from ART alone is 5.9% (IQR, 4.6%7.4%), from PrEP alone is
0.5% (IQR, 0.3%0.7%), from overlapping ART + PrEP is 7%
(IQR, 5.6%8.8%), and from nonoverlapping ART + PrEP is
6.5% (IQR, 5.2%8.1%). These nding are consistent with our
base-case scenarios.
Overlapping ART + PrEP compared to ART alone
(Figure 2C), increases the number of prevalent overall and
transmitted ART-resistant cases after 10 years by a median
8.8% (IQR, 5.8%13.1%) and 15.9% (IQR, 11.4%21.9%), re-
spectively, while modestly decreasing the number of acquired
ART-resistant cases (median, 0.9%; IQR, 1.8% to 0%). Non-
overlapping ART + PrEP decreases the overall drug resistance
prevalence at 20 years (median, 4%; IQR, 7.5% to 0.7%).
Inappropriate PrEP Use
Inappropriate PrEP use by persons infected at baseline increas-
es HIV drug resistance from PrEP. When measured against the
overlapping ART + PrEP base-case, an overlapping ART +
PrEP strategy that includes inappropriate window-use PrEP
prevents almost the same number of infections (1.63 million),
with a modest increase (8.3% vs 8.2%) in the prevalence of re-
sistance (data not shown). In contrast, overlapping ART + PrEP
with inappropriate general-use PrEP leads to an increase in the
overall resistance prevalence from 8.2% to over 10%, with ac-
quired PrEP resistance rising to 1.3% from 0.2% and transmit-
ted PrEP resistance to 0.4% from 0.1% (data not shown).
Nonoverlapping ART + PrEP with inappropriate general-use
PrEP raises the overall resistance prevalence to 8.5% (data not
shown).
Sensitivity Analyses
The results of the sensitivity analyses are described in detail in
Supplementary Text S1 and summarized in Table 3.
DISCUSSION
The important insights derived from our study are several.
First, an ART strategy of treatment initiation at a CD4 count
<200 cells/µL combined with PrEP prevents more infections
than either ART alone or PrEP alone; however, the incremental
benet of PrEP critically depends on PrEP efcacy, adherence,
and coverage. Second, the prevalence of HIV drug resistance is
largely driven by ART in both ART alone and ART + PrEP
strategies. Third, PrEP alone results in low prevalence of drug
resistance; high PrEP adherence leads to fewer infections and
less opportunity for acquired resistance, while low adherence
leads to predominantly wild-type breakthrough infections
because of low drug pressure for emergence of acquired resis-
tance. Fourth, use of overlapping ARVs for both ART and
PrEP could increase drug-resistance prevalence compared to
ART alone due to more frequent transmitted resistance. By
contrast, resistance prevalence falls with non-overlapping
ART + PrEP; however, this decrease is modest because the prin-
cipal driver of resistance is ART, not PrEP. Fifth, inappropriate
PrEP initiation among individuals with undetectable HIV in-
fection produces only a minor increase in the overall resistance
prevalence; however, inappropriate PrEP use among persons
with established HIV infection could signicantly increase
drug resistance from PrEP. Lastly, PrEP prevents many more
infections per case of resistance than ART does.
The extent of coverage and the degree of effectiveness against
HIV transmission are the principal determinants of the infec-
tions prevented with ART. Similarly, PrEP coverage and effec-
tiveness against HIV acquisition are the key determinants of
the additional preventive benet of ART + PrEP. The paradigm
of test and treat [42] has gained considerable momentum, and
the HPTN 052 trial [36] has provided the needed proof of
concept for ART-based prevention, though its population-level
impact may be limited by potential reluctance of asymptomatic
Figure 4. A, Ratio of cumulative infections prevented to prevalent drug-
resistant cases (20122022). B, Ratio of cumulative infections prevented
to incident drug-resistant cases (20122022). Window use refers to inap-
propriate PrEP initiation by persons in the preseroconversion phase of
acute HIV. General use refers to inappropriate PrEP initiation by persons
with established HIV infection at a per capita rate of 2.5%/year. Abbrevia-
tions: ART, antiretroviral therapy; HIV, human immunodeciency virus;
PrEP, pre-exposure prophylaxis.
230 JID 2013:208 (15 July) Abbas et al
by guest on September 10, 2016http://jid.oxfordjournals.org/Downloaded from
Table 3. Sensitivity Analysis of Outcomes With ARV Strategies Versus Naive Epidemic at 2022
a
Model Input
b
Standardized Regression Coefficients (% variance explained)
c
ART Alone PrEP Alone ART + PrEP
Infections prevented (%)
Reduction in WT infectivity on ART 0.70 (0.49) . . . 0.47 (0.22)
ART coverage 0.59 (0.35) . . . 0.37 (0.14)
Relative infectivity of virus with acquired ART resistance 0.24 (0.06) . . .
PrEP coverage . . . 0.67 (0.45) 0.50 (0.25)
PrEP proportion highly adherent . . . 0.45 (0.20) 0.33 (0.11)
PrEP adherence (low) . . . 0.40 (0.16) 0.30 (0.09)
PrEP efficacy against WT virus . . . 0.29 (0.08) . . .
Prevalence of overall drug resistance (%)
d
Survival time on ART with acquired resistance 0.57 (0.32) . . . 0.56 (0.31)
ART coverage 0.46 (0.22) . . . 0.44 (0.19)
WT virologic failure rate during first year on ART 0.36 (0.13) . . . 0.34 (0.11)
WT virologic failure rate during subsequent years on ART 0.31 (0.09) . . . 0.30 (0.09)
% failing subsequent years on ART due to nonadherence 0.25 (0.06) . . . 0.25 (0.06)
Persistence of transmitted ART resistance 0.23 (0.06) . . . 0.25 (0.06)
PrEP coverage . . . 0.55 (0.30) . . .
Frequency of HIV testing . . . 0.50 (0.25) . . .
PrEP adherence (low) . . . 0.30 (0.09) . . .
PrEP efficacy against WT virus . . . 0.26 (0.07) . . .
Development time for acquired PrEP resistance . . . 0.25 (0.06) . . .
