ArticlePDF Available

Macaque studies of vaccine and microbicide combinations for preventing HIV-1 sexual transmission

Authors:

Abstract and Figures

Vaccination and the application of a vaginal microbicide have traditionally been considered independent methods to prevent the sexual transmission of HIV-1 to women. Both techniques can be effective in macaque models, and limited efficacy has been observed in clinical trials for each. Here, we have addressed whether vaccines and microbicides can be used together to provide reinforced protection against virus challenge of rhesus macaques. In two separate experiments, four groups of animals were vaccinated with a T-cell-based adenovirus (Ad) vectored vaccine aimed at reducing postinfection viral loads and/or a partially effective dose of a vaginal microbicide aimed at blocking infection of a high-dose vaginal challenge with SIVmac251 or SHIV-162P3. In the first study, the only two protected animals were in the group that received Ad26/Ad5HVR48 vaccine vectors combined with the fusion inhibitor T-1249 as the vaginal microbicide before SIVmac251 challenge. In the second study, vaccination with Ad35/Ad26 vectors combined with the CCR5 inhibitor maraviroc as the vaginal microbicide led to significant reductions of both acquisition of infection and postinfection viral loads following SHIV-SF162P3 challenge. As expected, the vaccine by itself reduced viral loads but had no acquisition effect, whereas the microbicide had a partial acquisition effect but minimal impact on viral loads. For both measures of protective efficacy, the vaccine-microbicide combination differed more from controls than did either separate intervention. Overall, the data suggest that vaccines and microbicides are complementary techniques that may protect better when used together than separately.
Content may be subject to copyright.
Macaque studies of vaccine and microbicide
combinations for preventing HIV-1 sexual transmission
Dan H. Barouch
a,b,1
, Per Johan Klasse
c
, Jason Dufour
d
, Ronald S. Veazey
d
, and John P. Moore
c,1
a
Beth Israel Deaconess Medical Center, Boston, MA 02115;
b
Ragon Institute of MGH, MIT, and Harvard, Boston, MA 02114;
c
Department of Microbiology and
Immunology, Weill Cornell Medical College, New York, NY 10021; and
d
Tulane National Primate Research Center, Tulane University School of Medicine,
Covington, LA 70433
Edited* by Robert C. Gallo, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, and approved April 12, 2012 (received for
review February 22, 2012)
Vaccination and the application of a vaginal microbicide have
traditionally been considered independent methods to prevent
the sexual transmission of HIV-1 to women. Both techniques can
be effective in macaque models, and limited efcacy has been ob-
served in clinical trials for each. Here, we have addressed whether
vaccines and microbicides can be used together to provide rein-
forced protection against virus challenge of rhesus macaques. In
two separate experiments, four groups of animals were vacci-
nated with a T-cellbased adenovirus (Ad) vectored vaccine aimed
at reducing postinfection viral loads and/or a partially effective
dose of a vaginal microbicide aimed at blocking infection of a
high-dose vaginal challenge with SIVmac251 or SHIV-162P3. In
the rst study, the only two protected animals were in the group
that received Ad26/Ad5HVR48 vaccine vectors combined with
the fusion inhibitor T-1249 as the vaginal microbicide before SIV-
mac251 challenge. In the second study, vaccination with Ad35/
Ad26 vectors combined with the CCR5 inhibitor maraviroc as the
vaginal microbicide led to signicant reductions of both acquisi-
tion of infection and postinfection viral loads following SHIV-
SF162P3 challenge. As expected, the vaccine by itself reduced viral
loads but had no acquisition effect, whereas the microbicide had
a partial acquisition effect but minimal impact on viral loads. For
both measures of protective efcacy, the vaccinemicrobicide com-
bination differed more from controls than did either separate in-
tervention. Overall, the data suggest that vaccines and microbicides
are complementary techniques that may protect better when used
together than separately.
HIV type 1 (HIV-1) continues to spread globally through
sexual transmission, particularly among young women in
the developing world who have little power to insist on the use of
condoms to protect themselves (1, 2). In such circumstances,
biology-based interventions become of substantial importance
(2). Traditionally, the spread of infectious disease has been most
effectively controlled by vaccination, but this approach has had,
at best, limited success against HIV-1 in efcacy trials (3, 4). New
vaccines based on live viral vectors are under evaluation in the
macaque model of HIV-1 infection, with partial success at
blocking acquisition of infection and a more consistent ability to
reduce postinfection viral loads (5, 6). However, inducing neu-
tralizing antibodies of sufcient breadth and potency to prevent
virus transmission remains a major challenge (7).
An alternative approach to HIV-1 prevention involves the
application of antiretroviral drugs (ARVs), either systemically as
oral preexposure prophylaxis (PrEP) or locally at the mucosal
sites of entry of the virus into the body (8). In the latter method,
a gel or other formulation containing an ARV is administered
vaginally or rectally as a microbicide to prevent the initial stages
of entry of HIV-1 into the body (9). Studies with oral PrEP and
vaginal microbicides have shown partial efcacy against HIV-1
sexual transmission to humans in the CAPRISA and iPREX
trials but not in other studies (1013).
Within the HIV-1 prevention elds, vaccines and microbicides
have been regarded as independent, and in some respects, rival
technologies. And yet there is logic to considering the two ap-
proaches as complementary. On a very simple level, two partially
effective barriers to infection might be superior to either one
alone. A more sophisticated argument is that, by reducing the
extent to which an incoming virus replicates in mucosal tissues,
a microbicide might buy time for the maturation of vaccine-in-
duced immune responses that either eliminate the virus or fur-
ther counter its expansion such that the viral load set point is
reduced (14, 15). Another possibility is that the combined use
of a microbicide that partially blocks acquisition of infection
with a T-cellbased vaccine that reduces postinfection viral loads
will result in both protective effects following virus challenge. We
sought to test the underlying hypotheses by carrying out two
experiments in the rhesus macaque vaginal challenge model,
using inhibitors of HIV-1 entry as the microbicides and adeno-
virus (Ad)-vectorbased vaccines.
Results
Design of Combination Studies. Both studies were of broadly sim-
ilar design. The rhesus macaques were immunized by the intra-
muscular route with an Ad-vectorbased vaccine. Eight months
after the boost immunization, a vaginally applied inhibitor was
then given as the microbicide for 30 min before vaginal simian
immunodeciency virus (SIV) or simian-human immunode-
ciency virus (SHIV) challenge. For comparison, other animals
were not vaccinated or did not receive the microbicide. In both
experiments, the macaques were treated with progesterone 30
d before challenge, to ensure that most control animals would
become infected after a single exposure to virus (16). We con-
sidered other designs, particularly for the second experiment,
such as the use of multiple challenges without prior progesterone
treatment. However, our experience has been that infection of
control animals can be highly inconsistent under such conditions,
which adversely impacts how the outcome of the study can be
interpreted (17). The Ad-vectorbased vaccines were primarily
T-cellbased vaccines aimed at reducing viral loads post-
infection; they would not be expected to block acquisition of
infection based on the high-dose challenge in the SIVmac251
study and the mismatched Env in the SHIV-162P3 study (6, 18).
