SlideShare a Scribd company logo
1 of 11
Download to read offline
Published Ahead of Print 6 May 2009.
2009, 83(14):7166. DOI: 10.1128/JVI.00374-09.J. Virol.
Nabel
Schwartz, C. Richter King, Jason G. D. Gall and Gary J.
Kong, Masaru Kanekiyo, David Einfeld, Richard M.
Lingshu Wang, Cheng Cheng, Sung-Youl Ko, Wing-Pui
Enhanced Mucosal Cellular Immunity
Vaccine Vectors to the Intestine Induces
Delivery of Human Immunodeficiency Virus
http://jvi.asm.org/content/83/14/7166
Updated information and services can be found at:
These include:
SUPPLEMENTAL MATERIAL Supplemental material
REFERENCES
http://jvi.asm.org/content/83/14/7166#ref-list-1at:
This article cites 48 articles, 16 of which can be accessed free
CONTENT ALERTS
more»articles cite this article),
Receive: RSS Feeds, eTOCs, free email alerts (when new
http://journals.asm.org/site/misc/reprints.xhtmlInformation about commercial reprint orders:
http://journals.asm.org/site/subscriptions/To subscribe to to another ASM Journal go to:
onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
JOURNAL OF VIROLOGY, July 2009, p. 7166–7175 Vol. 83, No. 14
0022-538X/09/$08.00ϩ0 doi:10.1128/JVI.00374-09
Copyright © 2009, American Society for Microbiology. All Rights Reserved.
Delivery of Human Immunodeficiency Virus Vaccine Vectors to the
Intestine Induces Enhanced Mucosal Cellular Immunityᰔ
†
Lingshu Wang,1
‡ Cheng Cheng,1
‡ Sung-Youl Ko,1
Wing-Pui Kong,1
Masaru Kanekiyo,1
David Einfeld,2
Richard M. Schwartz,1
C. Richter King,2
Jason G. D. Gall,2
and Gary J. Nabel1
*
Vaccine Research Center, NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC-3005, 40 Convent Drive, Bethesda,
Maryland 20892-3005,1
and GenVec, Inc., 65 West Watkins Mill Rd., Gaithersburg, Maryland 208782
Received 19 February 2009/Accepted 30 April 2009
Effective vaccines for human immunodeficiency virus type 1 (HIV-1) will likely need to stimulate protective
immunity in the intestinal mucosa, where HIV-1 infection causes severe CD4؉
T-cell depletion. While repli-
cation-competent recombinant adenovirus (rAd) vectors can stimulate adenovirus-specific mucosal immunity
after replication, oral delivery of replication-defective rAd vectors encoding specific immunogens has proven
challenging. In this study, we have systematically identified barriers to effective gut delivery of rAd vectors and
identified sites and strategies to induce potent cellular and humoral immunity. Vector-mediated gene transfer
by rAd5 was susceptible to low-pH buffer, gastric and pancreatic proteases, and extracellular mucins. Using ex
vivo organ explants, we found that transduction with rAd5 was highest in the ileum and colon among all
intestinal segments. Transgene expression was 100-fold higher after direct surgical introduction into the ileum
than after oral gavage, with rAd5 showing greater potency than the rAd35 or the rAd41 vector. A single
immunization of rAd5 encoding HIV-1 gp140B to the ileum stimulated potent CD8؉
T-cell responses in the
intestinal and systemic compartments, and these responses were further enhanced by intramuscular rAd5
boosting. These studies suggest that induction of primary immune responses by rAd5 gut immunization and
subsequent systemic boosting elicits potent antigen-specific gut mucosal responses.
Human immunodeficiency virus type 1 (HIV-1) infection is
characterized by uncontrolled virus replication and cytopath-
icity in the intestinal mucosa, the site of major T-cell depletion
after primary infection. The gastrointestinal (GI) tract is the
predominant site of a pronounced CD4ϩ
T-cell loss in the
early stages of HIV infection and simian immunodeficiency
virus (SIV) infection in the nonhuman primate model (3, 23,
26, 43). It has been suggested that a mucosal vaccine which
generates HIV-specific CD8ϩ
T cells in the gut could prevent
the loss of CD4ϩ
cells in gut-associated lymphoid tissue, es-
tablishment of infection, or spread of virus (13, 34). Therefore,
targeted delivery of vaccines to the GI tract to stimulate mu-
cosal responses has the potential to improve the efficacy of
immune protection against HIV-1; however, the site of gene-
based transduction and the barriers to vaccine delivery have
not been well defined.
Adenoviruses (Ads) have been used extensively as vectors
for both gene transfer and vaccine development. They offer
several advantages as tools for vaccine delivery, such as the
ability to transduce both dividing and nondividing cells, rela-
tive safety and stability in vivo, ease of production in high
titers, and lack of integration (2, 35). These vectors are prom-
ising because parenteral administration in both animals and
humans has been shown to generate strong and long-lasting
humoral and cellular immune responses. The immune re-
sponses surpass those achieved with other types of gene vectors
and genetic vaccines (5, 38, 46). As a result, recombinant Ad
(rAd) vectors have been developed and tested as vaccine
vehicles to immunize against a number of pathogens (4, 10, 15,
18, 41).
Orally (p.o.) delivered vaccines are attractive in theory be-
cause of their ease of administration and potential to deliver
antigen to gut-associated lymphoid tissue, permitting induction
of immune responses in both mucosal and systemic compart-
ments. At the same time, p.o. delivery of replication-defective
rAd vectors has posed a challenge and has met with variable
levels of success. Immunization with rAd5 encoding rabies
virus antigens, influenza virus antigens, or other antigens has
generated some protection against infection in animal models
(9, 27, 31, 39, 41), but p.o. immunization has elicited much
lower CD8ϩ
T-cell responses than systemic delivery (33), and
a much higher dose is required to induce immune responses
(37). We have recently shown in an HIV vaccine model that
rAd41, a human enteric Ad-based vector, induced potent
CD8ϩ
T-cell responses in both systemic and mucosal compart-
ments when primed p.o. or in the ileum (17). The previous
study showed that rAd41 vectors delivered through direct ileal
injection elicited mucosal cell immunity, but whether other
rAd vectors could stimulate these responses and which factors
affected delivery and immunogenicity were unknown. In this
report, we have investigated the mechanisms associated with
the low immunogenicity of rAd5 dosed through the p.o. route
in mice. The purpose was to identify barriers to effective de-
livery of rAd vectors to gut tissues and to ascertain sites and
strategies for induction of potent cellular and humoral immu-
nity. To investigate the mechanism of the low immunogenicity
* Corresponding author. Mailing address: Vaccine Research Center,
NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC-
3005, 40 Convent Drive, Bethesda, MD 20892-3005. Phone: (301)
496-1852. Fax: (301) 480-0274. E-mail: gnabel@nih.gov.
† Supplemental material for this article may be found at http://jvi
.asm.org/.
‡ These authors contributed equally to this report.
ᰔ
Published ahead of print on 6 May 2009.
7166
onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
of rAd vectors through the p.o. route and develop effective
delivery of rAd5 and rare serotype rAd35 vectors as gut mu-
cosal HIV vaccines, we have analyzed the obstacles to p.o.
immunization, characterized vector transgene expression, and
systematically compared immune responses induced by p.o.
and local immunization strategies. These studies demonstrated
that the higher immune responses were strongly associated
with higher gene expression in the intestine and support fur-
ther study of gut mucosal immunization in SIV challenge mod-
els as a potential HIV vaccine strategy.
MATERIALS AND METHODS
Animals and viruses. Six- to 8-week-old female BALB/c mice were purchased
from Jackson Laboratories and housed in the experimental animal facility of the
Vaccine Research Center, NIAID, NIH (Bethesda, MD). All animal experi-
ments were reviewed and approved by the Animal Care and Use Committee,
VRC, NIAID, NIH, and performed in accordance with all relevant federal and
NIH guidelines and regulations.
rAd5 vectors are replication-defective E1-, E3-, and E4-deleted human Ads,
and rAd35 vectors are E1- and E3-deleted replication-defective vectors. rAd35/
41L and rAd35/41S vectors are rAd35-based vectors with chimeric Ad41 long
fiber or short fiber, respectively. rAd5-luc, rAd35-luc, rAd35/41L-luc, and rAd35/
41S-luc encode luciferase reporter genes under the control of the cytomegalo-
virus promoter/enhancer. rAd5-gp140B, rAd35-gp140B, and rAd35/41L- or
rAd35/41S-gp140B (rAd35 with Ad41 long or short fiber, respectively) encode
gp140⌬CFI⌬V1V2 of HIV-1 clade B (20). Ad vectors were propagated in 293-
ORF6 cells and purified by cesium chloride gradients (6).
Mouse intestinal explants. Explant culture medium consisted of Dulbecco’s
modified Eagle’s medium–F-12 medium supplemented with 5% fetal bovine
serum, L-glutamine (2 mM), penicillin (100 U/ml), streptomycin (100 ␮g/ml),
gentamicin (50 ␮g/ml), and epidermal growth factor (10 U/ml). All media and
supplements were purchased from Invitrogen, Inc. (Carlsbad, CA). Mouse in-
testines were isolated and cut open longitudinally. After being washed thor-
oughly, the tissues were cut into 5- by 5-mm pieces and placed on presoaked
Gelfoam (Pfizer, New York, NY), with the epithelium uppermost, in 24-well
plates containing 0.3 ml of culture medium. Four microliters of virus solution
containing 1 ϫ 108
to 5 ϫ 108
virus particles (VP) of treated or untreated rAd
vectors encoding luciferase was applied directly to the upper surface of each
explant. After overnight incubation, the tissues were rinsed in phosphate-buff-
ered saline (PBS) and placed in 0.1% Triton X-100. Following a 1-min ultra-
sound treatment and three freeze-thaw cycles, the lysates were centrifuged at
2,000 rpm for 10 min. The supernatants were used for a luciferase assay. The
luminescence was measured using a microplate scintillation and luminescence
counter (PerkinElmer, Shelton, CT). The protein concentration of the cleared
supernatant was determined using a DC protein microplate assay kit (Bio-Rad,
Hercules, CA). The results are shown as relative luminescence units per ␮g
protein.
For the acid and protease sensitivity study, rAd5-luc was (i) treated with 0.1 M
HCl and 2.5 mg/ml pepsin from porcine gastric mucosa (Sigma, St. Louis, MO)
for 5 min at 37°C and neutralized with 10ϫ THB (200 mM Tris, 500 mM HEPES,
1.5 M NaCl), (ii) treated with 25 ␮g/ml trypsin from bovine pancreas (Sigma, St.
Louis, MO) or 2.5 U/ml chymotrypsin (Sigma, St. Louis, MO) for 30 min at 37°C,
or (iii) exposed to the combination of treatments with HCl-pepsin and trypsin
and/or chymotrypsin and then applied to the tissue explants.
To test the susceptibility of intestinal segments to Ad vectors, mouse duode-
num (1 cm downstream of the stomach), jejunum (median part of the small
intestine), ileum (from the junction of the ileum and cecum to 2 cm upstream of
the cecum), and colon (2 cm downstream of the cecum) were isolated and either
washed thoroughly (native) or gently stripped to remove the mucus for culture.
Ten pieces from three mice for each segment were cultured with 4 ␮l virus
solution containing 1 ϫ 108
VP of rAd5-luc and harvested as described above.
Treatment of intestinal explants with DTT, hyaluronidase, and exoglycosi-
dases. The mouse intestinal explants were either not treated or treated with
dithiothreitol (DTT) (20 mM, catalogue number 43816; Sigma, St. Louis, MO) or
hyaluronidase (1 or 10 mg/ml, catalogue number H 3506; Sigma, St. Louis, MO)
for 30 min or exoglycosidases from Trimeresurus cornutus (1, 5, or 10 mg/ml;
Seikagaku Biobusiness, Tokyo, Japan) for 2 min at 37°C. The explants were then
rinsed in the prewarmed medium and cultured with 4 ␮l virus solution containing
1 ϫ 108
VP of rAd5-luc. The stripped jejunum and the untreated ileum were
used as the positive controls, while the medium only was used for the negative
controls. The tissue explants were harvested as described above.
Gene expression in mouse intestine transduced with rAd5-luc in vivo. Three
BALB/c mice per group were either administered p.o. 1010
VP of rAd5-luc in 500
␮l PBS or given an intraileal injection with 1010
VP of rAd5-luc in 100 ␮l PBS.
Twenty-four hours after injection, the mice were sacrificed, and intestinal seg-
ments, including duodenum, jejunum, ileum, colon, and mesenteric lymph nodes
(MLN), were lysed and assayed for luciferase activity. The group injected with
100 ␮l rAd5 empty vector via the ileum lumen was used as the control. The
intraileal-injection procedure was described previously (17).
Immunization. For the immunogenicity study, immune responses induced by
a single injection of rAd5 encoding HIV-1 gp140B into the ileum lumen were
tested. Five mice from each group were immunized with 1010
VP of rAd5-gp140B
vector in 500 ␮l PBS p.o. or with 1010
VP of rAd5-gp140B vector in 100 ␮l PBS
via ileal injection as described above. Mice that received rAd5 empty vector p.o.
or via ileal injections were used as the negative controls. Three weeks after
inoculation, peripheral blood mononuclear cells (PBMC), spleens, and small
intestines were collected for H-2Dd
/PA-9 tetramer staining. The spleens were
also used for detecting HIV-1 peptide-specific cytokine-producing CD4ϩ
or
CD8ϩ
T lymphocytes. The serum immunoglobulin G (IgG) antibodies were
determined by enzyme-linked immunosorbent assay (ELISA).
In the prime-boost experiments, five mice per group were primed with 1010
VP
of rAd5-gp140B via p.o. dosing or with rAd5, rAd35, Ad35/41L, and rAd35/41S,
all encoding gp140B, via intraileal injections. Three weeks after immunization,
blood was collected and PBMC were isolated and stained for H-2Dd
/PA-9
tetramer. These mice were then boosted intramuscularly (i.m.) with rAd5-
gp140B at 109
VP. Two weeks after the boost, PBMC, spleens, MLN, and small
intestines were harvested for H-2Dd
/PA-9 tetramer staining, and the spleens
were also used for detecting HIV-1 peptide-specific cytokine-producing CD4ϩ
or
CD8ϩ
T lymphocytes. The IgG and IgA antibodies in the sera and vaginal washes
were determined by ELISA.
Lymphocyte isolation. The isolation of lymphocytes from blood, spleen, and
MLN was described previously (17). For lymphocyte isolation from the gut, the
small intestines from individual mice were collected in cold medium. The intes-
tine was opened and washed thoroughly following excision of Peyer’s patches.
Intestinal sections were then minced and digested with 0.5 mg/ml collagenase II
(C-6885; Sigma) for 30 min at 37°C. The resulting supernatants were filtered
through a 40-␮m cell strainer. After centrifugation, the cell pellets were sus-
pended in 6 ml of 40% Percoll (Amersham Biosciences, Piscataway, NJ) and
overlaid on 4 ml of 75% Percoll. The samples were then subjected to centrifu-
gation at 800 ϫ g for 20 min at 22°C. The cells from the interface of 75% and
40% Percoll were collected, washed with a large volume of medium, and
counted.
Tetramer staining and ICS. Lymphocytes isolated from blood, spleen, MLN,
and the small intestine were used for tetramer staining. The splenic lymphocytes
from individual mice were used for intracellular cytokine staining (ICS), while
the combined intestinal cells from the group of five mice were also used for ICS
in the rAd35/41 chimeric fiber experiment, using ViViD dye (Invitrogen Life
Sciences, Carlsbad, CA) to exclude nonviable cells. The detailed methods for
tetramer staining and ICS were described previously (M. Honda, R. Wang, W.-P.
Kong, M. Kanekiyo, W. Akahata, L. Xu, K. Matsuo, K. Natarajan, H. Robinson,
T. E. Asher, D. A. Price, D. C. Douek, D. H. Margulies, and G. J. Nabel,
submitted for publication; 17).
ELISA for IgG and IgA antibodies to HIV Env. Levels of HIV gp140B-specific
IgG and IgA in the sera or vaginal washes of vaccinated mice were assessed using
ELISA (17). The sera were diluted at 1:1,000 for IgG and 1:50 for IgA, while the
vaginal washes were diluted at 1:3 for the detection of both IgG and IgA.
Absorbance at 450 nm was determined by a Spectra Max instrument (Molecular
Devices).
Data and statistical analysis. All results are presented as means with standard
errors. Statistical analyses were performed based upon comparisons between the
control groups and the treated groups or between differently treated groups by
using the two-tailed Student t test. P values of less than 0.05 were considered
statistically significant.
RESULTS
Determination of barriers to effective delivery of rAd5 vec-
tors to the GI tract. To identify potential mechanisms that
inhibit transduction of the GI tract after rAd gene transfer
after p.o. delivery, we first examined the transduction of mouse
VOL. 83, 2009 INTRAILEAL rAd5 VACCINE STIMULATES MUCOSAL IMMUNITY 7167
onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
intestinal explants by an rAd5 vector expressing luciferase.
Intestinal explants of the duodenum, jejunum, ileum, and co-
lon were cultured ex vivo, and the relative rAd transduction
efficiencies were determined. The ileum and the colon were
transduced at significantly higher levels than were the duode-
num and jejunum (Fig. 1A, open bars). To determine whether
the superficial layer played a role in the inhibition of transduc-
tion of intestinal cells by rAd, mucus was removed by mechan-
ical and biochemical means. Mechanical removal of the mucus
significantly enhanced the transduction activity in all segments
(P Ͻ 0.05 for the duodenum and jejunum, P Ͻ 0.001 for the
ileum) except for the colon (Fig. 1A, filled versus open bars).
The differences between transductions of native and mucus-
stripped explants were 100-fold in the duodenum and jejunum
and 10-fold in the ileum. Previous in vitro studies have shown
that intestinal mucus can also be dissolved by a variety of
agents, including enzymes, detergents, and sulfhydryl com-
pounds (1, 28). The effect of DTT, hyaluronidase, and glyco-
sidases on the transduction of intestinal explants by rAd5 was
investigated using explants treated and exposed to vector ex
vivo. Transduction of explants treated with DTT was signifi-
cantly higher than transduction of those that were untreated,
specifically in the upper intestinal segments, duodenum and