Prevalence of transmitted ART resistance (%)
Persistence of transmitted ART resistance 0.57 (0.32) . . . 0.55 (0.30)
Relative infectivity of virus with acquired ART resistance 0.45 (0.20) . . . 0.46 (0.21)
Survival time on ART with acquired resistance 0.35 (0.12) . . . 0.35 (0.12)
ART coverage 0.30 (0.09) . . . 0.29 (0.08)
WT virologic failure rate during first yr on ART 0.25 (0.06) . . . 0.24 (0.06)
Prevalence of acquired ART resistance (%)
Survival time on ART with acquired resistance 0.61 (0.38) . . . 0.62 (0.39)
ART coverage 0.49 (0.24) . . . 0.49 (0.24)
WT virologic failure rate during first year on ART 0.37 (0.14) . . . 0.35 (0.12)
WT virologic failure rate during subsequent years on ART 0.32 (0.10) . . . 0.32 (0.10)
% failing subsequent years on ART due to nonadherence 0.26 (0.07) . . . 0.26 (0.07)
Prevalence of transmitted PrEP resistance (%)
PrEP coverage . . . 0.51 (0.26) 0.48 (0.23)
Persistence of transmitted PrEP resistance . . . 0.39 (0.15) 0.39 (0.15)
Frequency of HIV testing . . . 0.29 (0.09) 0.30 (0.09)
PrEP adherence (low) . . . 0.29 (0.09) 0.28 (0.08)
Development time for acquired PrEP resistance . . . 0.24 (0.06) 0.25 (0.06)
Prevalence of acquired PrEP resistance (%)
Frequency of HIV Testing . . . 0.56 (0.32) 0.56 (0.31)
PrEP coverage . . . 0.52 (0.27) 0.50 (0.25)
PrEP adherence (low) . . . 0.28 (0.08) 0.27 (0.07)
PrEP efficacy against WT virus . . . 0.28 (0.08) 0.29 (0.08)
Development time for acquired PrEP resistance . . . 0.24 (0.06) 0.24 (0.06)
Abbreviations: ART, antiretroviral therapy; HIV, human immunodeficiency virus; PrEP, pre-exposure prophylaxis; SRC, standardized regression coefficient; WT, wild-type.
a
The results of sensitivity analyses are described in the Supplementary Text S1. Briefly, the principal determinants of infections prevented by PrEP alone and/or
ART + PrEP include PrEP coverage, reduction in WT viral infectivity by ART, the proportion of persons highly adherent to PrEP, and the level of PrEP adherence.
Drug resistance prevalence from ART alone and ART + PrEP is most influenced by the duration of survival on ART with acquired ART resistance and the WT
virologic failure rate during the first year of ART. PrEP coverage and the frequency of HIV testing are the key determinants of drug resistance from PrEP.
b
Parameters that contribute 5% or more of the variance in the model outcome are shown (SRC
2
0.05). The reported coefficients were significant (P.05).
c
Percentage of the variance in the predicted outcome explained by the regression model. The respective R
2
values were 0.93 (cumulative infections prevented),
0.94 (overall prevalence of resistance), 0.90 (prevalence of transmitted ART resistance), 0.95 (prevalence of acquired ART resistance), 0.79 (prevalence of
transmitted PrEP resistance), and 0.81 (prevalence of acquired PrEP resistance) for ART + PrEP scenario.
d
Proportion of cases with drug-resistant infection in the infected population.
Overlapping and Non-Overlapping ARVs JID 2013:208 (15 July) 231
by guest on September 10, 2016http://jid.oxfordjournals.org/Downloaded from
HIV-infected persons for ART initiation. Notwithstanding
scale-up efforts, there is considerable unmet need for ART in
resource-constrained settings; about 60% of those eligible did
not have access to ART at the end of 2010 [43]. Moreover, the
population-level effect of treatment as prevention could be
limited by the actual proportion of infected individuals opti-
mally and durably suppressed on ART. In 2010, of the 1.2
million infected persons in the United States, 80% were aware
of their status, but 41% were retained in care, and only 28% had
virologic suppression [44]. The situation is much worse in sub
Saharan Africa, where about two-thirds of HIV-infected
persons are unaware of their seropositive status [45]. In a sys-
tematic review [46], fewer than one-third of HIV-positive
persons were retained in care between HIV testing and ART
initiation. Furthermore, studies show high rates of loss to
follow-up among patients starting ART [16]. Thus, PrEP could
play an important additional role in controlling the HIV pan-
demic. Prioritized coverage with effective PrEP of individuals at
highest risk of HIV acquisition and spread could potentially
yield the optimal public health and cost benets [40]. ART
rollout is also limited by infrequent [18] access to second-line
regimens and CD4 cell count, rather than virological monitor-
ing [4]. As a result, there are high levels of drug resistance mu-
tations among individuals with prolonged virological failure
[22,32], which may compromise both rst-line [29,47] and the
limited second-line [48] ART regimens available. Our model
shows that ART drives the prevalence of HIV drug resistance in
both ART alone and ART + PrEP strategies. The principal de-
terminants of the prevalence of acquired resistance include
ART coverage, survival on ART with acquired resistance, and
the rate of treatment failure. For the prevalence of transmitted
resistance, determinants include the infectiousness of persons
with acquired ART resistance and the persistence time of trans-
mitted resistance. We nd that PrEP is about 6- to 7-fold more
efcient in HIV prevention than ART in terms of ratios of in-
fections prevented to incident/prevalent drug-resistant cases
generated. Thus, improving the effectiveness of rst- and
second-line ART is critical for preventing HIV infection and
controlling drug resistance.
Our model projects a low prevalence of drug resistance from
PrEP. Highly effective PrEP results in few breakthrough infec-
tions and a chance for emergence of acquired resistance. By
contrast, poorly effective PrEP fails to protect from acquisition
of wild-type HIV but also fails to exert selective pressure for
emergence of acquired resistance. Both of these phenomena
have been observed in recent PrEP trials [1,2]. However, drug
resistance from PrEP at the population level could rise with
inappropriate PrEP use among those with undiagnosed HIV
infection. While this increase is modest from inappropriate
PrEP use during the preseroconversion phase of acute infec-
tion, it becomes more pronounced with inappropriate use
among persons with established HIV. The latter may be of
concern in potential situations of unsupervised PrEP use (eg,
black-market drugs and drug sharing [49]) or inaccurate HIV
testing [50].
There are some important limitations of our model. The accu-
racy of our predictions will be affected by variations in the model
structure and sexual activity details, for which data are very
limited. We therefore employed a well-established template of
sexual behavior [40] with robust epidemiological and demo-
graphic parameterization, broadly applicable to South Africa.
