Significant protection against high-dose simian immunodeficiency virus challenge conferred by a new prime-boost vaccine regimen

Size: px
Start display at page:

Download "Significant protection against high-dose simian immunodeficiency virus challenge conferred by a new prime-boost vaccine regimen"

Transcription

1 JVI Accepts, published online ahead of print on 1 April 0 J. Virol. doi:./jvi.00- Copyright 0, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved Significant protection against high-dose simian immunodeficiency virus challenge conferred by a new prime-boost vaccine regimen John B. Schell, 1 Nina F. Rose, 1 Kapil Bahl, 1 Kathryn Diller, 1 Linda Buonocore, 1 Meredith Hunter, Preston A. Marx, Ratish Gahmbira, Haili Tang, David C. Montefiori, Welkin E. Johnson, and John K. Rose 1 1 Yale University School of Medicine, New Haven, CT Tulane National Primate Research Center, Covington, LA Duke University Medical Center, Durham, NC New England Regional Primate Research Center, Harvard University, Southborough, MA Running Head: Vaccine Protection from SIV challenge 1 0 Corresponding author: John K. Rose Department of Pathology Yale University School of Medicine Cedar St. New Haven, CT. 0 john.rose@yale.edu Tel 0-- Fax 0-- 1

2 We constructed vaccine vectors based on live recombinant vesicular stomatitis virus (VSV) and a Semliki Forest Virus replicon (SFVG) that propagates through expression of the VSV glycoprotein (G). These vectors expressing SIV Gag and Env proteins were used to vaccinate rhesus macaques in a new heterologous prime-boost regimen designed to optimize induction of antibody. Six vaccinated animals and six controls were then given a high-dose mucosal challenge with the diverse SIVsmE0 quasispecies. All control animals became infected and had peak viral RNA loads of - copies/ml. In contrast, four of the vaccinees showed significant (p=0.0) apparent sterilizing immunity and no detectable viral load. Subsequent CD + T cell depletion confirmed the absence of SIV infection in these animals. The two other vaccinees had peak viral loads of x and x copies/ml, levels below all of the controls, and showed undetectable virus loads by day post-challenge. The vaccine regimen induced high-titer pre-challenge serum neutralizing antibodies (nab) to some cloned SIVsmE0 Env proteins, but antibodies able to neutralize the challenge virus swarm were not detected. The cellular immune responses induced by the vaccine were generally weak and did not correlate with protection. Although the immune correlates of protection are not yet clear, the heterologous VSV/SFVG prime-boost is clearly a potent vaccine regimen for inducing virus nabs and protection against a heterogeneous viral swarm.

3 Development of an effective HIV-1 (human immunodeficiency virus type 1) vaccine is a critical global health priority and has been a major scientific challenge for over years. Initial clinical trials of an HIV-1 Env (Envelope) protein vaccine showed no efficacy (, ). This failure was likely due to the inability of the vaccine to generate neutralizing antibodies (nabs) to the diverse HIV-1 Env proteins present in the infecting strains and potentially to the high-risk population used in the clinical trial. Studies in non-human primates using defective adenovirus type vectors (Ad) indicated that induction of potent cellular immunity to the simian immunodeficiency (SIV) proteins could reduce viral loads following SIV infection and at least slow disease progression (,,, ). Such studies led to clinical trials of Ad vectors expressing HIV-1 Gag, Pol, and Nef proteins. Despite the induction of significant cellular immune responses in vaccinees, this vaccine failed to protect against HIV-1 infection or to reduce viral loads following infection (, ). In addition, vaccinees with pre-existing Ad-specific nabs exhibited an enhanced rate of HIV-1 acquisition. The latter finding has led to major concerns with the use of vaccine vectors for which there is significant pre-existing immunity in the human population. A more recent clinical trial in a low-risk population in Thailand generated renewed hope for HIV vaccine development because it showed marginally significant protection from infection (1). The vaccine employed a heterologous prime-boost strategy using the canary pox vector ALVAC-HIV, expressing HIV Gag, Pro, and Env, followed by boosting with purified HIV Env protein. The vaccine did not generate consistent cellular or

4 detectable nab responses to HIV-1 and the immune correlates of protection remain unknown. These results suggests that a more potent vaccine regimen will be required to generate an HIV-1 vaccine providing more significant protection. Our own studies have been directed toward development of two virus-derived vaccine vectors for which there is no significant pre-existing immunity in the human population. The two vector systems are based on attenuated vesicular stomatitis virus (0,,, ) and an alphavirus, Semliki Forest Virus, replicon (SFVG) that is packaged by a VSV glycoprotein into infectious vesicles (, ). VSV-based HIV vaccine vectors () are scheduled for clinical trials beginning in 0. In previous studies, VSV vectors expressing Env and Gag proteins have provided protection against disease following challenge with an SIV/HIV (SHIV) hybrid virus (). A heterologous prime-boost regimen employing VSV and modified vaccinia virus Ankara (MVA) vectors was also highly effective against this SHIV challenge (0) and provided protection lasting over five years (). Vaccine regimens using heterologous viral vectors for priming and boosting are highly effective at focusing the boost response on the vaccine antigens expressed in the vectors (1,, 1,,, ). In addition, the potency of the vector combinations may derive from the induction of a more diverse set of innate immune responses that act as adjuvants. An extensive comparison of six different vaccine vectors in heterologous primeboost combinations showed that a VSV vector combined with an alphavirus replicon was the most synergistic combination for induction of antibody to a viral protein (1).