jejunum (Fig. 1B, filled versus open bars). When mouse jeju-
num was treated with hyaluronidase or exoglycosidases (see
Materials and Methods), transduction was significantly higher
than that of the jejunum treated with medium (Fig. 1C and D,
1 mg/ml). However, high concentrations of these enzymes did
not enhance transduction, possibly due to nonspecific cell
damage.
We next examined the effect of acid and selected proteases
on rAd5 transduction of mouse intestinal explant cultures.
Gastric acid and pepsin are the major components secreted
into the stomach, and trypsin and chymotrypsin are the major
proteases produced by the pancreas. To mimic the passage of
rAd through the gastric environment and into the proximal
section of the small intestine, rAd5-luciferase vector was
treated with combinations of acid and protease and then ap-
plied to the tissue explants. Although we did not observe a
significant decrease in luciferase activity after acid-pepsin,
trypsin, or chymotrypsin treatment alone, the combination of
acid-pepsin treatment with either trypsin or chymotrypsin
treatment significantly reduced transgene expression (Fig. 2A,
lanes 5 and 6). The combination of acid-pepsin with both
trypsin and chymotrypsin further reduced luciferase activity, to
approximately 100-fold lower than transduction with untreated
rAd5-luc (Fig. 2A, lane 7). This result suggested that rAd5
vectors were susceptible to degradation by gastric proteases.
Taken together, these ex vivo experiments indicated that mul-
tiple barriers reduce transduction of intestinal epithelia by
rAd5 vectors and demonstrated differential transduction of
specific intestinal segments.
Development of intraileal delivery to increase gene delivery
to the small intestine. Since the untreated ileum was trans-
FIG. 1. The distal regions of the intestine were efficiently transduced by rAd5, and removal of luminal substances increased transduction of the
proximal intestine. (A) Efficient transduction of intestinal segments. Segments of intestine were left untreated (native), or the mucus was gently
removed (stripped) and then transduced with rAd5-luc. (B) Treatment of intestinal segments with DTT increased the transduction efficiency of
rAd5-Luc. (C and D) Effect of removal of the glycocalyx and hyaluronic acid on transduction. Mouse jejuna were treated with hyaluronidase (C) or
mixed glycosidases (D) prior to transduction with rAd5-luc. *, P Ͻ 0.05; **, P Ͻ 0.01; ***, P Ͻ 0.001. RLU, relative luminescence units.
7168 WANG ET AL. J. VIROL.
onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
duced most effectively in ex vivo organ culture, we developed
an in vivo route of administration that targeted the ileum and
bypassed the significant barriers encountered by rAd in the GI
tract. It has been reported that receptor binding and uptake of
rAd5 occur relatively quickly, with the majority (80 to 85%) of
the bound virions being taken up by permissive cells within 5 to
10 min (12). We found that a 10-min exposure of ileum ex-
plants to rAd5 resulted in levels of transgene expression similar
to those obtained with longer time exposures (see Fig. S1 in
the supplemental material). Therefore, for in vivo studies,
the ileum loop was formed to hold the virus solution for 20 min
to ensure efficient binding of viruses within the intestine and
the separation of virus from undigested food in the ileum.
To determine the relative efficiency of rAd5-mediated trans-
duction, mice were administered rAd5 (1010
VP) encoding
luciferase either p.o. or via the intraileal-injection method.
Twenty-four hours later, mice were sacrificed, and the intesti-
nal segments were assayed for luciferase activity. A low level of
luciferase activity was detected in the ileum after p.o. admin-
istration, but the luciferase activity was not significantly differ-
ent from that for the empty rAd5 control group (Fig. 2B) (P Ͼ
0.05). Following intraileal injection, there was approximately a
10- to 100-fold-higher luciferase activity in the ileum and MLN
than for the control group (Fig. 2B) (P Ͻ 0.05). Importantly,
the luciferase activity in the ilea from injected mice was signif-
icantly higher than that for the p.o. dosed mice (Fig. 2B) (P Ͻ
0.05).
Immunization via intraileal injection generated humoral
and cellular responses in both mucosal and systemic compart-
ments. The higher efficiency of intestinal cell transduction ob-
served with ileal injection than with p.o. dosing suggested the
possibility that intraileal administration of rAd vectors would
FIG. 2. Transduction of intestinal explant cultures by rAd5-luc was reduced by exposure to low pH and proteases and enhanced gene expression
by intraileal delivery of rAd5 in vivo. (A) The rAd vector encoding luciferase was treated as indicated (ϩ, treatment; Ϫ, no treatment) and applied
to explants of mouse ileum. *, P Ͻ 0.05. (B) Mice were administered rAd5-luc via p.o. or intraileal injection, and the levels of luciferase activity
in intestinal segments and MLN were determined 24 h later. *, P Ͻ 0.05. RLU, relative luminescence units.
VOL. 83, 2009 INTRAILEAL rAd5 VACCINE STIMULATES MUCOSAL IMMUNITY 7169
onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
induce a more potent immune response to the encoded trans-
gene. Mice received a single administration of rAd5-gp140B
(1010
VP) p.o. or via intraileal injection. Three weeks later, the
mice immunized with rAd5-gp140B via intraileal injection
showed significantly higher levels of H-2Dd
/PA-9ϩ
CD8ϩ
T
cells in both the small intestine and the systemic compartments
than did the mice immunized p.o. (Fig. 3A). Oral administra-
tion did not result in detectable responses higher than back-
ground (Fig. 3A) (P Ͼ 0.05). The average levels of H-2Dd
/
PA-9ϩ
CD8ϩ
T cells in the mice given an intraileal injection
FIG. 3. Mucosal and systemic immune responses induced by a single intraileal injection of rAd5-gp140B. Mice were immunized with
rAd5gp-140B (rAd5) or rAd5 empty vector (Ϫ) via the p.o. or the intraileal route as indicated. (A) Percentages of HIV-specific H-2Dd
/PA-9ϩ
CD8ϩ
T cells from PBMC, spleens, and small intestines. (B) IgG antibody in the serum. OD450nm, absorbance at 450 nm. (C) Percentages of
HIV-specific IL-2-, IFN-␥-, and TNF-␣-secreting CD4ϩ
or CD8ϩ
T cells from the spleens. The data from each individual mouse and the mean
values from the five mice (black bars) are shown. *, P Ͻ 0.05; **, P Ͻ 0.01.
7170 WANG ET AL. J. VIROL.
onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
were 7.11% (ranging from 2 to 11%) in PBMC, 3.47% (ranging
from 1.44 to 5.82%) in the spleen, and 5.53% (ranging from
2.13 to 9.65%) in the small intestine. HIV Env-specific IgG
responses were also detected in sera from mice immunized via
the intraileal injection but not in sera from the p.o. dosed mice
(Fig. 3B). ICS analysis of splenocytes was consistent with the
tetramer staining results; numbers of HIV-1-specific CD4ϩ
and CD8ϩ
T cells producing interleukin-2 (IL-2), gamma in-
terferon (IFN-␥), and tumor necrosis factor alpha (TNF-␣)
were significantly higher in the mice that received rAd5-
gp140B via intraileal injection than in the mice that received
equal amounts of rAd5-gp140B p.o. (Fig. 3C). In this analysis,
the magnitudes of CD8ϩ
T-cell response differed between tet-
ramer staining and ICS, suggesting that there was a dispropor-
tionate representation of the dominant tetramer response or
that not all of the tetramer-positive CD8ϩ
T cells were secret-
ing the measurable cytokines, a dichotomy described to occur
previously when using these techniques (42). Tetramer staining
measures CD8ϩ
T cells responding to an immunodominant
HIV-Env peptide regardless of function, including inactivated
cells and activated cells, while ICS detects only activated cells
or functional cells. Oral immunization generated only a back-
ground level of cytokine-producing CD4ϩ
or CD8ϩ
T cells.
These data demonstrate that the rAd5 vector induced substan-
tial antigen-specific cellular and humoral immune responses
after a single intraileal immunization, while p.o. immunization
of the same amount of rAd5 did not induce any detectable
immune responses.
A mucosal prime-systemic boost regimen elicited potent cel-
lular immunity in the small intestine and the systemic com-
partments. The relative potencies of p.o. and intraileal routes
of administration as primes for systemic boosting were next
examined. Mice were primed on day 0 with empty rAd5 or
rAd5-gp140B p.o. or via intraileal injection and were boosted
i.m. on day 21 with rAd5-gp140B. Before the boosting step,
PBMC were harvested for tetramer staining, and consistent
with the results described above, immunization with rAd5-
gp140B via intraileal injections induced significantly higher
responses than p.o. administration (Fig. 4A). After the mice
were boosted with rAd5-gp140B i.m., the mice primed via the
intraileal route showed a dramatic increase in antigen-specific
H-2Dd
/PA-9ϩ
CD8ϩ
T-cell responses in both systemic and
mucosal compartments compared to the responses of the
group primed p.o. or i.m. (Fig. 4B). HIV-1 peptide-specific
cytokine-producing CD8ϩ
T cells and CD4ϩ
T cells were also
boosted effectively in the mice given an ileal prime and an i.m.
boost. On average, 12.44% of CD8ϩ
splenic T cells produced
IFN-␥, and 10.65% of CD8ϩ
splenic T cells were TNF-␣ pos-
itive (Fig. 4C). The mice primed p.o. or via ileal injection and
boosted i.m. generated higher levels of HIV-1 peptide-specific
IFN-␥- and TNF-␣-producing CD4ϩ
T cells than the mice
immunized by i.m. injection only (Fig. 4C). There were no
significant differences in CD4 frequencies between the prime-
boost groups. The only differences were relative to the single-
immunization group, indicating that prime-boost regimens in-
duce higher frequencies of immune cells than a single
immunization. HIV-1-specific IgG and IgA antibodies were
also detected in sera and vaginal washes, but the differences
between the mice primed p.o. and the mice primed by ileal
injection were not significant (see Fig. S2 in the supplemental
FIG. 4. Immune responses induced by ileum prime and i.m. boost.
Mice were administered rAd5gp-140B (rAd5) or empty vector (Ϫ) via
the p.o. or intraileal route and boosted i.m. (ϩ) 3 weeks later. (A) Per-
centages of HIV-specific H-2Dd
/PA-9ϩ
CD8ϩ
T cells from the PBMC
immediately prior to the i.m. injection of rAd5-gp140B. (B) Percent-
ages of HIV-specific H-2Dd
/PA-9ϩ
CD8ϩ
T cells from the PBMC,
spleens, MLN, and small intestines at 2 weeks after the i.m. injection
of rAd5-gp140B. (C) HIV-1-specific IFN-␥- and TNF-␣-secreting
CD4ϩ
or CD8ϩ
T lymphocytes from the spleens 2 weeks after i.m.
injection of rAd5-gp140B. The data from each individual mouse and
the mean values from the five mice (black bars) are shown. *, P Ͻ 0.05;
**, P Ͻ 0.01; ***, P Ͻ 0.001.
VOL. 83, 2009 INTRAILEAL rAd5 VACCINE STIMULATES MUCOSAL IMMUNITY 7171
onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
material). These results demonstrate that priming directly in
the ileum and boosting i.m. induced the most potent CD8ϩ
T-cell immunity in the small intestine as well as in the systemic
compartments.
The fiber proteins of Ad41 did not increase the mucosal
immunogenicity of a rare serotype vector, rAd35. The swap-
ping of Ad5 fiber proteins into Ad35 has previously been
shown to increase immunogenicity (29). Since we have seen
intestinal and systemic immune responses induced by rAd41
vectors of a magnitude similar to that of responses induced by
rAd5 (17), we determined whether the swap of Ad41 proteins
into rAd35 would affect rAd35-mediated transduction and im-
mune responses. Explant cultures of murine intestine were
transduced with rAd35, rAd35/41L, and rAd35/41S, encoding
luciferase at a 1- to 2-log-lower efficiency than that for rAd5
(Fig. 5A). We also compared the relative potencies of the
rAd35 and rAd5 vectors in priming for an i.m. rAd5 boost.
While an intraileal prime-i.m. boost with rAd5-gp140B elicited
significantly higher levels of CD8ϩ
T-cell responses in the
spleen and in the small intestine than did rAd5 i.m. alone,
intraileal administration of rAd35, rAd35/41L, and rAd35/41S
failed to increase the rAd5 i.m boost (Fig. 5B and C, right, lane
3 versus lane 2, 4, 5, or 6). However, CD4ϩ
T-cell responses did
not show a difference after rAd5 boost, possibly due to the
lower magnitude of responders than CD8ϩ
cells typically seen
with rAd5 immunization. In addition, an ileal prime and i.m.
boost with rAd5-gp140B induced a twofold-greater IFN-␥ϩ
CD8ϩ
T-cell response in the small intestine than did rAd5 i.m.
alone. In contrast, an ileal prime with rAd35, rAd35/41L, and
rAd35/41S did not show a boost response in the small intestine
higher than that with rAd5 i.m. alone (see Fig. S3 in the
supplemental material). These results demonstrate that the
direct intraileal injection of rAd5, but not that of rAd35,
rAd35/41L, and rAd35/41S, could prime for an i.m. rAd5 boost
to generate potent immune responses in the systemic and mu-
cosal compartments.
DISCUSSION
The intestinal mucosa represents a primary site of HIV
replication during the peak of infection (3, 23, 26, 43). Vacci-
nation through the GI mucosa could potentially induce muco-
sal immunity to prevent virus entry and/or limit the spread of
infection. Oral immunization is the most convenient way to
deliver mucosal vaccines. Although several studies have re-
ported that the use of this route of immunization of Ad vectors
encoding various virus antigens generated antibody responses
in the serum and mucosal secretions (33, 37), the administra-
tion of Ads encoding Gag or Env from HIV-1 or SIV elicited
weak or undetectable intestinal mucosa responses (24, 30). The
major obstacles to successful induction of mucosal immunity
and/or systemic immunity via GI delivery of vaccine have not
been defined previously. In this study utilizing murine intesti-
nal explant cultures and in vivo gene expression assessments,
we demonstrate that the combination of a low-pH environ-
ment and the presence of proteases as well as the mucus/
glycocalyx in the GI tract prevents rAd5 from effectively trans-
ducing the intestinal cells when delivered p.o. Therefore, p.o.
immunization of vaccine vectors must overcome or bypass
these barriers in the GI tract in order to be effective. We also
show that the ileum is the most receptive site for rAd trans-
duction and that intraileal injection of rAd5 results in a signif-
icantly higher transgene expression in the ileum than does p.o.
delivery. In addition, intraileal immunization with rAd5 did not
induce significant systemic antivector neutralizing activity,
since the rAd5 i.m. administration was effective in the homol-
ogous rAd5 prime-rAd5 boost regimen. This is in agreement
with our previous work demonstrating the absence of circulat-
ing neutralizing antibodies following p.o. administration of
rAd5 and rAd41 (17). It has also been shown that preexisting
immunity to the vaccine carrier did not impair p.o. vaccination
with Ad vectors in mice (45). Thus, the ileum represents a valid
target for delivery of rAd vectors.
Utilization of a route of administration that bypassed the
barriers to ileal transduction revealed the potency of rAd5 as a
mucosal immunogen. A single immunization of mice with rAd5
encoding HIV-1 gp140B via intraileal injection generated sig-
nificantly higher numbers of H-2Dd
/PA-9ϩ
CD8ϩ
T cells in
both mucosal and systemic compartments and antigen-specific
IFN-␥-, TNF-␣-, and IL-2-producing CD4ϩ
and CD8ϩ
T cells
in the spleen than did p.o. administration (Fig. 3A and C). In
fact, p.o. immunization did not generate any detectable im-
mune response after a single dose in our study. These results
indicate that the higher transduction from intraileal injections
than from p.o. delivery (Fig. 2B) is associated with the stronger
immune responses to HIV-1 gp140B. Systemic boosting with
rAd5-gp140B dramatically increased CD8ϩ
T-cell responses in
the mice primed via the intraileal route in both systemic and
mucosal compartments compared to responses in the group
primed p.o. (Fig. 4B and C, right) as well as splenic CD4ϩ
T-cell responses compared to those in the mice primed p.o. or
via ileal injection (Fig. 4C, left). Whether the activation of
CD4ϩ
T cells would provide more target cells for HIV acqui-
sition will be addressed in nonhuman primate (NHP) studies,
though a previous study in which such mucosal responses were
detected showed that these responses led to protection (21,
25). Oral dosing with replication-competent Ad vectors gener-
ates systemic immune responses in nonhuman primates, but
multiple primes and protein boosting are required for protec-
tive efficacy against SIVmac251 challenge (32, 47). Oral vacci-
nation with replication-competent Ad4 and Ad7 has been
shown to generate protective Ad neutralizing antibodies in
humans (11), indicating that replication-competent vectors
elicit antivector neutralizing antibodies, which may compete
with transgene-specific responses or may prevent multiple p.o.
administrations of the same vector and affect efficacy. In addi-
tion, the magnitude and contribution of the transgene-specific
response in mediating protection by replication-competent
rAd remain unknown in this model because the gut mucosal
immune responses were not measured and compared to sys-
temic responses. Thus, there are potential efficacy and regula-
tory concerns with replication-competent rAds, and further
studies are needed to assess the potential efficacy of gut mu-
cosal immunization against lentivirus infection in NHP. Direct
mucosal introduction of replication-defective vectors offers an
alternative approach to address questions of immunogenicity
and efficacy in this model. Although cellular responses were
robust after intraileal injection, we did not detect a significant
level of IgG or IgA antibody from fecal extracts (data not
shown). Some animals showed IgA responses from the vaginal
7172 WANG ET AL. J. VIROL.
onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
washes, but the responses were low and inconsistent. Though
collection methods for mucosal secretions can be optimized
(19), it is likely that these responses are low in magnitude in
the GI tract.