Nevertheless, the HIV epidemic in South Africa is heterogeneous
and incompletely understood, with signicant differences
between the demographic and HIV/AIDS epidemiological esti-
mates predicted by different agencies. HIV incidence is also not
precisely known, even when measured directly at the population
level. Although there is uncertainty regarding ARV-related pa-
rameters, we employed ranges (within plausible bounds) and
performed extensive sensitivity and uncertainty analyses. We ex-
cluded population stratication by age and analysis of prioritized
ARV implementation, as this was addressed in previous work
[40]. Because of limited access to both second-line regimens [38]
and drug-resistance testing [39] in resource-limited settings, we
chose not to represent specic drug resistance mutations or
second- or third-line ART regimens, nor do we consider HIV
subtype polymorphism. We also did not explicitly include other
inuences on transmission. These and other renements will be
included in future work, although including such parameters
greatly increases model complexity.
A key conclusion of this study is that combined ART + PrEP
can have a greater public health impact than ART alone;
however, overlapping ARVs for both can increase drug resis-
tance in resource-limited settings. Drug resistance prevalence is
predominantly driven by ART and not PrEP; consequently,
nonoverlapping strategies will produce only modest declines in
resistance. Thus, it is critical to consider the impact of ARVs
not only on prevention but also drug resistance. Improved ef-
cacy of rst-line therapy and timely switching of ART to effec-
tive second-line regimens are critical for controlling HIV drug
resistance. In addition, frequent and accurate HIV testing could
minimize resistance consequences of PrEP. Our study also
highlights that poor adherence to PrEP will undermine its po-
tential impact on HIV prevention. Thus, prioritization of PrEP
to groups at most risk of HIV acquisition and counseling about
PrEP adherence are likely to maximize efciency of PrEP and
minimize drug resistance.
Supplementary Data
Supplementary materials are available at The Journal of Infectious Diseases
online (http://jid.oxfordjournals.org/). Supplementary materials consist of
data provided by the author that are published to benet the reader. The
posted materials are not copyedited. The contents of all supplementary data
are the sole responsibility of the authors. Questions or messages regarding
errors should be addressed to the author.
232 JID 2013:208 (15 July) Abbas et al
by guest on September 10, 2016http://jid.oxfordjournals.org/Downloaded from
Notes
Financial support. This work was supported by the Bill & Melinda
Gates Foundation (OPP1005974). The funders had no role in study design,
data collection and analysis, decision to publish, or preparation of the man-
uscript.
Potential conicts of interest. U. L. A. acknowledges grant support
from the Bill and Melinda Gates Foundation (OPP1005974). J. W. M. is a
member of the scientic advisory board of Gilead Sciences, has share
options of RFS Pharmaceuticals, and acknowledges grant support from the
AIDS Clinical Trials Group (National Institute of Allergy and Infectious
Diseases [NIAID] U01AI38858), the Microbicide Trials Network (NIAID
U01AI068633), the National Cancer Institute (Science Applications Inter-
national Corporation [SAIC] contract 20XS190A), and the Bill and Melinda
Gates Foundation (OPP1005974). All other authors report no potential
conicts.
All authors have submitted the ICMJE Form for Disclosure of Potential
Conicts of Interest. Conicts that the editors consider relevant to the
content of the manuscript have been disclosed.
References
1. Baeten JM, Donnell D, Ndase P, et al. Antiretroviral prophylaxis for
HIV prevention in heterosexual men and women. N Engl J Med 2012;
367:399410.
2. Grant RM, Lama JR, Anderson PL, et al. Preexposure chemoprophylax-
is for HIV prevention in men who have sex with men. N Engl J Med
2010; 363:258799.
3. Thigpen MC, Kebaabetswe PM, Paxton LA, et al. Antiretroviral preex-
posure prophylaxis for heterosexual HIV transmission in Botswana. N
Engl J Med 2012; 367:42334.
4. World Health Organization. Antiretroviral therapy for HIV infection
in adults and adolescents: recommendations for a public health ap-
proach, Geneva, Switzerland. 2010 revision. 2010.
5. Anderson RM, May RM. Infectious diseases of humans: dynamics and
control. Oxford: Oxford University Press, 1991.
6. Van Damme L, Corneli A, Ahmed K, et al. Preexposure prophylaxis for
HIV infection among African women. N Engl J Med 2012; 367:41122.
7. Marrazzo J, Ramjee G, Nair G, et al. Pre-exposure Prophylaxis for HIV
in Women: Daily Oral Tenofovir, Oral Tenofovir/Emtricitabine, or
Vaginal Tenofovir Gel in the VOICE Study (MTN 003). 20th Confer-
ence on Retroviruses and Opportunistic Infections; March 3-6, 2013;
Atlanta, GA.
8. Wandel S, Egger M, Rangsin R, et al. Duration from seroconversion to
eligibility for antiretroviral therapy and from ART eligibility to death in
adult HIV-infected patients from low and middle-income countries:
collaborative analysis of prospective studies. Sex Transm Infect 2008;
84(Suppl 1):i316.
9. Wawer MJ, Gray RH, Sewankambo NK, et al. Rates of HIV-1 transmis-
sion per coital act, by stage of HIV-1 infection, in Rakai, Uganda.
J Infect Dis 2005; 191:14039.
10. SACEMA. The modes of transmission of HIV in South Africa,
UNAIDS: Geneva, Switzerland. 2009.
11. Department of Health South Africa. HIV and AIDS and STI Strategic
Plan for South Africa, Pretoria. 20072011. 2007.
12. Department of Health South Africa. HIV and AIDS and STI Strategic
Plan for South Africa, Pretoria. 20122016. 2012.
13. Mahy M, Lewden C, Brinkhof MW, et al. Derivation of parameters
used in Spectrum for eligibility for antiretroviral therapy and survival
on antiretroviral therapy. Sex Transm Infect 2010; 86(Suppl 2):ii2834.
14. Nglazi MD, Lawn SD, Kaplan R, et al. Changes in programmatic out-
comes during 7 years of scale-up at a community-based antiretroviral
treatment service in South Africa. J Acquir Immune Dec Syndr 2011;
56:e18.
15. UNAIDS. Global report: UNAIDS report on the global AIDS epidemic
2012. 2012.
16. Wandeler G, Keiser O, Pfeiffer K, et al. Outcomes of antiretroviral treat-
ment programs in rural Southern Africa. J Acquir Immune Dec Syndr
2012; 59:e916.