We hypothesized that the acquisition and viral load effects of
each individual component would both occur when the two
methods were used together. Alternatively, a microbicide-medi-
ated reduction in the infectivity of the incoming virus, or in the rate
at which it expanded locally after deposition in the vagina, might
increase the protective capabilities of the vaccine.
A critical aspect of the experimental designwas to use a partially
protective microbicide concentration; too much inhibitor might
be so strongly protective as to prevent us from ascertaining any
Author contributions: D.H.B., P.J.K., and J.P.M. designed research; D.H.B., J.D., and R.S.V.
performed research; P.J.K. analyzed data; and D.H.B., P.J.K., and J.P.M. wrote the paper.
The authors declare no conict of interest.
*This Direct Submission article had a prearranged editor.
1
To whom correspondence may be addressed. E-mail: dbarouch@bidmc.harvard.edu or
jpm2003@med.cornell.edu.
86948698
|
PNAS
|
May 29, 2012
|
vol. 109
|
no. 22 www.pnas.org/cgi/doi/10.1073/pnas.1203183109
impact of the vaccine, but too little might have no measurable ef-
fect. Hence in preliminary experiments we titrated each microbicide
against the challenge virus in an attempt to dene a concentration
that would protect half of animals when used alone. In both
experiments, the animals were divided into four groups: Control
(i.e., no intervention), group C; vaccine only, group V; microbicide
only, group M; and vaccine + microbicide, group V+M.
Fusion-Inhibitor Microbicide and SIVmac251 Challenge. In the rst
study, the vaccine was an Ad26/Ad5HVR48 vector regimen
expressing Gag-Pol-Env-Nef (18). The microbicide was the fu-
sion inhibitory peptide, T-1249, given at 200 μg/mL (40 μM) in
4 mL of an aqueous gel 30 min before high-dose vaginal chal-
lenge with SIVmac251 (19, 20). Each group contained six ani-
mals, but one animal was omitted from group V when it was
found to be retrovirus infected before challenge. All of the
animals in groups C (n= 6) and V (n= 5) became infected, and
postinfection viral loads were consistently 10-fold lower in
vaccinees (group V) as expected (Fig. 1). This magnitude of viral
load reduction is comparable with what was observed using the
same vaccine in other challenge formats (18). Hence any effect
of progesterone treatment on vaccine efcacy and/or local im-
munity is not substantial (21, 22).
We did not achieve our intended goal of protecting half of the
animals in group M. Here, four of the animals became system-
ically viremic with typical viral load kinetics. However, the other
two did so only after a delay of 46 wk. In our experience, such a
delay is highly unusual and thus likely relates to partially pro-
tective effects of the T-1249 microbicide (Discussion). Two of the
six animals in the V+M group remained uninfected throughout
the period of monitoring (Fig. 1). This outcome was not statis-
tically signicant for acquisition (V+M vs. C, P= 0.23). However,
as the only two animals to remain uninfected in this experiment
were in the V+M combination, we considered that the data
pattern was sufciently encouraging as to warrant a further ex-
ploration of the vaccinemicrobicide combination concept.
CCR5-Inhibitor Microbicide and SHIV-SF162P3 Challenge. In the sec-
ond study, we changed the vaccine to the more potent Ad35/
Ad26 vector regimen expressing SIVsmE543 Gag-Pol-Env (6).
We also switched the microbicide to the CCR5 inhibitor mar-
aviroc as, unlike T-1249, this drug is used for treating HIV-1
infected people and is currently being evaluated for preventing
transmission (23, 24). Our original intent was to continue to use
SIVmac251 as the challenge virus; accordingly, the vaccine in-
cluded an SIV Env component. However, during the period
between vaccination and challenge, we concluded that we could
not reliably identify a maraviroc dose that provided 50% pro-
tection against SIVmac251. In contrast, we obtained a smooth
doseresponse curve for maraviroc-mediated protection against
SHIV-162P3 and decided to use this challenge virus instead
(24). We also considered adopting a multiple challenge protocol
without progesterone, but were unable to obtain a sufciently
consistent rate of infection of control animals under these con-
ditions (17). Given the necessity to infect almost all of the control
animals and approximately half of those in the microbicide-only
group, we elected to proceed with a single, high-dose challenge
with SHIV-162P3. Thus, the Env component of the vaccine (SIV)
and the challenge virus (HIV-1) were mismatched.
As a result, we predicted that the vaccine would reduce viral
loads but would not block acquisition of infection following virus
challenge (6). The study therefore allowed us to assess whether
the combined use of the partially protective microbicide and the
T-cellbased vaccine would prove superior to each individual
modality by affording both protection against acquisition of in-
fection and improved virologic control. The basic predictions
were that: (i) V+M would prove superior to V alone for blocking
acquisition of infection and (ii) V+M would prove superior to M
alone for virologic control. A more tangible advantage to the
combination would be evident if V+M also protected better
against acquisition than M and curbed viral loads more than V.
Each group in this study included eight animals to increase
statistical power, although one macaque was omitted from the
combination group when it was found to be retrovirus infected
Fig. 1. Viral load proles for animals given vaccine (V), T-1249 (M), the combination (V+M), or neither (C) before challenge with SIVmac251. The viral load
over time after challenge is shown in one diagram for each group (n= 5 for V; n= 6 for each of the other groups). Baseline values are offset from each other
for clarity.
Barouch et al. PNAS
|
May 29, 2012
|
vol. 109
|
no. 22
|
8695
MEDICAL SCIENCES
before challenge. The immunogenicity of the Ad35/Ad26 vac-
cine, as judged by IFN-γELISPOT assays, was comparable to
that observed previously in experiments involving the same
vaccine but different challenge conditions, e.g., intrarectal with
SIVmac251 (6).
After the high-dose vaginal SHIV-162P3 challenge (Fig. 2),
seven of eight control animals (group C) were infected. Similarly,
seven of eight animals in the vaccine-only group (group V) were
infected, as expected for this T-cellbased vaccine with no rele-
vant Env component. In the microbicide-only group (group M),
four of eight animals were infected. Thus, we achieved our goal
of using a microbicide dose that protected half of the animals. In
the combination group (group V+M), two of seven animals were
infected. The intervention efcacies in groups M and V+M were
43 and 67%, respectively. For V+M compared with V or C, the
infection rate was signicantly lower (P= 0.035), although the
rate for group M was not signicantly lower than for group C
(P= 0.14). V+M was, therefore, superior to V for blocking
acquisition of infection, in accordance with our rst prediction.
Although the infection rate was lowest in the combination group,
V+M was not, however, signicantly different from M for ac-
quisition (P= 0.38).
Plasma viral loads were assessed to determine postinfection
virologic control in the infected animals. Viral loads in the four
groups differed in terms of peak viral loads (Fig. 3A;P= 0.0006,
V vs. C, P<0.01; V+M vs. M, P<0.05; and V+M vs. C, P<
0.001), and of viral loads on day 28, which was the rst day on
which all infected monkeys had detectable viremia (Fig. 3B;P=
0.0001, V vs. C, P<0.01; V+M vs. M, P<0.01; and V+M vs. C,
P<0.001). The log viral load reductions measured as peaks and
on day 28 were, respectively, 1.2 and 1.1 for V; 0.70 and 0.45 for
M; and 2.4 and 2.3 for V+M. The magnitude of the viral load
reduction by the vaccine was again similar to what we have
reported in previous experiments with the Ad35/Ad26 vaccine
(6). Hence V+M was superior to M for virologic control, in
accordance with our second prediction.