While the results reported here describe the immunogenicity
of rAd5- and rAd35-based vectors, we have previously ana-
lyzed rAd41, an enteric tropic Ad, for its ability to generate
immune responses through direct ileum lumenal administra-
FIG. 5. Transduction efficiencies and immune responses induced by rAd35 and rAd35/41 chimeras. (A) Transduction of ileum explants by rAd
vectors. RLU, relative luminescence units. (B and C) Immune responses induced by rAd vectors in mice. Mice were administered the indicated
rAd vectors or empty rAd5 vector (Ϫ) via the intraileal route and boosted i.m. 3 weeks later. (B) Percentages of HIV-specific H-2Dd
/PA-9ϩ
CD8ϩ
T cells from spleens and small intestines 2 weeks after the i.m. rAd5-gp140B boost. (C) HIV-1-specific IFN-␥-secreting CD4ϩ
or CD8ϩ
T
lymphocytes from spleens 2 weeks after i.m. rAd5-gp140B boost. The data from each individual mouse and the mean values from the five mice
(black bars) are shown. -, empty vector. *, P Ͻ 0.05; **, P Ͻ 0.01; ***, P Ͻ 0.001.
VOL. 83, 2009 INTRAILEAL rAd5 VACCINE STIMULATES MUCOSAL IMMUNITY 7173
onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
tion (17). When rAd41 was administered directly to the lumen
of the ileum, it efficiently primed antigen-specific cellular im-
munity. Ad41, a member of the Ad species F, has been re-
ported to bind/uptake more efficiently than Ad5 in the isolated
intestinal loops of rats. Furthermore, gene delivery with Ad5 to
differentiated enterocytes in vitro and to the rat jejunum in
vivo is extremely inefficient (7, 8, 44). However, the immune
responses induced by Ad41 and Ad5 through intraileal injec-
tion priming and i.m. boosting were not significantly different,
which highlighted that vectors derived from mucosal patho-
gens, both enteric and respiratory, can be used in these regi-
mens. However, due to the prevalence of neutralizing antibod-
ies to Ad5 and Ad41 in humans, the development and use of
alternative serotypes of Ad vectors for virus vaccines remain a
priority. Ad35 is one relatively well-characterized rare serotype
vector and has been associated with infections of the kidney
and urinary tract. Therefore, the fiber swap study was done for
two reasons: (i) to define the contribution of fiber versus hexon
to transduction of mucosal epithelium and (ii) to generate
vectors resistant to anti-Ad41 hexon neutralizing antibodies,
should they be mediated by fiber. Here, we found that rAd35
did not transduce the organ cultures of intestinal segments as
efficiently as rAd5 and that rAd35 was less immunogenic in the
ileal priming regimen, thus allowing us to address these ques-
tions. Unexpectedly, grafting the long fiber or the short fiber of
Ad41 to the rAd35 vector did not improve the transduction
efficiency of intestinal explants or immunogenicity, suggesting
that the Ad41 fiber is not the only determinant of enteric
tropism. However, these results suggest that the immunoge-
nicity of rAd vectors in vivo correlates with the efficiency of
transduction of intestinal explants ex vivo. We conclude that
the lower small intestine represents a target for antigen deliv-
ery with certain rAd vectors. Further development of special
formulations to deliver rAd to the ileum should be emphasized
for practical vaccination.
Our results suggest that it is important to pursue a mucosal
route of administration of rAd vaccine vectors for HIV. The
strong cellular immune responses induced by intraileal injec-
tion, further strengthened by i.m. boost, suggest a strategy for
protection of gut-associated lymphoid tissue lymphocytes from
HIV-1 infection. Possible mechanisms of immune stimulation
include persistent antigen expression from lamina propria cells
transduced by the rAd vector (7, 40). Further characterization
of the immune responses induced by the vaccination regimens
employed in this work, such as the balance of CD8ϩ
and CD4ϩ
T-cell responses, the polyfunctionality of the T cells, and the
duration of the immune response, could provide a better pre-
diction as to which is the most important type of immune
response for protection against HIV-1 (22). Regarding the
applicability of these data to other species, as well as the
uncertain applicability to humans, this study represents one of
a stepwise series of studies to determine whether mucosal
immunity can be enhanced by different rAd vectors with alter-
native routes of administration. Having been demonstrated
with rodents, this will next be evaluated with nonhuman pri-
mate challenge models to determine its contribution to im-
mune protection. Should these results support the concept of a
mucosal vaccine, an effort will be made to formulate an Ad
vector for delivery to the ileum in humans, since the site-
specific delivery technology to deliver drugs and vaccines to the
ileum has been developed with partial success so far (14, 16,
27, 36).
The use of rAd5 vaccines in populations with moderate to
high neutralizing antibodies to Ad5 based on the STEP trial
(3a) results is a concern. It will be difficult to define mecha-
nisms responsible for this effect or even to document it defin-
itively. At the same time, rAd5 vectors are well characterized
and induce strong immune responses. There are numerous
differences in vector design, insert combination, and immuni-
zation regimen that may result in different performances in
future clinical studies. Regardless of its future clinical utility,
rAd5 remains a useful model vector for inducing mucosal im-
munity and for determining how mucosal immune responses
contribute to protection. Once these key questions are ad-
dressed, rAd5 vectors can be used if justified by efficacy/safety
considerations, or, if concerns remain, other vectors can be
substituted.
ACKNOWLEDGMENTS
We thank Ati Tislerics for help with manuscript preparation, Brenda
Hartman and Morteza Loghmani for assistance with figures, Srinivas
Rao and Saran Bao for help with surgical techniques, D. Margulies and
the NIAID Tetramer Core Facility for production of tetramers, and
members of the Nabel laboratory for helpful discussions.
This work was supported by the Intramural Research Program of the
National Institutes of Health, Vaccine Research Center, NIAID, and
by the Bill and Melinda Gates Foundation.
REFERENCES
1. Aoki, Y., M. Morishita, and K. Takayama. 2005. Role of the mucous/glyco-
calyx layers in insulin permeation across the rat ileal membrane. Int.
J. Pharm. 297:98–109.
2. Benihoud, K., P. Yeh, and M. Perricaudet. 1999. Adenovirus vectors for gene
delivery. Curr. Opin. Biotechnol. 10:440–447.
3. Brenchley, J. M., T. W. Schacker, L. E. Ruff, D. A. Price, J. H. Taylor, G. J.
Beilman, P. L. Nguyen, A. Khoruts, M. Larson, A. T. Haase, and D. C.
Douek. 2004. CD4ϩ T cell depletion during all stages of HIV disease occurs
predominantly in the gastrointestinal tract. J. Exp. Med. 200:749–759.
3a.Buchbinder, S. P., D. V. Mehrotra, A. Duerr, D. W. Fitzgerald, R. Mogg, D.
Li, P. B. Gilbert, J. R. Lama, M. Marmor, C. Del Rio, M. J. McElrath, D. R.
Casimiro, K. M. Gottesdiener, J. A. Chodakewitz, L. Corey, M. N. Robert-
son, and the Step Study Protocol Team. 2008. Efficacy assessment of a
cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind,
randomised, placebo-controlled, test-of-concept trial. Lancet 372:1881–1983.
4. Casimiro, D. R., A. J. Bett, T. M. Fu, M. E. Davies, A. Tang, K. A. Wilson,
M. Chen, R. Long, T. McKelvey, M. Chastain, S. Gurunathan, J. Tartaglia,
E. A. Emini, and J. Shiver. 2004. Heterologous human immunodeficiency
virus type 1 priming-boosting immunization strategies involving replication-
defective adenovirus and poxvirus vaccine vectors. J. Virol. 78:11434–11438.
5. Casimiro, D. R., L. Chen, T. M. Fu, R. K. Evans, M. J. Caulfield, M. E.
Davies, A. Tang, M. Chen, L. Huang, V. Harris, D. C. Freed, K. A. Wilson,
S. Dubey, D. M. Zhu, D. Nawrocki, H. Mach, R. Troutman, L. Isopi, D.
Williams, W. Hurni, Z. Xu, J. G. Smith, S. Wang, X. Liu, L. Guan, R. Long,
W. Trigona, G. J. Heidecker, H. C. Perry, N. Persaud, T. J. Toner, Q. Su, X.
Liang, R. Youil, M. Chastain, A. J. Bett, D. B. Volkin, E. A. Emini, and J. W.
Shiver. 2003. Comparative immunogenicity in rhesus monkeys of DNA plas-
mid, recombinant vaccinia virus, and replication-defective adenovirus vec-
tors expressing a human immunodeficiency virus type 1 gag gene. J. Virol.
77:6305–6313.
6. Cheng, C., J. G. Gall, W. P. Kong, R. L. Sheets, P. L. Gomez, C. R. King, and
G. J. Nabel. 2007. Mechanism of ad5 vaccine immunity and toxicity: fiber
shaft targeting of dendritic cells. PLoS Pathog. 3:e25.
7. Croyle, M. A., M. Stone, G. L. Amidon, and B. J. Roessler. 1998. In vitro and
in vivo assessment of adenovirus 41 as a vector for gene delivery to the
intestine. Gene Ther. 5:645–654.
8. Croyle, M. A., E. Walter, S. Janich, B. J. Roessler, and G. L. Amidon. 1998.
Role of integrin expression in adenovirus-mediated gene delivery to the
intestinal epithelium. Hum. Gene Ther. 9:561–573.
9. Ertl, H. C. 2005. Immunological insights from genetic vaccines. Virus Res.
111:89–92.
10. Gao, W., A. C. Soloff, X. Lu, A. Montecalvo, D. C. Nguyen, Y. Matsuoka, P. D.
Robbins, D. E. Swayne, R. O. Donis, J. M. Katz, S. M. Barratt-Boyes, and A.
Gambotto. 2006. Protection of mice and poultry from lethal H5N1 avian
7174 WANG ET AL. J. VIROL.
onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
influenza virus through adenovirus-based immunization. J. Virol. 80:1959–
1964.
11. Gaydos, C. A., and J. C. Gaydos. 1995. Adenovirus vaccines in the U.S.
military. Mil. Med. 160:300–304.
12. Greber, U. F. 2002. Signalling in viral entry. Cell. Mol. Life Sci. 59:608–626.
13. Haase, A. T. 2005. Perils at mucosal front lines for HIV and SIV and their
hosts. Nat. Rev. Immunol. 5:783–792.
14. Hamman, J. H., G. M. Enslin, and A. F. Kotze. 2005. Oral delivery of peptide
drugs: barriers and developments. BioDrugs 19:165–177.
15. Jaiswal, S., N. Khanna, and S. Swaminathan. 2003. Replication-defective
adenoviral vaccine vector for the induction of immune responses to dengue
virus type 2. J. Virol. 77:12907–12913.
16. Klein, S., J. Stein, and J. Dressman. 2005. Site-specific delivery of anti-
inflammatory drugs in the gastrointestinal tract: an in-vitro release model.
J. Pharm. Pharmacol. 57:709–719.
17. Ko, S. Y., C. Cheng, W. P. Kong, L. Wang, M. Kanekiyo, D. Einfeld, C. R.
King, J. G. Gall, and G. J. Nabel. 2009. Enhanced induction of intestinal
cellular immunity by oral priming with enteric adenovirus 41 vectors. J. Vi-
rol. 83:748–756.
18. Kobinger, G. P., H. Feldmann, Y. Zhi, G. Schumer, G. Gao, F. Feldmann, S.
Jones, and J. M. Wilson. 2006. Chimpanzee adenovirus vaccine protects
against Zaire Ebola virus. Virology 346:394–401.
19. Kozlowski, P. A., R. M. Lynch, R. R. Patterson, S. Cu-Uvin, T. P. Flanigan,
and M. R. Neutra. 2000. Modified wick method using Weck-Cel sponges for
collection of human rectal secretions and analysis of mucosal HIV antibody.
J. Acquir. Immune Defic. Syndr. 24:297–309.
20. Lemiale, F., H. Haddada, G. J. Nabel, D. E. Brough, C. R. King, and J. G.
Gall. 2007. Novel adenovirus vaccine vectors based on the enteric-tropic
serotype 41. Vaccine 25:2074–2084.
21. Letvin, N. L., J. R. Mascola, Y. Sun, D. A. Gorgone, A. P. Buzby, L. Xu, Z. Y.
Yang, B. Chakrabarti, S. S. Rao, J. E. Schmitz, D. C. Montefiori, B. R.
Barker, F. L. Bookstein, and G. J. Nabel. 2006. Preserved CD4ϩ central
memory T cells and survival in vaccinated SIV-challenged monkeys. Science
312:1530–1533.
22. Letvin, N. L., S. S. Rao, V. Dang, A. P. Buzby, B. Korioth-Schmitz, D.
Dombagoda, J. G. Parvani, R. H. Clarke, L. Bar, K. R. Carlson, P. A.
Kozlowski, V. M. Hirsch, J. R. Mascola, and G. J. Nabel. 2007. No evidence
for consistent virus-specific immunity in simian immunodeficiency virus-
exposed, uninfected rhesus monkeys. J. Virol. 81:12368–12374.
23. Li, Q., L. Duan, J. D. Estes, Z. M. Ma, T. Rourke, Y. Wang, C. Reilly,
J. Carlis, C. J. Miller, and A. T. Haase. 2005. Peak SIV replication in resting
memory CD4ϩ T cells depletes gut lamina propria CD4ϩ T cells. Nature
434:1148–1152.
24. Lin, S. W., A. S. Cun, K. Harris-McCoy, and H. C. Ertl. 2007. Intramuscular
rather than oral administration of replication-defective adenoviral vaccine
vector induces specific CD8ϩ T cell responses in the gut. Vaccine 25:2187–
2193.
25. Mattapallil, J. J., D. C. Douek, A. Buckler-White, D. Montefiori, N. L.
Letvin, G. J. Nabel, and M. Roederer. 2006. Vaccination preserves CD4
memory T cells during acute simian immunodeficiency virus challenge. J.
Exp. Med. 203:1533–1541.
26. Mattapallil, J. J., D. C. Douek, B. Hill, Y. Nishimura, M. Martin, and M.
Roederer. 2005. Massive infection and loss of memory CD4ϩ T cells in
multiple tissues during acute SIV infection. Nature 434:1093–1097.
27. Mercier, G. T., P. N. Nehete, M. F. Passeri, B. N. Nehete, E. A. Weaver, N. S.
Templeton, K. Schluns, S. S. Buchl, K. J. Sastry, and M. A. Barry. 2007. Oral
immunization of rhesus macaques with adenoviral HIV vaccines using en-
teric-coated capsules. Vaccine 25:8687–8701.
28. Monteiro-Riviere, N. A., and J. A. Popp. 1986. Ultrastructural evaluation of
acute nasal toxicity in the rat respiratory epithelium in response to formal-
dehyde gas. Fundam. Appl. Toxicol. 6:251–262.
29. Nanda, A., D. M. Lynch, J. Goudsmit, A. A. Lemckert, B. A. Ewald, S. M.
Sumida, D. M. Truitt, P. Abbink, M. G. Kishko, D. A. Gorgone, M. A. Lifton,
L. Shen, A. Carville, K. G. Mansfield, M. J. Havenga, and D. H. Barouch.
2005. Immunogenicity of recombinant fiber-chimeric adenovirus serotype 35
vector-based vaccines in mice and rhesus monkeys. J. Virol. 79:14161–14168.
30. Oomura, K., K. Q. Xin, M. Takakura, K. Shinoda, N. Jounai, and K. Okuda.
2006. Oral administration of the adenovirus vector induces systemic immu-
nity rather than intestinal mucosal immunity. Vaccine 24:1045–1046.
31. Patel, A., Y. Zhang, M. Croyle, K. Tran, M. Gray, J. Strong, H. Feldmann,
J. M. Wilson, and G. P. Kobinger. 2007. Mucosal delivery of adenovirus-
based vaccine protects against Ebola virus infection in mice. J. Infect. Dis.
196(Suppl. 2):S413–S420.
32. Patterson, L. J., N. Malkevitch, D. Venzon, J. Pinczewski, V. R. Gomez-
Roman, L. Wang, V. S. Kalyanaraman, P. D. Markham, F. A. Robey, and M.
Robert-Guroff. 2004. Protection against mucosal simian immunodeficiency
virus SIVmac251 challenge by using replicating adenovirus-SIV multigene
vaccine priming and subunit boosting. J. Virol. 78:2212–2221.
33. Pinto, A. R., J. C. Fitzgerald, G. P. Gao, J. M. Wilson, and H. C. Ertl. 2004.
Induction of CD8ϩ T cells to an HIV-1 antigen upon oral immunization of
mice with a simian E1-deleted adenoviral vector. Vaccine 22:697–703.
34. Pope, M., and A. T. Haase. 2003. Transmission, acute HIV-1 infection and
the quest for strategies to prevent infection. Nat. Med. 9:847–852.
35. Robbins, P. D., and S. C. Ghivizzani. 1998. Viral vectors for gene therapy.
Pharmacol. Ther. 80:35–47.
36. Schellekens, R. C., F. Stellaard, D. Mitrovic, F. E. Stuurman, J. G. Kos-
terink, and H. W. Frijlink. 2008. Pulsatile drug delivery to ileo-colonic
segments by structured incorporation of disintegrants in pH-responsive poly-
mer coatings. J. Control. Release 132:91–98.
37. Sharpe, S., A. Fooks, J. Lee, K. Hayes, C. Clegg, and M. Cranage. 2002.
Single oral immunization with replication deficient recombinant adenovirus
elicits long-lived transgene-specific cellular and humoral immune responses.
Virology 293:210–216.
38. Shiver, J. W., and E. A. Emini. 2004. Recent advances in the development of
HIV-1 vaccines using replication-incompetent adenovirus vectors. Annu.
Rev. Med. 55:355–372.
39. Stahl-Hennig, C., S. Kuate, M. Franz, Y. S. Suh, H. Stoiber, U. Sauermann,
K. Tenner-Racz, S. Norley, K. S. Park, Y. C. Sung, R. Steinman, P. Racz, and
K. Uberla. 2007. Atraumatic oral spray immunization with replication-defi-
cient viral vector vaccines. J. Virol. 81:13180–13190.
40. Tatsis, N., J. C. Fitzgerald, A. Reyes-Sandoval, K. C. Harris-McCoy, S. E.
Hensley, D. Zhou, S. W. Lin, A. Bian, Z. Q. Xiang, A. Iparraguirre, C.
Lopez-Camacho, E. J. Wherry, and H. C. Ertl. 2007. Adenoviral vectors
persist in vivo and maintain activated CD8ϩ T cells: implications for their
use as vaccines. Blood 110:1916–1923.
41. Thorner, A. R., and D. H. Barouch. 2007. HIV-1 vaccine development:
progress and prospects. Curr. Infect. Dis. Rep. 9:71–75.
42. Tobery, T. W., S. A. Dubey, K. Anderson, D. C. Freed, K. S. Cox, J. Lin, M. T.
Prokop, K. J. Sykes, R. Mogg, D. V. Mehrotra, T. M. Fu, D. R. Casimiro, and
J. W. Shiver. 2006. A comparison of standard immunogenicity assays for
monitoring HIV type 1 gag-specific T cell responses in Ad5 HIV type 1 gag
vaccinated human subjects. AIDS Res. Hum. Retrovir. 22:1081–1090.
43. Veazey, R. S., M. DeMaria, L. V. Chalifoux, D. E. Shvetz, D. R. Pauley, H. L.
Knight, M. Rosenzweig, R. P. Johnson, R. C. Desrosiers, and A. A. Lackner.
1998. Gastrointestinal tract as a major site of CD4ϩ T cell depletion and
viral replication in SIV infection. Science 280:427–431.
44. Walter, E., M. A. Croyle, B. J. Roessler, and G. L. Amidon. 1997. The
absence of accessible vitronectin receptors in differentiated tissue hinders
adenoviral-mediated gene transfer to the intestinal epithelium in vitro.
Pharm. Res. 14:1216–1222.
45. Xiang, Z. Q., G. P. Gao, A. Reyes-Sandoval, Y. Li, J. M. Wilson, and H. C.
Ertl. 2003. Oral vaccination of mice with adenoviral vectors is not impaired
by preexisting immunity to the vaccine carrier. J. Virol. 77:10780–10789.
46. Xiang, Z. Q., Y. Yang, J. M. Wilson, and H. C. Ertl. 1996. A replication-
defective human adenovirus recombinant serves as a highly efficacious vac-
cine carrier. Virology 219:220–227.
47. Zhou, Q., R. Hidajat, B. Peng, D. Venzon, M. K. Aldrich, E. Richardson, E. M.
Lee, V. S. Kalyanaraman, G. Grimes, V. R. Gomez-Roman, L. E. Summers, N.
Malkevich, and M. Robert-Guroff. 2007. Comparative evaluation of oral and
intranasal priming with replication-competent adenovirus 5 host range mutant
(Ad5hr)-simian immunodeficiency virus (SIV) recombinant vaccines on immu-
nogenicity and protective efficacy against SIV(mac251). Vaccine 25:8021–8035.
VOL. 83, 2009 INTRAILEAL rAd5 VACCINE STIMULATES MUCOSAL IMMUNITY 7175
onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom

More Related Content

What's hot

Coreceptor usage
Coreceptor usageCoreceptor usage
Coreceptor usagemakarandptl
 
Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...
Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...
Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...ILRI
 
Presentation 18: Problems other than AHPND in EMS ponds, including the micros...
Presentation 18: Problems other than AHPND in EMS ponds, including the micros...Presentation 18: Problems other than AHPND in EMS ponds, including the micros...
Presentation 18: Problems other than AHPND in EMS ponds, including the micros...ExternalEvents
 
Recent advances in African swine fever vaccine development at the Internation...
Recent advances in African swine fever vaccine development at the Internation...Recent advances in African swine fever vaccine development at the Internation...
Recent advances in African swine fever vaccine development at the Internation...ILRI
 
Beltwide poster main
Beltwide poster mainBeltwide poster main
Beltwide poster mainSameer Khanal
 
Capnocytophaga canimorsus
Capnocytophaga canimorsus Capnocytophaga canimorsus
Capnocytophaga canimorsus fateh11
 
Research articl e
Research articl eResearch articl e
Research articl egisa_legal
 
WGHA Discovery Series: Robert Sinden
WGHA Discovery Series: Robert SindenWGHA Discovery Series: Robert Sinden
WGHA Discovery Series: Robert SindenUWGlobalHealth
 
Steines CFTR gene transfer
Steines CFTR gene transferSteines CFTR gene transfer
Steines CFTR gene transferBenjamin Steines
 
Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild ...
Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild ...Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild ...
Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild ...Guy Boulianne
 
SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...
SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...
SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...Guy Boulianne
 
A Highly Intensified ART Regimen Induces Long-Term Viral Suppression and Rest...
A Highly Intensified ART Regimen Induces Long-Term Viral Suppression and Rest...A Highly Intensified ART Regimen Induces Long-Term Viral Suppression and Rest...
A Highly Intensified ART Regimen Induces Long-Term Viral Suppression and Rest...Gruppo Salute Arcigay Torino
 
2013_CarterEtal_MultiplexPCR-Cronobacter_ AEM
2013_CarterEtal_MultiplexPCR-Cronobacter_ AEM2013_CarterEtal_MultiplexPCR-Cronobacter_ AEM
2013_CarterEtal_MultiplexPCR-Cronobacter_ AEMMonica Pava-Ripoll
 
Intracellular Reverse Transcription of Pfizer BioNTech COVID-19 mRNA Vaccine ...
Intracellular Reverse Transcription of Pfizer BioNTech COVID-19 mRNA Vaccine ...Intracellular Reverse Transcription of Pfizer BioNTech COVID-19 mRNA Vaccine ...
Intracellular Reverse Transcription of Pfizer BioNTech COVID-19 mRNA Vaccine ...Guy Boulianne
 
Antiviral Hammerhead ribozymes are ef
Antiviral Hammerhead ribozymes are efAntiviral Hammerhead ribozymes are ef
Antiviral Hammerhead ribozymes are efSiddhesh Sapre
 

What's hot (19)

Coreceptor usage
Coreceptor usageCoreceptor usage
Coreceptor usage
 
Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...
Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...
Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...
 
GKA deel 2 college 6
GKA deel 2 college 6GKA deel 2 college 6
GKA deel 2 college 6
 
1584-09
1584-091584-09
1584-09
 
Presentation 18: Problems other than AHPND in EMS ponds, including the micros...
Presentation 18: Problems other than AHPND in EMS ponds, including the micros...Presentation 18: Problems other than AHPND in EMS ponds, including the micros...
Presentation 18: Problems other than AHPND in EMS ponds, including the micros...
 
Recent advances in African swine fever vaccine development at the Internation...
Recent advances in African swine fever vaccine development at the Internation...Recent advances in African swine fever vaccine development at the Internation...
Recent advances in African swine fever vaccine development at the Internation...
 
Candidiasis immunity
Candidiasis immunityCandidiasis immunity
Candidiasis immunity
 
Beltwide poster main
Beltwide poster mainBeltwide poster main
Beltwide poster main
 
Capnocytophaga canimorsus
Capnocytophaga canimorsus Capnocytophaga canimorsus
Capnocytophaga canimorsus
 
5.4 Caerina
5.4 Caerina5.4 Caerina
5.4 Caerina
 
Research articl e
Research articl eResearch articl e
Research articl e
 
WGHA Discovery Series: Robert Sinden
WGHA Discovery Series: Robert SindenWGHA Discovery Series: Robert Sinden
WGHA Discovery Series: Robert Sinden
 
Steines CFTR gene transfer
Steines CFTR gene transferSteines CFTR gene transfer
Steines CFTR gene transfer
 
Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild ...
Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild ...Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild ...
Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild ...
 
SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...
SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...
SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...
 
A Highly Intensified ART Regimen Induces Long-Term Viral Suppression and Rest...
A Highly Intensified ART Regimen Induces Long-Term Viral Suppression and Rest...A Highly Intensified ART Regimen Induces Long-Term Viral Suppression and Rest...
A Highly Intensified ART Regimen Induces Long-Term Viral Suppression and Rest...
 
