ArticlePDF Available

Comparative Evaluation of Prophylactic SIV Vaccination Modalities Administered to the Oral Cavity

Authors:

Abstract and Figures

Attempts to develop a protective HIV vaccine have had limited success, especially in terms of inducing protective antibodies capable of neutralizing different viral strains. As HIV transmission occurs mainly via mucosal surfaces, HIV replicates significantly in the gastrointestinal tract, and the oral route of vaccination is a convenient one to implement worldwide, we explored three SIV vaccine modalities administered orally and composed of SIV DNA priming with different boosting immunogens, with the goal of evaluating whether they could provide lasting humoral and cellular responses, including at mucosal surfaces that are sites of HIV entry. Twenty-four Cynomolgus macaques (CyM), were primed with replication-incompetent SIV DNA provirus and divided into three groups for the following booster vaccinations, all administered in the oral cavity: Group 1 with recombinant SIV gp140 and E. coli heat-labile toxin adjuvant dmLT, Group 2 with recombinant SIV-Oral Poliovirus (SIV-OPV), and Group 3 with recombinant SIV-MVA. Cell-mediated responses were measured using blood, lymph node, rectal and vaginal mononuclear cells. Significant levels of systemic and mucosal T-cell responses against Gag and Env were observed in all groups. Some SIV-specific plasma IgG, rectal and salivary IgA antibodies were generated, mainly in animals that received SIV DNA + SIV-MVA, but no vaginal IgA was detected. Susceptibility to infection after SIVmac251 challenge was similar in vaccinated and non-vaccinated animals, but acute infection viremia levels were lower in the group that received SIV DNA + SIV-MVA. Non-vaccinated CyM macaques maintained central memory and total CD4+ T cell levels in the normal range during the 5 months of post-infection follow-up as did the vaccinated animals, precluding evaluation of vaccine impact on disease progression. We conclude that the oral cavity vaccination tested in these regimens can stimulate cell-mediated immunity systemically and mucosally, but humoral response stimulation was limited with the doses and the vaccine platforms used.
Content may be subject to copyright.
FULL MANUSCRIPT
C
AU1 comparative Evaluation of Prophylactic SIV Vaccination
Modalities Administered to the Oral Cavity
AU2 cOmkar Chaudhary,
1,2
Lingyun Wang,
1,2
Deepanwita Bose,
1,2
Vivek Narayan,
1,2
Ming Te Yeh,
3
Angela Carville,
4
John D. Clements,
5
Raul Andino,
3
Pamela A. Kozlowski,
6
and Anna Aldovini
1,2
A
AU3 cbstract
Attempts to develop a protective HIV vaccine have had limited success, especially in terms of inducing
protective antibodies capable of neutralizing different viral strains. As HIV transmission occurs mainly via
mucosal surfaces, HIV replicates significantly in the gastrointestinal tract, and the oral route of vaccination is a
very convenient one to implement worldwide, we explored three SIV vaccine modalities administered orally
and composed of SIV DNA priming with different boosting immunogens, with the goal of evaluating whether
they could provide lasting humoral and cellular responses, including at mucosal surfaces that are sites of HIV
entry. Twenty-four Cynomolgus macaques (CyM) were primed with replication-incompetent SIV DNA pro-
virus and divided into three groups for the following booster vaccinations, all administered in the oral cavity:
Group 1 with recombinant SIV gp140 and Escherichia coli heat-labile toxin adjuvant dmLT, Group 2 with
recombinant SIV-Oral Poliovirus (SIV-OPV), and Group 3 with recombinant SIV-MVA. Cell-mediated re-
sponses were measured using blood, lymph node, rectal and vaginal mononuclear cells. Significant levels of
systemic and mucosal T-cell responses against Gag and Env were observed in all groups. Some SIV-specific
plasma IgG, rectal and salivary IgA antibodies were generated, mainly in animals that received SIV DNA +
SIV-MVA, but no vaginal IgA was detected. Susceptibility to infection after SIV
mac251
challenge was similar in
vaccinated and nonvaccinated animals, but acute infection viremia levels were lower in the group that received
SIV DNA +SIV-MVA. Nonvaccinated CyM maintained central memory and total CD4
+
T-cell levels in the
normal range during the 5 months of postinfection follow-up as did the vaccinated animals, precluding eval-
uation of vaccine impact on disease progression. We conclude that the oral cavity vaccination tested in these
regimens can stimulate cell-mediated immunity systemically and mucosally, but humoral response stimulation
was limited with the doses and the vaccine platforms used.
Keywords: AIDS, SIV vaccine, DNA vaccine, mucosal immunity
Introduction
N
AU4 catural transmission of HIV and SIV occurs pre-
dominantly via mucosal surfaces. Systemic dissemina-
tion occurs usually within a few days and, at that point, the
intestinal mucosa is a site of major virus replication and CD4
+
T-cell depletion in addition to lymphoid organs.
1–5
To control
both entry and systemic dissemination, an effective HIV
vaccine may need to stimulate both arms of the adaptive
immune system, eliciting cellular and humoral immunity
systemically as well as at mucosal surfaces. Among mucosal
routes, the oral route of vaccination is particularly appealing
because of its ease of administration, an issue particularly
important in settings with limited health care resources.
A few oral vaccines are in use: the attenuated oral poliovirus
vaccine (OPV), the most widely used, the typhoid, the rota-
virus, and three cholera vaccines, one based on a live-
attenuated organism, and live vaccines have been shown to be
1
Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA.
2
Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.
3
Department of Microbiology and Immunology, UCSF, San Francisco, California, USA.
4
Biomere, Worcester, Massachusetts, USA.
5
Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA.
6
Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans,
Louisiana, USA.
AIDS RESEARCH AND HUMAN RETROVIRUSES
Volume 00, Number 00, 2020
ªMary Ann Liebert, Inc.
DOI: 10.1089/aid.2020.0157
1
AID-2020-0157-ver9-Chaudhary_1P
Type: research-article
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 1
on average more immunogenic than vaccines based on killed
pathogens.
6–11
These vaccines are administered in the oral
cavity, but they are swallowed and induce immunity in both
the oral cavity and the GI tract. They are safe, provide a high
level of protection, and provide evidence for oral vaccination
as an important approach to disease prevention, especially
when a pathogen enters via mucosal surfaces as is the case in
the vast majority of HIV transmissions.
It is unclear to what extent oral vaccination could protect in
the context of HIV/SIV infection.
12–14
Data from mucosal
immunization in humans indicate that responses are maximal
at the site of antigen exposure and present to a lesser degree at
other mucosal sites as well, supporting the notion of limited
compartmentalization of the mucosal immune system.
15–17
Immunization at one mucosal site can lead to an immune
response at other mucosal effector sites, as immunologically
competent cells with homing receptors specific for mucosal
sites circulate among different sites, but there are differences
in the magnitude observed at different sites. The highest
antibody (Ab) titers are usually achieved at the mucosal site
of antigen exposure and decrease at distant sites.
17–21
Al-
though HIV replicates at significant levels in the intestinal
mucosa,
1,22–24
it enters the body predominantly via the gen-
ital tract or the rectum, likely requiring more disseminated
immunity than pathogens that infect exclusively orally.
12–14
OPV is one of the most successful oral vaccines currently
used in the world.
25,26
It is very safe—in extremely rare
cases, *1 in every 2.7 million first doses of the vaccine, OPV
can cause paralysis
26
- and using a HIV-recombinant OPV
could provide a cheap way to simultaneously immunize
against HIV and poliovirus. While live-attenuated OPV has
been replaced by the less efficacious but safer, inactivated
poliovirus vaccine (IPV) in most countries, it is still in use in
many countries. Previous testing of recombinant SIV-OPV,
using a mixture of 20 recombinants expressing small SIV
fragments, indicated that excellent antibody responses can be
obtained with this approach, and partial protection from in-
fection and disease was also achieved in Cynomolgus ma-
caques (CyM).
27–29
Furthermore, pre-existing immunity to
OPV did not prevent development of immunity to a recom-
binant protein subsequently delivered by OPV.
30
One of the
shortcomings of the SIV-OPV was the apparent limited
stimulation of cell-mediated immunity, at least in CyM, but
technical limitations in analysis of T-cell responses at the
time of these experiments may have affected the evaluation.
In previous preclinical trials, we achieved significant sys-
temic and mucosal T-cell responses with a SIV or SHIV
DNA-rMVA approach after oral and intestinal immuniza-
tions.
31–35
However, IgG and IgA responses were sporadic,
short lived, and usually undetectable by the day of challenge.
Differences in systemic and vaginal anti-SIV responses were
observed in animals vaccinated orally vs. intestinally, and
these immunizations provided some protection from infec-
tion and some from disease progression. Our candidate vac-
cine, given via oral routes, resulted in two significant
outcomes: intestinal immunization provided significant pro-
tection from infection but no protection from disease pro-
gression; oral cavity immunization provided significant
protection from AIDS with >50% of the animals controlling
viremia to undetectable levels after a peak of viremia.
35
We
reasoned that it could be interesting to investigate whether a
strategy that simultaneously stimulates immunity in the oral
cavity and in the intestine could provide protection bAU5
at the
level of both infection and disease progression. The OPV is
suited for this goal, as when given orally it infects cells both
in the oral cavity and in the intestine, therefore we opted to
investigate an approach based on the combination of re-
combinant OPV and DNA immunization, where the latter is
known to be effective to induce cell-mediated immunity. This
platform was compared with two platforms, SIV DNA/MVA
and SIV DNA/protein, that we previously utilized via mu-
cosal routes.
31–35
The nonhuman primate CyM species was
selected because, at the time of this study, poliovirus, which
provides the vector used in this vaccine approach, was
thought not to be able to infect Rhesus macaques (RM)
orally.
36–38
We compared the immunogenicity and protection
of oral SIV DNA vaccine regimens boosted with either the
gp140 SIV Env protein, a SIV-recombinant attenuated oral
poliovirus (SIV-OPV) expressing SIV Gag and SIV Env,
based on an attenuated OPV vector,
39
or SIV Gag, Pol, Env
rMVA. We found that the most immunogenic approach was
the SIV DNA/MVA, while the recombinant SIV DNA/OPV
did not achieve the expected humoral responses we had an-
ticipated, although it did stimulate significant cell-mediated
immunity.