17. Department of Health South Africa. Operational plan for comprehen-
sive HIV and AIDS care, management and treatment for South Africa,
Pretoria. 2003.
18. World Health Organization. HIV in the WHO African Region: pro-
gress towards achieving universal access to priority health sector inter-
ventions, Geneva, Switzerland: 2011 update. 2011.
19. El-Khatib Z, Ekstrom AM, Ledwaba J, et al. Viremia and drug resis-
tance among HIV-1 patients on antiretroviral treatment: a cross-
sectional study in Soweto, South Africa. AIDS 2010; 24:167987.
20. Hance AJ, Lemiale V, Izopet J, et al. Changes in human immunode-
ciency virus type 1 populations after treatment interruption in patients
failing antiretroviral therapy. J Virol 2001; 75:64107.
21. Harrison L, Castro H, Cane P, et al. The effect of transmitted HIV-1
drug resistance on pre-therapy viral load. AIDS 2010; 24:191722.
22. Hosseinipour MC, van Oosterhout JJ, Weigel R, et al. The public health
approach to identify antiretroviral therapy failure: high-level nucleoside
reverse transcriptase inhibitor resistance among Malawians failing rst-
line antiretroviral therapy. AIDS 2009; 23:112734.
23. Kearney M, Maldarelli F, Shao W, et al. Human immunodeciency
virus type 1 population genetics and adaptation in newly infected indi-
viduals. J Virol 2009; 83:271527.
24. Little S. Transmission of HIV drug resistance and treatment response.
In: Program and Abstracts of the 14th Conference on Retroviruses and
Opportunistic Infections, Los Angeles, CA, 2528 February 2007.
25. Modjarrad K, Chamot E, Vermund SH. Impact of small reductions in
plasma HIV RNA levels on the risk of heterosexual transmission and
disease progression. AIDS 2008; 22:217985.
26. Pingen M, Nijhuis M, de Bruijn JA, Boucher CA, Wensing AM. Evolu-
tionary pathways of transmitted drug-resistant HIV-1. J Antimicrob
Chemother 2011; 66:146780.
27. Wainberg MA, Moisi D, Oliveira M, Toni TD, Brenner BG. Transmis-
sion dynamics of the M184V drug resistance mutation in primary HIV
infection. J Antimicrob Chemother. 2011; 66:23469.
28. Hamers RL, Wallis CL, Kityo C, et al. HIV-1 drug resistance in antire-
troviral-naive individuals in sub-Saharan Africa after rollout of antiretro-
viral therapy: a multicentre observational study. Lancet Infect Dis 2011;
11:7509.
29. Hamers RL, Schuurman R, Sigaloff KC, et al. Effect of pretreatment
HIV-1 drug resistance on immunological, virological, and drug-resis-
tance outcomes of rst-line antiretroviral treatment in sub-Saharan
Africa: a multicentre cohort study. Lancet Infect Dis 2012; 12:30717.
30. Barth RE, van der Loeff MF, Schuurman R, Hoepelman AI, Wensing
AM. Virological follow-up of adult patients in antiretroviral treatment
programmes in subSaharan Africa: a systematic review. Lancet Infect
Dis 2010; 10:15566.
31. Deeks SG,Wrin T, Liegler T, et al. Virologic and immunologic consequen-
ces of discontinuing combination antiretroviral-drug therapy in HIV-
infected patients with detectable viremia. N Engl J Med 2001; 344:47280.
32. Gupta RK, Hill A, Sawyer AW, et al. Virological monitoring and resis-
tance to rst-line highly active antiretroviral therapy in adults infected
with HIV-1 treated under WHO guidelines: a systematic review and
meta-analysis. Lancet Infect Dis 2009; 9:40917.
33. Jain V, Sucupira MC, Bacchetti P, et al. Differential persistence of trans-
mitted HIV-1 drug resistance mutation classes. J Infect Dis 2011; 203:
117481.
34. Rehle TM, Hallett TB, Shisana O, et al. A decline in new HIV infections
in South Africa: estimating HIV incidence from three national HIV
surveys in 2002, 2005 and 2008. PLOS One 2010; 5:e11094.
35. Nowak MA, May R. Virus dynamics: mathematical principles of immu-
nology and virology. New York: Oxford University Press, 2001.
36. Cohen MS, Chen YQ, McCauley M, et al. Prevention of HIV-1
infection with early antiretroviral therapy. N Engl J Med 2011; 365:
493505.
Overlapping and Non-Overlapping ARVs JID 2013:208 (15 July) 233
by guest on September 10, 2016http://jid.oxfordjournals.org/Downloaded from
37. Hofer E. Sensitivity analysis in the context of uncertainty analysis for
computationally intensive models. Comput Phys Commun 1999; 117:
2134.
38. Long L, Fox M, Sanne I, Rosen S. The high cost of second-line antire-
troviral therapy for HIV/AIDS in South Africa. AIDS 2010; 24:9159.
39. Keiser O, Chi BH, Gsponer T, et al. Outcomes of antiretroviral treat-
ment in programmes with and without routine viral load monitoring in
Southern Africa. AIDS 2011; 25:17619.
40. Abbas UL, Anderson RM, Mellors JW. Potential impact of antiretrovi-
ral chemoprophylaxis on HIV-1 transmission in resource-limited set-
tings. PLOS ONE 2007; 2:e875.
41. Miller DK, Homan SM. Determining transition probabilities: confusion
and suggestions. Med Decis Making 1994; 14:528.
42. Granich RM, Gilks CF, Dye C, De Cock KM, Williams BG. Universal
voluntary HIV testing with immediate antiretroviral therapy as a strat-
egy for elimination of HIV transmission: a mathematical model. Lancet
2009; 373:4857.
43. UNAIDS. AIDS at 30: Nations at the crossroads. 2011.
44. Vital signs: HIV prevention through care and treatmentUnited
States. MMWR Morb Mortal Wkly Rep 2011; 60:161823.
45. World Health Organization. Towards universal access: Scaling up pri-
ority HIV/AIDS interventions in the health sector. Geneva, WHO.
2010.
46. Rosen S, Fox MP. Retention in HIV care between testing and treatment
in sub-Saharan Africa: a systematic review. PLOS Med 2011;8:
e1001056.