2013_CarterEtal_MultiplexPCR-Cronobacter_ AEM
2013_CarterEtal_MultiplexPCR-Cronobacter_ AEM2013_CarterEtal_MultiplexPCR-Cronobacter_ AEM
2013_CarterEtal_MultiplexPCR-Cronobacter_ AEM
 
Intracellular Reverse Transcription of Pfizer BioNTech COVID-19 mRNA Vaccine ...
Intracellular Reverse Transcription of Pfizer BioNTech COVID-19 mRNA Vaccine ...Intracellular Reverse Transcription of Pfizer BioNTech COVID-19 mRNA Vaccine ...
Intracellular Reverse Transcription of Pfizer BioNTech COVID-19 mRNA Vaccine ...
 
Antiviral Hammerhead ribozymes are ef
Antiviral Hammerhead ribozymes are efAntiviral Hammerhead ribozymes are ef
Antiviral Hammerhead ribozymes are ef
 

Similar to J. Virol.-2009-Wang-7166-75

journal.pone.0106004
journal.pone.0106004journal.pone.0106004
journal.pone.0106004Huma Qureshi
 
Seminario Biología Molecular
Seminario Biología Molecular Seminario Biología Molecular
Seminario Biología Molecular JulianaAceros
 
Intracellular highways in the plants: the role of the cytoskeleton in camv i...
Intracellular highways in the plants:  the role of the cytoskeleton in camv i...Intracellular highways in the plants:  the role of the cytoskeleton in camv i...
Intracellular highways in the plants: the role of the cytoskeleton in camv i...CIAT
 
Cerullo_verona2010_AdenovirusGT
Cerullo_verona2010_AdenovirusGTCerullo_verona2010_AdenovirusGT
Cerullo_verona2010_AdenovirusGTVincenzo Cerullo
 
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...Dr. Md. Ehsanul Haque
 
Recombinant low-seroprevalent adenoviral vectors Ad26 and Ad35
Recombinant low-seroprevalent adenoviral vectors Ad26 and Ad35Recombinant low-seroprevalent adenoviral vectors Ad26 and Ad35
Recombinant low-seroprevalent adenoviral vectors Ad26 and Ad35Arun kumar
 
vaccine development against malaria parasite
vaccine development against malaria parasitevaccine development against malaria parasite
vaccine development against malaria parasiteswarnendu basak
 
Dr. Tanja Opriessnig - Update on novel experimental pig vaccine approaches
Dr. Tanja Opriessnig - Update on novel experimental pig vaccine approachesDr. Tanja Opriessnig - Update on novel experimental pig vaccine approaches
Dr. Tanja Opriessnig - Update on novel experimental pig vaccine approachesJohn Blue
 
art%3A10.1186%2Fs12936-016-1304-8
art%3A10.1186%2Fs12936-016-1304-8art%3A10.1186%2Fs12936-016-1304-8
art%3A10.1186%2Fs12936-016-1304-8wagatua njoroge
 
The 'omics' revolution: How will it improve our understanding of infections a...
The 'omics' revolution: How will it improve our understanding of infections a...The 'omics' revolution: How will it improve our understanding of infections a...
The 'omics' revolution: How will it improve our understanding of infections a...WAidid
 
Expression and Purification of the Plasmodium berghei Apical Membrane Antigen-1
Expression and Purification of the Plasmodium berghei Apical Membrane Antigen-1Expression and Purification of the Plasmodium berghei Apical Membrane Antigen-1
Expression and Purification of the Plasmodium berghei Apical Membrane Antigen-1Nathan Jones
 
JPT_Poster_EPS_2016_final
JPT_Poster_EPS_2016_finalJPT_Poster_EPS_2016_final
JPT_Poster_EPS_2016_finalPaul Von Hoegen
 

Similar to J. Virol.-2009-Wang-7166-75 (20)

journal.pone.0106004
journal.pone.0106004journal.pone.0106004
journal.pone.0106004
 
Seminario Biología Molecular
Seminario Biología Molecular Seminario Biología Molecular
Seminario Biología Molecular
 
9 Malaria Vaccine (1)
9 Malaria Vaccine (1)9 Malaria Vaccine (1)
9 Malaria Vaccine (1)
 
Malarial vaccine
Malarial vaccineMalarial vaccine
Malarial vaccine
 
Intracellular highways in the plants: the role of the cytoskeleton in camv i...
Intracellular highways in the plants:  the role of the cytoskeleton in camv i...Intracellular highways in the plants:  the role of the cytoskeleton in camv i...
Intracellular highways in the plants: the role of the cytoskeleton in camv i...
 
Cerullo_verona2010_AdenovirusGT
Cerullo_verona2010_AdenovirusGTCerullo_verona2010_AdenovirusGT
Cerullo_verona2010_AdenovirusGT
 
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
 
Recombinant low-seroprevalent adenoviral vectors Ad26 and Ad35
Recombinant low-seroprevalent adenoviral vectors Ad26 and Ad35Recombinant low-seroprevalent adenoviral vectors Ad26 and Ad35
Recombinant low-seroprevalent adenoviral vectors Ad26 and Ad35
 
vaccine development against malaria parasite
vaccine development against malaria parasitevaccine development against malaria parasite
vaccine development against malaria parasite
 
Dr. Tanja Opriessnig - Update on novel experimental pig vaccine approaches
Dr. Tanja Opriessnig - Update on novel experimental pig vaccine approachesDr. Tanja Opriessnig - Update on novel experimental pig vaccine approaches
Dr. Tanja Opriessnig - Update on novel experimental pig vaccine approaches
 
Janse_2013_EID_pla
Janse_2013_EID_plaJanse_2013_EID_pla
Janse_2013_EID_pla
 
Antimalarial vaccines
Antimalarial vaccinesAntimalarial vaccines
Antimalarial vaccines
 
art%3A10.1186%2Fs12936-016-1304-8
art%3A10.1186%2Fs12936-016-1304-8art%3A10.1186%2Fs12936-016-1304-8
art%3A10.1186%2Fs12936-016-1304-8
 
The 'omics' revolution: How will it improve our understanding of infections a...
The 'omics' revolution: How will it improve our understanding of infections a...The 'omics' revolution: How will it improve our understanding of infections a...
The 'omics' revolution: How will it improve our understanding of infections a...
 
Expression and Purification of the Plasmodium berghei Apical Membrane Antigen-1
Expression and Purification of the Plasmodium berghei Apical Membrane Antigen-1Expression and Purification of the Plasmodium berghei Apical Membrane Antigen-1
Expression and Purification of the Plasmodium berghei Apical Membrane Antigen-1
 
My CV_final
My CV_finalMy CV_final
My CV_final
 
JPT_Poster_EPS_2016_final
JPT_Poster_EPS_2016_finalJPT_Poster_EPS_2016_final
JPT_Poster_EPS_2016_final
 
Tetanus toxoid paper
Tetanus toxoid paperTetanus toxoid paper
Tetanus toxoid paper
 
2012 hla and ama1 dbp csp msp1
2012 hla and ama1 dbp csp msp12012 hla and ama1 dbp csp msp1
2012 hla and ama1 dbp csp msp1
 
2012 hla and ama1 dbp csp msp1
2012 hla and ama1 dbp csp msp12012 hla and ama1 dbp csp msp1
2012 hla and ama1 dbp csp msp1
 