Materials and Methods
Vaccine formulations, vaccination arms, and SIV
mac251
low-dose vaginal challenge
Twenty-four female CyM, used in this study, were housed
at Biomere Biomedical Research Models, Worcester, MA,
according to an approved protocol under the guidelines es-
tablished by the Animal Welfare Act and the National In-
stitute of Health Guide for the Care and Use of Laboratory
Animals, and were divided into three groups. One animal in
Group 3 died of unrelated causes during the vaccine phase
leaving 7 animals in this group. Each animal received a total
of three DNA doses on day 1, week 8, and week 16 that
consisted of 1 mg of pVacc7 DNA ( bF1
Fig. 1A). The DNA
plasmid pVacc7 used for priming is a derivative of pVacc6 in
which SIV
mac239
Env was replaced by SIV
smE543
env and
includes a full SIVmac239 genome with multiple mutations
in the NC basic domain, in the functional domains of RT, INT,
and PR, and a stop codon at the beginning of the vpr gene.
Gene expression is under the control of the CMV promoter,
replacing the 5¢LTR, while the 3¢is replaced by a poly-
adenylation signal. The DNA sequence was confirmed by se-
quencing, and the profile of viral particles produced was
evaluated by 293T transfection and subsequent Western blot
using macaque SIV-positive sera.
35
DNA was formulated in
1 mL of 20 mM DOTAP (1,2-dioleoyl-3-trimethylammonium-
propane, cholesterol (1:1)(Encapsula Nano Sciences). On
weeks 8, 16, and 24, Group 1 animals (n=8) were immunized
with 1 mg of SIV rgp140smE543 (a.a. 23–671; Genebank No.
AAC56565; Immune Technology Corp, New York, NY) and
100 lgofE. coli double mutant heat-labile toxin (dmLT)
adjuvant.
40
Group 2 (n=8) received 5 ·10
7
pfu SIV-OPV,
and Group 3 (n=8) received 5 ·10
8
pfu SIV gag,pol,env-
expressing MVA (DR2 vector created by Dr. Bernie Moss,
National Cancer Institute).
41–44
SIV-OPV was constructed by
inserting the sequence of the entire SIV gag (nucleotides
1309–2841 of SIV
mac239
) or SIV Env (nucleotides 6860–
9499)
45
in the plasmid pSabin2-eGFP,
39
replacing GFP
2 CHAUDHARY ET AL.
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 2
between two poliovirus protease cleavage sites. SIV gag and
Env proteins are cleaved from the OPV polyprotein by the
poliovirus protease and independently expressed intracellu-
larly. The two corresponding viruses were mixed in equal
amounts (2.5 ·10
7
each), and the mix was designated SIV-
OPV. SIV-OPV and SIV-MVA recombinant vaccines were
formulated in PBS in a final volume of 500 mL. Recombinant
MVA and OPV doses were selected based on what is the
optimal dose for each vector. DNA and boosting vaccines
were administered to the animals in the oral cavity, applied to
the mucosa between the gum and the cheek while the animals
were sedated. In the case of SIV-MVA, it was applied to the
oral cavity mucosa by scarification, as normally done for
poxvirus vaccination. Eight weeks after the last vaccination,
FIG. 1. Study overview and SIV-specific systemic IgG responses in vaccinated animals. (A) Vaccination scheme and
animal groups. Levels of antibodies to (B) SIV lysate and (C) SIV gp140 Env measured in plasma using ELISA. The left
panels show the concentrations in individual animals before vaccination and at weeks 26 and 28 (corresponding to 2 and 4
weeks after the last immunization). Bars denote the median. Panels on the right show the fold increase over prevaccination
level for each animal on week 26 when peak responses were typically observed.
AU11 cPostimmunization concentrations had to be
threefold higher than the preimmune concentration to be considered significant.
4C c
SIV ORAL IMMUNIZATION 3
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 3
vaccinated animals and controls were inoculated with a low
dose (*0.2 AID
50
in a RM vaginal titration, corresponding to
100 TCID
50
) of the pathogenic SIV
mac251
virus grown in RM
PBMC (a gift from Dr. Nancy Miller and Dr. Ron Desrosiers,
stock 2010 Day 8 SIV
mac251
), administered nontraumatically
with a needleless tuberculin syringe as cell-free virus in the
vagina.
46
This virus stock is a highly diverse swarm of many
different quasispecies of CCR5-tropic viruses.
47
Challenge
was repeated weekly until RT-PCR tests in two independent
laboratories were positive for virus in plasma.
Specimens and sample processing
Blood and secretions were collected 2- and 4-weeks
postvaccination, followed by monthly collection. Rectal,
vaginal, and iliac lymph node tissues were biopsied on the
day of the first vaccination and 2 weeks after each vaccina-
tion. Plasma and peripheral blood mononuclear cell (PBMCs)
were isolated from EDTA anti-coagulated whole blood ac-
cording to the instructions of the manufacturer (Amersham
Pharmacia, Uppsala, Sweden). Mononuclear cells (MNC)
from lymph nodes and mucosal tissues were obtained from
tissue biopsies. Briefly, after Telazol anesthesia, seven to
eight biopsies/animal/time points were obtained from the
rectum and vagina and lymph nodes using sterile forceps (for
rectal and vaginal) and a small pinch biopsy device (Olympus
endoscopic biopsy forceps). MNC from tissues were obtained
with mechanical dissociation using GentleMACS dissociator
(Miltenyi Biotech, Paris, France). Suspensions were passed
through a 70 mm pore size cell-strainer and washed with 10%
FBS in RPMI.
48
PBMC and MNC are cryopreserved in FBS
with 10% DMSO. Rectal, salivary and vaginal secretions
were collected using Weck-Cel sponges (Beaver Visitec,
Waltham, MA) premoistened with 50 lL of Dulbecco’s
phosphate-buffered saline as described previously.
49
Evaluation of SIV-specific IgG in plasma samples
and IgA in rectal and vaginal secretions
Antiviral and total IgA and IgG in secretions and IgG in
plasma were measured by ELISA as described.
33,34,48,49
Briefly, for the SIV ELISAs, microtiter plates were coated
overnight with 100 ng/well SIV rgp140smE543 (Immune
Technology) or 100 lL per well of 1/400 aldrithiol-2-
inactivated SIV particles (kindly provided by Dr. Jeff Lifson,
Leidos Biomedical Research, Frederick, MD) that had been
lysed with TritonX-100 detergent. Using monoclonal anti-
bodies, the SIV lysate was found to contain gp41 but not
gp120 at the 1/400 coating dilution used. For gp140 and SIV
lysate assays, the standards were pooled serum or purified
serum IgA from SIV-infected macaques, calibrated as previ-
ously described.
32
Plates were developed using biotinylated
goat antimonkey IgA or IgG (Rockland Immunochemicals,
Gaithersburg, MD), neutravidin-labeled peroxidase and tetra-
methylbenzidene substrate (SouthenBiotech, Birmingham,
AL). Before performing IgA assays, all samples were depleted
of IgG using Protein G Sepharose as described because the
goat antimonkey IgA was found to cross-react with monkey
IgG.
49
The concentration of anti-SIV IgA or IgG in each se-
cretion was subsequently divided by the concentration of total
IgA or IgG to obtain the specific activity (ng anti-SIV antibody
per lg immunoglobulin). Specific activity or concentrations of
SIV-specific IgG in plasma were considered significant if they
were three-fold greater than those in pre-immune plasma. IgG
neutralization titers were measured as a function of Tat-
induced luciferase reporter gene expression after single round
of infection in either M7-Luc or TZM-bl cells.
50
The viruses
used in the neutralization assay were a TCLA stock of SIV-
mac251 (M7-Luc assay) and SIVmac239 (TZM-bl assay).
Titers of SIV-specific neutralizing antibodies are the plasma
dilution at which relative luminescence units were reduce
50% compared with virus control wells after subtraction of
background.
Immunophenotyping and intracellular cytokine staining
10
5
MNC and 10
6
PBMCs were incubated for 14 h with
medium (Unstimulated), 1 lg/mL pools of 15-mer SIV Gag
or SIV Env peptides. Cells incubated with 10 ng/mL PMA
(4-a-phorbol 12-myristate 13-acetate; Sigma) and 1 lg/mL
ionomycin (Sigma) or without any stimulation provides re-
spectively positive and negative controls. Cultures contained
Brefeldin A (BD GolgiPlug Cat. # 555029; BD Biosciences)
and 1 mg/mL of anti-CD49d and anti-CD28. PBMCs and
MNC were washed, stained for surface markers in the dark,
followed by fixation, permeabilization. Characterization of
CD4
+
and CD8
+
T cells in PBMC was conducted according to
previously published procedures.
51
After the permeabiliza-
tion, cells were intracellularly stained for cytokine expression
with anticytokine antibodies for 1 h in the dark according to
previously described procedures.
52
The following antibodies
were used in this study: anti-CD3-pacific blue/PerCp-Cy5.5
(clone SP34–2), anti-CD4-Amcyan (clone L200), anti-CD8-
APC-Cy7 (clone RPA-T8), anti-TNFa-PE (clone MAb11),
anti-IFNc-Alexa Fluor-700 (clone B27), IL-2-APC (clone
MQ1–17H12), anti-CD95-FITC (clone DX2), and anti-
CD28-Pe-Cy5 (clone CD28.2). Gates for all antibodies were
estimated in Fluorescence Minus One (FMO) staining sam-
ples. A viability dye (VIVID, LIVE/DEAD kit; Invitrogen)
was added to the antibody cocktail to exclude dead cells. The
acquisition of cells was done on LSRII flow cytometry using
FACSDIVA software. The data were analyzed using FlowJO
version 10.5.2 software (TreeStar, Ashland, OR). Data for
peptide-stimulated populations are reported as percentage,
determined after subtracting the percentage of positive cells
detected in unstimulated cells for each sample. Evaluation of
single, double, or triple-positive cells was carried out using
FlowJo Boolean gate.
Viral load quantitation
Plasma SIV RNA levels were measured by real-time RT-
PCR assay in Dr. Lifson’s facility as described.
53
The Lif-
son assay has a threshold sensitivity of 30 copy equivalents
per milliliter. Interassay variation is <25% (coefficient of
variation). Mean viral loads were calculated by transform-
ing the number in its logarithmic value and averaging the
logarithmic values of all the animals of the group at one
specific time point.