Furthermore, the viral loads in the V+M group were the
lowest among the four groups in both comparisons and they
differed more from controls than did either V or M.
Discussion
In this study, we used a rhesus macaque vaginal transmission
model to test the concept that vaccines and microbicides are two
prevention methods that might work better together than in-
dividually. In both experiments, the fewest infected animals were
found in the V+M combination group, and the difference in
acquisition between the V and V+M groups was statistically
signicant in the second experiment. Moreover, the difference in
viral loads between groups M and V+M was statistically signif-
icant in the second experiment. Thus, these data provide overall
support to the concept that combining vaccines and microbicides
may be superior to using either by itself. Whether a vaccine
containing a relevant Env component combined with a micro-
bicide might provide synergistic protection against acquisition of
infection remains to be determined.
We observed a trend toward augmented protection against
acquisition of infection in the V+M group, compared with group
M, in both studies (two of six vs. zero of six animals uninfected
for V+M vs. M in the rst study, ve of seven vs. four of eight
animals in the second). One interpretation of this trend is that
the partially protective microbicide dose slowed down the local
expansion of the incoming virus in the mucosal tissues of a subset
of the animals, and hence the effective size of the inoculum (see
below). We note that, in the rst experiment, two animals in
group M (i.e., T-1249 recipients) become infected only after an
unusually long delay of 24 wk (Fig. 1). We have only previously
seen such a long delay in a single animal, one that was also given
a partially protective dose of T-1249 before SIVmac251 chal-
lenge. In the combination groups, we hypothesize that the
microbicide-mediated delay in virus expansion may have allowed
vaccine-elicited immune responses enough additional time to
either eliminate the infection entirely or to restrict replication
Fig. 2. Viral load proles for animals given vaccine (V), maraviroc (M), the combination (V+M), or neither (C) before challenge with SHIV-162P3. The viral load
over time after challenge is shown in one diagram for each group (n= 7 for V+M; n= 8 for each of the other groups). Baseline values are offset from each
other for clarity.
8696
|
www.pnas.org/cgi/doi/10.1073/pnas.1203183109 Barouch et al.
and lower the set point. In the second experiment, maraviroc
may reduce the number of target cells in the vaginal mucosa that
express sufcient free CCR5 for virus entry, noting that the
abundance of CCR5
+
target cells is an important inuence on
the establishment of infection (25). We think it is unlikely that
vaccine-induced antibodies are involved in these effects, partic-
ularly given the complete mismatch of the Env component of the
vaccine and challenge virus in the second study. Instead, cellular
immunity, such as CD8
+
T-cell responses, is likely responsi-
ble (6). Consistent with this possibility is our observation that
these Ad vectors, delivered by the intramuscular route to rhesus
macaques, elicit potent and durable effector memory CD8
+
T
lymphocytes at mucosal surfaces, including in the cervicovaginal
mucosa (26). We note that if a vaccinemicrobicide combination
approach were used in humans, the Env components of the
vaccine strain and the incoming virus would probably also be
mismatched, but not to the same extent as in our experiment
(SIV vs. HIV-1). Accordingly, at least some additional protective
contribution from the Env component of the vaccine could
reasonably be anticipated (6).
The combined effects of the vaccine and the microbicide on
acquisition and viral loads are compatible with our emerging
understanding of the early events in mucosal SIV/SHIV infection
(15). In a simple model, the crucial event is the productive in-
fection of the rst target cell, with successive generations of
progeny virus then irrevocably colonizing the rest of the organism.
However, postexposure prophylaxis with a reverse-transcriptase
inhibitor can be started as late as 2448 h after inoculation and
still completely prevent SIV infection (27), and the earliest foci of
infection appear during the rst few days after intravaginal in-
oculation (28). A more sophisticated view of mucosal infection is
that the events preceding systemic breakout involve a branching
process (29); a limited number of replication foci are rst estab-
lished but they can be extinguished in a critical labile phase before
they spread the virus to increasingly distant lymphoid tissue sites
(15). Sometimes, the initial foci may be eradicated by CD8
+
T
lymphocytes within the mucosa, leading to a failed infection at
the level of the whole organism (30). This benecial outcome may
be more probable when the inoculum size is low or is lowered by
an intervention (e.g., a topically or orally delivered ARV). We
therefore suggest that, in our experiments, the partially active
microbicide doses reduced the infectivity of the inoculum to this
labile zone where the outcome of infection could go either way
(systemic breakout or extinction), perhaps stochastically. The
vaccine-induced immune responses may therefore contribute to
reducing acquisition when the number of infectious foci is close
to the critical threshold level.
In the only previous study of combining a vaccine (DNA/
rAd5) and a microbicide (a zinc-nger inhibitor) in macaques,
there were modest differences in the rate of infection and viral
loads between the combination and the no-intervention control
group (31). However, as neither the vaccine nor the microbicide
caused detectable reductions in viral loads or acquisition, re-
spectively, the outcome was inconclusive as to the benetof
the combination; the KaplanMeier curves for the vaccine and
combination groups were largely superimposed, as were those
for the microbicide and control groups (31). Any interpretation
is further complicated by two escalations of the challenge virus
dose [eventually to 3,000 TCID
50
(50% tissue culture infectious
dose)] to try to overcome inconsistent infections of the control
animals (17, 31).
In summary, our data support the concept that vaccines and
microbicides might be useful when used in combination for
protection against sexual HIV-1 transmission to women by the
vaginal route. By extrapolation, the same benets might apply to
rectal microbicides for men and women, although this supposi-
tion will need to be conrmed experimentally given that trans-
mission conditions may be tissue dependent. Further research in
these areas is important for the HIV-1 prevention eld, because
if microbicides do become licensed for clinical use they would
likely be used in the context of HIV-1 vaccine efcacy trials.
Although we used inhibitors of virus-cell entry as the micro-
bicides, blockers of other stages in the HIV-1 replication cycle
might have similar effects and thus warrant further investigation
(9, 10). Any benecial effect of a microbicide on vaccine efcacy
would, of course, only come into play when the product is ac-
tually used but does not fully prevent HIV-1 transmission.
Microbicide failures due to nonuse require different solutions
such as sustained release devices (vaginal rings) or long-lasting
gels (3234). Whether these coitally independent delivery
methods, and also orally delivered ARVs, can be combined with
vaccination for reinforced protection remains to be determined.
A conceptually related combination approach would be to use
a vaccine designed to induce mucosal antibodies of appropriate
quality and quantity to block acquisition of infection together
with a T-cell vaccine that limits replication postinfection. All of
these hypotheses could, in principle, be tested in the macaque
model using various challenge formats. The availability of larger
numbers of animals would increase the statistical power of tests
of vaccinemicrobicide combinations to reduce acquisition,
particularly when the microbicide itself has, by design, a partial
effect by itself, as was the case in our second experiment.