J. Virol.-2009-Wang-7166-75

  • 1. Published Ahead of Print 6 May 2009. 2009, 83(14):7166. DOI: 10.1128/JVI.00374-09.J. Virol. Nabel Schwartz, C. Richter King, Jason G. D. Gall and Gary J. Kong, Masaru Kanekiyo, David Einfeld, Richard M. Lingshu Wang, Cheng Cheng, Sung-Youl Ko, Wing-Pui Enhanced Mucosal Cellular Immunity Vaccine Vectors to the Intestine Induces Delivery of Human Immunodeficiency Virus http://jvi.asm.org/content/83/14/7166 Updated information and services can be found at: These include: SUPPLEMENTAL MATERIAL Supplemental material REFERENCES http://jvi.asm.org/content/83/14/7166#ref-list-1at: This article cites 48 articles, 16 of which can be accessed free CONTENT ALERTS more»articles cite this article), Receive: RSS Feeds, eTOCs, free email alerts (when new http://journals.asm.org/site/misc/reprints.xhtmlInformation about commercial reprint orders: http://journals.asm.org/site/subscriptions/To subscribe to to another ASM Journal go to: onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
  • 2. JOURNAL OF VIROLOGY, July 2009, p. 7166–7175 Vol. 83, No. 14 0022-538X/09/$08.00ϩ0 doi:10.1128/JVI.00374-09 Copyright © 2009, American Society for Microbiology. All Rights Reserved. Delivery of Human Immunodeficiency Virus Vaccine Vectors to the Intestine Induces Enhanced Mucosal Cellular Immunityᰔ † Lingshu Wang,1 ‡ Cheng Cheng,1 ‡ Sung-Youl Ko,1 Wing-Pui Kong,1 Masaru Kanekiyo,1 David Einfeld,2 Richard M. Schwartz,1 C. Richter King,2 Jason G. D. Gall,2 and Gary J. Nabel1 * Vaccine Research Center, NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC-3005, 40 Convent Drive, Bethesda, Maryland 20892-3005,1 and GenVec, Inc., 65 West Watkins Mill Rd., Gaithersburg, Maryland 208782 Received 19 February 2009/Accepted 30 April 2009 Effective vaccines for human immunodeficiency virus type 1 (HIV-1) will likely need to stimulate protective immunity in the intestinal mucosa, where HIV-1 infection causes severe CD4؉ T-cell depletion. While repli- cation-competent recombinant adenovirus (rAd) vectors can stimulate adenovirus-specific mucosal immunity after replication, oral delivery of replication-defective rAd vectors encoding specific immunogens has proven challenging. In this study, we have systematically identified barriers to effective gut delivery of rAd vectors and identified sites and strategies to induce potent cellular and humoral immunity. Vector-mediated gene transfer by rAd5 was susceptible to low-pH buffer, gastric and pancreatic proteases, and extracellular mucins. Using ex vivo organ explants, we found that transduction with rAd5 was highest in the ileum and colon among all intestinal segments. Transgene expression was 100-fold higher after direct surgical introduction into the ileum than after oral gavage, with rAd5 showing greater potency than the rAd35 or the rAd41 vector. A single immunization of rAd5 encoding HIV-1 gp140B to the ileum stimulated potent CD8؉ T-cell responses in the intestinal and systemic compartments, and these responses were further enhanced by intramuscular rAd5 boosting. These studies suggest that induction of primary immune responses by rAd5 gut immunization and subsequent systemic boosting elicits potent antigen-specific gut mucosal responses. Human immunodeficiency virus type 1 (HIV-1) infection is characterized by uncontrolled virus replication and cytopath- icity in the intestinal mucosa, the site of major T-cell depletion after primary infection. The gastrointestinal (GI) tract is the predominant site of a pronounced CD4ϩ T-cell loss in the early stages of HIV infection and simian immunodeficiency virus (SIV) infection in the nonhuman primate model (3, 23, 26, 43). It has been suggested that a mucosal vaccine which generates HIV-specific CD8ϩ T cells in the gut could prevent the loss of CD4ϩ cells in gut-associated lymphoid tissue, es- tablishment of infection, or spread of virus (13, 34). Therefore, targeted delivery of vaccines to the GI tract to stimulate mu- cosal responses has the potential to improve the efficacy of immune protection against HIV-1; however, the site of gene- based transduction and the barriers to vaccine delivery have not been well defined. Adenoviruses (Ads) have been used extensively as vectors for both gene transfer and vaccine development. They offer several advantages as tools for vaccine delivery, such as the ability to transduce both dividing and nondividing cells, rela- tive safety and stability in vivo, ease of production in high titers, and lack of integration (2, 35). These vectors are prom- ising because parenteral administration in both animals and humans has been shown to generate strong and long-lasting humoral and cellular immune responses. The immune re- sponses surpass those achieved with other types of gene vectors and genetic vaccines (5, 38, 46). As a result, recombinant Ad (rAd) vectors have been developed and tested as vaccine vehicles to immunize against a number of pathogens (4, 10, 15, 18, 41). Orally (p.o.) delivered vaccines are attractive in theory be- cause of their ease of administration and potential to deliver antigen to gut-associated lymphoid tissue, permitting induction of immune responses in both mucosal and systemic compart- ments. At the same time, p.o. delivery of replication-defective rAd vectors has posed a challenge and has met with variable levels of success. Immunization with rAd5 encoding rabies virus antigens, influenza virus antigens, or other antigens has generated some protection against infection in animal models (9, 27, 31, 39, 41), but p.o. immunization has elicited much lower CD8ϩ T-cell responses than systemic delivery (33), and a much higher dose is required to induce immune responses (37). We have recently shown in an HIV vaccine model that rAd41, a human enteric Ad-based vector, induced potent CD8ϩ T-cell responses in both systemic and mucosal compart- ments when primed p.o. or in the ileum (17). The previous study showed that rAd41 vectors delivered through direct ileal injection elicited mucosal cell immunity, but whether other rAd vectors could stimulate these responses and which factors affected delivery and immunogenicity were unknown. In this report, we have investigated the mechanisms associated with the low immunogenicity of rAd5 dosed through the p.o. route in mice. The purpose was to identify barriers to effective de- livery of rAd vectors to gut tissues and to ascertain sites and strategies for induction of potent cellular and humoral immu- nity. To investigate the mechanism of the low immunogenicity * Corresponding author. Mailing address: Vaccine Research Center, NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC- 3005, 40 Convent Drive, Bethesda, MD 20892-3005. Phone: (301) 496-1852. Fax: (301) 480-0274. E-mail: gnabel@nih.gov. † Supplemental material for this article may be found at http://jvi .asm.org/. ‡ These authors contributed equally to this report. ᰔ Published ahead of print on 6 May 2009. 7166 onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
  • 3. of rAd vectors through the p.o. route and develop effective delivery of rAd5 and rare serotype rAd35 vectors as gut mu- cosal HIV vaccines, we have analyzed the obstacles to p.o. immunization, characterized vector transgene expression, and systematically compared immune responses induced by p.o. and local immunization strategies. These studies demonstrated that the higher immune responses were strongly associated with higher gene expression in the intestine and support fur- ther study of gut mucosal immunization in SIV challenge mod- els as a potential HIV vaccine strategy. MATERIALS AND METHODS Animals and viruses. Six- to 8-week-old female BALB/c mice were purchased from Jackson Laboratories and housed in the experimental animal facility of the Vaccine Research Center, NIAID, NIH (Bethesda, MD). All animal experi- ments were reviewed and approved by the Animal Care and Use Committee, VRC, NIAID, NIH, and performed in accordance with all relevant federal and NIH guidelines and regulations. rAd5 vectors are replication-defective E1-, E3-, and E4-deleted human Ads, and rAd35 vectors are E1- and E3-deleted replication-defective vectors. rAd35/ 41L and rAd35/41S vectors are rAd35-based vectors with chimeric Ad41 long fiber or short fiber, respectively. rAd5-luc, rAd35-luc, rAd35/41L-luc, and rAd35/ 41S-luc encode luciferase reporter genes under the control of the cytomegalo- virus promoter/enhancer. rAd5-gp140B, rAd35-gp140B, and rAd35/41L- or rAd35/41S-gp140B (rAd35 with Ad41 long or short fiber, respectively) encode gp140⌬CFI⌬V1V2 of HIV-1 clade B (20). Ad vectors were propagated in 293- ORF6 cells and purified by cesium chloride gradients (6). Mouse intestinal explants. Explant culture medium consisted of Dulbecco’s modified Eagle’s medium–F-12 medium supplemented with 5% fetal bovine serum, L-glutamine (2 mM), penicillin (100 U/ml), streptomycin (100 ␮g/ml), gentamicin (50 ␮g/ml), and epidermal growth factor (10 U/ml). All media and supplements were purchased from Invitrogen, Inc. (Carlsbad, CA). Mouse in- testines were isolated and cut open longitudinally. After being washed thor- oughly, the tissues were cut into 5- by 5-mm pieces and placed on presoaked Gelfoam (Pfizer, New York, NY), with the epithelium uppermost, in 24-well plates containing 0.3 ml of culture medium. Four microliters of virus solution containing 1 ϫ 108 to 5 ϫ 108 virus particles (VP) of treated or untreated rAd vectors encoding luciferase was applied directly to the upper surface of each explant. After overnight incubation, the tissues were rinsed in phosphate-buff- ered saline (PBS) and placed in 0.1% Triton X-100. Following a 1-min ultra- sound treatment and three freeze-thaw cycles, the lysates were centrifuged at 2,000 rpm for 10 min. The supernatants were used for a luciferase assay. The luminescence was measured using a microplate scintillation and luminescence counter (PerkinElmer, Shelton, CT). The protein concentration of the cleared supernatant was determined using a DC protein microplate assay kit (Bio-Rad, Hercules, CA). The results are shown as relative luminescence units per ␮g protein. For the acid and protease sensitivity study, rAd5-luc was (i) treated with 0.1 M HCl and 2.5 mg/ml pepsin from porcine gastric mucosa (Sigma, St. Louis, MO) for 5 min at 37°C and neutralized with 10ϫ THB (200 mM Tris, 500 mM HEPES, 1.5 M NaCl), (ii) treated with 25 ␮g/ml trypsin from bovine pancreas (Sigma, St. Louis, MO) or 2.5 U/ml chymotrypsin (Sigma, St. Louis, MO) for 30 min at 37°C, or (iii) exposed to the combination of treatments with HCl-pepsin and trypsin and/or chymotrypsin and then applied to the tissue explants. To test the susceptibility of intestinal segments to Ad vectors, mouse duode- num (1 cm downstream of the stomach), jejunum (median part of the small intestine), ileum (from the junction of the ileum and cecum to 2 cm upstream of the cecum), and colon (2 cm downstream of the cecum) were isolated and either washed thoroughly (native) or gently stripped to remove the mucus for culture. Ten pieces from three mice for each segment were cultured with 4 ␮l virus solution containing 1 ϫ 108 VP of rAd5-luc and harvested as described above. Treatment of intestinal explants with DTT, hyaluronidase, and exoglycosi- dases. The mouse intestinal explants were either not treated or treated with dithiothreitol (DTT) (20 mM, catalogue number 43816; Sigma, St. Louis, MO) or hyaluronidase (1 or 10 mg/ml, catalogue number H 3506; Sigma, St. Louis, MO) for 30 min or exoglycosidases from Trimeresurus cornutus (1, 5, or 10 mg/ml; Seikagaku Biobusiness, Tokyo, Japan) for 2 min at 37°C. The explants were then rinsed in the prewarmed medium and cultured with 4 ␮l virus solution containing 1 ϫ 108 VP of rAd5-luc. The stripped jejunum and the untreated ileum were used as the positive controls, while the medium only was used for the negative controls. The tissue explants were harvested as described above. Gene expression in mouse intestine transduced with rAd5-luc in vivo. Three BALB/c mice per group were either administered p.o. 1010 VP of rAd5-luc in 500 ␮l PBS or given an intraileal injection with 1010 VP of rAd5-luc in 100 ␮l PBS. Twenty-four hours after injection, the mice were sacrificed, and intestinal seg- ments, including duodenum, jejunum, ileum, colon, and mesenteric lymph nodes (MLN), were lysed and assayed for luciferase activity. The group injected with 100 ␮l rAd5 empty vector via the ileum lumen was used as the control. The intraileal-injection procedure was described previously (17). Immunization. For the immunogenicity study, immune responses induced by a single injection of rAd5 encoding HIV-1 gp140B into the ileum lumen were tested. Five mice from each group were immunized with 1010 VP of rAd5-gp140B vector in 500 ␮l PBS p.o. or with 1010 VP of rAd5-gp140B vector in 100 ␮l PBS via ileal injection as described above. Mice that received rAd5 empty vector p.o. or via ileal injections were used as the negative controls. Three weeks after inoculation, peripheral blood mononuclear cells (PBMC), spleens, and small intestines were collected for H-2Dd /PA-9 tetramer staining. The spleens were also used for detecting HIV-1 peptide-specific cytokine-producing CD4ϩ or CD8ϩ T lymphocytes. The serum immunoglobulin G (IgG) antibodies were determined by enzyme-linked immunosorbent assay (ELISA). In the prime-boost experiments, five mice per group were primed with 1010 VP of rAd5-gp140B via p.o. dosing or with rAd5, rAd35, Ad35/41L, and rAd35/41S, all encoding gp140B, via intraileal injections. Three weeks after immunization, blood was collected and PBMC were isolated and stained for H-2Dd /PA-9 tetramer. These mice were then boosted intramuscularly (i.m.) with rAd5- gp140B at 109 VP. Two weeks after the boost, PBMC, spleens, MLN, and small intestines were harvested for H-2Dd /PA-9 tetramer staining, and the spleens were also used for detecting HIV-1 peptide-specific cytokine-producing CD4ϩ or CD8ϩ T lymphocytes. The IgG and IgA antibodies in the sera and vaginal washes were determined by ELISA. Lymphocyte isolation. The isolation of lymphocytes from blood, spleen, and MLN was described previously (17). For lymphocyte isolation from the gut, the small intestines from individual mice were collected in cold medium. The intes- tine was opened and washed thoroughly following excision of Peyer’s patches. Intestinal sections were then minced and digested with 0.5 mg/ml collagenase II (C-6885; Sigma) for 30 min at 37°C. The resulting supernatants were filtered through a 40-␮m cell strainer. After centrifugation, the cell pellets were sus- pended in 6 ml of 40% Percoll (Amersham Biosciences, Piscataway, NJ) and overlaid on 4 ml of 75% Percoll. The samples were then subjected to centrifu- gation at 800 ϫ g for 20 min at 22°C. The cells from the interface of 75% and 40% Percoll were collected, washed with a large volume of medium, and counted. Tetramer staining and ICS. Lymphocytes isolated from blood, spleen, MLN, and the small intestine were used for tetramer staining. The splenic lymphocytes from individual mice were used for intracellular cytokine staining (ICS), while the combined intestinal cells from the group of five mice were also used for ICS in the rAd35/41 chimeric fiber experiment, using ViViD dye (Invitrogen Life Sciences, Carlsbad, CA) to exclude nonviable cells. The detailed methods for tetramer staining and ICS were described previously (M. Honda, R. Wang, W.-P. Kong, M. Kanekiyo, W. Akahata, L. Xu, K. Matsuo, K. Natarajan, H. Robinson, T. E. Asher, D. A. Price, D. C. Douek, D. H. Margulies, and G. J. Nabel, submitted for publication; 17). ELISA for IgG and IgA antibodies to HIV Env. Levels of HIV gp140B-specific IgG and IgA in the sera or vaginal washes of vaccinated mice were assessed using ELISA (17). The sera were diluted at 1:1,000 for IgG and 1:50 for IgA, while the vaginal washes were diluted at 1:3 for the detection of both IgG and IgA. Absorbance at 450 nm was determined by a Spectra Max instrument (Molecular Devices). Data and statistical analysis. All results are presented as means with standard errors. Statistical analyses were performed based upon comparisons between the control groups and the treated groups or between differently treated groups by using the two-tailed Student t test. P values of less than 0.05 were considered statistically significant. RESULTS Determination of barriers to effective delivery of rAd5 vec- tors to the GI tract. To identify potential mechanisms that inhibit transduction of the GI tract after rAd gene transfer after p.o. delivery, we first examined the transduction of mouse VOL. 83, 2009 INTRAILEAL rAd5 VACCINE STIMULATES MUCOSAL IMMUNITY 7167 onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