5 In the SIV vaccine study reported here, we have tested VSV and SFVG alphavirus replicon vectors expressing SIV Env and Gag proteins in a heterologous prime-boost regimen. Our goal was to induce optimal nab responses to the SIV challenge virus and determine if the vaccine regimen might be sufficient to provide protection against challenge with a highly pathogenic SIV quasispecies, SIVsmE0. We report that this vaccine regimen provided statistically significant and apparently sterilizing protection from high-dose mucosal challenge. MATERIALS AND METHODS Vaccine vector construction. To construct a DNA vector expressing an SFV RNA replicon with two SFV subgenomic promoters, we started with the pbk-t-sfv plasmid that has a CMV promoter preceding the SFV replicon sequence (0). The single Apa I site in this vector was eliminated with T DNA polymerase. To generate a plasmid designated pbksfvg-gp10, an Spe I-Sac I fragment containing the CMV promoter and the beginning of the SFV replicon (, nucleotides) from this plasmid was ligated to a, nucleotide Spe I-SacI fragment from psfvg-gp10 (). This vector encodes the SFV replicon with one subgenomic promoter driving expression of VSV G and a second SFV subgenomic promoter driving expression of HIV gp10. Apa I sites flank the gp10 sequence in this plasmid. E0 env and gag genes were obtained by PCR of DNA from CEMx1 cells infected with SIVsmE0, and cloned into pbssk +. An E0 envg gene was then generated by overlap extension PCR (1). The E0 envg gene encodes an Env protein with its cytoplasmic domain replaced with the VSV G cytoplasmic domain for efficient

6 incorporation into VSV particles. E0 envg was amplified by PCR from the plasmid pbse0envg using the forward primer: GATCGATCGGGCCCTTAATTAACTCGAGAAGATGGGATGTCTTGGGAATCAGC- and the reverse primer: GATCCCCCCCGGGCCCTGCAGGCTAGCTTACTTTCCAAGTCGGTTCATCTC-. Both primers contained Apa I sites (underlined) and also introduced unique Pac I and Sbf I sites for use in subsequent directional cloning. The PCR product was digested with Apa I and then cloned into pbksfvg-gp10 following excision of the gp10 insert with Apa I. The plasmid obtained was designated pbksfvg-e0envg. To construct pbksfvg-e0gag, the E0 gag gene was amplified by PCR using the forward primer: -GATCGATCGGGCCCTTAATTAAGGATCCAAGATGGGCGCGAGAAACTCCGTC- containing an underlined Pac I site, and a reverse primer: -GATCGATCGGGCCCTGCAGGGATCCCTACTGGTCTTCTCCAAAGAGAG- containing an underlined Sbf I site. The PCR product was digested with Pac I and Sbf I and cloned into pbksfvg-e0envg after excising the E0EnvG with Pac I and Sbf I. The E0 gag and EnvG genes were also inserted into the fifth position of the pvsv- XN plasmids expressing either the VSV Indiana or New Jersey serotype G proteins (,, 1). VSV recombinants expressing the proteins were then derived by standard procedures (). Correct protein expression was verified by [ S] methionine labeling and SDS-PAGE of lysates from infected cells and by immunofluorescence microscopy. The vectors are shown in Fig. 1A.

7 1 1 Preparation of vaccine stocks. To prepare SFVG replicon particles, BHK-1 cells on cm diameter dishes ( x cells/dish) were transfected with µg of pbksfvg- E0Gag or pbksfvg-e0envg using Lipofectamine (Invitrogen) and OptiMEM medium according to the manufacturers protocol. After five hours, the transfection medium was removed and replaced with ml DMEM + % FBS. After hours, the cells were sonicated to obtain optimal release of the infectious particles and particles were then concentrated and titered as described previously for cells transfected directly with SFVG replicon RNA (). Serum-free stocks of VSV vectors were grown on BHK-1 cells and titered as described previously (). Vaccinations. Indian rhesus macaques were housed and cared for at the Tulane National Primate Research Center (TNPRC). Vaccinations followed the schedule shown in Fig. 1B. For the VSV prime and second boost, a total of 1x plaque forming units (pfu) of both VSV-E0Gag and VSV-E0EnvG viruses were delivered intramuscularly (IM; x 1 pfu) and intranasally (IN; x pfu) to each monkey. The SFVG replicon particle boost was delivered by intramuscular injection only. Each animal received 1x infectious units (i.u.) of both SFVG-Gag and SFVG-EnvG. Animals in the control group were primed and boosted with VSV vectors expressing an influenza hemagglutinin protein. Samples and viral load assays. All tissue and serum samples were shipped cryogenically frozen to Yale University and were stored in liquid nitrogen upon arrival. Branched DNA (bdna) analysis on serum samples was done by Siemens Diagnostics, Clinical Laboratory (Berkeley, CA).

8 Neutralization Assays. Neutralization was measured as a function of reductions in luciferase reporter gene expression after either a single round of infection in TZM-bl cells () or multiple rounds of replication in..egfp.luc.m (M-Luc) cells (). Neutralization titers in both assays are the dilution at which relative luminescence units (RLU) were reduced by 0% compared to virus control wells after subtraction of background RLUs. Assay stocks of molecularly cloned Env-pseudotyped virus E0/CR-PK-A and SIVsmE0. were prepared by transfection in T cells and were titrated in TZM-bl cells as described (). E0/CR-PK-A and BR-CGV.IR are SGA-derived plasma clones obtained at peak viremia during primary infection after intrarectal challenge () and exhibit a resistant tier neutralization phenotype (D. Montefiori, unpublished data). E0. is a functional rev-env cassette cloned from DNA of PBMC infected in vitro and exhibits a highly sensitive tier 1A neutralization phenotype. An uncloned stock of SIVsmE0 was prepared in CEMx1 cells and titrated in M-Luc cells () for use in neutralization assays. The VSV G-E0EnvG-GFP and VSV G neutralization assays were described previously (, ). The VSV GE0EnvG-GFP neutralization assay used HOS-CD-CCR cells obtained from the NIH AIDS research and reference reagent program and were described previously (). An endpoint of ninety percent neutralization of infection was scored by counting infected (GFP + ) cells by fluorescence microscopy. VSV vector neutralization assays were described previously (). ELISA for serum antibody to oligomeric SIV Env. Enzyme-linked immunosorbent assays (ELISAs) were performed essentially as described previously for HIV Env (),with