Materials and Methods
Challenge Viruses. The R5 virus SHIV-162P3, derived from the HIV-1 SF162
primary isolate as described elsewhere (35) and propagated in phytohe-
magglutin in (PHA)-activated rhesus macaque peripheral blood mono-
nuclear cells (PBMC), was obtained through the National Institutes of Health
(NIH) AIDS Research and Reference Reagent Program, Division of AIDS,
National Institute of Allergy and Infectious Diseases, NIH (cat. no. 6526;
contributors: Janet Harouse, Cecilia Cheng-Mayer, and Ranajit Pal). The
SIVmac251 stock we used has been described previously (18).
Fig. 3. Peak and set-point viral loads for animals given vaccine (V), maraviroc (M), the combination (V+M), or neither (C) before challenge with SHIV-162P3.
Measurements of viral loads are depicted on the yaxis for each of the four groups as indicated on the category axis. Means are marked by horizontal bars ±
SEM for (A) the peak viral loads and (B) the viral loads on day 28, the rst day on which all of the animals that became infected were detectably viremic.
Barouch et al. PNAS
|
May 29, 2012
|
vol. 109
|
no. 22
|
8697
MEDICAL SCIENCES
Microbicides. T-1249 was a gift from Michael Greenberg and Dani Bolognesi
who, at the time, worked for Trimeris (Durham, NC) (19, 20). Maraviroc
was obtained via John Pottage (ViiV Healthcare, Research Triangle Park, NC)
(23, 24).
Vaccine Delivery. Ad26, Ad35, and Ad5HVR48 vectors expressing SIV Gag/Pol/
Env antigens were produced as previously described (36, 37). Indian-origin
rhesus monkeys were immunized twice by the intramuscular route with 10
10
viral particles per vector in Ad26/Ad5HVR48 or Ad35/Ad26 heterologous
prime-boost regimens (6). Vaccine immunogenicity was conrmed by IFN-γ
ELISPOT assays. Viral challenges were performed at least 6 mo following the
nal vaccination.
Microbicide Delivery and Virus Challenge. A single intramuscular injection
of Depo-Provera (progesterone) was given to female Indian rhesus macaques
30 d before challenge, to synchronize the menstrual cycle, thin the vaginal
epithelium, and facilitate virus transmission (16). Virus challenge and
microbicide delivery protocols are more fully described elsewhere (20, 24,
38). On the day of challenge, 4 mL of the microbicide formulated in a
hydroxyethylcellulose (HEC) gel, or a placebo HEC gel, were applied atrau-
matically to the vagina, 30 min before SHIV-162P3 or SIVmac251 was added
in a 1-mL volume containing 500 TCID
50
or 4 ×10
7
RNA copies/mL, re-
spectively. Infection status was determined by measuring plasma viral load
at 7, 14, 21, 28, 42, 56, and 70 d postchallenge, using a commercially avail-
able branched DNA (bDNA) assay with a sensitivity limit of 125 RNA copies/
mL (Siemens). All protocols were approved by the institutional animal care
and use committees. The animals were housed in accordance with the
American Association for Accreditation of Laboratory Animal Care stand-
ards. All 55 animals described in Figs. 1 and 2 were experimentally naïve at
the start of the studies and were negative for antibodies against SIV and
type D retrovirus.
Statistical Analysis. Acquisition frequencies were analyzed by Fishers exact
test. Because the interventions reduced acquisitions in the hypothesized
direction, and any marginal increase in acquisition over that for controls
(possible only in the second experiment) would have been entirely attrib-
utable to random effects, the Pvalues given are for one-tail comparisons.
The αlevel was set to P= 0.05 for all tests. Measurements of viral loads
among infected animals (peak and set points) were compared overall for the
four groups by one-way ANOVA, followed by Tukeys posttest for individual
group pairs, with Kramers extension for unequal group sizes. Although the
group sizes were too small to test for Gaussian distribution or homosce-
dasticity, we chose the parametric test because the high frequency of pro-
tection by V+M left so few infected animals that a nonparametric approach
would have been too blunt to make some important comparisons. In Results,
the Pvalue for the four-group comparison is given rst, followed by any
signicant outcome of Tukeys posttest. Thus, out of the six pairwise com-
binations of the four groups, three results by Tukeys test turned out to be
nonsignicant and are not listed. ANOVA and Tukey tests were performed
in Prism (GraphPad).
ACKNOWLEDGMENTS. We thank Dani Bolognesi and Michael Greenberg for
the provision of T-1249 and Kelsi Rasmussen, Megan Gardner, Meagan
Watkins, and Thomas Ketas for technical support. This work was funded by
a supplement to National Institutes of Health (NIH) Cooperative Agreement
U19 AI76982 and by NIH Grants AI078526 and AI066924, the Ragon Institute,
and the Bill and Melinda Gates Foundation.
1. Quinn TC, Overbaugh J (2005) HIV/AIDS in women: An expanding epidemic. Science
308:15821583.
2. Padian NS, et al. (2011) HIV prevention transformed: The new prevention research
agenda. Lancet 378:269278.
3. Kim JH, Rerks-Ngarm S, Excler JL, Michael NL (2010) HIV vaccines: Lessons learned and
the way forward. Curr Opin HIV AIDS 5:428434.
4. Barouch DH, Korber B (2010) HIV-1 vaccine development after STEP. Annu Rev Med
61:153167.
5. Hansen SG, et al. (2011) Profound early control of highly pathogenic SIV by an ef-
fector memory T-cell vaccine. Nature 473:523527.
6. Barouch DH, et al. (2012) Vaccine protection against acquisition of neutralization-
resistant SIV challenges in rhesus monkeys. Nature 482:8993.
7. Walker LM, Burton DR (2010) Rational antibody-based HIV-1 vaccine design: Current
approaches and future directions. Curr Opin Immunol 22:358366.
8. Wainberg MA (2011) AIDS: Drugs that prevent HIV infection. Nature 469:306307.
9. Minces LR, McGowan I (2010) Advances in the development of microbicides for the
prevention of HIV infection. Curr Infect Dis Rep 12:5662.
10. Abdool Karim Q, et al.; CAPRISA 004 Trial Group (2010) Effectiveness and safety of
tenofovir gel, an antiretroviral microbicide, for the prevention of HIV infection in
women. Science 329:11681174.
11. Grant RM, et al.; iPrEx Study Team (2010) Preexposure chemoprophylaxis for HIV
prevention in men who have sex with men. N Engl J Med 363:25872599.
12. Celum C, Baeten JM (2012) Tenofovir-based pre-exposure prophylaxis for HIV pre-
vention: Evolving evidence. Curr Opin Infect Dis 25:5157.
13. van der Straten A, van Damme L, Haberer JE, Bangsberg DR (2012) How well does
PREP work? Unraveling the divergent results of PrEP trials for HIV prevention. AIDS
26:F13-F19.
14. Haase AT (2005) Perils at mucosal front lines for HIV and SIV and their hosts. Nat Rev
Immunol 5:783792.
15. Haase AT (2010) Targeting early infection to prevent HIV-1 mucosal transmission.
Nature 464:217223.
16. Marx PA, et al. (1996) Progesterone implants enhance SIV vaginal transmission and
early virus load. Nat Med 2:10841089.
17. Veazey RS, Shattock RJ, Klasse PJ, Moore JP (2012) Animal models for microbicide
studies. Curr HIV Res 10:7987.