Euthanasia
Animals were euthanized because of closure of the study,
or earlier if they developed signs and symptoms consistent
with the definition of AIDS. AIDS was defined as being SIV
+
(detectable viremia) and experiencing one of the following
4 CHAUDHARY ET AL.
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 4
criteria: 1—weight loss >15% in 2 weeks or >30% in 2
months; 2—documented opportunistic infection; 3—per-
sistent anorexia >3 days without explicable cause; 4—severe,
intractable diarrhea, 5—progressive neurologic signs, 6—
significant cardiac and/or pulmonary signs, 7—loss of CD4
+
T cells <200 or 10%.
Statistical analysis
Calculations and statistical analyses were performed
using the GraphPad Prism version 7 software. Two-tailed
Fisher’s exact test was used to compare the frequency of
IgA responses between groups. Between-group compari-
sons were carried out by two-tailed, t-test or Mann–
Whitney test, and among four groups one-way ANOVA and
Mann–Whitney test were used. Results of statistical ana-
lyses were considered significant if they produced pvalues
£.05. Display of multicomponent distributions was per-
formed with SPICE v5.2 (freely available from http://exon
.niaid.nih.gov/spice/).
54
Results
Vaccine data
Oral vaccination can stimulate systemic and mucosal anti-
SIV responses. We reasoned that the combination of DNA,
an excellent stimulator of T-cell responses, and SIV-OPV
could provide a very interesting vaccine platform to explore
and compare to what we accomplished using SIV DNA+
SIV-MVA via the oral route and with a vaccine composed of
SIV DNA+SIVgp140, which constitutes
AU6 ca more heavily
explored approach to SIV and HIV immunization. During the
vaccination phase of the study, the systemic and mucosal
antibody responses were measured in plasma and secretions.
The oral vaccination elicited systemic IgG responses to SIV
antigens in only two of eight Group 1 and one of eight Group
2 animals on weeks 26 and 28, corresponding to 2 and 4
weeks after the last vaccination (Fig. 1B, C). Plasma IgG
antibodies against SIV lysate or gp140 Env were more fre-
quently observed in Group 3 animals immunized with SIV
DNA/rMVA (Fig. 1B, C). These IgG responses were nega-
tive in neutralization assays. A similar trend was observed for
rectal IgA responses (
F2 cFig. 2A, B). The oral cavity vaccination
stimulated rectal IgA responses to SIV antigens in only two
Group 1 and Group 2 animals, whereas five of the seven
animals in Group 3 had detectable anti-SIV or -gp140 Env
IgA in rectal secretions. The same number of Group 3 ani-
mals also developed salivary IgA antibodies to SIV antigens,
and these responses were detected in 50% of Group 1 animals
that were orally boosted with SIV gp140 and dmLT adjuvant.
However, only three of the eight Group 2 animals boosted
with SIV-OPV had SIV-specific salivary IgA antibodies
(Fig. 2A, B), and none of the animals in any group were found
to have SIV-specific IgA in vaginal secretions (not shown).
Thus, the SIV-OPV boost failed to stimulate the significant
antibody responses we expected from an OPV-based ap-
proach. These results indicate that the oral cavity immuni-
zation is a suitable route to induce systemic, salivary, and
rectal humoral responses using diverse approaches, but ap-
propriate doses and formulations need to be systematically
evaluated to identify those that more consistently generate
antibodies.
Vaccine-induced systemic and mucosal SIV-specific
CD4
+
and CD8
+
T-cell responses
As anti-SIV cell-mediated responses, in particular CD8
+
T-cell responses, are critical in viremia control and long-term
protection from disease progression, we evaluated the levels
and breath of vaccine-induced cell-mediated immunity.
Virus-specific T-cell responses were measured by intracel-
lular staining and flow cytometric analysis at multiple time
points, and are reported as percentage of antigen-specific
CD4
+
and CD8
+
T-cells producing TNFa, IFNc, and IL-2 in
MNC after stimulation with SIV Gag or Env peptide. The
analysis of the systemic SIV-specific immune responses,
measured during immunization in PBMC and reported as the
sum of anti-Gag and anti-Env percentages, revealed that all
three vaccine modalities could stimulate significant anti-SIV
cell-mediated responses in PBMCs and that, although con-
sistently slightly higher in Group 3, these differences were
not statistically significant ( bF3
Fig. 3A).
We also evaluated bAU7
long-term magnitude and functionality
of these anti-SIV responses, measuring the fraction of SIV-
specific CD4
+
and CD8
+
T cells producing TNFa, IFNc, and
IL-2 as single, double, or triple-positive cells, on week 32, 2
months after the last immunization, when the immune re-
sponse has usually contracted to the memory level (Fig. 3B).
On the day of challenge, Group 3 maintained levels of anti-
SIV CD4
+
T-cell immune responses *2-fold higher than the
other groups; the same was true for anti-SIV CD8
+
T-cell
responses when both Groups 2 and 3 were compared with
Group 1. We found that these responses were mainly
monofunctional and predominantly made of cells producing
IFNcin all groups (Fig. 3B). However, variable percentages
of multifunctional responses, varying between 8.7% and
38.15% of the total when both CD4
+
and CD8
+
T-cells were
considered, were present in the different groups.
We evaluated the ability of the vaccine candidate to induce
SIV-specific T-cell responses in tissues during the time
course of the immunization regimen by investigating the
percentages of SIV-specific mucosal MNC present in ingui-
nal lymph nodes, rectal and vaginal biopsies, collected during
immunization. Samples were collected at five time points, on
day of first immunization, 2 weeks after second, third and
fourth immunization, when the responses were likely to reach
peak, and 8 weeks after the last immunization, when the
effector component of the response is usually substantially
reduced. Oral vaccine administration stimulated CD4
+
and
CD8
+
T-cell responses at all examined sites with variable
magnitude that increased over the course of the immuniza-
tions. When SIV-specific responses were analyzed in rectal,
vaginal, and lymph node MNC on week 26, 2 weeks after the
last immunization, significant levels of Gag and Env specific
for CD4
+
and CD8
+
T-cell responses were observed in all
vaccinated groups in all tissues ( bF4
Fig. 4A). The differences in
SIV-specific immune responses did not reach significance
when compared between the groups, although rectal and
vaginal responses were consistently higher than those in
lymph node MNC. On week 32, 8 weeks after last immuni-
zation, the responses had contracted to a frequency varying
from *0.2% to 0.4% for the individual cytokines with T cells
producing IL-2, or IFNcor TNFasimilarly represented in
SIV-specific rectal and vaginal CD4
+
and CD8
+
T cells and
values varying between 0.6% and 1.2% (Fig. 4A, B). When
SIV ORAL IMMUNIZATION 5
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 5
multifunctionality of antigen-specific responses was inves-
tigated with Boolean analysis on the same time point sam-
ples, a large fraction of the response, ranging from 70% to
90% of the total, was characterized by monofunctional cells
producing each of the three cytokines tested. However,
polyfunctional responses were detectable in all tissues with
frequency varying from *10% to 32% of the total (Fig. 4C).
Analysis of SIV-specific CD4
+
and CD8
+
T-cell
responses memory and effector subsets
We compared the central memory (C
M
) and effector
memory (E
M
) subset fractions of the cell-mediated response
in blood and tissue MNC at peak after the last immunization
(week 26) and 2 months after the last immunization (week
32). On week 26, C
M
or E
M
varied within a twofold range
among the three immunization regimens, both being consis-
tently lower in Group 1 that received boosting immunizations
not including a SIV-Gag antigen but only SIV Env (
F5 cFig. 5A).
However, differences between groups did not reach statistical
significance, probably because of the small size of the groups
and of the variability in the magnitude of the responses
among animals within the same groups. On week 32, the SIV
responses were also of comparable magnitude in all three
groups, although reduced compared with week 26 peak re-
sponses (Fig. 5A). When the distribution between the C
M
and
E
M
CD4
+
and CD8
+
T cells was evaluated for each animal
within each vaccination group, the average group C
M
/E
M
ratio of SIV-specific cells present in tissue compartments on
week 32 was higher compared with week 26 for most com-
parisons ( p<.05), supporting a larger contraction of the E
M
than the C
M
immune response since its peak and the persis-
tence of significant C
M
antigen-specific responses.
When the fraction of antigen-specific responses producing
the same cytokine alone or in combination was compared
with each of the other two cytokines in C
M
and E
M
of SIV-
specific CD4
+
and CD8
+
T cells, IL-2 was the predominantly
expressed cytokine in CD4
+
/E
M
+
and CD8
+
/E
M
T cells of
FIG. 2. Rectal and salivary IgA responses in vaccinated animals. Shown are the postimmunization fold increases in
specific activity (ng of specific antibody per lg total IgA) to (A) SIV lysate and (B) SIV gp140 Env measured in rectal and
salivary secretions. Fold increases were calculated by dividing the week 26 or 28 specific activity by that measured on week
0. To be considered significant, the specific activity had to exceed the dashed line (representing the mean +3SD of negative
controls) in the graphs. Bars represent the median for each group.
4C c
6 CHAUDHARY ET AL.
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 6
Group 2 (Fig. 5B, p<.05) and the multifunctional C
M
CD4
+
T-cell SIV Gag+Env-specific responses were a significantly
larger percentage than that of the E
M
CD4
+
fraction ( p<.05).
The other two groups had similar levels of C
M
and E
M
antigen-specific multifunctional responses, supporting a
more prolonged survival of E
M
responses in these groups and
in CD8
+
C
M
and E
M
responses the largest fraction of antigen-
specific cells secreted IFNc(p<.05).
When all the above data are considered together, we
concluded that all vaccine modalities given in the oral cavity
were similarly effective in stimulating cell-mediated T-cell
responses at multiple sites, including sites of HIV exposure,
but much less effective at stimulating humoral responses. The
vaccination utilizing the SIV DNA-MVA modality produced
more consistent humoral results, although some low-level
responses were observed in a couple of animals of the other
two groups as well. These data indicate that the oral cavity
can be explored as a site to stimulate broadly distributed
responses but higher doses of vaccine, more immunizations,
or different formulations might be necessary.