9 modifications published previously (). SIV gp10 was obtained from the medium of cells infected with a vaccinia virus recombinant (vpaul) encoding the secreted SIVmac gp10 (a gift from Dr. R. Doms, University of Pennsylvania). Optical densities were determined at a wavelength of 1 nm in a Bio-Rad ELISA plate reader. Titers were determined from graphs of the data and are given as the reciprocal of the serum dilution that gave an absorbance of 0. after subtraction of background values obtained from an identical dilution series with preimmune sera. Flow cytometry. For cell surface marker and tetramer analysis, cryo-preserved cells were washed in staining buffer (PBS containing % fetal calf serum) and stained for surface marker expression. Directly conjugated monoclonal antibodies (BD; San Jose, CA) CD-V0 (SP-), CD-PerCP (L00), and CD-Alexa Fluor-00 (RPA-T) were used to discern T cell subsets. Cells were co-stained with both Mamu-A*01 specific APC and PE conjugated tetramer containing the SIV gag peptide pc (Beckman Coulter; Fullerton, CA). Cell viability was assessed with the LIVE/DEAD Fixable Aqua dead cell stain kit (Molecular Probes; Eugene, OR). Following surface staining, cells were washed in staining buffer and fixed using Cytofix fixation buffer (BD). Cells were then resuspended in staining buffer prior to data acquisition using a multicolor LSR II flow cytometer (BD). Collected data was analyzed using FlowJo software version.. (Treestar; Ashland, OR). ELISpot assays. The ELISpot protocol has been described previously in greater detail (0). Cryo-preserved PBMCs were thawed and pelleted by centrifugation. Cells were infected with recombinant vaccinia virus (rvv) expressing either SIV Gag, Env, or bacteriophage T RNA polymerase (negative control) at a multiplicity of infection (MOI)

10 of. In parallel, cells were either treated with PMA (0 ng/ml) and ionomycin (0 ng/ml) or mock infected, and served as positive and negative controls, respectively. Cultures were stimulated for 1 hours in a humidified incubator before being used in the ELISpot assay. MultiScreen HTS -well plates (Millipore; Bedford, MA) were coated with mouse anti-human interferon gamma antibody at a final concentration of 1µg/well (BD). After overnight infection or stimulation described above, x PBMCs were transferred to the antibody coated plates. After a five-hour incubation at o C, the cells were washed off the plates using PBS-Tween (0.0%) and then treated with an anti-mouse IgG biotinylated antibody (Invitrogen; Camarillo, CA) for one hour. Upon washing, plates were then treated with streptavadin-hrp (Southern Biotech; Birmingham, AL) and then exposed to one-step chromogenic substrate (Thermo Scientific; Rockford, IL). Plates were washed, allowed to air dry and finally counted for spot forming cells (SFC). Data was analyzed and plotted using Instat software, version.0b (GraphPad Software; San Diego, CA). CD + T cell depletion For CD + T cell depletion experiments, animals were injected 1 days post-challenge with a rhesus recombinant anti-cdα antibody from the NIH Nonhuman Primate Reagent Resource (M-T0). The initial dose of antibody ( mg/kg body weight) was delivered subcutaneously, whereas the three subsequent doses ( mg/kg) were delivered intravenously RESULTS

11 Study design and antibody responses to vectors. In this study two groups of rhesus macaques were vaccinated, boosted, and challenged according to the schedule shown in Fig. 1B. In each group, we included three animals carrying the MamuA*01 MHC I allele to allow enumeration of SIV Gag-specific, CD T cells using MHC I tetramers. Animals with other MHC I alleles that have been associated with better control of SIV replication (B0, B1) were excluded from the vaccine group. The six vaccine group animals were primed with VSV-based vectors expressing EnvG and Gag proteins derived from SIVsmE0 quasispecies (Fig. 1A). The EnvG protein is an E0 Env with its cytoplasmic tail replaced with the VSV G cytoplasmic tail. This modification enhances EnvG protein incorporation into VSV particles (1). The six control group animals received VSV vectors expressing an unrelated influenza antigen. The priming vector expressed the VSV New Jersey (NJ) serotype glycoprotein and nabs to VSV NJ were detected in all animals by day (Fig. 1C). The vaccine group animals were then boosted at day with SFVG propagating replicons expressing the same EnvG and Gag proteins (Fig. 1A), but expressing the Indiana (I) serotype glycoprotein to avoid nabs generated to the priming vector. At day, the animals had all developed nabs to VSV(I), but these were between and -fold lower than we have previously seen in macaques vaccinated with VSV(I) vectors (Fig. 1C, and (0)). We then boosted all vaccine group animals at day with a VSV(I) vector expressing the E0 EnvG and Gag proteins. Following the boost, the nab responses to VSV(I) increased more than -fold by day 1. This result suggested that the boost was effective despite the exposure to the VSV G(I) protein encoded by the SFVG boosting vector. Control and vaccine group animals

12 were all given a high-dose (000 TCID0), intrarectal challenge of a SIVsmE0 stock at day 1 (Fig. 1B). Viral loads following SIVsmE0 challenge. For the initial evaluation of the vaccine effectiveness we measured viral loads in the vaccine and control groups (Figs. A, B). After challenge, all control animals became infected and had peak viral loads between 1x and x copies/ml of plasma (Fig. A). The three control animals with the highest loads all progressed to AIDS within three to eight months following challenge. Interestingly, two of these animals, EN and N carried the MamuA*01 MHC allele that is associated with better control of SIVmac/1 challenge (,, ), but was not sufficient to provide control of SIVsmE0 loads. The three other control animals were able to reduce their viral loads to below detection within eight months. The viral load results in the vaccine group were significantly different. No viral loads were detected at any time in four out of six vaccine group animals (Fig. B). The two vaccinees that became infected, had lower peak viral loads than any of the control animals. Animal DG1 had a peak viral load of x copies/ml of plasma, and had an undetectable load by day 1 days following challenge. Animal DF had a peak viral load of.x copies/ml of plasma, and controlled the infection to below the limit of detection days after the challenge. The control and vaccinee peak viral loads (geometric means) at 1 days post-infection were 1x and x copies/ml, respectively and the difference is significant (P<0.001, two-tailed Mann-Whitney test). Effects of SIV challenge on CD T cells in the gut and peripheral blood. Pathogenic infection with SIV causes rapid depletion of the CCR + memory CD + T cells residing in 1