18. Liu J, et al. (2009) Immune control of an SIV challenge by a T-cell-based vaccine in
rhesus monkeys. Nature 457:8791.
19. Eron JJ, et al. (2004) Short-term safety and antiretroviral activity of T-1249, a second-
generation fusion inhibitor of HIV. J Infect Dis 189:10751083.
20. Veazey RS, et al. (2008) Tropism-independent protection of macaques against vaginal
transmission of three SHIVs by the HIV-1 fusion inhibitor T-1249. Proc Natl Acad Sci
USA 105:1053110536.
21. Genescà M, McChesney MB, Miller CJ (2010) Depo-provera treatment does not ab-
rogate protection from intravenous SIV challenge in female macaques immunized
with an attenuated AIDS virus. PLoS ONE 5:e9814.
22. Sanders-Beer B, et al. (2010) Depo-Provera does not alter disease progression in
SIVmac-infected female Chinese rhesus macaques. AIDS Res Hum Retroviruses 26:
433443.
23. MacArthur RD, Novak RM (2008) Reviews of anti-infective agents: Maraviroc: The rst
of a new class of antiretroviral agents. Clin Infect Dis 47:236241.
24. Veazey RS, et al. (2010) Protection of rhesus macaques from vaginal infection by
vaginally delivered maraviroc, an inhibitor of HIV-1 entry via the CCR5 co-receptor.
J Infect Dis 202:739744.
25. Pandrea I, et al. (2012) Mucosal SIV transmission in African Green Monkeys: Suscep-
tibility to infection is proportional to target cell availability at mucosal sites. J Virol.
26. Li H, et al. (2011) Durable mucosal simian immunodeciency virus-specic effector
memory T lymphocyte responses elicited by recombinant adenovirus vectors in rhesus
monkeys. J Virol 85:1100711015.
27. Tsai CC, et al. (1998) Effectiveness of postinoculation (R)-9-(2-phosphonylmethox-
ypropyl) adenine treatment for prevention of persistent simian immunodeciency
virus SIVmne infection depends critically on timing of initiation and duration of
treatment. J Virol 72:42654273.
28. Miller CJ, et al. (2005) Propagation and dissemination of infection after vaginal
transmission of simian immunodeciency virus. J Virol 79:92179227.
29. Wick D, Self SG (2000) Early HIV infection in vivo: Branching-process model for
studying timing of immune responses and drug therapy. Math Biosci 165:115134.
30. Blancou P, et al. (2001) The inltration kinetics of simian immunodeciency virus-
specic T cells drawn to sites of high antigenic stimulation determines local in vivo
viral escape. Proc Natl Acad Sci USA 98:1323713242.
31. Cheng-Mayer C, et al. (2011) Delay of SHIV infection and control of viral replication in
vaccinated macaques challenged in the presence of a topical microbicide. AIDS 25:
18331841.
32. Malcolm RK, Edwards KL, Kiser P, Romano J, Smith TJ (2010) Advances in microbicide
vaginal rings. Antiviral Res 88(Suppl 1):S30S39.
33. Forbes CJ, et al. (2011) Non-aqueous silicone elastomer gels as a vaginal microbicide
delivery system for the HIV-1 entry inhibitor maraviroc. J Control Release 156:
161169.
34. Malcolm RK, et al. (2012) Sustained release of the CCR5 inhibitors CMPD167 and
maraviroc from vaginal rings in rhesus macaques. Antimicrob Agents Chemother 56:
22512258.
35. Harouse JM, et al. (2001) Mucosal transmission and induction of simian AIDS by CCR5-
specic simian/human immunodeciency virus SHIV(SF162P3). J Virol 75:19901995.
36. Abbink P, et al. (2007) Comparative seroprevalence and immunogenicity of six rare
serotype recombinant adenovirus vaccine vectors from subgroups B and D. J Virol 81:
46544663.
37. Roberts DM, et al. (2006) Hexon-chimaeric adenovirus serotype 5 vectors circumvent
pre-existing anti-vector immunity. Nature 441:239243.
38. Veazey RS, et al. (2005) Protection of macaques from vaginal SHIV challenge by
vaginally delivered inhibitors of virus-cell fusion. Nature 438:99102.
8698
|
www.pnas.org/cgi/doi/10.1073/pnas.1203183109 Barouch et al.
... This has led to alternative antigen design and delivery approaches including, among others, sequential multi-immunogen strategies with germline targeting [4,5]; B cell lineage-based vaccine design [6]; formulations for sustained priming of germinal centers (such as osmotic pumps [7], nanomaterials [8], or microneedle patches [9,10]); the use of cytomegalovirus as viral vectors, which have shown long-term efficacy in macaques [11,12]; and vaccine and non-vaccine pre-exposure prophylaxis combinatorial regimes, currently tested in the PrEPVacc trial (ClinicalTrials.gov Identifier: NCT04066881) following proof-of-concept in macaque studies [13][14][15]. Despite mosaic antigen vaccines showing promising results in macaques [16], the HVTN 705/HPX2008/Imbokodo study (ClinicalTrials.gov ...
Article
Full-text available
Non-human primates (NHPs) remain the most relevant challenge model for the evaluation of HIV vaccine candidates; however, discrepancies with clinical trial results have emphasized the need to further refine the NHP model. Furthermore, classical evaluation of vaccine candidates is based on endpoints measured systemically. We assessed the mucosal responses elicited upon vaccination with ALVAC and AIDSVAX using ex vivo Rhesus macaque mucosal tissue explant models. Following booster immunization with ALVAC/AIDSVAX, anti-gp120 HIV-1CM244-specific IgG and IgA were detected in culture supernatant cervicovaginal and colorectal tissue explants, as well as systemically. Despite protection from ex vivo viral challenge, no neutralization was observed with tissue explant culture supernatants. Priming with ALVAC induced distinct cytokine profiles in cervical and rectal tissue. However, ALVAC/AIDSVAX boosts resulted in similar modulations in both mucosal tissues with a statistically significant decrease in cytokines linked to inflammatory responses and lymphocyte differentiation. With ALVAC/AIDSVAX boosts, significant correlations were observed between cytokine levels and specific IgA in cervical explants and specific IgG and IgA in rectal tissue. The cytokine secretome revealed differences between vaccination with ALVAC and ALVAC/AIDSVAX not previously observed in mucosal tissues and distinct from the systemic response, which could represent a biosignature of the vaccine combination.
... Among them are envelope inhibitors such as griffithsin (GRFT) and cyanovirin-N (CV-N) (K. B. Alexandre et al., 2012), the reverse transcriptase inhibitor tenofovir, the CCR5 inhibitor maraviroc (Barouch et al., 2012); and an extract of the plant Cistus incanus (Rebensburg et al., 2016). Human clinical trials with HIV-1 microbicide candidates have shown varying levels of success; in some trials they prevented transmission of the virus while in others they were not effective (Abdool Karim et al., 2011;Marrazzo et al., 2015). ...