FIG. 3. Circulating cell-mediated total percentages of SIV Gag and SIV Env-specific T-cell responses during immuni-
zation phase. (A) Geometric means of each vaccinated group is shown during the immunization phase as percentage of
CD4
+
T-cells and CD8
+
T-cells producing IFNc, TNFa, and IL-2, measured in PBMC by ICS upon stimulation with Gag or
Env peptide pools. The graphs show the total SIV-specific T-cell responses (Gag plus Env) for vaccinated groups: SIV-
DNA+SIV-gp140 vaccinated animals are represented in blue (Group 1), SIV-DNA+SIV-OPV in brown (Group 2), SIV-
DNA+SIV-MVA in green (Group 3). Statistical significance between groups was tested with one-way ANOVA assay
(p<.05). (B) Qualitative analysis of systemic SIV Gag and Env-specific cell-mediated responses for each vaccinated group
2 weeks on the first day of challenge (week 32), 8 weeks after last immunization. Pie graphs representing the diversity of
CD4
+
(left panels) and CD8
+
T-cell responses (right panels) with different colors representing proportionally to percentages
of the production of IFNc, TNFa, and IL-2 alone and the simultaneous production of 2 cytokines combined are shown. The
total mean percentage of SIV-specific multifunctional responses for each group is shown on the upper left side of each pie.
ICS, intracellular cytokine staining; PBMC, peripheral blood mononuclear cell.
4C c
SIV ORAL IMMUNIZATION 7
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 7
FIG. 4. Tissue SIV-specific cell-mediated
T-cell responses during immunization. SIV
Gag+Env-specific (A) CD4
+
and (B) CD8
+
T-cell responses were detected in MNC
isolated from vaginal and rectal and lymph
node biopsies during the immunization
protocol, and the group average is re-
presented as a bar graph for each group.
Each bar represents the sum of Gag- and
Env-specific T-cells producing IFNc, IL-2,
and TNFaevaluated as individual cytokines
after stimulation with Gag or Env peptide
pools. (C) Functionality of lymph node,
vaginal and rectal SIV-specific T-cell re-
sponses on the first day of challenge (week
32). Pie graphs illustrate the diversity of
Gag+Env-specific CD4
+
T-cells and CD8
+
T-cells producing IFNc, TNFa, and IL-2 and
colors are proportionate to the percentages
of single, double, or triple positives for each
group. The SIV-specific T-cell mean per-
centage of the total CD4
+
or CD8
+
T cells is
shown for each group beneath each pie. The
total mean percentage of SIV-specific mul-
tifunctional responses for each group is
shown on the upper left side of each pie.
MNC, mononuclear cell.
b4C
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 8
8
FIG. 5. Central memory (CD95
+
/CD28
+
) and effector memory (CD95
+
/CD28
-
) SIV-specific cell-mediated T-cell re-
sponses during the immunization protocol. (A) SIV Gag+Env-specific CD4
+
and CD8
+
T-cell responses were detected in
PBMC and in MNC isolated from lymph node, vaginal and rectal biopsies 2 weeks after last immunization (week 26) and on
first day of challenge (week 32), and the group average is represented as a bar graph for each group. Each bar represents the
sum of anti-C
M
and E
M
Gag +Env-specific T-cells producing IFNc, IL-2, and TNFaevaluated as individual cytokines.
CD3
+
T cells were gated within total PMBC, CD4
+
and CD8
+
T cells were gated within the CD3
+
population, and
percentages of cytokine
+
cells were estimated in these populations for each cytokine and added to provide the total.
(B)
AU12 cFunctionality of PBMC SIV-specific T-cell responses on first day of challenge (week 32). Pie graphs illustrate the
diversity of Gagmen-specific CD4
+
T-cells and CD8
+
T-cells producing IFNc, TNFa, and IL-2 as percentages of single,
double, or triple positives for each group. The SIV-specific T-cell mean percentage of the total CD4
+
or CD8
+
T cells is
shown for each group beneath each pie.
4C c
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 9
9
Resistance to challenge and disease progression
To evaluate levels of protection provided by the vaccina-
tion, animals were vaginally challenged with repeated low
doses of SIV
mac251
(
F6 cFig. 6). No significant differences were
observed when the median number of challenges was com-
pared among groups, indicating that the levels of immune
responses present in the vaginal tissues were not sufficient to
provide protection (Fig. 6A). When viral loads were evalu-
ated, early control of viremia from peak levels to week 8 was
observed in Group 3 compared with other groups. However,
after that, all three groups equally controlled viremia
(Fig. 6B). The level of viremia control observed in the control
group was significantly higher than that previously observed
in infected RM
32,33,35
and viremia was at least half log lower
than what is normally observed for CyM and *2 logs of what
was observed in another study where SIV-OPV was inves-
tigated in CyM.
29,55,56
C
M
CD4
+
T-cell counts, which usually
decline earlier after infection than total CD4
+
T-cell counts
in RM, recovered after the declined observed 2 week post-
infection to levels comparable with those preinfection and
remained stable during the postinfection follow-up in all
groups (Fig. 6C, group average, Fig. 6E, individual absolute
counts/mm
3
). Total CD4
+
T-cell counts declined *10% 2
weeks after infection in all groups and remained at that level
for the remaining of the follow-up (Fig. 6D, group percentage
FIG. 6. Low-dose vaginal SIV
mac251
challenge outcome. (A)
AU13 cThe Kaplan–Meier graph represents the number of non-
traumatic challenges received for each vaccinated group until a positive infection was detected in plasma. The nontraumatic
challenges were stopped after 26 challenges when all the animals had become positive. The dotted line intersects the point
in each curve corresponding to 50% of the animals being infected. (B) Average viral loads in the groups. (C) PBMC C
M
(CD95
+
/CD28
+
) CD4
+
T-cell percentages postinfection. (D) PBMC total CD4
+
T-cell percentages postinfection. (E, F)
Absolute number of PBMCs C
M
(E) and total
78
CD4
+
T-cells during the course of the infection.
4C c
10 CHAUDHARY ET AL.
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 10
averages, Fig. 6F, individual absolute counts/mm
3
,p=ns
when time 0 values were compared with those of week 20
postinfection), indicating that the levels of viremia were
compatible with preservation of these cells for the length of
the follow-up in vaccinated group and controls. Although a
milder course of SIV infection in CyM versus RM has been
reported, preservation of CD4
+
T cells occurred in these
animals at levels that were not statistically different than
when noninfected, unlike what was observed by others in
SIV-infected CyM, where significant decrease to <50% of
preinfection value in both C
M
and total CD4
+
T cells has been
observed [Fig. 2 in
55
]. The unusual resistance of these control
animals to SIV-mediated CD4
+
T-cell decline reduced the
ability to reveal any level protection from the vaccination, if
any had been induced.
Discussion
As of today, only one vaccine modality tested in clinical
trials and administered intramuscularly (i.m.) achieved par-
tial protection (31.2% efficacy), the RV-144 ALVAC-HIV
(v CP1521) plus AIDSVAX,
57
AU8 cnot only supporting the fea-
sibility of achieving protection but also requiring further
improvement. However, the recent trial HVTN702 in South
Africa, based on the same modality of vaccination, was ter-
minated because of lack of efficacy.
58
This trial utilized
MF59 instead of Alum, and the difference in outcome was
predicted in a preclinical SIV
mac251
trial in RM that compared
side by side the two adjuvants, supporting the different
contributions of each vaccine adjuvant to the vaccine out-
come and the need to test each variable independently.
59
Achievement of appropriate antibody responses, in particular
neutralizing antibody, is considered the holy grail of the
successful HIV vaccine [
60–62
and references therein].
In previous studies in RM, we have shown that mucosal
immunization stimulating cell-mediated immune responses
can achieve protection from disease progression but did not
protect from acquisition of infection, although in some cases
infection was delayed.
32–34,63
Intranasal vaccination was
more efficient in eliciting cellular and humoral virus-specific
responses at mucosal sites than the same regime administered
systemically (i.m.) and provided better protection from dis-
ease progression after rectal or vaginal challenge. SIV-
specific CD4
+
and CD8
+
IFNcproducing T cells present at
the time of challenge correlated with the subsequent control
of the viremia and longer survival of these animals. However,
it did not stimulate significant humoral responses in the cir-
culation or vaginal mucosa. In a recent study, where HIV-
MVA followed by recombinant trimeric HIV gp120 with
dmLT was administered with a pressurized, needle-free in-
jection devise both buccally and sublingually in RM, Jones
et al. found that this immunization strategy provided an ef-
fective route to induce immunity and partial protection
against rectal SHIV challenge.
64
The study reported here was initiated with the goal of using
a vector based on OPV, a virus known to be excellent at
stimulating long-lived antibody responses and in particular
mucosal IgA, to improve the stimulation of humoral immu-
nity and the oral cavity route as easily accessible and prac-
tical in resource-limited settings. The importance of mucosal
IgA responses is highlighted in humans by the detection of
HIV-specific IgA in semen or vaginal secretions of some
cohorts of HIV-1 resistant, heavily exposed but seronegative
sex workers, which has been interpreted as indication that
local IgA, induced by viral exposure, can protect during
subsequent exposures.
65–68
Secretory IgA antibodies have
been shown to inhibit host entry and dissemination of path-
ogens into systemic compartment, and this mechanism,
combined with others, such as antibody-dependent cellular
cytotoxicity (ADCC) by FcaR
+
tissue macrophages, could
work for HIV as well.
69–71
An additional modality capable of inducing significant
antibodies is the use of a purified protein as priming and/or
boosting tool of antipathogen responses. Comparative studies
in humans and NHP that utilized protein immunogens effi-
ciently internalized at all mucosal surfaces established that
local immunization stimulates mucosal IgA responses often
limited to the vaccination site or of less magnitude at more
distant sites (reviewed in
72
). These differential antibody re-
sponses may be less apparent with replicating vaccines. For
example, a replication-competent adenovirus type 5 vector
with SIV genes was recently reported to induce SIV-specific
mucosal IgA responses in multiple mucosal tissues regardless
of whether it was administered to female macaques by the
sublingual, rectal, vaginal, or nasal +intratracheal mucosal
routes.
73
The protein dose used here did not provide satis-
factory results and may have been too low, as uptake of to-
pically applied proteins in the oral cavity is inefficient.
Antigens formulated in conjunction with adjuvants, as we did
in this immunization, avoid inducing tolerance and failure to
stimulate an immune response is dose- or antigen depen-
dent.
74–76
Achieving immune responses with oral protein
immunization may require testing multiple doses, formula-
tions, and methods of application to find the dose high enough
to induce the optimal response.