13 the gut lamina propria without causing substantial depletion of the CD + peripheral blood T cells that are largely naïve (, ). To examine the effects of the SIV challenge on these cells in the vaccine and control groups, we analyzed the percentage of total T cells in the gut that were CD + at day of challenge (day 0), and and 0 days following challenge (Fig. A, B). CD + T cell counts were also measured in the peripheral blood (expressed as percent pre-challenge CD + cell counts) at these same time points and at later times throughout the study (Fig. C, D). Five of the six control animals showed major depletion of the gut CD + T cells by day following challenge while the CD + T cells counts in the blood showed at most a 0% decrease. The animals showing the highest peak loads (indicated by asterisks) also showed the largest decreases in gut CD + T cells. Large decreases in the blood CD + T cell counts were seen in three control animals at later times as they developed AIDS. In contrast, the vaccine group showed good preservation of the gut CD + T cell population as would be expected from the apparent lack of infection in four animals and the greatly reduced loads in the two animals that became infected. DF, the animal with the highest peak viral load did show an early decrease in gut CD + T cells followed by a recovery. Peripheral blood CD + T cell counts in the vaccine group animals showed no significant decrease over time. Antibodies to SIV Envs. The SIVsmE0 quasispecies is relatively resistant to nab although antibodies capable of neutralizing it do develop following infection of macaques. These antibodies are, however, insufficient to control the virus (, ). To elucidate the immune correlates of protection in the vaccine group animals, we first tested for the 1

14 presence of nab to the SIVsmE0 quasispecies following priming, boosting, the day of challenge, and up to 0 days post-challenge. No nab to the quasispecies was detectable even at serum dilutions of 1:0 in CEMx1 cells. Because results of a nab assay using a complex quasispecies is difficult to interpret, we next measured nabs using two different Env pseudotypes derived from cloned Env sequences of SIVsmE0. No nabs to tier Envs (CR-PK-A and BR-CGV.IR) were detected even at serum dilutions of 1:0. However, we did detect very high levels of nab to the tier 1A E0. envelope (Fig. A). All vaccine group animals showed nab to E0. after the prime, and a dramatic increase in neutralizing antibody (nab) titers (approximately 0-fold) was seen after the SFVG replicon boost. The second VSV boost was less effective, but did increase waning Ab titers an average of about -fold. At the time of challenge, all animals had antibodies capable of neutralizing the E0. envelope. We did not detect a correlation between the magnitude of the nab titers to the E0. Env and protection. The two animals that became infected (DF and DG1) had higher pre-challenge nab titers than two of the protected animals (CM1 and FP). Following challenge, nab titers continued to increase in DF and DG1, consistent with an anamnestic immune response following infection. In the four protected animals, nab titers decreased or stayed the same, consistent with these animals being protected from SIV infection. We also measured nabs to a VSV recombinant (VSV G-EGFP) expressing the E0- EnvG protein used in the vaccine. These surrogate viruses lack VSV G protein, but have the EnvG protein incorporated into the VSV envelope, and infect cells via CD and CCR 1

15 1 1 (). In this assay, the nab titers were lower than, but largely paralleled what was seen in the E0. pseudotype assay. Total serum antibody to oligomeric SIV Env gp10 was also measured by ELISA assay at the time of challenge and post-challenge (Fig. S1). The titers decreased following challenge in the four uninfected vaccinees consistent with lack of infection, while anamnestic increases occurred in the two vaccinees that became infected. However, the pre-challenge titers did not correlate with protection since the animal with the highest prechallenge titer (DF) was one of the two vaccinees that became infected. CD + T cell responses did not correlate with protection. MHC class I tetramers were used to quantify the percentage of CD + CD + T cells capable of recognizing the immunodominant gag epitope, pc, from the three MamuA*01 + animals in the vaccine group. Fig. A shows the results of tetramer analysis in these animals at one week after prime (day ), one week after the second boost (day ), and also at days post- 1 challenge (day 1 post-prime). Two of the animals (DG1 and DT0) showed approximately 0.% pc + cells after the second boost, while the third animal, CJ, had a tetramer response that was less than 0.1%. Following challenge, the one MamuA*01 vaccine animal that was infected (DG1), showed a six-fold increase in pc + CD cells consistent with an anamnestic response to infection. By day post-challenge, pc + CD cells were low to undetectable in the uninfected animals and had decreased by six-fold in DG1 consistent with effective control of the infection (data not shown). Therefore, pc tetramer positivity was not predictive of protection since an animal with the lowest 1

16 response (CJ) was protected from infection, while DG1, an animal with a much greater response, was infected. We next measured functional responses of the T cells isolated from these animals to Gag by using an interferon-gamma (IFN-γ) ELISpot assay (Fig. B). Gag-specific responses were minimal in all animals, but were generally consistent with the tetramer results. While an anamnestic response in DG1 was apparent post-challenge, none of the animals made significant pre- or post-challenge responses to SIV Env in ELISpot assays (data not shown). T cell responses in the non-mamua*01 animals were also analyzed using Gag and Env ELISpot assays. The responses to Gag and Env were at the background level in these animals and also did not correlate with protection from infection (data not shown). CD depletion in the vaccine group animals confirms apparent sterilizing immunity. The lack of detectable viral loads and absence of anamnestic antibody and T cell responses in the four protected animals in the vaccine group was consistent with complete protection, but did not rule out a very low level of infection. To determine if the four protected animals in the vaccine group had a low level infection that was being controlled by CD T cells, we depleted this population in all six vaccine group animals by four subcutaneous injections of rhesus anti-cd Ab given at three day intervals. Depletion was started 1 days after the SIV challenge. By three days following the first SC injection, all animals showed complete depletion of CD + T cells in the peripheral blood (Fig. A). The two animals that were definitely infected initially, DF and DG1, showed a rapid reappearance of viral RNA in the blood by three and seven days post- 1