Article
Ethnopharmacological relevance There have been different methods proposed to prevent the sexual transmission of HIV-1 and many of them have centered on the use of anti-retrovirals as microbicides. Given that a large section of the African population still relies on herbal medicine, Lobostemon trigonus (L. trigonus), a traditionally used medicinal plant in South Africa to treat HIV-1 was further investigated for its potential as a natural microbicide to prevent the sexual transmission of HIV-1. Methods The aerial parts of L. trigonus were oven-dried at 80°C, ground, extracted with boiling water for 30 minutes and then filtered. The aqueous extract produced was then bioassayed using different HIV-1 inhibition assays. The active components were purified and chemically profiled using ultra-performance liquid chromatography/quadrupole time-of flight mass spectrometry (UPLC-qTOF-MS). The mechanism of HIV-1 inhibition was determined by fusion arrest assay and time of addition assay. Molecular modelling and molecular dynamic simulations, using Schrödinger, were used to better understand the molecule’s mechanism of entry inhibition by evaluating their docking affinity and stability against the gp120 of HIV-1. Results The aqueous extract of this plant had a broad spectrum of activity against different subtypes of the virus; neutralizing subtype A, B and C in the TZM-bl cells, with IC50 values ranging from 0.10 to 7.21μg/mL. The extract was also inhibitory to the virus induced cytopathic effects in CEM-SS cells with an EC50 of 8.9μg/mL; it also inhibited infection in peripheral blood mononuclear cells (PBMC) and macrophages with IC50 values of 0.97 and 4.4μg/mL, respectively. In the presence of vaginal and seminal simulants, and in human semen it retained its inhibitory activity albeit with a decrease in efficiency, by about 3-fold. Studies of the mode of action suggested that the extract blocked HIV-1 attachment to target cells. No toxicity was observed when the Lactobacilli strains, L. acidophilus, L. jensenii, and L. crispatus that populate the female genital tract were cultured in the presence of L. trigonus extract. UPLC-qTOF-MS analyses of the purified fraction of the extract, confirmed the presence of six compounds of which four were identified as rosmarinic acid, salvianolic acids B and C and lithospermic acid. The additional molecular dynamic simulations provided further insight into the entry inhibitory characteristics of salvianolic acid B against the HIV-1 gp120, with a stable pose being found within the CD4 binding site. Conclusion The data suggests that the inhibitory effect of L. trigonus may be due to the presence of organic acids which are known to possess anti-HIV-1 properties. The molecules salvianolic acids B and C have been identified for the first time in L. trigonus species. Our study also showed that the L. trigonus extract blocked HIV-1 attachment to target cells, and that it has a broad spectrum of activity against different subtypes of the virus; thus, justifying further investigation as a HIV-1 microbicide.
... Although prophylactic vaccines have been considered the best strategy for controlling the HIV pandemic, combined implementation of vaccination and vaginal microbicide as preexposure prophylaxis have suggested improved defense against SIV or SHIV exposures (7,23). Delayed SHIV acquisition and viremia control were also obtained by combined SIV Gag/Pol DNA prime/ adenovirus type 5 (Ad5)gag/pol boost immunization and vaginal administration of a 0.1% SAMT-247 gel formulation prior to vaginal SHIV challenge (24). ...
Article
Because of microbicide noncompliance and lack of a durable, highly effective vaccine, a combined approach might improve HIV prophylaxis. We tested whether a vaccine-microbicide combination would enhance protection against SIV infection in rhesus macaques. Four macaque groups included vaccine only, vaccine-microbicide, microbicide only, and controls. Vaccine groups were primed twice mucosally with replicating adenovirus type 5 host range mutant SIV env/rev, gag, and nef recombinants and boosted twice i.m. with SIV gp120 proteins in alum. Controls and the microbicide-only group received adenovirus type 5 host range mutant empty vector and alum. The microbicide was SAMT-247, a 2-mercaptobenzamide thioester that targets the viral nucleocapsid protein NCp7, causing zinc ejection and preventing RNA encapsidation. Following vaccination, macaques were challenged intravaginally with repeated weekly low doses of SIVmac251 administered 3 h after application of 0.8% SAMT-247 gel (vaccine-microbicide and microbicide groups) or placebo gel (vaccine-only and control groups). The microbicide-only group exhibited potent protection; 10 of 12 macaques remained uninfected following 15 SIV challenges. The vaccine-only group developed strong mucosal and systemic humoral and cellular immunity but did not exhibit delayed acquisition compared with adjuvant controls. However, the vaccine-microbicide group exhibited significant acquisition delay compared with both control and vaccine-only groups, indicating further exploration of the combination strategy is warranted. Impaired protection in the vaccine-microbicide group compared with the microbicide-only group was not attributed to a vaccine-induced increase in SIV target cells. Possible Ab-dependent enhancement will be further investigated. The potent protection provided by SAMT-247 encourages its movement into human clinical trials.
... The SIVmac251 rhesus macaque model of HIV-1 infection and pathogenesis has been one of the most frequently used models to study HIV-1 viral pathogenesis, vaccine research, drug development, and eradication (27)(28)(29)(30)(31)(32)(33)(34). The model recapitulates HIV-1 infection and progression to AIDS in humans as well as infection of myeloid cells in the CNS (35)(36)(37). ...
Article
Full-text available
This study provides further evidence that the latent reservoir is comprised of both CD4 ⁺ T cells and myeloid cells. The data presented here suggest that CD4 ⁺ T cells and macrophages found throughout tissues in the body can contain replication-competent SIV and contribute to rebound of the virus after treatment interruption. Additionally, we have shown that monocytes in blood contain latent virus and, though not considered a reservoir themselves due to their short life span, could contribute to the size of the latent reservoir upon entering the tissue and differentiating into long-lived macrophages. These new insights into the size and location of the SIV reservoir using a model that is heavily studied in the HIV field could have great implications for HIV-infected individuals and should be taken into consideration with the development of future HIV cure strategies.
Article
Full-text available
Adenovirus was first identified in the 1950s and since then this pathogenic group of viruses has been explored and transformed into a genetic transfer vehicle. Modification or deletion of few genes are necessary to transform it into a conditionally or non-replicative vector, creating a versatile tool capable of transducing different tissues and inducing high levels of transgene expression. In the early years of vector development, the application in monogenic diseases faced several hurdles, including short-term gene expression and even a fatality. On the other hand, an adenoviral delivery strategy for treatment of cancer was the first approved gene therapy product. There is an increasing interest in expressing transgenes with therapeutic potential targeting the cancer hallmarks, inhibiting metastasis, inducing cancer cell death or modulating the immune system to attack the tumor cells. Replicative adenovirus as vaccines may be even older and date to a few years of its discovery, application of non-replicative adenovirus for vaccination against different microorganisms has been investigated, but only recently, it demonstrated its full potential being one of the leading vaccination tools for COVID-19. This is not a new vector nor a new technology, but the result of decades of careful and intense work in this field.