  • 4. intestinal explants by an rAd5 vector expressing luciferase. Intestinal explants of the duodenum, jejunum, ileum, and co- lon were cultured ex vivo, and the relative rAd transduction efficiencies were determined. The ileum and the colon were transduced at significantly higher levels than were the duode- num and jejunum (Fig. 1A, open bars). To determine whether the superficial layer played a role in the inhibition of transduc- tion of intestinal cells by rAd, mucus was removed by mechan- ical and biochemical means. Mechanical removal of the mucus significantly enhanced the transduction activity in all segments (P Ͻ 0.05 for the duodenum and jejunum, P Ͻ 0.001 for the ileum) except for the colon (Fig. 1A, filled versus open bars). The differences between transductions of native and mucus- stripped explants were 100-fold in the duodenum and jejunum and 10-fold in the ileum. Previous in vitro studies have shown that intestinal mucus can also be dissolved by a variety of agents, including enzymes, detergents, and sulfhydryl com- pounds (1, 28). The effect of DTT, hyaluronidase, and glyco- sidases on the transduction of intestinal explants by rAd5 was investigated using explants treated and exposed to vector ex vivo. Transduction of explants treated with DTT was signifi- cantly higher than transduction of those that were untreated, specifically in the upper intestinal segments, duodenum and jejunum (Fig. 1B, filled versus open bars). When mouse jeju- num was treated with hyaluronidase or exoglycosidases (see Materials and Methods), transduction was significantly higher than that of the jejunum treated with medium (Fig. 1C and D, 1 mg/ml). However, high concentrations of these enzymes did not enhance transduction, possibly due to nonspecific cell damage. We next examined the effect of acid and selected proteases on rAd5 transduction of mouse intestinal explant cultures. Gastric acid and pepsin are the major components secreted into the stomach, and trypsin and chymotrypsin are the major proteases produced by the pancreas. To mimic the passage of rAd through the gastric environment and into the proximal section of the small intestine, rAd5-luciferase vector was treated with combinations of acid and protease and then ap- plied to the tissue explants. Although we did not observe a significant decrease in luciferase activity after acid-pepsin, trypsin, or chymotrypsin treatment alone, the combination of acid-pepsin treatment with either trypsin or chymotrypsin treatment significantly reduced transgene expression (Fig. 2A, lanes 5 and 6). The combination of acid-pepsin with both trypsin and chymotrypsin further reduced luciferase activity, to approximately 100-fold lower than transduction with untreated rAd5-luc (Fig. 2A, lane 7). This result suggested that rAd5 vectors were susceptible to degradation by gastric proteases. Taken together, these ex vivo experiments indicated that mul- tiple barriers reduce transduction of intestinal epithelia by rAd5 vectors and demonstrated differential transduction of specific intestinal segments. Development of intraileal delivery to increase gene delivery to the small intestine. Since the untreated ileum was trans- FIG. 1. The distal regions of the intestine were efficiently transduced by rAd5, and removal of luminal substances increased transduction of the proximal intestine. (A) Efficient transduction of intestinal segments. Segments of intestine were left untreated (native), or the mucus was gently removed (stripped) and then transduced with rAd5-luc. (B) Treatment of intestinal segments with DTT increased the transduction efficiency of rAd5-Luc. (C and D) Effect of removal of the glycocalyx and hyaluronic acid on transduction. Mouse jejuna were treated with hyaluronidase (C) or mixed glycosidases (D) prior to transduction with rAd5-luc. *, P Ͻ 0.05; **, P Ͻ 0.01; ***, P Ͻ 0.001. RLU, relative luminescence units. 7168 WANG ET AL. J. VIROL. onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
  • 5. duced most effectively in ex vivo organ culture, we developed an in vivo route of administration that targeted the ileum and bypassed the significant barriers encountered by rAd in the GI tract. It has been reported that receptor binding and uptake of rAd5 occur relatively quickly, with the majority (80 to 85%) of the bound virions being taken up by permissive cells within 5 to 10 min (12). We found that a 10-min exposure of ileum ex- plants to rAd5 resulted in levels of transgene expression similar to those obtained with longer time exposures (see Fig. S1 in the supplemental material). Therefore, for in vivo studies, the ileum loop was formed to hold the virus solution for 20 min to ensure efficient binding of viruses within the intestine and the separation of virus from undigested food in the ileum. To determine the relative efficiency of rAd5-mediated trans- duction, mice were administered rAd5 (1010 VP) encoding luciferase either p.o. or via the intraileal-injection method. Twenty-four hours later, mice were sacrificed, and the intesti- nal segments were assayed for luciferase activity. A low level of luciferase activity was detected in the ileum after p.o. admin- istration, but the luciferase activity was not significantly differ- ent from that for the empty rAd5 control group (Fig. 2B) (P Ͼ 0.05). Following intraileal injection, there was approximately a 10- to 100-fold-higher luciferase activity in the ileum and MLN than for the control group (Fig. 2B) (P Ͻ 0.05). Importantly, the luciferase activity in the ilea from injected mice was signif- icantly higher than that for the p.o. dosed mice (Fig. 2B) (P Ͻ 0.05). Immunization via intraileal injection generated humoral and cellular responses in both mucosal and systemic compart- ments. The higher efficiency of intestinal cell transduction ob- served with ileal injection than with p.o. dosing suggested the possibility that intraileal administration of rAd vectors would FIG. 2. Transduction of intestinal explant cultures by rAd5-luc was reduced by exposure to low pH and proteases and enhanced gene expression by intraileal delivery of rAd5 in vivo. (A) The rAd vector encoding luciferase was treated as indicated (ϩ, treatment; Ϫ, no treatment) and applied to explants of mouse ileum. *, P Ͻ 0.05. (B) Mice were administered rAd5-luc via p.o. or intraileal injection, and the levels of luciferase activity in intestinal segments and MLN were determined 24 h later. *, P Ͻ 0.05. RLU, relative luminescence units. VOL. 83, 2009 INTRAILEAL rAd5 VACCINE STIMULATES MUCOSAL IMMUNITY 7169 onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
  • 6. induce a more potent immune response to the encoded trans- gene. Mice received a single administration of rAd5-gp140B (1010 VP) p.o. or via intraileal injection. Three weeks later, the mice immunized with rAd5-gp140B via intraileal injection showed significantly higher levels of H-2Dd /PA-9ϩ CD8ϩ T cells in both the small intestine and the systemic compartments than did the mice immunized p.o. (Fig. 3A). Oral administra- tion did not result in detectable responses higher than back- ground (Fig. 3A) (P Ͼ 0.05). The average levels of H-2Dd / PA-9ϩ CD8ϩ T cells in the mice given an intraileal injection FIG. 3. Mucosal and systemic immune responses induced by a single intraileal injection of rAd5-gp140B. Mice were immunized with rAd5gp-140B (rAd5) or rAd5 empty vector (Ϫ) via the p.o. or the intraileal route as indicated. (A) Percentages of HIV-specific H-2Dd /PA-9ϩ CD8ϩ T cells from PBMC, spleens, and small intestines. (B) IgG antibody in the serum. OD450nm, absorbance at 450 nm. (C) Percentages of HIV-specific IL-2-, IFN-␥-, and TNF-␣-secreting CD4ϩ or CD8ϩ T cells from the spleens. The data from each individual mouse and the mean values from the five mice (black bars) are shown. *, P Ͻ 0.05; **, P Ͻ 0.01. 7170 WANG ET AL. J. VIROL. onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
  • 7. were 7.11% (ranging from 2 to 11%) in PBMC, 3.47% (ranging from 1.44 to 5.82%) in the spleen, and 5.53% (ranging from 2.13 to 9.65%) in the small intestine. HIV Env-specific IgG responses were also detected in sera from mice immunized via the intraileal injection but not in sera from the p.o. dosed mice (Fig. 3B). ICS analysis of splenocytes was consistent with the tetramer staining results; numbers of HIV-1-specific CD4ϩ and CD8ϩ T cells producing interleukin-2 (IL-2), gamma in- terferon (IFN-␥), and tumor necrosis factor alpha (TNF-␣) were significantly higher in the mice that received rAd5- gp140B via intraileal injection than in the mice that received equal amounts of rAd5-gp140B p.o. (Fig. 3C). In this analysis, the magnitudes of CD8ϩ T-cell response differed between tet- ramer staining and ICS, suggesting that there was a dispropor- tionate representation of the dominant tetramer response or that not all of the tetramer-positive CD8ϩ T cells were secret- ing the measurable cytokines, a dichotomy described to occur previously when using these techniques (42). Tetramer staining measures CD8ϩ T cells responding to an immunodominant HIV-Env peptide regardless of function, including inactivated cells and activated cells, while ICS detects only activated cells or functional cells. Oral immunization generated only a back- ground level of cytokine-producing CD4ϩ or CD8ϩ T cells. These data demonstrate that the rAd5 vector induced substan- tial antigen-specific cellular and humoral immune responses after a single intraileal immunization, while p.o. immunization of the same amount of rAd5 did not induce any detectable immune responses. A mucosal prime-systemic boost regimen elicited potent cel- lular immunity in the small intestine and the systemic com- partments. The relative potencies of p.o. and intraileal routes of administration as primes for systemic boosting were next examined. Mice were primed on day 0 with empty rAd5 or rAd5-gp140B p.o. or via intraileal injection and were boosted i.m. on day 21 with rAd5-gp140B. Before the boosting step, PBMC were harvested for tetramer staining, and consistent with the results described above, immunization with rAd5- gp140B via intraileal injections induced significantly higher responses than p.o. administration (Fig. 4A). After the mice were boosted with rAd5-gp140B i.m., the mice primed via the intraileal route showed a dramatic increase in antigen-specific H-2Dd /PA-9ϩ CD8ϩ T-cell responses in both systemic and mucosal compartments compared to the responses of the group primed p.o. or i.m. (Fig. 4B). HIV-1 peptide-specific cytokine-producing CD8ϩ T cells and CD4ϩ T cells were also boosted effectively in the mice given an ileal prime and an i.m. boost. On average, 12.44% of CD8ϩ splenic T cells produced IFN-␥, and 10.65% of CD8ϩ splenic T cells were TNF-␣ pos- itive (Fig. 4C). The mice primed p.o. or via ileal injection and boosted i.m. generated higher levels of HIV-1 peptide-specific IFN-␥- and TNF-␣-producing CD4ϩ T cells than the mice immunized by i.m. injection only (Fig. 4C). There were no significant differences in CD4 frequencies between the prime- boost groups. The only differences were relative to the single- immunization group, indicating that prime-boost regimens in- duce higher frequencies of immune cells than a single immunization. HIV-1-specific IgG and IgA antibodies were also detected in sera and vaginal washes, but the differences between the mice primed p.o. and the mice primed by ileal injection were not significant (see Fig. S2 in the supplemental FIG. 4. Immune responses induced by ileum prime and i.m. boost. Mice were administered rAd5gp-140B (rAd5) or empty vector (Ϫ) via the p.o. or intraileal route and boosted i.m. (ϩ) 3 weeks later. (A) Per- centages of HIV-specific H-2Dd /PA-9ϩ CD8ϩ T cells from the PBMC immediately prior to the i.m. injection of rAd5-gp140B. (B) Percent- ages of HIV-specific H-2Dd /PA-9ϩ CD8ϩ T cells from the PBMC, spleens, MLN, and small intestines at 2 weeks after the i.m. injection of rAd5-gp140B. (C) HIV-1-specific IFN-␥- and TNF-␣-secreting CD4ϩ or CD8ϩ T lymphocytes from the spleens 2 weeks after i.m. injection of rAd5-gp140B. The data from each individual mouse and the mean values from the five mice (black bars) are shown. *, P Ͻ 0.05; **, P Ͻ 0.01; ***, P Ͻ 0.001. VOL. 83, 2009 INTRAILEAL rAd5 VACCINE STIMULATES MUCOSAL IMMUNITY 7171 onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
  • 8. material). These results demonstrate that priming directly in the ileum and boosting i.m. induced the most potent CD8ϩ T-cell immunity in the small intestine as well as in the systemic compartments. The fiber proteins of Ad41 did not increase the mucosal immunogenicity of a rare serotype vector, rAd35. The swap- ping of Ad5 fiber proteins into Ad35 has previously been shown to increase immunogenicity (29). Since we have seen intestinal and systemic immune responses induced by rAd41 vectors of a magnitude similar to that of responses induced by rAd5 (17), we determined whether the swap of Ad41 proteins into rAd35 would affect rAd35-mediated transduction and im- mune responses. Explant cultures of murine intestine were transduced with rAd35, rAd35/41L, and rAd35/41S, encoding luciferase at a 1- to 2-log-lower efficiency than that for rAd5 (Fig. 5A). We also compared the relative potencies of the rAd35 and rAd5 vectors in priming for an i.m. rAd5 boost. While an intraileal prime-i.m. boost with rAd5-gp140B elicited significantly higher levels of CD8ϩ T-cell responses in the spleen and in the small intestine than did rAd5 i.m. alone, intraileal administration of rAd35, rAd35/41L, and rAd35/41S failed to increase the rAd5 i.m boost (Fig. 5B and C, right, lane 3 versus lane 2, 4, 5, or 6). However, CD4ϩ T-cell responses did not show a difference after rAd5 boost, possibly due to the lower magnitude of responders than CD8ϩ cells typically seen with rAd5 immunization. In addition, an ileal prime and i.m. boost with rAd5-gp140B induced a twofold-greater IFN-␥ϩ CD8ϩ T-cell response in the small intestine than did rAd5 i.m. alone. In contrast, an ileal prime with rAd35, rAd35/41L, and rAd35/41S did not show a boost response in the small intestine higher than that with rAd5 i.m. alone (see Fig. S3 in the supplemental material). These results demonstrate that the direct intraileal injection of rAd5, but not that of rAd35, rAd35/41L, and rAd35/41S, could prime for an i.m. rAd5 boost to generate potent immune responses in the systemic and mu- cosal compartments. DISCUSSION The intestinal mucosa represents a primary site of HIV replication during the peak of infection (3, 23, 26, 43). Vacci- nation through the GI mucosa could potentially induce muco- sal immunity to prevent virus entry and/or limit the spread of infection. Oral immunization is the most convenient way to deliver mucosal vaccines. Although several studies have re- ported that the use of this route of immunization of Ad vectors encoding various virus antigens generated antibody responses in the serum and mucosal secretions (33, 37), the administra- tion of Ads encoding Gag or Env from HIV-1 or SIV elicited weak or undetectable intestinal mucosa responses (24, 30). The major obstacles to successful induction of mucosal immunity and/or systemic immunity via GI delivery of vaccine have not been defined previously. In this study utilizing murine intesti- nal explant cultures and in vivo gene expression assessments, we demonstrate that the combination of a low-pH environ- ment and the presence of proteases as well as the mucus/ glycocalyx in the GI tract prevents rAd5 from effectively trans- ducing the intestinal cells when delivered p.o. Therefore, p.o. immunization of vaccine vectors must overcome or bypass these barriers in the GI tract in order to be effective. We also show that the ileum is the most receptive site for rAd trans- duction and that intraileal injection of rAd5 results in a signif- icantly higher transgene expression in the ileum than does p.o. delivery. In addition, intraileal immunization with rAd5 did not induce significant systemic antivector neutralizing activity, since the rAd5 i.m. administration was effective in the homol- ogous rAd5 prime-rAd5 boost regimen. This is in agreement with our previous work demonstrating the absence of circulat- ing neutralizing antibodies following p.o. administration of rAd5 and rAd41 (17). It has also been shown that preexisting immunity to the vaccine carrier did not impair p.o. vaccination with Ad vectors in mice (45). Thus, the ileum represents a valid target for delivery of rAd vectors. Utilization of a route of administration that bypassed the barriers to ileal transduction revealed the potency of rAd5 as a mucosal immunogen. A single immunization of mice with rAd5 encoding HIV-1 gp140B via intraileal injection generated sig- nificantly higher numbers of H-2Dd /PA-9ϩ CD8ϩ T cells in both mucosal and systemic compartments and antigen-specific IFN-␥-, TNF-␣-, and IL-2-producing CD4ϩ and CD8ϩ T cells in the spleen than did p.o. administration (Fig. 3A and C). In fact, p.o. immunization did not generate any detectable im- mune response after a single dose in our study. These results indicate that the higher transduction from intraileal injections than from p.o. delivery (Fig. 2B) is associated with the stronger immune responses to HIV-1 gp140B. Systemic boosting with rAd5-gp140B dramatically increased CD8ϩ T-cell responses in the mice primed via the intraileal route in both systemic and mucosal compartments compared to responses in the group primed p.o. (Fig. 4B and C, right) as well as splenic CD4ϩ T-cell responses compared to those in the mice primed p.o. or via ileal injection (Fig. 4C, left). Whether the activation of CD4ϩ T cells would provide more target cells for HIV acqui- sition will be addressed in nonhuman primate (NHP) studies, though a previous study in which such mucosal responses were detected showed that these responses led to protection (21, 25). Oral dosing with replication-competent Ad vectors gener- ates systemic immune responses in nonhuman primates, but multiple primes and protein boosting are required for protec- tive efficacy against SIVmac251 challenge (32, 47). Oral vacci- nation with replication-competent Ad4 and Ad7 has been shown to generate protective Ad neutralizing antibodies in humans (11), indicating that replication-competent vectors elicit antivector neutralizing antibodies, which may compete with transgene-specific responses or may prevent multiple p.o. administrations of the same vector and affect efficacy. In addi- tion, the magnitude and contribution of the transgene-specific response in mediating protection by replication-competent rAd remain unknown in this model because the gut mucosal immune responses were not measured and compared to sys- temic responses. Thus, there are potential efficacy and regula- tory concerns with replication-competent rAds, and further studies are needed to assess the potential efficacy of gut mu- cosal immunization against lentivirus infection in NHP. Direct mucosal introduction of replication-defective vectors offers an alternative approach to address questions of immunogenicity and efficacy in this model. Although cellular responses were robust after intraileal injection, we did not detect a significant level of IgG or IgA antibody from fecal extracts (data not shown). Some animals showed IgA responses from the vaginal 7172 WANG ET AL. J. VIROL. onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