17 depletion. In contrast, there was no detectable viral RNA in the four protected animals consistent with lack of any initial infection. The rate of reappearance of viral RNA in DF and DG1 correlated with their initial peak viral loads. Interestingly, while DF was able to control its viral load within a month following the last injection, DG1 was not. The control by DF corresponded to a rapid reappearance of CD + T cells in this animal. CD + T cells in DG1 did not return to normal levels and this animal maintained high levels of viremia. DISCUSSION Virus based vaccine vectors are often strong inducers of immune responses, and 1 can be particularly potent when used in a heterologous prime-boost regimen. The goal of our current study was to test a new vaccine vector prime-boost combination for its ability to induce immune responses and protect macaques against a single, high-dose mucosal challenge with a highly pathogenic SIV quasispecies, SIVsmE0 (1). We chose to challenge with SIVsmE0 because this virus swarm shows a moderate degree of resistance to nab, however some nabs to the swarm typically develop in infected animals within - months post-infection, much like in HIV-1 infection (). In contrast, the more commonly used SIVmac1 quasispecies is extremely resistant to nab (). Therefore, E0 might be a more relevant model for obtaining sterilizing immunity based on induction of nab. As with other pathogenic SIV challenges, high-dose SIVsmE0 challenge has proven difficult to protect against. Several vaccine studies have employed IV challenges 1

18 with SIVsmE0 with only partial success at obtaining control of virus replication (, 1, 1,, ). A vaccine study employing a high-dose rectal challenge with SIVsmE0 was reported by Johnston et al. (00). In that study, they observed better control of peak virus load in vaccinees compared to controls, and one out of six vaccinees appeared not to be infected after challenge (1). A more recent vaccine study employed multiple low-dose rectal challenges with the E0 virus swarm (). A significant reduction in rate of acquisition of E0 infection was seen in animals vaccinated with the heterologous live-attenuated SIVmac nef. In addition, reductions in viral loads were seen in animals that became infected. However, much like the study presented here, the immune correlates of protection were not clear. In the present study we used a mucosal challenge because this mode of transmission accounts for the majority of human HIV infections. Although a multiple lowdose challenge might better mimic typical human HIV transmission, we used a high-dose challenge to ensure infection of all control animals after a single challenge. Protection from high-dose challenge could also provide stronger evidence of vaccine effectiveness because multiple distinct viral genotypes should be transmitted simultaneously (). The level of protection we observed was striking with all six controls infected, and only two of six vaccinees infected. The protection from infection was statistically significant (p=0.0, Fisher s exact test) and was verified by CD + cell depletion, which showed no virus rebound in the four completely protected animals. Virus rebound in the two initially infected animals indicated an important role for CD + cells in control of replication. The ability of CD + T cells to control SIV infection is well established (0,, 0). The results 1

19 also show that any antibodies that may have been present at the time of depletion were insufficient to prevent a rapid rebound of virus replication. The immune correlates of apparently sterilizing protection in our study are not yet clear. Cellular immune responses to both Gag and Env antigens in the vaccine were modest prior to challenge and their magnitudes did not correlate with protection. It is, of course, possible that cellular immune responses at the site of infection were generated and were sufficient to clear brief transient infection that went undetected (). Such a mechanism has been reported for a rhesus cytomegalovirus-vectored SIV vaccine inducing strong effector-memory T cells (1). There was clear evidence of anamnestic antibody and cellular immune responses in the vaccinees that became infected in that study. However, the lack of any anamnestic T cell or antibody responses in our protected animals argues against infection in these animals. Determining the possible contribution of virus neutralizing antibody to protection is especially complicated when multiple genotypes are present in the quasispecies challenge. After the priming vaccination, all vaccinees made detectable serum nabs to a specific Env clone (E0.) derived from the quasispecies and to the cloned E0 EnvG envelope used in the vaccine. There was a striking increase in these nab titers (0-fold) after the SFVG vector boost with titers exceeding in one animal. However, we did not detect nabs to the E0 quasispecies itself or to other E0 Env clones (CR-PK-A and BR-CGV.IR). Despite the absence of nab to the quasispecies in the in vitro assay, it is still possible that the majority of the viruses infecting in vivo are nab sensitive and that 1

20 mucosal nab responses could explain the protection. Recent studies indicate that even relatively low levels of mucosal nabs can protect against mucosal SHIV challenge (1). Detection of nabs in fecal samples or rectal washes is difficult and has not yet given us consistent results even in SIVsmE0 infected animals with high serum nab titers. It is also possible that non-neutralizing mucosal antibodies played a role in protection through clearance of immune complexes or antibody-dependent cell-mediated cytotoxicity. We analyzed the E0 challenge stock by single genome amplification (SGA) and sequencing of sixty env genes (1, ). The results showed a mixture of sequences (Fig. S A, B). The complexity was high with a maximum difference of 1.% between individual sequences, similar to the result of 1.% obtained by Keele et al, 00 for a related SIVsmE0 stock (). The portion of the env gene used in the vaccine (lacking the Env cytoplasmic tail coding sequence) was an outlier sequence (Fig. S C) and differed from the consensus sequence of the quasispecies at 1 amino acid positions out of (%). In the future it should be possible using SGA to determine the sequences the founder virus env genes from the control and vaccinated animals. Analysis of the nab sensitivities of these viruses could provide support for a role of nab in protection if the majority of control animals were infected with founder viruses sensitive to nab induced in the protected vaccinees. A recent study assessed the effect of TRIMα on viral load in rhesus macaques infected with SIVsmE- (), a clone of a virus related to the SIVsmE0 quasi-species. That study showed a trend toward better control of SIVsmE- replication in animals homozygous for the TRIM TFP/TFP allele compared to animals heterozygous for the 0