Preprint
Full-text available
Human immunodeficiency virus (HIV) infection continues to promote neurocognitive impairment, mood disorders, and brain atrophy even in the modern era of viral suppression. Brain lipids are vulnerable to HIV-associated energetic strain and contribute to HIV-associated neurologic dysfunction due to alterations in lipid breakdown and structural lipid composition. HIV neuropathology is region dependent, yet there has not been comprehensive characterization of the spatial heterogeneity of brain lipids during infection that may impact neurologic function. To address this gap, we evaluated the spatial lipid distribution using matrix laser desorption/ionization imaging mass spectrometry (MALDI-IMS) across four brain regions (parietal cortex, midbrain, thalamus, and temporal cortex), as well as kidney for a peripheral tissue control, in a virally suppressed simian immunodeficiency virus (SIV)-infected rhesus macaque. We assessed lipids indicative of fat breakdown [acylcarnitines (CARs)] and critical structural lipids [phosphatidylcholines (PCs) and phosphatidylethanolamines (PEs)] across fatty acid chain lengths and degrees of unsaturation. CARs with very long-chain, polyunsaturated fatty acids (PUFAs) were more abundant across all brain regions than shorter chain, saturated or monounsaturated species. We observed distinct brain lipid distribution patterns for CARs and PCs. However, no clear expression patterns emerged for PEs. Surprisingly, kidney was nearly devoid of ions corresponding to PUFAs common in brain. PE’s and PC’s with PUFAs had little intensity and less density than other species and, only one CAR species was observed in kidney at high intensity. Overall, our study provides substantial evidence for persistent bioenergetic changes to the brain despite viral suppression, including region-dependent mobilization of CARs for oxidation and disparities in the presence of key phospholipids necessary for maintaining proper brain structure and function. These data indicate that region-specific interventions to restore proper lipid metabolism are essential for treating HIV neurologic disease in the era of antiretroviral therapy. Graphical Abstract
Article
The CHARM-03 study was a randomized, open label, crossover Phase 1 safety and pharmacokinetic study of oral maraviroc and maraviroc 1% Gel. At a single site, healthy HIV-uninfected men and women were enrolled and randomized to an open label crossover sequence of eight consecutive daily exposures to MVC 300 mg dosed orally, MCV 1% gel dosed rectally, and MVC 1% gel dosed vaginally. Assessments were undertaken at baseline and following each eight-day period and included collection of plasma, rectal/cervical tissue, and rectal/endocervical/vaginal fluids. Eleven men and nine women were enrolled. Two participants withdrew from the study before receiving study product. There were 25 adverse events (AEs), of which 24 were Grade 1 (G1) and one was G2 (unrelated). After eight doses, MVC was quantifiable in all samples following oral, rectal, or vaginal product administration. The highest drug concentrations in plasma, rectal tissue, and cervical tissue were associated with oral, rectal, and vaginal drug delivery respectively. There were significant reductions in tissue drug concentrations when biopsies were incubated in media prior to tissue processing for PK (p<0.0001). Only oral MVC was associated with limited protection in the rectal explant HIV challenge model (p<0.05). There were no immunological changes in rectal tissue, and all products were acceptable to participants. In conclusion, all products were found to be safe and acceptable and did not induce local inflammation. The lack of ex vivo efficacy demonstrated in study samples may be due to rapid disassociation of MVC from the explant tissue.
Preprint
Full-text available
There are no known cures or vaccines for COVID-19, the defining pandemic of this era. Animal models are essential to fast track new interventions and nonhuman primate (NHP) models of other infectious diseases have proven extremely valuable. Here we compare SARS-CoV-2 infection in three species of experimentally infected NHPs (rhesus macaques, baboons, and marmosets). During the first 3 days, macaques developed clinical signatures of viral infection and systemic inflammation, coupled with early evidence of viral replication and mild-to-moderate interstitial and alveolar pneumonitis, as well as extra-pulmonary pathologies. Cone-beam CT scans showed evidence of moderate pneumonia, which progressed over 3 days. Longitudinal studies showed that while both young and old macaques developed early signs of COVID-19, both groups recovered within a two-week period. Recovery was characterized by low-levels of viral persistence in the lung, suggesting mechanisms by which individuals with compromised immune systems may be susceptible to prolonged and progressive COVID-19. The lung compartment contained a complex early inflammatory milieu with an influx of innate and adaptive immune cells, particularly interstitial macrophages, neutrophils and plasmacytoid dendritic cells, and a prominent Type I-interferon response. While macaques developed moderate disease, baboons exhibited prolonged shedding of virus and extensive pathology following infection; and marmosets demonstrated a milder form of infection. These results showcase in critical detail, the robust early cellular immune responses to SARS-CoV-2 infection, which are not sterilizing and likely impact development of antibody responses. Thus, various NHP genera recapitulate heterogeneous progression of COVID-19. Rhesus macaques and baboons develop different, quantifiable disease attributes making them immediately available essential models to test new vaccines and therapies.
Article
Full-text available
Application of highly active antiretroviral drugs (ARDs) effectively reduces morbidity and mortality in HIV-infected individuals. However, the emergence of multiple drug-resistant strains has led to the increased failure of ARDs, thus calling for the development of anti-HIV drugs with targets or mechanisms of action different from those of the current ARDs. The first peptide-based HIV entry inhibitor, enfuvirtide, was approved by the U.S. FDA in 2003 for treatment of HIV/AIDS patients who have failed to respond to the current ARDs, which has stimulated the development of several series of protein- and peptide-based HIV entry inhibitors in preclinical and clinical studies. In this review, we highlighted the properties and mechanisms of action for those promising protein- and peptide-based HIV entry inhibitors targeting the HIV-1 gp120 or gp41 and discussed their advantages and disadvantages, compared with the current ARDs.
Article
Full-text available
Simian immunodeficiency virus (SIV) can cross the intact vaginal epithelium to establish a systemic infection in macaques (mac). Using this SIVmac model, we found that subcutaneous progesterone implants, which could mimic hormonally based contraceptives, thinned the vaginal epithelium and enhanced SIV vaginal transmission 7.7−fold over that observed in macaques treated with placebo implants and exposed to SIV in the follicular phase of the menstrual cycle. Progesterone treatment also increased the number of SIV DNA−positive cells in the vaginal lamina propria as detected by in situ polymerase chain reaction analysis. Moreover, plasma viral RNA was elevated for the first three months in macaques with progesterone implants, and three of the progesterone−treated macaques developed relatively rapid disease courses. This study shows that SIV genital infection and disease course are enhanced by subcutaneous implants containing progesterone when compared with the rate of vaginal transmission in the follicular phase.
Article
Full-text available
Antiretroviral entry inhibitors are now being considered as vaginally administered microbicide candidates for the prevention of the sexual transmission of human immunodeficiency virus. Previous studies testing the entry inhibitors maraviroc and CMPD167 in aqueous gel formulations showed efficacy in the macaque challenge model, although protection was highly dependent on the time period between initial gel application and subsequent challenge. In this paper, we describe the sustained release of maraviroc and CMPD167 from matrix-type silicone elastomer vaginal rings both in vitro and in vivo. Both inhibitors were released continuously during 28 days from rings in vitro at rates of 100 to 2,500 μg/day. In 28-day pharmacokinetic studies in rhesus macaques, the compounds were measured in the vaginal fluid and vaginal tissue; steady-state fluid concentrations were ∼106-fold greater than the 50% inhibitory concentrations (IC50s) for simian human immunodeficiency virus 162P3 inhibition in macaque lymphocytes in vitro. Plasma concentrations for both compounds were very low. The pretreatment of macaques with Depo-Provera (DP), which is commonly used in macaque challenge studies, was shown to significantly modify the biodistribution of the inhibitors but not the overall amount released. Vaginal fluid and tissue concentrations were significantly decreased while plasma levels increased with DP pretreatment. These observations have implications for designing macaque challenge experiments and also for ring performance during the human female menstrual cycle.