  • 9. washes, but the responses were low and inconsistent. Though collection methods for mucosal secretions can be optimized (19), it is likely that these responses are low in magnitude in the GI tract. While the results reported here describe the immunogenicity of rAd5- and rAd35-based vectors, we have previously ana- lyzed rAd41, an enteric tropic Ad, for its ability to generate immune responses through direct ileum lumenal administra- FIG. 5. Transduction efficiencies and immune responses induced by rAd35 and rAd35/41 chimeras. (A) Transduction of ileum explants by rAd vectors. RLU, relative luminescence units. (B and C) Immune responses induced by rAd vectors in mice. Mice were administered the indicated rAd vectors or empty rAd5 vector (Ϫ) via the intraileal route and boosted i.m. 3 weeks later. (B) Percentages of HIV-specific H-2Dd /PA-9ϩ CD8ϩ T cells from spleens and small intestines 2 weeks after the i.m. rAd5-gp140B boost. (C) HIV-1-specific IFN-␥-secreting CD4ϩ or CD8ϩ T lymphocytes from spleens 2 weeks after i.m. rAd5-gp140B boost. The data from each individual mouse and the mean values from the five mice (black bars) are shown. -, empty vector. *, P Ͻ 0.05; **, P Ͻ 0.01; ***, P Ͻ 0.001. VOL. 83, 2009 INTRAILEAL rAd5 VACCINE STIMULATES MUCOSAL IMMUNITY 7173 onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
  • 10. tion (17). When rAd41 was administered directly to the lumen of the ileum, it efficiently primed antigen-specific cellular im- munity. Ad41, a member of the Ad species F, has been re- ported to bind/uptake more efficiently than Ad5 in the isolated intestinal loops of rats. Furthermore, gene delivery with Ad5 to differentiated enterocytes in vitro and to the rat jejunum in vivo is extremely inefficient (7, 8, 44). However, the immune responses induced by Ad41 and Ad5 through intraileal injec- tion priming and i.m. boosting were not significantly different, which highlighted that vectors derived from mucosal patho- gens, both enteric and respiratory, can be used in these regi- mens. However, due to the prevalence of neutralizing antibod- ies to Ad5 and Ad41 in humans, the development and use of alternative serotypes of Ad vectors for virus vaccines remain a priority. Ad35 is one relatively well-characterized rare serotype vector and has been associated with infections of the kidney and urinary tract. Therefore, the fiber swap study was done for two reasons: (i) to define the contribution of fiber versus hexon to transduction of mucosal epithelium and (ii) to generate vectors resistant to anti-Ad41 hexon neutralizing antibodies, should they be mediated by fiber. Here, we found that rAd35 did not transduce the organ cultures of intestinal segments as efficiently as rAd5 and that rAd35 was less immunogenic in the ileal priming regimen, thus allowing us to address these ques- tions. Unexpectedly, grafting the long fiber or the short fiber of Ad41 to the rAd35 vector did not improve the transduction efficiency of intestinal explants or immunogenicity, suggesting that the Ad41 fiber is not the only determinant of enteric tropism. However, these results suggest that the immunoge- nicity of rAd vectors in vivo correlates with the efficiency of transduction of intestinal explants ex vivo. We conclude that the lower small intestine represents a target for antigen deliv- ery with certain rAd vectors. Further development of special formulations to deliver rAd to the ileum should be emphasized for practical vaccination. Our results suggest that it is important to pursue a mucosal route of administration of rAd vaccine vectors for HIV. The strong cellular immune responses induced by intraileal injec- tion, further strengthened by i.m. boost, suggest a strategy for protection of gut-associated lymphoid tissue lymphocytes from HIV-1 infection. Possible mechanisms of immune stimulation include persistent antigen expression from lamina propria cells transduced by the rAd vector (7, 40). Further characterization of the immune responses induced by the vaccination regimens employed in this work, such as the balance of CD8ϩ and CD4ϩ T-cell responses, the polyfunctionality of the T cells, and the duration of the immune response, could provide a better pre- diction as to which is the most important type of immune response for protection against HIV-1 (22). Regarding the applicability of these data to other species, as well as the uncertain applicability to humans, this study represents one of a stepwise series of studies to determine whether mucosal immunity can be enhanced by different rAd vectors with alter- native routes of administration. Having been demonstrated with rodents, this will next be evaluated with nonhuman pri- mate challenge models to determine its contribution to im- mune protection. Should these results support the concept of a mucosal vaccine, an effort will be made to formulate an Ad vector for delivery to the ileum in humans, since the site- specific delivery technology to deliver drugs and vaccines to the ileum has been developed with partial success so far (14, 16, 27, 36). The use of rAd5 vaccines in populations with moderate to high neutralizing antibodies to Ad5 based on the STEP trial (3a) results is a concern. It will be difficult to define mecha- nisms responsible for this effect or even to document it defin- itively. At the same time, rAd5 vectors are well characterized and induce strong immune responses. There are numerous differences in vector design, insert combination, and immuni- zation regimen that may result in different performances in future clinical studies. Regardless of its future clinical utility, rAd5 remains a useful model vector for inducing mucosal im- munity and for determining how mucosal immune responses contribute to protection. Once these key questions are ad- dressed, rAd5 vectors can be used if justified by efficacy/safety considerations, or, if concerns remain, other vectors can be substituted. ACKNOWLEDGMENTS We thank Ati Tislerics for help with manuscript preparation, Brenda Hartman and Morteza Loghmani for assistance with figures, Srinivas Rao and Saran Bao for help with surgical techniques, D. Margulies and the NIAID Tetramer Core Facility for production of tetramers, and members of the Nabel laboratory for helpful discussions. This work was supported by the Intramural Research Program of the National Institutes of Health, Vaccine Research Center, NIAID, and by the Bill and Melinda Gates Foundation. REFERENCES 1. Aoki, Y., M. Morishita, and K. Takayama. 2005. Role of the mucous/glyco- calyx layers in insulin permeation across the rat ileal membrane. Int. J. Pharm. 297:98–109. 2. Benihoud, K., P. Yeh, and M. Perricaudet. 1999. Adenovirus vectors for gene delivery. Curr. Opin. Biotechnol. 10:440–447. 3. Brenchley, J. M., T. W. Schacker, L. E. Ruff, D. A. Price, J. H. Taylor, G. J. Beilman, P. L. Nguyen, A. Khoruts, M. Larson, A. T. Haase, and D. C. Douek. 2004. CD4ϩ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J. Exp. Med. 200:749–759. 3a.Buchbinder, S. P., D. V. Mehrotra, A. Duerr, D. W. Fitzgerald, R. Mogg, D. Li, P. B. Gilbert, J. R. Lama, M. Marmor, C. Del Rio, M. J. McElrath, D. R. Casimiro, K. M. Gottesdiener, J. A. Chodakewitz, L. Corey, M. N. Robert- son, and the Step Study Protocol Team. 2008. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet 372:1881–1983. 4. Casimiro, D. R., A. J. Bett, T. M. Fu, M. E. Davies, A. Tang, K. A. Wilson, M. Chen, R. Long, T. McKelvey, M. Chastain, S. Gurunathan, J. Tartaglia, E. A. Emini, and J. Shiver. 2004. Heterologous human immunodeficiency virus type 1 priming-boosting immunization strategies involving replication- defective adenovirus and poxvirus vaccine vectors. J. Virol. 78:11434–11438. 5. Casimiro, D. R., L. Chen, T. M. Fu, R. K. Evans, M. J. Caulfield, M. E. Davies, A. Tang, M. Chen, L. Huang, V. Harris, D. C. Freed, K. A. Wilson, S. Dubey, D. M. Zhu, D. Nawrocki, H. Mach, R. Troutman, L. Isopi, D. Williams, W. Hurni, Z. Xu, J. G. Smith, S. Wang, X. Liu, L. Guan, R. Long, W. Trigona, G. J. Heidecker, H. C. Perry, N. Persaud, T. J. Toner, Q. Su, X. Liang, R. Youil, M. Chastain, A. J. Bett, D. B. Volkin, E. A. Emini, and J. W. Shiver. 2003. Comparative immunogenicity in rhesus monkeys of DNA plas- mid, recombinant vaccinia virus, and replication-defective adenovirus vec- tors expressing a human immunodeficiency virus type 1 gag gene. J. Virol. 77:6305–6313. 6. Cheng, C., J. G. Gall, W. P. Kong, R. L. Sheets, P. L. Gomez, C. R. King, and G. J. Nabel. 2007. Mechanism of ad5 vaccine immunity and toxicity: fiber shaft targeting of dendritic cells. PLoS Pathog. 3:e25. 7. Croyle, M. A., M. Stone, G. L. Amidon, and B. J. Roessler. 1998. In vitro and in vivo assessment of adenovirus 41 as a vector for gene delivery to the intestine. Gene Ther. 5:645–654. 8. Croyle, M. A., E. Walter, S. Janich, B. J. Roessler, and G. L. Amidon. 1998. Role of integrin expression in adenovirus-mediated gene delivery to the intestinal epithelium. Hum. Gene Ther. 9:561–573. 9. Ertl, H. C. 2005. Immunological insights from genetic vaccines. Virus Res. 111:89–92. 10. Gao, W., A. C. Soloff, X. Lu, A. Montecalvo, D. C. Nguyen, Y. Matsuoka, P. D. Robbins, D. E. Swayne, R. O. Donis, J. M. Katz, S. M. Barratt-Boyes, and A. Gambotto. 2006. Protection of mice and poultry from lethal H5N1 avian 7174 WANG ET AL. J. VIROL. onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom
  • 11. influenza virus through adenovirus-based immunization. J. Virol. 80:1959– 1964. 11. Gaydos, C. A., and J. C. Gaydos. 1995. Adenovirus vaccines in the U.S. military. Mil. Med. 160:300–304. 12. Greber, U. F. 2002. Signalling in viral entry. Cell. Mol. Life Sci. 59:608–626. 13. Haase, A. T. 2005. Perils at mucosal front lines for HIV and SIV and their hosts. Nat. Rev. Immunol. 5:783–792. 14. Hamman, J. H., G. M. Enslin, and A. F. Kotze. 2005. Oral delivery of peptide drugs: barriers and developments. BioDrugs 19:165–177. 15. Jaiswal, S., N. Khanna, and S. Swaminathan. 2003. Replication-defective adenoviral vaccine vector for the induction of immune responses to dengue virus type 2. J. Virol. 77:12907–12913. 16. Klein, S., J. Stein, and J. Dressman. 2005. Site-specific delivery of anti- inflammatory drugs in the gastrointestinal tract: an in-vitro release model. J. Pharm. Pharmacol. 57:709–719. 17. Ko, S. Y., C. Cheng, W. P. Kong, L. Wang, M. Kanekiyo, D. Einfeld, C. R. King, J. G. Gall, and G. J. Nabel. 2009. Enhanced induction of intestinal cellular immunity by oral priming with enteric adenovirus 41 vectors. J. Vi- rol. 83:748–756. 18. Kobinger, G. P., H. Feldmann, Y. Zhi, G. Schumer, G. Gao, F. Feldmann, S. Jones, and J. M. Wilson. 2006. Chimpanzee adenovirus vaccine protects against Zaire Ebola virus. Virology 346:394–401. 19. Kozlowski, P. A., R. M. Lynch, R. R. Patterson, S. Cu-Uvin, T. P. Flanigan, and M. R. Neutra. 2000. Modified wick method using Weck-Cel sponges for collection of human rectal secretions and analysis of mucosal HIV antibody. J. Acquir. Immune Defic. Syndr. 24:297–309. 20. Lemiale, F., H. Haddada, G. J. Nabel, D. E. Brough, C. R. King, and J. G. Gall. 2007. Novel adenovirus vaccine vectors based on the enteric-tropic serotype 41. Vaccine 25:2074–2084. 21. Letvin, N. L., J. R. Mascola, Y. Sun, D. A. Gorgone, A. P. Buzby, L. Xu, Z. Y. Yang, B. Chakrabarti, S. S. Rao, J. E. Schmitz, D. C. Montefiori, B. R. Barker, F. L. Bookstein, and G. J. Nabel. 2006. Preserved CD4ϩ central memory T cells and survival in vaccinated SIV-challenged monkeys. Science 312:1530–1533. 22. Letvin, N. L., S. S. Rao, V. Dang, A. P. Buzby, B. Korioth-Schmitz, D. Dombagoda, J. G. Parvani, R. H. Clarke, L. Bar, K. R. Carlson, P. A. Kozlowski, V. M. Hirsch, J. R. Mascola, and G. J. Nabel. 2007. No evidence for consistent virus-specific immunity in simian immunodeficiency virus- exposed, uninfected rhesus monkeys. J. Virol. 81:12368–12374. 23. Li, Q., L. Duan, J. D. Estes, Z. M. Ma, T. Rourke, Y. Wang, C. Reilly, J. Carlis, C. J. Miller, and A. T. Haase. 2005. Peak SIV replication in resting memory CD4ϩ T cells depletes gut lamina propria CD4ϩ T cells. Nature 434:1148–1152. 24. Lin, S. W., A. S. Cun, K. Harris-McCoy, and H. C. Ertl. 2007. Intramuscular rather than oral administration of replication-defective adenoviral vaccine vector induces specific CD8ϩ T cell responses in the gut. Vaccine 25:2187– 2193. 25. Mattapallil, J. J., D. C. Douek, A. Buckler-White, D. Montefiori, N. L. Letvin, G. J. Nabel, and M. Roederer. 2006. Vaccination preserves CD4 memory T cells during acute simian immunodeficiency virus challenge. J. Exp. Med. 203:1533–1541. 26. Mattapallil, J. J., D. C. Douek, B. Hill, Y. Nishimura, M. Martin, and M. Roederer. 2005. Massive infection and loss of memory CD4ϩ T cells in multiple tissues during acute SIV infection. Nature 434:1093–1097. 27. Mercier, G. T., P. N. Nehete, M. F. Passeri, B. N. Nehete, E. A. Weaver, N. S. Templeton, K. Schluns, S. S. Buchl, K. J. Sastry, and M. A. Barry. 2007. Oral immunization of rhesus macaques with adenoviral HIV vaccines using en- teric-coated capsules. Vaccine 25:8687–8701. 28. Monteiro-Riviere, N. A., and J. A. Popp. 1986. Ultrastructural evaluation of acute nasal toxicity in the rat respiratory epithelium in response to formal- dehyde gas. Fundam. Appl. Toxicol. 6:251–262. 29. Nanda, A., D. M. Lynch, J. Goudsmit, A. A. Lemckert, B. A. Ewald, S. M. Sumida, D. M. Truitt, P. Abbink, M. G. Kishko, D. A. Gorgone, M. A. Lifton, L. Shen, A. Carville, K. G. Mansfield, M. J. Havenga, and D. H. Barouch. 2005. Immunogenicity of recombinant fiber-chimeric adenovirus serotype 35 vector-based vaccines in mice and rhesus monkeys. J. Virol. 79:14161–14168. 30. Oomura, K., K. Q. Xin, M. Takakura, K. Shinoda, N. Jounai, and K. Okuda. 2006. Oral administration of the adenovirus vector induces systemic immu- nity rather than intestinal mucosal immunity. Vaccine 24:1045–1046. 31. Patel, A., Y. Zhang, M. Croyle, K. Tran, M. Gray, J. Strong, H. Feldmann, J. M. Wilson, and G. P. Kobinger. 2007. Mucosal delivery of adenovirus- based vaccine protects against Ebola virus infection in mice. J. Infect. Dis. 196(Suppl. 2):S413–S420. 32. Patterson, L. J., N. Malkevitch, D. Venzon, J. Pinczewski, V. R. Gomez- Roman, L. Wang, V. S. Kalyanaraman, P. D. Markham, F. A. Robey, and M. Robert-Guroff. 2004. Protection against mucosal simian immunodeficiency virus SIVmac251 challenge by using replicating adenovirus-SIV multigene vaccine priming and subunit boosting. J. Virol. 78:2212–2221. 33. Pinto, A. R., J. C. Fitzgerald, G. P. Gao, J. M. Wilson, and H. C. Ertl. 2004. Induction of CD8ϩ T cells to an HIV-1 antigen upon oral immunization of mice with a simian E1-deleted adenoviral vector. Vaccine 22:697–703. 34. Pope, M., and A. T. Haase. 2003. Transmission, acute HIV-1 infection and the quest for strategies to prevent infection. Nat. Med. 9:847–852. 35. Robbins, P. D., and S. C. Ghivizzani. 1998. Viral vectors for gene therapy. Pharmacol. Ther. 80:35–47. 36. Schellekens, R. C., F. Stellaard, D. Mitrovic, F. E. Stuurman, J. G. Kos- terink, and H. W. Frijlink. 2008. Pulsatile drug delivery to ileo-colonic segments by structured incorporation of disintegrants in pH-responsive poly- mer coatings. J. Control. Release 132:91–98. 37. Sharpe, S., A. Fooks, J. Lee, K. Hayes, C. Clegg, and M. Cranage. 2002. Single oral immunization with replication deficient recombinant adenovirus elicits long-lived transgene-specific cellular and humoral immune responses. Virology 293:210–216. 38. Shiver, J. W., and E. A. Emini. 2004. Recent advances in the development of HIV-1 vaccines using replication-incompetent adenovirus vectors. Annu. Rev. Med. 55:355–372. 39. Stahl-Hennig, C., S. Kuate, M. Franz, Y. S. Suh, H. Stoiber, U. Sauermann, K. Tenner-Racz, S. Norley, K. S. Park, Y. C. Sung, R. Steinman, P. Racz, and K. Uberla. 2007. Atraumatic oral spray immunization with replication-defi- cient viral vector vaccines. J. Virol. 81:13180–13190. 40. Tatsis, N., J. C. Fitzgerald, A. Reyes-Sandoval, K. C. Harris-McCoy, S. E. Hensley, D. Zhou, S. W. Lin, A. Bian, Z. Q. Xiang, A. Iparraguirre, C. Lopez-Camacho, E. J. Wherry, and H. C. Ertl. 2007. Adenoviral vectors persist in vivo and maintain activated CD8ϩ T cells: implications for their use as vaccines. Blood 110:1916–1923. 41. Thorner, A. R., and D. H. Barouch. 2007. HIV-1 vaccine development: progress and prospects. Curr. Infect. Dis. Rep. 9:71–75. 42. Tobery, T. W., S. A. Dubey, K. Anderson, D. C. Freed, K. S. Cox, J. Lin, M. T. Prokop, K. J. Sykes, R. Mogg, D. V. Mehrotra, T. M. Fu, D. R. Casimiro, and J. W. Shiver. 2006. A comparison of standard immunogenicity assays for monitoring HIV type 1 gag-specific T cell responses in Ad5 HIV type 1 gag vaccinated human subjects. AIDS Res. Hum. Retrovir. 22:1081–1090. 43. Veazey, R. S., M. DeMaria, L. V. Chalifoux, D. E. Shvetz, D. R. Pauley, H. L. Knight, M. Rosenzweig, R. P. Johnson, R. C. Desrosiers, and A. A. Lackner. 1998. Gastrointestinal tract as a major site of CD4ϩ T cell depletion and viral replication in SIV infection. Science 280:427–431. 44. Walter, E., M. A. Croyle, B. J. Roessler, and G. L. Amidon. 1997. The absence of accessible vitronectin receptors in differentiated tissue hinders adenoviral-mediated gene transfer to the intestinal epithelium in vitro. Pharm. Res. 14:1216–1222. 45. Xiang, Z. Q., G. P. Gao, A. Reyes-Sandoval, Y. Li, J. M. Wilson, and H. C. Ertl. 2003. Oral vaccination of mice with adenoviral vectors is not impaired by preexisting immunity to the vaccine carrier. J. Virol. 77:10780–10789. 46. Xiang, Z. Q., Y. Yang, J. M. Wilson, and H. C. Ertl. 1996. A replication- defective human adenovirus recombinant serves as a highly efficacious vac- cine carrier. Virology 219:220–227. 47. Zhou, Q., R. Hidajat, B. Peng, D. Venzon, M. K. Aldrich, E. Richardson, E. M. Lee, V. S. Kalyanaraman, G. Grimes, V. R. Gomez-Roman, L. E. Summers, N. Malkevich, and M. Robert-Guroff. 2007. Comparative evaluation of oral and intranasal priming with replication-competent adenovirus 5 host range mutant (Ad5hr)-simian immunodeficiency virus (SIV) recombinant vaccines on immu- nogenicity and protective efficacy against SIV(mac251). Vaccine 25:8021–8035. VOL. 83, 2009 INTRAILEAL rAd5 VACCINE STIMULATES MUCOSAL IMMUNITY 7175 onFebruary25,2013byguesthttp://jvi.asm.org/Downloadedfrom