47. Gupta RK, Jordan MR, Sultan BJ, et al. Global trends in antiretroviral
resistance in treatment-naive individuals with HIV after rollout of anti-
retroviral treatment in resource-limited settings: a global collaborative
study and meta-regression analysis. Lancet 2012; 380:12508.
48. Hamers RL, Sigaloff KC, Wensing AM, et al. Patterns of HIV-1 drug re-
sistance after rst-line antiretroviral therapy (ART) failure in 6 sub
Saharan African countries: implications for second-line ART strategies.
Clin Infect Dis 2012; 54:16609.
49. Inciardi JA, Surratt HL, Kurtz SP, Cicero TJ. Mechanisms of prescrip-
tion drug diversion among drug-involved club- and street-based popu-
lations. Pain Med 2007; 8:17183.
50. Wolpaw BJ, Mathews C, Chopra M, et al. The failure of routine rapid
HIV testing: a case study of improving low sensitivity in the eld. BMC
Health Serv Res 2010; 10:73.
234 JID 2013:208 (15 July) Abbas et al
by guest on September 10, 2016http://jid.oxfordjournals.org/Downloaded from
... 20−23 Supervie et al. 20 developed a mathematical model to predict how PrEP might affect population-level HIV resistance. Abbas et al. 21 evaluated the effects of implementing ART alone, PrEP alone, or PrEP plus ART on HIV incidence and drug resistance incidence in South Africa. Drabo et al. 22 estimated the costs and benefits of PrEP combined with test-and-treat among MSM in Los Angeles and Shen et al. 23 evaluated the cost-effectiveness of earlier ART plus PrEP among MSM in San Francisco. ...
... Our results showed that ART implementation alone would prevent 19% of HIV infections but increase drug resistance prevalence by 5¢4% over a 30-year period, ART plus PrEP would futher decrease the incidence. These results were consistent with the study by Abbas et al. 21 But they found that expanded ART plus PrEP would increase resistance prevalence, which may be attributed to the different initial DR prevalence between Africa and China. Despite the increasing proportion of new TDR infections, the incidence of TDR infections decreased. ...
Article
Full-text available
Background Oral pre-exposure prophylaxis (PrEP) and antiretroviral therapy (ART) can effectively prevent HIV infections among men who have sex with men (MSM), but the emergence and transmission of HIV drug-resistance (HIVDR) may compromise their benefits. The costs and benefits of expanding PrEP and ART coverage in the presence of HIVDR in China remain unknown. Methods We developed a comprehensive dynamic transmission model incorporating the transmitted (TDR) and acquired (ADR) HIV drug resistance. The model was calibrated by the HIV surveillance data from 2009 to 2019 among MSM in Jiangsu Province, China, and validated by the dynamic prevalence of ADR and TDR. We aimed to investigate the impact of eight intervention scenarios (no PrEP, 20%, 50% or 80% of PrEP, without (77% coverage) or with (90% coverage) expanded ART) on the HIV epidemic trend and cost-effectiveness of PrEP over the next 30 years. Findings 20% or 50% PrEP + 90% ART would be cost-effective, with an incremental cost-effectiveness ratio (ICER) of 25,417 (95% confidence interval [CI]: 12,390–38,445) or 47,243 (23,756–70,729), and would yield 154,949 (89,662–220,237) or 179,456 (102,570–256,342) incremental quality-adjusted life-years (QALYs) over the next 30 years. No PrEP + 90% ART would yield 125,211 (73,448–176,974) incremental QALYs and be cost-saving. However, 20–80% PrEP + 77% ART and 80% PrEP + 90% ART with ICER of $77,862–$98,338 and $63,332, respectively, and were not cost-effective. A reduction of 64% in the annual cost of oral PrEP would make it highly cost-effective for 50% PrEP + 90% ART. Interpretation 20% or 50% PrEP + 90% ART is cost-effective for HIV control in the presence of HIVDR. Expanded ART alone may be the optimal policy under the current limited budgets. Funding National Natural Science Foundation of China, the National S&T Major Project Foundation of China.
... Secondly, studies using mathematical models indicate that combining PrEP and test and treat strategy is better than using either alone. 35,36,105 This is because they can reach two distinct subpopulations: infected people and susceptible people. However, our study indicates that the synergy between these two could be more complicated given individual risk behavior volatility. ...
... One of the relevant research question is the how effective the UT&T strategy needs to be in testing and treating individuals to eliminate HIV infections.Granich et al suggest that testing every individual once a year and immediately initiating treatment once the individual is diagnosed HIV positive can eliminate HIV infections. 8 UNAIDS proposed that HIV will be eliminated if 90% of infected individuals are tested, 90% of people who are tested HIV positive are linked to care and 90% of infected people who are linked to care Given the complexity of HIV epidemics, eliminating HIV infections may require combined efforts of multiple HIV interventions rather than relying on a single strategy.Mathematical studies suggest that UT&T can be combined with PrEP to reach the maximum prevention effects because two strategies can reach two distinct population: infected people and uninfected people.[35][36][37] However, the population effect that could be achieved with the combination of the two strategies vary greatly among different studies.25,36,38-40 ...
Thesis
Men who have sex with men (MSM) are heavily affected by HIV infections. However, controlling HIV transmission among MSM population remains a challenging task due to the complexity of the transmission dynamics of HIV. Mathematical models can facilitate understanding such dynamics. They also provide a basis for estimating important epidemiological parameters that can guide public health decision. This thesis advances methods to achieve both of these objectives and makes a substantive advance in the first area. In this dissertation, we relaxed the constancy of individual risk behavior assumption, by allowing individual risk behavior to fluctuate among different levels over time, namely individual risk behavior volatility (risk volatility). We found that increasing risk volatility considerably increases fraction of transmission from acute HIV infection and prevalence at endemic equilibrium. In addition, we found that increasing risk volatility considerably reduces the minimum required individual effectiveness to eliminate HIV infections of Universal Test and Treat or universally applied pre-exposure prophylaxis (PrEP). Furthermore, our results suggest that increasing risk volatility reduces the extent that a case’s risk level at HIV acquisition determines this case’s capacity to cause onward transmission later during infection. Consequently, assuming no risk volatility may cause one to overestimate the benefit of prioritizing PrEP efforts to susceptible individuals experiencing high risk as a strategy to eliminate HIV infections. Finally, we explored the possibility of using HIV phylogeny to indicate when risk volatility affects HIV transmission dynamics. Our results suggest that risk volatility has unique and strong impact on phylogeny imbalance and clustering pattern. This implies that risk volatility is potentially identifiable from HIV sequence data. Research in this thesis contributions to the field of study that uses mathematical models to estimate epidemiological parameters of HIV transmission from two perspectives. Firstly, research in this thesis reveals the importance of evaluating individual risk behavior volatility to enhance robustness of model inference of epidemiological parameters and quantities. Secondly, research in this thesis suggests HIV sequence data is potentially valuable to improve the identifiability of risk volatility parameter. Therefore, research in this thesis takes significant steps forward to improve mathematical model inference of HIV transmission parameters.