Article
Full-text available
African green monkeys (AGMs) are naturally infected with a simian immunodeficiency virus (SIVagm) that is nonpathogenic in its host. Although SIVagm is common and widespread, little is known about the mechanisms that govern its transmission. Since the earliest virus-host interactions may provide key insights into the nonpathogenic phenotype of SIVagm, we developed a mucosal transmission model for this virus. Using plasma from an acutely infected AGM as the virus inoculum, we exposed adult and juvenile AGMs, as well as pigtailed macaques (PTMs) as a nonnatural host control, by mucosal routes to increasing titers of virus and compared the doses needed to establish a productive infection. Four juvenile and four adult AGMs as well as two PTMs were intrarectally (IR) exposed, while two additional adult female AGMs were intravaginally (IVAG) exposed. No animal became infected following exposure to 10(5) RNA copies. Both PTMs but none of the AGMs became infected following exposure to 10(6) RNA copies. Finally, all adult AGMs and two of the four juvenile AGMs became infected following exposure to 10(7) RNA copies, acquiring either one (2 IR infected juveniles, 1 IR infected adult, 2 IVAG infected adults) or two (3 IR infected adults) transmitted founder viruses. These results were consistent with immunophenotypic data, which revealed a significant correlation between the percentage of CD4(+) T cells expressing CCR5 in the mucosa and the susceptibility to infection, in terms of both the viral dose and the numbers of transmitted founder viruses. Moreover, studies of uninfected AGMs showed that the fraction of CCR5-expressing CD4(+) T cells increased significantly with age. These results indicate that (i) AGMs are readily infected with SIVagm by both intrarectal and intravaginal routes, (ii) susceptibility to infection is proportional to the number of available CCR5(+) CD4(+) target cells in the mucosa, and (iii) the paucity of CCR5(+) CD4(+) target cells in infant and juvenile AGMs may explain the near absence of vertical transmission.
Article
Full-text available
There have been encouraging recent successes in the development of safe and effective topical microbicides to prevent vaginal or rectal HIV-1 transmission, based on the use of anti-retroviral drugs. However, much work remains to be accomplished before a microbicide becomes a standard element of prevention science strategies. Animal models should continue to play an important role in pre-clinical testing, with emphasis on safety, pharmacokinetic and efficacy testing.
Article
Full-text available
Preclinical studies of human immunodeficiency virus type 1 (HIV-1) vaccine candidates have typically shown post-infection virological control, but protection against acquisition of infection has previously only been reported against neutralization-sensitive virus challenges. Here we demonstrate vaccine protection against acquisition of fully heterologous, neutralization-resistant simian immunodeficiency virus (SIV) challenges in rhesus monkeys. Adenovirus/poxvirus and adenovirus/adenovirus-vector-based vaccines expressing SIV(SME543) Gag, Pol and Env antigens resulted in an 80% or greater reduction in the per-exposure probability of infection against repetitive, intrarectal SIV(MAC251) challenges in rhesus monkeys. Protection against acquisition of infection showed distinct immunological correlates compared with post-infection virological control and required the inclusion of Env in the vaccine regimen. These data demonstrate the proof-of-concept that optimized HIV-1 vaccine candidates can block acquisition of stringent, heterologous, neutralization-resistant virus challenges in rhesus monkeys.
Article
Preclinical studies of HIV-1 vaccine candidates have shown post-infection virologic control, but protection against acquisition of infection has to date only been reported against neutralization-sensitive virus challenges. Here we demonstrate vaccine protection against acquisition of fully heterologous, neutralization-resistant virus challenges in rhesus monkeys. Adenovirus/poxvirus and adenovirus/adenovirus vector-based vaccines expressing SIVsmE543 Gag, Pol, and Env antigens afforded per-exposure vaccine efficacy (VE) of >=80% against repetitive, intrarectal SIVmac251 challenges in rhesus monkeys. Protection against acquisition of infection required the inclusion of Env in the vaccine regimen and exhibited distinct immunologic correlates as compared with post-infection virologic control. These data demonstrate the first proof-of-concept that optimized HIV-1 vaccine candidates can block acquisition of stringent, heterologous, neutralization-resistant virus challenges in rhesus monkeys.
Article
Herpes simplex virus type 2 (HSV-2) increases the risk of HIV acquisition in men and overall CD4 T cell density in the foreskin. Using tissues obtained during routine male circumcision, we examined the impact of HSV-2 on the function and phenotype of foreskin T cells in Ugandan men. HSV-2 infection was predominantly associated with a compartmentalized increase in CCR5 expression by foreskin CD4 T cells, which may contribute to HIV susceptibility.
Article
We propose a stochastic, branching-process model of early events in vivo in human or simian immunodeficiency virus (HIV or SIV) infection and study the influence that the time of appearance of virus-specific antibodies or cytotoxic cells, or of administration of antiretroviral drugs, has on the probability of progression to a chronic infection. In some biological scenarios, our model predicts that a few days' delay in response or intervention would make little difference, while in others it would be highly deleterious. We show that prophylactic efficacy does not require perfect efficiency at neutralizing infectious virus. Data from a trial of PMPA, a potent antiretroviral drug, as post-exposure therapy for SIV infection in macaques, reported by C.-C. Tsai, P. Emau, K.E. Follis, T.W. Beck, R.E. Beneveniste, N. Bischofberger, J.D. Lifson, W.R. Morton (J. Virol. 72 (1998) 4265), provides a test of the model. We show that their observations are consistent with a branching-process without invoking supplementary viral- or host-variability. Finally, most animal trials of antiviral drugs or vaccines use very high viral inoculums; our model demonstrates that in such experiments we risk greatly underestimating the efficacy of these agents.
Article
Although the balance of recent evidence supports the efficacy of antiretroviral (ARV)-based pre-exposure prophylaxis (PrEP) against HIV-1 infection, recent negative trial results are perplexing. Of seven trials with available HIV endpoints, three different products have been tested: tenofovir 1% vaginal gel, oral tenofovir disoproxil fumarate (TDF) tablets, and TDF/emtricitabine tablets. Six of these trials were conducted exclusively in sub-Saharan Africa; all found the products to be well tolerated, and four demonstrated effectiveness. Furthermore, the HIV Prevention Trial Network (HPTN) 052 trial recently confirmed that antiretroviral treatment leads to 96% reduction in transmission to HIV-negative partners in HIV-serodiscordant couples. These results, along with human and animal data, provide substantial evidence for the efficacy of antiretroviral-based HIV prevention. Yet assessment of oral TDF/emtricitabine in the FEM-PrEP study and of oral and vaginal tenofovir in the Microbicide Trial Network (MTN)-003 trial (VOICE) was stopped for futility. How do we make sense of these discrepant results? We believe that adherence is a key factor, although it cannot be the only factor. Expanding upon a recent editorial in the Lancet, we discuss the impact of suboptimal product adherence on PrEP efficacy in the context of variable drug concentration at the exposure site, integrity of the vaginal epithelium, and the role of acute infection.