21 TRIM TFP/Q allele. Although it is not known if the SIVsmE0 quasispecies are subject to TRIMα restriction, we retrospectively determined the TRIMα status of the animals in this study. The results are shown in Table S1. All animals were either heterozygous TFP/ Q, or homozygous TFP/TFP at the TRIMα locus. The only TRIM TFP/TFP animal in the control group (FT0) had a low viral setpoint at day 1, but this setpoint was no lower than other control animals that were TRIM TFP/Q (DD0 and EP). Three control animals developed high viral loads by day 1 and were also predicted to be moderate controllers (TFP/Q). Therefore it is not clear from our limited data if SIVsmE0 replication is affected by TRIMα. A larger study of more than one hundred control animals (including the animals from this study) infected with SIVsmE0 has not revealed statistically significant effects of TRIMα alleles on SIVsmE0 replication (Welkin Johnson, unpublished results). In the vaccine group there were three TFP/TFP animals and all of these as well as one TFP/Q animal were protected from infection. The two vaccine group animals that became infected were TFP/Q and exhibited rapid control of viral loads. Although vaccinated animals were protected from infection independent of their TRIMα allele status, it remains possible that the TRIMα status could limit initial virus replication and thereby enhance vaccine efficacy. This issue could be avoided in future studies by eliminating potentially protective TRIMα alleles in control and vaccine groups. Recent results from the Thai vaccine trial suggest that a modest level of protection from HIV infection is possible (1). The consistent immune response seen in vaccinees was induction of anti-hiv Env antibody suggesting that antibody may have played a role 1

22 in protection. Given the marginal effects of that vaccine, it would be useful to have an animal model where consistent protection from SIV infection can be achieved. The experimental vaccine that we describe here combining VSV and SFVG replicon particles generates robust nab responses and provides a high level of protection. It could prove useful for understanding mechanisms of sterilizing protection from immunodeficiency virus challenge as well as control of virus replication following infection. This vector system should also be considered a strong candidate for future SIV and HIV vaccine studies attempting to elicit broadly neutralizing antibodies with novel antigens ACKNOWLEDGEMENTS We thank Dr. Philip Johnson for a generous supply of the SIVsmE0 challenge stock and for helpful suggestions. We thank Drs. Gunilla Karlsson and Peter Lilejstrom for providing the pbk-t-sfv vector and Dr. Robert Doms for providing vaccinia virus recombinants expressing SIV Env proteins. This work was supported by NIH grants AI and AI-0, the Tulane National Primate Research Center base grant RR0001, and NIAID contract AI.

23 Barefoot, B., N. J. Thornburg, D. H. Barouch, J. S. Yu, C. Sample, R. E. Johnston, H. X. Liao, T. B. Kepler, B. F. Haynes, and E. Ramsburg. 00. Comparison of multiple vaccine vectors in a single heterologous prime-boost trial. Vaccine :-1.. Barouch, D. H., J. Kunstman, M. J. Kuroda, J. E. Schmitz, S. Santra, F. W. Peyerl, G. R. Krivulka, K. Beaudry, M. A. Lifton, D. A. Gorgone, D. C. Montefiori, M. G. Lewis, S. M. Wolinsky, and N. L. Letvin. 00. Eventual AIDS vaccine failure in a rhesus monkey by viral escape from cytotoxic T lymphocytes. Nature 1:-.. Boritz, E., J. Gerlach, J. E. Johnson, and J. K. Rose. 1. Replication-competent rhabdoviruses with human immunodeficiency virus type 1 coats and green fluorescent protein: entry by a ph-independent pathway. J Virol :-.. 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, and M. N. Robertson. 00. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet :-.. Casimiro, D. R., F. Wang, W. A. Schleif, X. Liang, Z. Q. Zhang, T. W. Tobery, M. E. Davies, A. B. McDermott, D. H. O'Connor, A. Fridman, A. Bagchi, L. G. Tussey, A. J. Bett, A. C. Finnefrock, T. M. Fu, A. Tang, K. A. Wilson, M. Chen, H. C. Perry, G. J. Heidecker, D. C. Freed, A. Carella, K. S. Punt, K. J. Sykes, L. Huang, V. I. Ausensi, M. Bachinsky, U. Sadasivan-Nair, D. I. Watkins, E. A. Emini, and J. W. Shiver. 00. Attenuation of simian immunodeficiency virus SIVmac infection by prophylactic immunization with dna and recombinant adenoviral vaccine vectors expressing Gag. J Virol :1-.. Cooper, D., K. J. Wright, P. C. Calderon, M. Guo, F. Nasar, J. E. Johnson, J. W. Coleman, M. Lee, C. Kotash, I. Yurgelonis, R. J. Natuk, R. M. Hendry, S. A. Udem, and D. K. Clarke. 00. Attenuation of recombinant vesicular stomatitis virus-human immunodeficiency virus type 1 vaccine vectors by gene translocations and g gene truncation reduces neurovirulence and enhances immunogenicity in mice. J Virol :0-1.. Davis, N. L., I. J. Caley, K. W. Brown, M. R. Betts, D. M. Irlbeck, K. M. McGrath, M. J. Connell, D. C. Montefiori, J. A. Frelinger, R. Swanstrom, P. R. Johnson, and R. E. Johnston Vaccination of macaques against pathogenic simian immunodeficiency virus with Venezuelan equine encephalitis virus replicon particles. J Virol :1-.. Davis, N. L., A. West, E. Reap, G. MacDonald, M. Collier, S. Dryga, M. Maughan, M. Connell, C. Walker, K. McGrath, C. Cecil, L. H. Ping, J. Frelinger, R. Olmsted, P. Keith, R. Swanstrom, C. Williamson, P. Johnson, D. Montefiori, and R. E. Johnston. 00. Alphavirus replicon particles as candidate HIV vaccines. IUBMB Life :0-.. Deng, H., R. Liu, W. Ellmeier, S. Choe, D. Unutmaz, M. Burkhart, P. Di Marzio, S. Marmon, R. E. Sutton, C. M. Hill, C. B. Davis, S. C. Peiper, T. J. Schall, D. R. Littman, and N. R. Landau. 1. Identification of a major co-receptor for primary isolates of HIV- 1. Nature 1:1-.. Flynn, N. M., D. N. Forthal, C. D. Harro, F. N. Judson, K. H. Mayer, and M. F. Para. 00. Placebo-controlled phase trial of a recombinant glycoprotein 10 vaccine to prevent HIV-1 infection. J Infect Dis 11:-.