... Despite the cART being potent and life-prolonging, a significant limitation is that it is not curative and does not eradicate HIV-1 infection since treatment interruption inevitably results in a rapid rebound of viremia [105]. The rebound of HIV is due to the presence of HIV reservoirs, mainly in the latently infected resting CD4+ memory T cells and myeloid cells such as macrophages and microglia that are difficult to target by cART or immune effector mechanisms [106][107][108][109]. HIV latency is defined as a reversibly, non-productive state of infection of individual cells that retain the capacity to produce infectious virus particles but allow the virus to evade the host's immune response [110]. ...
... Despite the cART being potent and life-prolonging, a significant limitation is that it is not curative and does not eradicate HIV-1 infection since treatment interruption inevitably results in a rapid rebound of viremia [105]. The rebound of HIV is due to the presence of HIV reservoirs, mainly in the latently infected resting CD4+ memory T cells and myeloid cells such as macrophages and microglia that are difficult to target by cART or immune effector mechanisms [106][107][108][109]. HIV latency is defined as a reversibly, non-productive state of infection of individual cells that retain the capacity to produce infectious virus particles but allow the virus to evade the host's immune response [110]. ...
Article
Full-text available
The emergence of drug-resistant Human Immunodeficiency Virus-1 strains against anti-HIV therapies in the clinical pipeline, and the persistence of HIV in cellular reservoirs remains a significant concern. Therefore, there is a continuous need to discover and develop new, safer, and effective drugs targeting novel sites to combat HIV-1. The fungal species are gaining increasing attention as alternative sources of anti-HIV compounds or immunomodulators that can escape the current barriers to cure. Despite the potential of the fungal kingdom as a source for diverse chemistries that can yield novel HIV therapies, there are few comprehensive reports on the progress made thus far in the search for fungal species with the capacity to produce anti-HIV compounds. This review provides insights into the recent research developments on natural products produced by fungal species, particularly fungal endophytes exhibiting immunomodulatory or anti-HIV activities. In this study, we first explore currently existing therapies for various HIV-1 target sites. Then we assess the various activity assays developed for gauging antiviral activity production from microbial sources since they are crucial in the early screening phases for discovering novel anti-HIV compounds. Finally, we explore fungal secondary metabolites compounds that have been characterized at the structural level and demonstrate their potential as inhibitors of various HIV-1 target sites.
Article
Full-text available
Background Kenya included oral PrEP in the national guidelines as part of combination HIV prevention, and subsequently began providing PrEP to individuals who are at elevated risk of HIV infection in 2017. However, as scale-up continued, there was a recognized gap in knowledge on the cost of delivering oral PrEP. This gap limited the ability of the Government of Kenya to budget for its PrEP scale-up and to evaluate PrEP relative to other HIV prevention strategies. The following study calculated the actual costs of oral PrEP scale-up as it was being delivered in ten counties in Kenya. This costing also allowed for a comparison of various models of service delivery in different geographic regions from the perspective of service providers in Kenya. In addition, the analysis was also conducted to understand factors that indicate why some individuals place a greater value on PrEP than others, using a contingent valuation technique. Methods Data collection was completed between November 2017 and September 2018. Costing data was collected from 44 Kenyan health facilities, consisting of 23 public facilities, 5 private facilities and 16 drop-in centers (DICEs) through a cross-sectional survey in ten counties. Financial and programmatic data were collected from financial and asset records and through interviewer administered questionnaires. The costs associated with PrEP provision were calculated using an ingredients-based costing approach which involved identification and costing of all the economic inputs (both direct and indirect) used in PrEP service delivery. In addition, a contingent valuation study was conducted at the same 44 facilities to understand factors that reveal why some individuals place a greater value on PrEP than others. Interviews were conducted with 2,258 individuals (1,940 current PrEP clients and 318 non-PrEP clients). A contingent valuation method using a “payment card approach” was used to determine the maximum willingness to pay (WTP) of respondents regarding obtaining access to oral PrEP services. Results The weighted cost of providing PrEP was $253 per person year, ranging from $217 at health centers to $283 at dispensaries. Drop-in centers (DICEs), which served about two-thirds of the client volume at surveyed facilities, had a unit cost of $276. The unit cost was highest for facilities targeting MSM ($355), while it was lowest for those targeting FSW ($248). The unit cost for facilities targeting AGYW was $323 per person year. The largest percentage of costs were attributable to personnel (58.5%), followed by the cost of drugs, which represented 25% of all costs. The median WTP for PrEP was $2 per month (mean was $4.07 per month). This covers only one-third of the monthly cost of the medication (approximately $6 per month) and less than 10% of the full cost of delivering PrEP ($21 per month). A sizable proportion of current clients (27%) were unwilling to pay anything for PrEP. Certain populations put a higher value on PrEP services, including: FSW and MSM, Muslims, individuals with higher education, persons between the ages of 20 and 35, and households with a higher income and expenditures. Discussion This is the most recent and comprehensive study on the cost of PrEP delivery in Kenya. These results will be used in determining resource requirements and for resource mobilization to facilitate sustainable PrEP scale-up in Kenya and beyond. This contingent valuation study does have important implications for Kenya's PrEP program. First, it indicates that some populations are more motivated to adopt oral PrEP, as indicated by their higher WTP for the service. MSM and FSW, for example, placed a higher value on PrEP than AGYW. Higher educated individuals, in turn, put a much higher value on PrEP than those with less education (which may also reflect the higher “ability to pay” among those with more education). This suggests that any attempt to increase demand or improve PrEP continuation should consider these differences in client populations. Cost recovery from existing PrEP clients would have potentially negative consequences for uptake and continuation.