The approaches used here and compared with the
DNA/MVA platform previously used did not consistently
stimulate the desired humoral responses but were able to
stimulate cell-mediated responses. As the last boost did not
include the SIV DNA component and managed to increase
the levels of cell-mediated responses, it provided the evi-
dence that these different boosting modalities were immu-
nogenic on their own. In the case of the gp140 boost,
increasing the amount of protein, and perhaps adjuvant, could
work in a more significant and consistent way. In the case of
the SIV-OPV vectors, a few issues could have affected the
outcome. The vaccination was done with sedated animals that
were not swallowing, possibly limiting the spreading of it to
the entire gastrointestinal tract. Alternatively, the recombi-
nant SIV-OPV stability in vivo may have been more limited
than what was observed in vitro, with reduced in vivo viral
replication, and the possibility that a higher dose could have
provided better results. The OPV recombinant previously
used carried much smaller inserts than those employed here,
and that feature may have favored better in vivo replica-
tion.
27–29
Despite these shortcomings, this study shows that
the oral cavity can be considered a useful route of vaccination,
as all platforms achieved significant levels of cell-mediated
immunity and sporadic levels of humoral immunity, leaving
open the possibility of improvement for the latter if more
doses, formulations, and schedules are evaluated. Additional
studies should aim at improving vaccine platforms used via
this route, as it is highly amenable to its employment in
resource-limited environments. An unexpected result at the
SIV ORAL IMMUNIZATION 11
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 11
time of challenge was the viremia control observed in the naive
animals, being better than that normally observed in CyM.
This species was selected because previous results indicated
that RM could not be infected orally with OPV. No significant
decline of C
M
CD4
+
T cells or total CD4
+
T cells was observed
during the 5 months after infection. This occurrence prevented
the evaluation of the vaccination on the preservation of the
immune system. In addition to species-specific issues,
AU9 cit is
possible that the virus stock used in this study may not have
had the same virulence as other related similar stocks. As RMs
have been recently shown to be infectable orally
77
and post-
infection data in these species have been more consistent in our
hands, future experiments in this species with additional re-
combinant SIV- and SHIV-OPV will be pursued.
Acknowledgments
We thank Dr. David Montefiori (Duke University, Dur-
ham, NC) for carrying out the neutralization assay on plasma
samples, Dr. Jeff Lifson (Leidos Biomedical Research, Inc.,
Frederick National Laboratory, Frederick, MD 21702) for
evaluating plasma SIV viral loads, Robert L. Wilson for
analyses of antibody responses, and Olga Nichols for pro-
duction of the SIV-MVA in the LSU Health Sciences Vector
Core Facility.
Authors’ Contribution
Omkar Chaudhary obtained PBMC and MNC from blood
and tissues, carried out analysis of immune response in
samples, prepared the first partial draft of the article and
graphs for the figures reporting the cell-mediated immune
responses, contributed to some statistical analysis, and re-
vised final draft of the article.
Lingyun Wang obtained PBMC and MNC from blood and
tissues, carried out qualitative evaluation of SIV plasma viral
loads weekly after challenge, prepared graphs for some fig-
ures, contributed to some statistical analysis, and revised final
draft of the article.
Deepanwita Bose obtained PBMC and MNC from blood
and tissues, carried out analysis of immune responses in a
subset of samples and evaluated CD4+T cell counts, pre-
pared graphs for some figures, contributed to some statistical
analysis, and revised final draft of the article.
Vivek Narayan constructed and tested SIV-OPV recom-
binants, and revised final draft of the article.
Ming Te Yeh provided the pSabin2-eGFP vector, advised all
technical aspects of oral poliovirus vaccine work and titrated
the SIV-OPV virus stocks, revised final draft of the article.
Angela Carville supervised vaccinations, sampling and
animal care at the Biomere facility, revised final draft of the
article.
John D. Clements provided the double mutant heat-labile
toxin (dmLT) adjuvant, critically contributed to the inter-
pretation of the technical and intellectual content and to the
editing of the article.
Raul Andino provided the OPV vector, supervised Dr
Yeh’s work, and critically revised final version of the article.
Pamela A. Kozlowski provided the SIV-MVA, supervised
evaluation of antibody responses, composed the corre-
sponding figures, critically contributed to the interpretation
of the technical and intellectual content and to the editing of
the article.
Anna Aldovini designed the experiments, secured funding,
coordinated, supervised, and troubleshooted all aspects of the
execution, interpreted the data, wrote the article, and edited
the figures.
Author Disclosure Statement
No competing financial interests exist.
Funding Information
This work was supported by Grant NIH-R01DE026325
to AA.
References
1. Mattapallil JJ, Douek DC, Hill B, Nishimura Y, Martin M,
Roederer M: Massive infection and loss of memory CD4+
T cells in multiple tissues during acute SIV infection.
Nature 2005;434:1093–1097.
2. Veazey RS, DeMaria M, Chalifoux LV, et al.:Gastro-
intestinal tract as a major site of CD4+T cell depletion and
viral replication in SIV infection. Science 1998;280:427–431.
3. Barratt-Boyes SM, Soloff AC, Gao W, et al.: Broad cellular
immunity with robust memory responses to simian immu-
nodeficiency virus following serial vaccination with ade-
novirus 5- and 35-based vectors. J Gen Virol 2006;87(Pt 1):
139–149.
4. Gordon SN, Cervasi B, Odorizzi P, et al.: Disruption of
intestinal CD4+T cell homeostasis is a key marker of
systemic CD4+T cell activation in HIV-infected individ-
uals. J Immunol 2010;185:5169–5179.
5. Belyakov IM, Berzofsky JA: Immunobiology of mucosal HIV
infection and the basis for development of a new generation of
mucosal AIDS vaccines. Immunity 2004;20:247–253.
6. Cryz SJ, Jr., Que JU, Levine MM, Wiedermann G, Kol-
laritsch H: Safety and immunogenicity of a live oral biva-
lent typhoid fever (Salmonella typhi Ty21a)-cholera
(Vibrio cholerae CVD 103-HgR) vaccine in healthy adults.
Infect Immun 1995;63:1336–1339.
7. Guzman CA, Borsutzky S, Griot-Wenk M, et al.: Vaccines
against typhoid fever. Vaccine 2006;24:3804–3811.
8. O’Ryan M: Rotarix (RIX4414): An oral human rotavirus
vaccine. Expert Rev Vaccines 2007;6:11–19.
9. Ruiz-Palacios GM, Perez-Schael I, Velazquez FR, et al.:
Safety and efficacy of an attenuated vaccine against severe
rotavirus gastroenteritis. N Engl J Med 2006;354:11–22.
10. Ryan ET, Calderwood SB, Qadri F: Live attenuated oral
cholera vaccines. Expert Rev Vaccines 2006;5:483–494.
11. Taylor DN, Tacket CO, Losonsky G, et al.: Evaluation of a
bivalent (CVD 103-HgR/CVD 111) live oral cholera vac-
cine in adult volunteers from the United States and Peru.
Infect Immun 1997;65:3852–3856.
12. Aldovini A: Mucosal Vaccination for Prevention of HIV
Infection and AIDS. Curr HIV Res 2016;14:247–259.
13. Kozlowski PA, Aldovini A: Mucosal Vaccine Approaches
for Prevention of HIV and SIV Transmission. Curr Im-
munol Rev 2019;15:102–122.
14. Xu H, Wang X, Veazey RS: Mucosal immunology of HIV
infection. Immunol Rev 2013;254:10–33.
15. Kantele A, Hakkinen M, Moldoveanu Z, et al.: Differences
in immune responses induced by oral and rectal immuni-
zations with Salmonella typhi Ty21a: Evidence for com-
partmentalization within the common mucosal immune
system in humans. Infect Immun 1998;66:5630–5635.
12 CHAUDHARY ET AL.
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 12
16. Kozlowski PA, Cu-Uvin S, Neutra MR, Flanigan TP:
Comparison of the oral, rectal, and vaginal immunization
routes for induction of antibodies in rectal and genital tract
secretions of women. Infect Immun 1997;65:1387–1394.
17. Kozlowski PA, Williams SB, Lynch RM, et al.: Differential
induction of mucosal and systemic antibody responses in
women after nasal, rectal, or vaginal immunization: Influ-
ence of the menstrual cycle. J Immunol 2002;169:566–574.
18. Johansson EL, Wassen L, Holmgren J, Jertborn M, Rudin
A: Nasal and vaginal vaccinations have differential effects
on antibody responses in vaginal and cervical secretions in
humans. Infect Immun 2001;69:7481–7486.
19. Rudin A, Johansson EL, Bergquist C, Holmgren J: Dif-
ferential kinetics and distribution of antibodies in serum
and nasal and vaginal secretions after nasal and oral vac-
cination of humans. Infect Immun 1998;66:3390–3396.
20. Rudin A, Riise GC, Holmgren J: Antibody responses in the
lower respiratory tract and male urogenital tract in humans
after nasal and oral vaccination with cholera toxin B sub-
unit. Infect Immun 1999;67:2884–2890.
21. Russell MW, Moldoveanu Z, White PL, Sibert GJ, Mestecky
J, Michalek SM: Salivary, nasal, genital, and systemic an-
tibody responses in monkeys immunized intranasally with a
bacterial protein antigen and the Cholera toxin B subunit.
Infect Immun 1996;64:1272–1283.
22. Asiedu CK, Goodwin KJ, Balgansuren G, et al.: Elevated T
Regulatory Cells in Long-Term Stable Transplant Toler-
ance in Rhesus Macaques Induced by Anti-CD3 Im-
munotoxin and Deoxyspergualin. J Immunol 2005;175:
8060–8068.
23. Brenchley JM, Schacker TW, Ruff LE, et al.: CD4+T cell
depletion during all stages of HIV disease occurs pre-
dominantly in the gastrointestinal tract. J Exp Med 2004;
200:749–759.
24. Mehandru S, Poles MA, Tenner-Racz K, et al.: Primary
HIV-1 infection is associated with preferential depletion of
CD4+T lymphocytes from effector sites in the gastroin-
testinal tract. J Exp Med 2004;200:761–770.
25. Kew OM, Sutter RW, de Gourville EM, Dowdle WR, Pal-
lansch MA: Vaccine-derived polioviruses and the endgame
strategy for global polio eradication. Annu Rev Microbiol
2005;59:587–635.
26.
AU10 cAvailable at www.polioeradication.org/Polioandprevention/
Thevaccines/Oralpoliovaccine(OPV).aspx.