24 Haglund, K., I. Leiner, K. Kerksiek, L. Buonocore, E. Pamer, and J. K. Rose. 00. Robust recall and long-term memory T-cell responses induced by prime-boost regimens with heterologous live viral vectors expressing human immunodeficiency virus type 1 Gag and Env proteins. J Virol : Hansen, S. G., C. Vieville, N. Whizin, L. Coyne-Johnson, D. C. Siess, D. D. Drummond, A. W. Legasse, M. K. Axthelm, K. Oswald, C. M. Trubey, M. Piatak, Jr., J. D. Lifson, J. A. Nelson, M. A. Jarvis, and L. J. Picker. 00. Effector memory T cell responses are associated with protection of rhesus monkeys from mucosal simian immunodeficiency virus challenge. Nat Med 1:-. 1. Hessell, A. J., P. Poignard, M. Hunter, L. Hangartner, D. M. Tehrani, W. K. Bleeker, P. W. Parren, P. A. Marx, and D. R. Burton. 00. Effective, low-titer antibody protection against low-dose repeated mucosal SHIV challenge in macaques. Nat Med 1: Hirsch, V. M., R. A. Olmsted, M. Murphey-Corb, R. H. Purcell, and P. R. Johnson. 1. An African primate lentivirus (SIVsm) closely related to HIV-. Nature :-. 1. Johnson, J. E., W. Rodgers, and J. K. Rose. 1. A plasma membrane localization signal in the HIV-1 envelope cytoplasmic domain prevents localization at sites of vesicular stomatitis virus budding and incorporation into VSV virions. Virology 1:-. 1. Johnson, J. E., M. J. Schnell, L. Buonocore, and J. K. Rose. 1. Specific targeting to CD+ cells of recombinant vesicular stomatitis viruses encoding human immunodeficiency virus envelope proteins. J Virol 1: Johnson, P. R., B. C. Schnepp, M. J. Connell, D. Rohne, S. Robinson, G. R. Krivulka, C. I. Lord, R. Zinn, D. C. Montefiori, N. L. Letvin, and K. R. Clark. 00. Novel adenoassociated virus vector vaccine restricts replication of simian immunodeficiency virus in macaques. J Virol :-. 1. Johnston, M. I., and A. S. Fauci. 00. An HIV vaccine--challenges and prospects. N Engl J Med : Johnston, R. E., P. R. Johnson, M. J. Connell, D. C. Montefiori, A. West, M. L. Collier, C. Cecil, R. Swanstrom, J. A. Frelinger, and N. L. Davis. 00. Vaccination of macaques with SIV immunogens delivered by Venezuelan equine encephalitis virus replicon particle vectors followed by a mucosal challenge with SIVsmE0. Vaccine :-. 0. Karlsson, G. B., and P. Liljestrom. 00. Delivery and expression of heterologous genes in mammalian cells using self-replicating alphavirus vectors. Methods Mol Biol :-. 1. Keele, B. F., E. E. Giorgi, J. F. Salazar-Gonzalez, J. M. Decker, K. T. Pham, M. G. Salazar, C. Sun, T. Grayson, S. Wang, H. Li, X. Wei, C. Jiang, J. L. Kirchherr, F. Gao, J. A. Anderson, L. H. Ping, R. Swanstrom, G. D. Tomaras, W. A. Blattner, P. A. Goepfert, J. M. Kilby, M. S. Saag, E. L. Delwart, M. P. Busch, M. S. Cohen, D. C. Montefiori, B. F. Haynes, B. Gaschen, G. S. Athreya, H. Y. Lee, N. Wood, C. Seoighe, A. S. Perelson, T. Bhattacharya, B. T. Korber, B. H. Hahn, and G. M. Shaw. 00. Identification and characterization of transmitted and early founder virus envelopes in primary HIV-1 infection. Proc Natl Acad Sci U S A :-.. Keele, B. F., H. Li, G. H. Learn, P. Hraber, E. E. Giorgi, T. Grayson, C. Sun, Y. Chen, W. W. Yeh, N. L. Letvin, J. R. Mascola, G. J. Nabel, B. F. Haynes, T. Bhattacharya, A. S. Perelson, B. T. Korber, B. H. Hahn, and G. M. Shaw. 00. Low-dose rectal inoculation of rhesus macaques by SIVsmE0 or SIVmac1 recapitulates human mucosal infection by HIV-1. J Exp Med 0:1-.

25 Kim, E. Y., R. S. Veazey, R. Zahn, K. J. McEvers, S. H. Baumeister, G. J. Foster, M. D. Rett, M. H. Newberg, M. J. Kuroda, E. P. Rieber, M. Piatak, Jr., J. D. Lifson, N. L. Letvin, S. M. Wolinsky, and J. E. Schmitz. 00. Contribution of CD+ T cells to containment of viral replication and emergence of mutations in Mamu-A*01-restricted epitopes in Simian immunodeficiency virus-infected rhesus monkeys. J Virol :1-.. Kirmaier, A., F. Wu, R. M. Newman, L. R. Hall, J. S. Morgan, S. O'Connor, P. A. Marx, M. Meythaler, S. Goldstein, A. Buckler-White, A. Kaur, V. M. Hirsch, and W. E. Johnson. 0. TRIM suppresses cross-species transmission of a primate immunodeficiency virus and selects for emergence of resistant variants in the new species. PLoS Biol.. Lawson, N. D., E. A. Stillman, M. A. Whitt, and J. K. Rose. 1. Recombinant vesicular stomatitis viruses from DNA. Proc Natl Acad Sci U S A :-1.. 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. 00. Preserved CD+ central memory T cells and survival in vaccinated SIV-challenged monkeys. Science 1:10-.. Li, M., F. Gao, J. R. Mascola, L. Stamatatos, V. R. Polonis, M. Koutsoukos, G. Voss, P. Goepfert, P. Gilbert, K. M. Greene, M. Bilska, D. L. Kothe, J. F. Salazar-Gonzalez, X. Wei, J. M. Decker, B. H. Hahn, and D. C. Montefiori. 00. Human immunodeficiency virus type 1 env clones from acute and early subtype B infections for standardized assessments of vaccine-elicited neutralizing antibodies. J Virol :-.. 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. 00. Peak SIV replication in resting memory CD+ T cells depletes gut lamina propria CD+ T cells. Nature :-.. Li, Q., P. J. Skinner, S. J. Ha, L. Duan, T. L. Mattila, A. Hage, C. White, D. L. Barber, L. O'Mara, P. J. Southern, C. S. Reilly, J. V. Carlis, C. J. Miller, R. Ahmed, and A. T. Haase. 00. Visualizing antigen-specific and infected cells in situ predicts outcomes in early viral infection. Science : Lifson, J. D., J. L. Rossio, M. Piatak, Jr., T. Parks, L. Li, R. Kiser, V. Coalter, B. Fisher, B. M. Flynn, S. Czajak, V. M. Hirsch, K. A. Reimann, J. E. Schmitz, J. Ghrayeb, N. Bischofberger, M. A. Nowak, R. C. Desrosiers, and D. Wodarz Role of CD(+) lymphocytes in control of simian immunodeficiency virus infection and resistance to rechallenge after transient early antiretroviral treatment. J Virol : Liu, J., K. L. O'Brien, D. M. Lynch, N. L. Simmons, A. La Porte, A. M. Riggs, P. Abbink, R. T. Coffey, L. E. Grandpre, M. S. Seaman, G. Landucci, D. N. Forthal, D. C. Montefiori, A. Carville, K. G. Mansfield, M. J. Havenga, M. G. Pau, J. Goudsmit, and D. H. Barouch. 00. Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys. Nature :-1.. Metzner, K. J., X. Jin, F. V. Lee, A. Gettie, D. E. Bauer, M. Di Mascio, A. S. Perelson, P. A. Marx, D. D. Ho, L. G. Kostrikis, and R. I. Connor Effects of in vivo CD(+) T cell depletion on virus replication in rhesus macaques immunized with a live, attenuated simian immunodeficiency virus vaccine. J Exp Med 11: Montefiori, D. C. 00. Evaluating neutralizing antibodies against HIV, SIV, and SHIV in luciferase reporter gene assays. Curr Protoc Immunol Chapter 1:Unit 1.. Nilsson, C., B. Makitalo, P. Berglund, F. Bex, P. Liljestrom, G. Sutter, V. Erfle, P. ten Haaft, J. Heeney, G. Biberfeld, and R. Thorstensson Enhanced simian