Article
Purpose: To summarize the most noteworthy infectious diseases (ID) pharmacotherapy articles published in peer-reviewed literature in 2021. Summary: Members of the Houston Infectious Diseases Network (HIDN) nominated articles that were deemed to have significant contributions to ID pharmacotherapy in 2021. These nominations included articles pertaining to both general ID, including coronavirus disease 2019 (COVID-19), and human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS) pharmacotherapy. A total of 35 articles were nominated by HIDN: 30 articles pertaining to general ID pharmacotherapy and 5 articles with HIV/AIDS focus. To select the most influential articles of 2021, a survey was created and distributed to members of the Society of Infectious Diseases Pharmacists (SIDP). Of the 239 SIDP members who responded to the survey, there were 192 recorded votes for the top 10 general ID pharmacotherapy articles and 47 recorded votes for the top HIV/AIDS article, respectively. The top publications are summarized. Conclusion: Antimicrobial stewardship and the optimal management of infectious disease states continues to be a priority in the midst of the ongoing coronavirus disease 2019 (COVID-19) global pandemic. In light of the sheer volume of ID-related articles published in the past year, this review aims to aid clinicians in remaining up-to-date on key practice-changing ID pharmacotherapy publications from 2021.
Article
Full-text available
Latently infected cells harboring replication‐competent proviruses represent a major barrier to HIV‐1 cure. One major effort to purge these cells has focused on developing the “shock and kill” approach for forcing provirus reactivation to induce cell killing by viral cytopathic effects, host immune responses, or both. We conducted kinetic and mechanistic studies of HIV‐1 protein expression, virion production, and cell‐to‐cell virus transmission during provirus reactivation. Provirus‐activated ACH‐2 cells stimulated with romidepsin (RMD) or PMA produced Nef early, and then Env and Gag in parallel with the appearance of virions. Env on the surface of provirus‐activated cells and cellular F‐actin were critical in the formation of virological synapses to mediate cell‐to‐cell transmission of HIV‐1 from provirus‐activated cells to uninfected cells. This HIV‐1 cell‐to‐cell transmission was substantially more efficient than transmission seen via cell‐free virus spread and required F‐actin remodeling and CD4, but not chemokine receptors. Resting human primary CD4+ T cells including naïve and memory subpopulations and, especially the memory CD4+ T cells, were highly susceptible to HIV‐1 infection via cell‐to‐cell transmission. Cell‐to‐cell transmission of HIV‐1 from provirus‐activated cells was profoundly decreased by protease inhibitors (PIs) and neutralizing antibodies (nAbs) that recognize the CD4‐binding site (CD4bs) such as VRC01, but not by reverse transcriptase (RT) inhibitor Emtricitabine (FTC). Therefore, our results suggest that PIs with potent blocking abilities should be used in clinical application of the “shock and kill” approach, most likely in combination with CD4bs nAbs, to prevent new HIV‐1 infections. This article is protected by copyright. All rights reserved.
Chapter
The chapter reconsiders the idea of PrEP is a programme rather than simply a pill. It explores the meaning of PrEP is a programme by reconsidering what exactly PrEP trials demonstrate in terms of efficacy and what that efficacy means for getting that result to work as policy in non-trial contexts. The chapter introduces an analytical approach, installation theory, to guide a way of thinking about PrEP as a policy in different contexts.
Article
Adolescent girls and young women (aged 15–24 years; AGYW) continue to carry a disproportionate burden of HIV in sub‐Saharan Africa. Pre‐exposure prophylaxis (PrEP) helps reduce the risk of acquiring HIV for persons at substantial risk, including AGYW. As countries plan for the rollout of PrEP across sub‐Saharan Africa, PrEP policies and programs could address the unique needs of AGYW. The purpose of this analysis was to identify policy considerations to improve AGYW access to PrEP. After reviewing the literature, we identified 13 policy considerations that policymakers and stakeholders could evaluate when developing or reviewing PrEP‐related policies. We sorted these considerations into five categories, which together comprise an AGYW Access to PrEP Framework: AGYW‐friendly delivery systems, clinical eligibility and adherence support, legal barriers and facilitators, affordability, and community and AGYW outreach. We also reviewed policies in three countries (Kenya, South Africa, and Uganda) to explore how PrEP‐related policies addressed these considerations. Some of these policies addressed some of the 13 policy considerations, but none of the policies directly addressed the unique needs of AGYW for accessing PrEP. To improve access to PrEP for AGYW, country policies could include specific components that address these 13 considerations. To reach AGYW effectively, each country could use the 13 considerations we have identified to analyze current policies to identify existing programmatic barriers to AGYW accessing HIV services and address these barriers in PrEP‐related policies.
Article
This book combines mathematical models with extensive use of epidemiological and other data, to achieve a better understanding of the overall dynamics of populations of pathogens or parasites and their human hosts. The authors thus provide an analytical framework for evaluating public health strategies aimed at controlling or eradicating particular infections. With rising concern for programmes of primary health care against such diseases as measles, malaria, river blindness, sleeping sickness, and schistosomiasis in developing countries, and the advent of HIV/AIDS and other `emerging viruses', such a framework is increasingly important. Throughout, the mathematics is used as a tool for thinking clearly about fundamental and applied problems relating to infectious diseases. The book is divided into two major parts, one dealing with microparasites (viruses, bacteria, and protozoans) and the other with macroparasites (helminths and parasitic arthropods). Each part begins with simple models, developed in a biologically intuitive way, and then goes on to develop more complicated and realistic models as tools for public health planning. A major contribution by two of the leaders in the field, this book synthesizes previous work in this rapidly growing area with much new material, combining work scattered between the ecological and medical literature.
Article
Background: An estimated 1.2 million persons in the United States were living with human immunodeficiency virus (HIV) infection in 2008. Improving survival of persons with HIV and reducing transmission involve a continuum of services that includes diagnosis (HIV testing), linkage to and retention in HIV medical care, and ongoing HIV prevention interventions, including appropriately timed antiretroviral therapy (ART). Methods: CDC used three surveillance datasets to estimate recent HIV testing and HIV prevalence among U.S. adults by state, and the percentages of HIV-infected adults receiving HIV care for whom ART was prescribed, who achieved viral suppression, and who received prevention counseling from health-care providers. Published data were used to estimate the numbers of persons in the United States living with and diagnosed with HIV and, based on viral load and CD4 laboratory reports, linked to and retained in HIV care. Results: In 2010, 9.6% of adults had been tested for HIV during the preceding 12 months (range by state: 4.9%-29.8%). Of the estimated 942,000 persons with HIV who were aware of their infection, approximately 77% were linked to care, and 51% remained in care. Among HIV-infected adults in care, 45% received prevention counseling, and 89% were prescribed ART, of whom 77% had viral suppression. Thus, an estimated 28% of all HIV-infected persons in the United States have a suppressed viral load. Conclusions: Prevalence of HIV testing and linkage to care are high but warrant continued effort. Increasing the percentages of HIV-infected persons who remain in HIV care, achieve viral suppression, and receive prevention counseling requires additional effort. Implications for Public Health Practice: Public health officials and HIV care providers should improve engagement at each step in the continuum of HIV care and monitor progress in every community using laboratory reports of viral load and CD4 test results.