27. Crotty S, Andino R: Poliovirus vaccine strains as mucosal
vaccine vectors and their potential use to develop an AIDS
vaccine. Adv Drug Deliv Rev 2004;56:835–852.
28. Crotty S, Lohman BL, Lu FX, Tang S, Miller CJ, Andino
R: Mucosal immunization of cynomolgus macaques with
two serotypes of live poliovirus vectors expressing simian
immunodeficiency virus antigens: Stimulation of humoral,
mucosal, and cellular immunity. J Virol 1999;73:9485–
9495.
29. Crotty S, Miller CJ, Lohman BL, et al.: Protection against
simian immunodeficiency virus vaginal challenge by using
Sabin poliovirus vectors. J Virol 2001;75:7435–7452.
30. Mandl S, Hix L, Andino R: Preexisting immunity to po-
liovirus does not impair the efficacy of recombinant po-
liovirus vaccine vectors. J Virol 2001;75:622–627.
31. Manrique M, Kozlowski PA, Cobo-Molinos A, et al.: Im-
munogenicity of a vaccine regimen composed of simian
immunodeficiency virus DNA, rMVA, and viral particles
administered to female rhesus macaques via four different
mucosal routes. Journal of Virology 2013;87:4738–4750.
32. Manrique M, Kozlowski PA, Cobo-Molinos A, et al.:
Long-term control of simian immunodeficiency virus
mac251 viremia to undetectable levels in half of infected
female rhesus macaques nasally vaccinated with simian
immunodeficiency virus DNA/recombinant modified vac-
cinia virus Ankara. J Immunol 2011;186:3581–3593.
33. Manrique M, Kozlowski PA, Wang SW, et al.: Nasal DNA-
MVA SIV vaccination provides more significant protection
from progression to AIDS than a similar intramuscular
vaccination. Mucosal Immunol 2009;2:536–550.
34. Manrique M, Micewicz E, Kozlowski PA, et al.: DNA-
MVA vaccine protection after X4 SHIV challenge in
macaques correlates with day-of-challenge antiviral CD4+
cell-mediated immunity levels and postchallenge pres-
ervation of CD4+T cell memory. AIDS Res Hum
Retroviruses 2008;24:505–519.
35. Manrique M, Kozlowski PA, Cobo-Molinos A, et al.:Re-
sistance to infection, early and persistent suppression of simian
immunodeficiency virus SIVmac251 viremia, and significant
reduction of tissue viral burden after mucosal vaccination in
female rhesus macaques. J Virol 2014;88:212–224.
36. Iwasaki A, Welker R, Mueller S, Linehan M, Nomoto A,
Wimmer E: Immunofluorescence analysis of poliovirus
receptor expression in Peyer’s patches of humans, primates,
and CD155 transgenic mice: Implications for poliovirus
infection. J Infect Dis 2002;186:585–592.
37. Sabin AB: Pathogenesis of poliomyelitis; reappraisal in the
light of new data. Science 1956;123:1151–1157.
38. Takahashi Y, Misumi S, Muneoka A, et al.: Nonhuman
primate intestinal villous M-like cells: An effective polio-
virus entry site. Biochem Biophys Res Commun 2008;368:
501–507.
39. Yeh MT, Bujaki E, Dolan PT, et al.: Engineering the live-
attenuated polio vaccine to prevent reversion to virulence.
Cell Host Microbe 2020;27:736–751 e738.
40. Clements JD, Norton EB: The Mucosal Vaccine Adjuvant
LT(R192G/L211A) or dmLT. mSphere 2018;3.
41. Moss B, Carroll MW, Wyatt LS, et al.: Host range re-
stricted, non-replicating vaccinia virus vectors as vaccine
candidates. Adv Exp Med Biol 1996;397:7–13.
42. Sutter G, Moss B: Nonreplicating vaccinia vector effi-
ciently expresses recombinant genes. Proc Natl Acad Sci
U S A 1992;89:10847–10851.
43. Curtis AD, 2nd, Jensen K, Van Rompay KKA, Amara RR,
Kozlowski PA, De Paris K: A simultaneous oral and in-
tramuscular prime/sublingual boost with a DNA/Modified
Vaccinia Ankara viral vector-based vaccine induces simian
immunodeficiency virus-specific systemic and mucosal
immune responses in juvenile rhesus macaques. J Med
Primatol 2018;47:288–297.
44. Curtis AD, 2nd, Walter KA, Nabi R, et al.: Oral Coadmi-
nistration of an Intramuscular DNA/Modified Vaccinia
Ankara Vaccine for Simian Immunodeficiency Virus Is
Associated with Better Control of Infection in Orally Ex-
posed Infant Macaques. AIDS Res Hum Retroviruses 2019;
35:310–325.
45. Kestler H, Kodama T, Ringler D, et al.: Induction of AIDS
in rhesus monkeys by molecularly cloned simian immu-
nodeficiency virus. Science 1990;248:1109–1112.
46. Regoes RR: The role of exposure history on HIV acquisi-
tion: Insights from repeated low-dose challenge studies.
PLoS Comput Biol 2012;8:e1002767.
47. Keele BF, Li H, Learn GH, et al.: Low-dose rectal inocu-
lation of rhesus macaques by SIVsmE660 or SIVmac251
SIV ORAL IMMUNIZATION 13
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 13
recapitulates human mucosal infection by HIV-1. J Exp
Med 2009;206:1117–1134.
48. Guadalupe M, Reay E, Sankaran S, et al.: Severe CD4+
T-cell depletion in gut lymphoid tissue during primary
human immunodeficiency virus type 1 infection and sub-
stantial delay in restoration following highly active anti-
retroviral therapy. J Virol 2003;77:11708–11717.
49. Kozlowski PA, Lynch RM, Patterson RR, Cu-Uvin S,
Flanigan TP, Neutra MR: Modified wick method using
Weck-Cel sponges for collection of human rectal secretions
and analysis of mucosal HIV antibody. J Acquir Immune
Defic Syndr 2000;24:297–309.
50. Montefiori DC: Evaluating neutralizing antibodies against
HIV, SIV and SHIV in luciferase reporter gene assays. In:
Current Protocols in Immunology (Coligan JE, Kruisbeek
AM, Margulies DH, Shevach EM, Strober W, Coico R,
eds.) Vol 1. Hoboken, NJ, John Wiley & Sons, 2004,
pp. 12.11.11–12.11.15.
51. Mansfield KG, Veazey RS, Hancock A, et al.: Induction of
disseminated Mycobacterium avium in simian AIDS is
dependent upon simian immunodeficiency virus strain and
defective granuloma formation. Am J Pathol 2001;159:
693–702.
52. Manrique M, Micewicz E, Kozlowski PA, et al.:
DNA-MVA vaccine protection after X4 SHIV challenge
in macaques correlates with day-of-challenge antiviral
CD4+cell-mediated immunity levels and postchallenge
preservation of CD4+T cell memory. AIDS Res Hum
Retroviruses 2008;24:505–519.
53. Lifson JD, Rossio JL, Piatak M, Jr., et al.: Role of CD8(+)
lymphocytes in control of simian immunodeficiency virus
infection and resistance to rechallenge after transient early
antiretroviral treatment. J Virol 2001;75:10187–10199.
54. Roederer M, Nozzi JL, Nason MC: SPICE: Exploration and
analysis of post-cytometric complex multivariate datasets.
Cytometry A 2011;79:167–174.
55. Chebloune Y, Moussa M, Arrode-Bruses G, et al.: A single
lentivector DNA based immunization contains a late het-
erologous SIVmac251 mucosal challenge infection. Vac-
cine 2020;38:3729–3739.
56. Karlsson I, Malleret B, Brochard P, et al.: Dynamics of
T-cell responses and memory T cells during primary simian
immunodeficiency virus infection in cynomolgus ma-
caques. J Virol 2007;81:13456–13468.
57. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, et al.: Vac-
cination with ALVAC and AIDSVAX to prevent HIV-1
infection in Thailand. N Engl J Med 2009;361:2209–2220.
58. Cohen J: Combo of two HIV vaccines fails its big test.
Science 2020;367:611–612.
59. Vaccari M, Gordon SN, Fourati S, et al.: Adjuvant-
dependent innate and adaptive immune signatures of risk of
SIVmac251 acquisition. Nat Med 2016;22:762–770.
60. Cheng HD, Grimm SK, Gilman MS, et al.: Fine epitope
signature of antibody neutralization breadth at the HIV-1
envelope CD4-binding site. JCI Insight 2018;3.
61. Karuna ST, Corey L: Broadly Neutralizing Antibodies for
HIV Prevention. Annu Rev Med 2020;71:329–346.
62. Sok D, Burton DR: Recent progress in broadly neutralizing
antibodies to HIV. Nat Immunol 2018;19:1179–1188.
63. Wang SW, Kozlowski PA, Schmelz G, et al.: Effective in-
duction of simian immunodeficiency virus-specific systemic
and mucosal immune responses in primates by vaccination
with proviral DNA producing intact but noninfectious viri-
ons. J Virol 2000;74:10514–10522.
64. Jones AT, Shen X, Walter KL, et al.: HIV-1 vaccination by
needle-free oral injection induces strong mucosal immunity
and protects against SHIV challenge. Nat Commun 2019;
10:798.
65. Beyrer C, Artenstein AW, Rugpao S, et al.: Epidemiologic
and biologic characterization of a cohort of human immu-
nodeficiency virus type 1 highly exposed, persistently se-
ronegative female sex workers in northern Thailand.
Chiang Mai HEPS Working Group. J Infect Dis 1999;179:
59–67.
66. Clerici M, Barassi C, Devito C, et al.: Serum IgA of HIV-
exposed uninfected individuals inhibit HIV through rec-
ognition of a region within the alpha-helix of gp41. Aids
2002;16:1731–1741.
67. Clerici M, Salvi A, Trabattoni D, et al.: A role for mucosal
immunity in resistance to HIV infection. Immunol Lett
1999;66:21–25.
68. Lo Caputo S, Trabattoni D, Vichi F, et al.: Mucosal and
systemic HIV-1-specific immunity in HIV-1-exposed but
uninfected heterosexual men. Aids 2003;17:531–539.
69. Corthesy B: Multi-faceted functions of secretory IgA at
mucosal surfaces. Front Immunol 2013;4:185.