Article
BACKGROUND: Antiretroviral pre-exposure prophylaxis reduces sexual transmission of HIV. We assessed whether daily oral use of tenofovir disoproxil fumarate (tenofovir), an antiretroviral, can reduce HIV transmission in injecting drug users. METHODS: In this randomised, double-blind, placebo-controlled trial, we enrolled volunteers from 17 drug-treatment clinics in Bangkok, Thailand. Participants were eligible if they were aged 20-60 years, were HIV-negative, and reported injecting drugs during the previous year. We randomly assigned participants (1:1; blocks of four) to either tenofovir or placebo using a computer-generated randomisation sequence. Participants chose either daily directly observed treatment or monthly visits and could switch at monthly visits. Participants received monthly HIV testing and individualised risk-reduction and adherence counselling, blood safety assessments every 3 months, and were offered condoms and methadone treatment. The primary efficacy endpoint was HIV infection, analysed by modified intention-to-treat analysis. This trial is registered with ClinicalTrials.gov, number NCT00119106. FINDINGS: Between June 9, 2005, and July 22, 2010, we enrolled 2413 participants, assigning 1204 to tenofovir and 1209 to placebo. Two participants had HIV at enrolment and 50 became infected during follow-up: 17 in the tenofovir group (an incidence of 0·35 per 100 person-years) and 33 in the placebo group (0·68 per 100 person-years), indicating a 48·9% reduction in HIV incidence (95% CI 9·6-72·2; p=0·01). The occurrence of serious adverse events was much the same between the two groups (p=0·35). Nausea was more common in participants in the tenofovir group than in the placebo group (p=0·002). INTERPRETATION: In this study, daily oral tenofovir reduced the risk of HIV infection in people who inject drugs. Pre-exposure prophylaxis with tenofovir can now be considered for use as part of an HIV prevention package for people who inject drugs.
Article
The emergence and spread of high levels of HIV-1 drug resistance in resource-limited settings where combination antiretroviral treatment has been scaled up could compromise the effectiveness of national HIV treatment programmes. We aimed to estimate changes in the prevalence of HIV-1 drug resistance in treatment-naive individuals with HIV since initiation of rollout in resource-limited settings. We did a systematic search for studies and conference abstracts published between January, 2001, and July, 2011, and included additional data from the WHO HIV drug resistance surveillance programme. We assessed the prevalence of drug-resistance mutations in untreated individuals with respect to time since rollout in a series of random-effects meta-regression models. Study-level data were available for 26,102 patients from sub-Saharan Africa, Asia, and Latin America. We recorded no difference between chronic and recent infection on the prevalence of one or more drug-resistance mutations for any region. East Africa had the highest estimated rate of increase at 29% per year (95% CI 15 to 45; p=0·0001) since rollout, with an estimated prevalence of HIV-1 drug resistance at 8 years after rollout of 7·4% (4·3 to 12·7). We recorded an annual increase of 14% (0% to 29%; p=0·054) in southern Africa and a non-significant increase of 3% (-0·9 to 16; p=0·618) in west and central Africa. There was no change in resistance over time in Latin America, and because of much country-level heterogeneity the meta-regression analysis was not appropriate for Asia. With respect to class of antiretroviral, there were substantial increases in resistance to non-nucleoside reverse transcriptase inhibitors (NNRTI) in east Africa (36% per year [21 to 52]; p<0·0001) and southern Africa (23% per year [7 to 42]; p=0·0049). No increase was noted for the other drug classes in any region. Our findings suggest a significant increase in prevalence of drug resistance over time since antiretroviral rollout in regions of sub-Saharan Africa; this rise is driven by NNRTI resistance in studies from east and southern Africa. The findings are of concern and draw attention to the need for enhanced surveillance and drug-resistance prevention efforts by national HIV treatment programmes. Nevertheless, estimated levels, although increasing, are not unexpected in view of the large expansion of antiretroviral treatment coverage seen in low-income and middle-income countries--no changes in antiretroviral treatment guidelines are warranted at the moment. Bill & Melinda Gates Foundation and the European Community's Seventh Framework Programme.
Article
Preexposure prophylaxis with antiretroviral drugs has been effective in the prevention of human immunodeficiency virus (HIV) infection in some trials but not in others. In this randomized, double-blind, placebo-controlled trial, we assigned 2120 HIV-negative women in Kenya, South Africa, and Tanzania to receive either a combination of tenofovir disoproxil fumarate and emtricitabine (TDF-FTC) or placebo once daily. The primary objective was to assess the effectiveness of TDF-FTC in preventing HIV acquisition and to evaluate safety. HIV infections occurred in 33 women in the TDF-FTC group (incidence rate, 4.7 per 100 person-years) and in 35 in the placebo group (incidence rate, 5.0 per 100 person-years), for an estimated hazard ratio in the TDF-FTC group of 0.94 (95% confidence interval, 0.59 to 1.52; P=0.81). The proportions of women with nausea, vomiting, or elevated alanine aminotransferase levels were significantly higher in the TDF-FTC group (P=0.04, P<0.001, and P=0.03, respectively). Rates of drug discontinuation because of hepatic or renal abnormalities were higher in the TDF-FTC group (4.7%) than in the placebo group (3.0%, P=0.051). Less than 40% of the HIV-uninfected women in the TDF-FTC group had evidence of recent pill use at visits that were matched to the HIV-infection window for women with seroconversion. The study was stopped early, on April 18, 2011, because of lack of efficacy. Prophylaxis with TDF-FTC did not significantly reduce the rate of HIV infection and was associated with increased rates of side effects, as compared with placebo. Despite substantial counseling efforts, drug adherence appeared to be low. (Supported by the U.S. Agency for International Development and others; FEM-PrEP ClinicalTrials.gov number, NCT00625404.).