70. Duchemin M, Khamassi M, Xu L, Tudor D, Bomsel M:
IgA Targeting Human Immunodeficiency Virus-1 Envelope
gp41 Triggers Antibody-Dependent Cellular Cytotoxicity
Cross-Clade and Cooperates with gp41-Specific IgG to
Increase Cell Lysis. Front Immunol 2018;9:244.
71. Ruprecht RM, Marasini B, Thippeshappa R: Mucosal An-
tibodies: Defending Epithelial Barriers against HIV-1 In-
vasion. Vaccines (Basel) 2019;7:194.
72. Neutra MR, Kozlowski PA: Mucosal vaccines: The prom-
ise and the challenge. Nat Rev Immunol 2006;6:148–158.
73. Xiao P, Patterson LJ, Kuate S, et al.: Replicating adenovirus-
simian immunodeficiency virus (SIV) recombinant priming
and envelope protein boosting elicits localized, mucosal IgA
immunity in rhesus macaques correlated with delayed ac-
quisition following a repeated low-dose rectal SIV(mac251)
challenge. J Virol 2012;86:4644–4657.
74. Coffman RL, Sher A, Seder RA: Vaccine adjuvants: Put-
ting innate immunity to work. Immunity 2010;33:492–503.
75. Coquet JM, Rausch L, Borst J: The importance of co-
stimulation in the orchestration of T helper cell differenti-
ation. Immunol Cell Biol 2015;93:780–788.
76. Mestecky J, Russell MW, Elson CO: Perspectives on mu-
cosal vaccines: Is mucosal tolerance a barrier? J Immunol
2007;179:5633–5638.
77. Shen L, Chen CY, Huang D, et al.: Pathogenic events in a
nonhuman primate model of oral poliovirus infection leading
to paralytic poliomyelitis. J Virol 2017;91:e02310-16.
78. Tracey I, Lane J, Chang I, Navia B, Lackner A, Gonzalez
RG: 1H magnetic resonance spectroscopy reveals neuronal
injury in a simian immunodeficiency virus macaque model.
J Acquir Immune Defic Syndr Hum Retrovirol 1997;15:
21–27.
Address correspondence to:
Anna Aldovini
Department of Medicine
Boston Children’s Hospital
Enders 861, 300 Longwood Avenue
Boston, MA 02115
USA
E-mail: anna.aldovini@childrens.harvard.edu
14 CHAUDHARY ET AL.
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 14
AUTHOR QUERY FOR AID-2020-0157-VER9-CHAUDHARY_1P
AU1: Please note that gene symbols in any article should be formatted as per the gene nomenclature. Thus, please make
sure that gene symbols, if any in this article, are italicized.
AU2: Please identify (highlight or circle) all authors’ surnames for accurate indexing citations.
AU3: Include IRB approval or waiver statement in the Materials and Methods section. The Clinical Trial Registration
number, if applicable, should be included at the end of the abstract.
AU4: The Publisher requests for readability that no paragraph exceeds 15 typeset lines. Please check for long paragraphs
and divide where needed.
AU5: The phrase ‘‘at the level of both infection and disease progression’’ has been edited for clarity. Please check.
AU6: The phrase ‘‘a more heavily explored approach to SIV and HIV immunization’’ has been edited for clarity. Please
check.
AU7: The phrase ‘‘long-term magnitude and functionality of these anti-SIV responses’’ has been edited for clarity. Please
check.
AU8: The phrase ‘‘not only supporting the feasibility of achieving protection but also requiring further improvement’ has
been edited for clarity. Please check.
AU9: The phrase ‘‘it is possible that the virus stock used in this study may not have had the same virulence as other related
similar stocks’’ has been edited for clarity. Please check.
AU10: In Ref. 26, please mention the authors’ names, title of the work, and date you last viewed the website.
AU11: Please check the sentence ‘‘Postimmunization concentrations had to be threefold higher than the preimmune
concentration to be considered significant.’’ with respect to clarity.
AU12: The phrase ‘‘Functionality of PBMC SIV-specific T-cell responses on first day of challenge’’ has been edited for
clarity. Please check.
AU13: The phrase ‘‘The Kaplan–Meier graph represents the number of nontraumatic challenges received for each
vaccinated group until a positive infection was detected in plasma.’’ has been edited for clarity. Please check.
AID-2020-0157-ver9-Chaudhary_1P.3d 10/16/20 8:22am Page 15
... Assessment of CCR2 and CCR5 cell surface levels was conducted according to previously published procedures (33). After permeabilization, cells were intracellularly stained for JAK and/or STAT expression with anti-JAK and/or anti-STAT antibodies for 1 hour in the dark at 4°C, followed by IC fixation buffer fix for 20 min at 4°C in the dark, according to previously described procedures (10,59). The following antibodies were used in this study: anti-CD195-PE (CCR5; clone HEK/1/85a; BioLegend, San Diego, CA) or CD195-APC (allophycocyanin) (clone J418F1; BioLegend), anti-CD192-APC or BV605 (CCR2; clone K036C2; BioLegend), anti-JAK2-Alexa Fluor 647 polyclonal (Thermo Fisher Scientific), anti-pSTAT3-Alexa Fluor 647 or PE/ CY7 (clone A160898, Thermo Fisher Scientific), and anti-pSTAT5-PE-Cy7 (clone A17016B.Rec, Thermo Fisher Scientific), anti-CD4-APC (clone: RPA-T4, BioLegend), and anti-human CXCR4-PE (clone:12C5, BioLegend). ...
Article
Full-text available
CCR5 serves as R5-tropic HIV co-receptor. Knocking out CCR5 in HIV patients, which has occurred <10 times, is believed important for cure. JAK/STAT inhibitors tofacitinib and ruxolitinib inhibit CCR5 expression in HIV + viremic patients. We investigated the association of JAK/STAT signaling pathway with CCR5/CCR2 expression in human primary CD4 + T cells and confirmed its importance. Six of nine JAK/STAT inhibitors that reduced CCR5/CCR2 expression were identified. Inhibitor-treated CD4 + T cells were relatively resistant, specifically to R5-tropic HIV infection. Furthermore, single JAK2, STAT3, STAT5A, and STAT5B knockout and different combinations of JAK/STAT knockout significantly reduced CCR2/CCR5 expression of both RNA and protein levels, indicating that CCR5/CCR2 expression was positively regulated by JAK-STAT pathway in CD4 + T cells. Serum and glucocorticoid-regulated kinase 1 (SGK1) knockout affected CCR2/CCR5 gene expression, suggesting that SGK1 is involved in CCR2/CCR5 regulation. If cell surface CCR5 levels can be specifically and markedly down-regulated without adverse effects, that may have a major impact on the HIV cure agenda.
... Besides, the ability of sublingual vaccines to trigger mucosal IgA relies on the use of effective vaccine adjuvants or supplementation [14;15] Liquid sublingual administration of HIV-1 model antigens has permitted to highlight the induction of a robust immune response and has provided a better understanding of the cellular mechanisms involved in the dissemination of the immune response from the delivery site to the distant effector sites. However, though promising, vaccine formulations as liquid dosage led to high heterogeneity of the induced immune responses, as exemplified by non-human primates immunized with a sublingual gp41-based subunit vaccine [16] or with replicationincompetent SIV DNA provirus [17]. ...
... A previous trial where we explored a SIV immunization with 3 doses of SIV DNA plus SIV-OPV in Cynomolgus monkeys provided significant T-cell responses but very limited anti-SIV antibodies (82). We hypothesized that, among other factors, the intracellular expression of the recombinant fragment expressed by OPV, which does not become part of the virion, may be a limiting factor in the development of a significant antibody response and opted to modify the vector to achieve secretion of the recombinant Env fragment from the poliovirus-infected cells by engineering an OPV vector that could secret recombinant antigens. ...
Article
Full-text available
We modified a Sabin Oral Poliovirus Vaccine (OPV) vector to permit secretion of the antigens of interest with the goal of improving anti-HIV Env humoral responses in a SHIV mucosal immunization composed of DNA and recombinant OPVs. We evaluated stimulation of systemic and mucosal cell-mediated and humoral immunity in Rhesus macaques by two regimens, both involving a prime with a SHIVBG505 DNA construct producing non-infectious particles formulated in lipid nanoparticles, administered in the oral cavity, and two different viral vector boostings, administered in the oral cavity and intestinally. Group 1 was boosted with rMVA-SHIVBG505, expressing SIV Gag/Pol and HIVBG505 Env. Group 2 was boosted with a SHIVBG505-OPV vaccine including a non-secreting SIVmac239CA-p6-OPV, expressing Gag CA, NC and p6 proteins, and a HIVBG505C1-V2-OPV, secreting the C1-V2 fragment of HIV EnvBG505, recognized by the broadly neutralizing antibody PG16. A time course analysis of anti-SHIV Gag and Env CD4+ and CD8+ T-cell responses in PBMC and in lymph node, rectal, and vaginal MNC was carried out. Both regimens stimulated significant cell-mediated responses in all compartments, with SHIVBG505-OPV immunization stimulating more significant levels of responses than rMVA- SHIVBG505. Boolean analysis of these responses revealed predominantly monofunctional responses with multifunctional responses also present in all tissues. Stimulation of antibody responses was disappointing in both groups with negative anti-SHIV IgG in plasma, and IgA in salivary, rectal and vaginal secretions being restricted to a few animals. After repeated rectal challenge with SHIVBG505, two Group 1 animals remained uninfected at challenge termination. No significant differences were observed in post-infection viral loads between groups. After the acute phase decline, CD4+ T cell percentages returned to normal levels in vaccinated as well as control animals. However, when compared to controls, vaccinate groups had more significant preservation of PBMC and rectal MNC Th17/Treg ratios, considered the strongest surrogate marker of progression to AIDS. We conclude that the vaccine platforms used in this study are insufficient to stimulate significant humoral immunity at the tested doses and schedule but sufficient to stimulate significant mucosal and systemic cell-mediated immunity, impacting the preservation of key Th17 CD4+ T cells in blood and rectal mucosa.
Article
Vaccination is an urgently needed and effective option to address epidemic, cancers, allergies, and other diseases. Nasal administration of vaccines offers many benefits over needle-based injection including high compliance and less risk of infection. Inactivated or attenuated vaccines as convention vaccine present potential risks of pathogenic virulence reversal, the focus of nasal vaccine development has shifted to the use of next-generation (subunit and nucleic acid) vaccines. However, subunit and nucleic acid vaccine intranasally have numerous challenges in development and utilization due to mucociliary clearance, mucosal epithelial tight junction, and enzyme/pH degradation. Nanoplatforms as ideal delivery systems, with the ability to enhance the retention, penetration, and uptake of nasal mucosa, shows great potential in improving immunogenic efficacy of nasal vaccine. This review provides an overview of delivery strategies for overcoming nasal barrier, including mucosal adhesion, mucus penetration, targeting of antigen presenting cells (APCs), enhancement of paracellular transportation. We discuss methods of enhancing antigen immunogenicity by nanoplatforms as immune-modulators or multi-antigen co-delivery. Meanwhile, we describe the application status and development prospect of nanoplatforms for nasal vaccine administration. Development of nanoplatforms for vaccine delivery via nasal route will facilitate large-scale and faster global vaccination, helping to address the threat of epidemics.
Article
Full-text available
The live-attenuated oral poliovirus vaccine (OPV or Sabin vaccine) replicates in gut-associated tissues, eliciting mucosa and systemic immunity. OPV protects from disease and limits poliovirus spread. Accordingly, vaccination with OPV is the primary strategy used to end the circulation of all polioviruses. However, the ability of OPV to regain replication fitness and establish new epidemics represents a significant risk of polio re-emergence should immunization cease. Here, we report the development of a poliovirus type 2 vaccine strain (nOPV2) that is genetically more stable and less likely to regain virulence than the original Sabin2 strain. We introduced modifications within at the 5′ untranslated region of the Sabin2 genome to stabilize attenuation determinants, 2C coding region to prevent recombination, and 3D polymerase to limit viral adaptability. Prior work established that nOPV2 is immunogenic in preclinical and clinical studies, and thus may enable complete poliovirus eradication.
Article
Full-text available
The power of mucosal anti-HIV-1 envelope immunoglobulins (Igs) to block virus transmission is underappreciated. We used passive immunization, a classical tool to unequivocally prove whether antibodies are protective. We mucosally instilled recombinant neutralizing monoclonal antibodies (nmAbs) of different Ig classes in rhesus macaques (RMs) followed by mucosal simian–human immunodeficiency virus (SHIV) challenge. We gave anti-HIV-1 IgM, IgG, and dimeric IgA (dIgA) versions of the same human nmAb, HGN194 that targets the conserved V3 loop crown. Surprisingly, dIgA1 with its wide-open, flat hinge protected 83% of the RMs against intrarectal R5-tropic SHIV-1157ipEL-p challenge, whereas dIgA2, with its narrow hinge, only protected 17% of the animals—despite identical epitope specificities and in vitro neutralization curves of the two dIgA isotypes (Watkins et al., AIDS 2013 27(9):F13-20). These data imply that factors in addition to neutralization determine in vivo protection. We propose that this underlying protective mechanism is immune exclusion, which involves large nmAb/virion aggregates that prevent virus penetration of mucosal barriers. Future studies need to find biomarkers that predict effective immune exclusion in vivo. Vaccine development strategies against HIV-1 and/or other mucosally transmissible pathogens should include induction of strong mucosal Abs of different Ig classes to defend epithelial barriers against pathogen invasion.
Article
Full-text available
The oral mucosa is an attractive site for mucosal vaccination, however the thick squamous epithelium limits antigen uptake. Here we utilize a modified needle-free injector to deliver immunizations to the sublingual and buccal (SL/B) tissue of rhesus macaques. Needle-free SL/B vaccination with modified vaccinia Ankara (MVA) and a recombinant trimeric gp120 protein generates strong vaccine-specific IgG responses in serum as well as vaginal, rectal and salivary secretions. Vaccine-induced IgG responses show a remarkable breadth against gp70-V1V2 sequences from multiple clades of HIV-1. In contrast, topical SL/B immunizations generates minimal IgG responses. Following six intrarectal pathogenic SHIV-SF162P3 challenges, needle-free but not topical immunization results in a significant delay of acquisition of infection. Delay of infection correlates with non-neutralizing antibody effector function, Env-specific CD4⁺ T-cell responses, and gp120 V2 loop specific antibodies. These results demonstrate needle-free MVA/gp120 oral vaccination as a practical and effective route to induce protective immunity against HIV-1.
Article
Full-text available
Perhaps the best-studied mucosal adjuvants are the bacterially derived ADP-ribosylating enterotoxins. This adjuvant family includes heat-labile enterotoxin of Escherichia coli (LT), cholera toxin (CT), and mutants or subunits of LT and CT. These proteins promote a multifaceted antigen-specific response, including inflammatory Th1, Th2, Th17, cytotoxic T lymphocytes (CTLs), and antibodies. However, more uniquely among adjuvant classes, they induce antigen-specific IgA antibodies and long-lasting memory to coadministered antigens when delivered mucosally or even parenterally. The purpose of this minireview is to describe the general properties, history and creation, preclinical studies, clinical studies, mechanisms of action, and considerations for use of the most promising enterotoxin-based adjuvant to date, LT(R192G/L211A) or dmLT. This review is timely due to completed, ongoing, and planned clinical investigations of dmLT in multiple vaccine formulations by government, nonprofit, and industry groups in the United States and abroad.
Article
Variety of conventional vaccine strategies tested against HIV-1 have failed to induce protection against HIV acquisition or durable control of viremia. Therefore, innovative strategies that can induce long lasting protective immunity against HIV chronic infection are needed. Recently, we developed an integration-defective HIV lentiDNA vaccine that undergoes a single cycle of replication in target cells in which most viral antigens are produced. A single immunization with such lentiDNA induced long-lasting T-cell and modest antibody responses in cynomolgus macaques. Here eighteen months after this single immunization, all animals were subjected to repeated low dose intra-rectal challenges with a heterologous pathogenic SIVmac251 isolate. Although the viral set point in SIVmac-infected cynomolgus is commonly lower than that seen in Indian rhesus macaques, the vaccinated group of macaques displayed a two log reduction of peak of viremia followed by a progressive and sustained control of virus replication relative to control animals. This antiviral control correlated with antigen-specific CD4+ and CD8+ T cells with high capacity of recall responses comprising effector and central memory T cells but also memory T cell precursors. This is the first description of SIV control in NHP model infected at 18 months following a single immunization with a non-integrative single cycle lentiDNA HIV vaccine. While not delivering sterilizing immunity, our single immunization strategy with a single-cycle lentivector DNA vaccine appears to provide an interesting and safe vaccine platform that warrants further exploration.
Article
South African trial halted early because of "futility."
Article
In the last decade, over a dozen potent broadly neutralizing antibodies (bnAbs) to several HIV envelope protein epitopes have been identified, and their in vitro neutralization profiles have been defined. Many have demonstrated prevention efficacy in preclinical trials and favorable safety and pharmacokinetic profiles in early human clinical trials. The first human prevention efficacy trials using 10 sequential, every-two-month administrations of a single anti-HIV bnAb are anticipated to conclude in 2020. Combinations of complementary bnAbs and multi-specific bnAbs exhibit improved breadth and potency over most individual antibodies and are entering advanced clinical development. Genetic engineering of the Fc regions has markedly improved bnAb half-life, increased mucosal tissue concentrations of antibodies (especially in the genital tract), and enhanced immunomodulatory and Fc effector functionality, all of which improve antibodies' preventative and therapeutic potential. Human-derived monoclonal antibodies are likely to enter the realm of primary care prevention and therapy for viral infections in the near future.
Article
In this Review, we highlight some recent developments in the discovery and application of broadly neutralizing antibodies (bnAbs) to human immunodeficiency virus (HIV); i.e., antibodies able to neutralize diverse isolates of HIV. We consider the characterization of novel bnAbs, recent data on the effects of bnAbs in vivo in humans and animal models, and the importance of both kinds of data for the application of Abs to prophylaxis and therapy and to guide vaccine design. We seek to place newly discovered bnAbs in the context of existing bnAbs, and we explore the various characteristics of the antibodies that are most desirable for different applications.
Article
The majority of human immunodeficiency virus (HIV) type 1 infections in infants are acquired orally through breastfeeding. Toward development of a pediatric HIV vaccine to prevent breastmilk transmission, we tested the efficacy of a simultaneous oral and intramuscular (IM) vaccination regimen for preventing oral simian immunodeficiency virus (SIV) transmission in infant rhesus macaques. Two groups of neonatal macaques were immunized with DNA encoding SIV virus-like particles (DNA-SIV) on weeks 0 and 3, then boosted with modified vaccinia Ankara (MVA) virus expressing SIV antigens (MVA-SIV) on weeks 6 and 9. One group was prime/boosted by the IM route only. Another group was immunized with DNA by both the IM and topical oral (O) buccal routes, and boosted with MVA-SIV by both the IM and sublingual (SL) routes. A third group of control animals received saline by O + IM routes on weeks 0 and 3, and empty MVA by SL + IM routes on weeks 6 and 9. On week 12, infants were orally challenged once weekly with SIVmac251 until infected. The vaccine regimen that included oral routes resulted in reduced peak viremia. The rate of infection acquisition in vaccinated infants was found to be associated with prechallenge intestinal immunoglobulin G (IgG) responses to SIV gp120 and V1V2. Peak viremia was inversely correlated with postinfection intestinal IgG responses to gp120, gp41, and V1V2. These results suggest that codelivery of a pediatric HIV vaccine by an oral route may be superior to IM-only regimens for generating mucosal antibodies and preventing HIV breastmilk transmission in neonates.
Article
Background A pediatric vaccine to prevent breast milk transmission of human immunodeficiency virus (HIV) may generate greater immune responses at viral entry sites if given by an oral route. Methods We compared immune responses induced in juvenile macaques by prime/boosting with simian immunodeficiency virus (SIV)‐expressing DNA/modified vaccinia Ankara virus (MVA) by the intramuscular route (IM), the oral (O)/tonsillar routes (T), the O/sublingual (SL) routes, and O+IM/SL routes. Results O/T or O/SL immunization generated SIV‐specific T cells in mucosal tissues but failed to induce SIV‐specific IgA in saliva or stool or IgG in plasma. IM/IM or O+IM/SL generated humoral and cellular responses to SIV. IM/IM generated greater frequencies of TFH in spleen, but O+IM/SL animals had higher avidity plasma IgG and more often demonstrated mucosal IgA responses. Conclusion These results suggest that codelivery of HIV DNA/MVA vaccines by the oral and IM routes might be optimal for generating both systemic and mucosal antibodies.