26 immunodeficiency virus-specific immune responses in macaques induced by priming with recombinant Semliki Forest virus and boosting with modified vaccinia virus Ankara. Vaccine 1:-.. O'Connor, D. H., B. R. Mothe, J. T. Weinfurter, S. Fuenger, W. M. Rehrauer, P. Jing, R. R. Rudersdorf, M. E. Liebl, K. Krebs, J. Vasquez, E. Dodds, J. Loffredo, S. Martin, A. B. McDermott, T. M. Allen, C. Wang, G. G. Doxiadis, D. C. Montefiori, A. Hughes, D. R. Burton, D. B. Allison, S. M. Wolinsky, R. Bontrop, L. J. Picker, and D. I. Watkins. 00. Major histocompatibility complex class I alleles associated with slow simian immunodeficiency virus disease progression bind epitopes recognized by dominant acute-phase cytotoxic-t-lymphocyte responses. J Virol :0-0.. Ourmanov, I., M. Bilska, V. M. Hirsch, and D. C. Montefiori Recombinant modified vaccinia virus ankara expressing the surface gp10 of simian immunodeficiency virus (SIV) primes for a rapid neutralizing antibody response to SIV infection in macaques. J Virol :0-.. Ourmanov, I., T. Kuwata, R. Goeken, S. Goldstein, R. Iyengar, A. Buckler-White, B. Lafont, and V. M. Hirsch. 00. Improved survival in rhesus macaques immunized with modified vaccinia virus Ankara recombinants expressing simian immunodeficiency virus envelope correlates with reduction in memory CD+ T-cell loss and higher titers of neutralizing antibody. J Virol :-00.. Pitisuttithum, P., P. Gilbert, M. Gurwith, W. Heyward, M. Martin, F. van Griensven, D. Hu, J. W. Tappero, and K. Choopanya. 00. Randomized, double-blind, placebocontrolled efficacy trial of a bivalent recombinant glycoprotein 10 HIV-1 vaccine among injection drug users in Bangkok, Thailand. J Infect Dis 1:-1.. Priddy, F. H., D. Brown, J. Kublin, K. Monahan, D. P. Wright, J. Lalezari, S. Santiago, M. Marmor, M. Lally, R. M. Novak, S. J. Brown, P. Kulkarni, S. A. Dubey, L. S. Kierstead, D. R. Casimiro, R. Mogg, M. J. DiNubile, J. W. Shiver, R. Y. Leavitt, M. N. Robertson, D. V. Mehrotra, and E. Quirk. 00. Safety and immunogenicity of a replication-incompetent adenovirus type HIV-1 clade B gag/pol/nef vaccine in healthy adults. Clin Infect Dis : Ramsburg, E., N. F. Rose, P. A. Marx, M. Mefford, D. F. Nixon, W. J. Moretto, D. Montefiori, P. Earl, B. Moss, and J. K. Rose. 00. Highly effective control of an AIDS virus challenge in macaques by using vesicular stomatitis virus and modified vaccinia virus Ankara vaccine vectors in a single-boost protocol. J Virol : Rerks-Ngarm, S., P. Pitisuttithum, S. Nitayaphan, J. Kaewkungwal, J. Chiu, R. Paris, N. Premsri, C. Namwat, M. de Souza, E. Adams, M. Benenson, S. Gurunathan, J. Tartaglia, J. G. McNeil, D. P. Francis, D. Stablein, D. L. Birx, S. Chunsuttiwat, C. Khamboonruang, P. Thongcharoen, M. L. Robb, N. L. Michael, P. Kunasol, and J. H. Kim. 00. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med 1:0-0.. Reynolds, M. R., A. M. Weiler, S. M. Piaskowski, H. L. Kolar, A. J. Hessell, M. Weiker, K. L. Weisgrau, E. J. Leon, W. E. Rogers, R. Makowsky, A. B. McDermott, R. Boyle, N. A. Wilson, D. B. Allison, D. R. Burton, W. C. Koff, and D. I. Watkins. 0. Macaques vaccinated with simian immunodeficiency virus SIVmacDelta nef delay acquisition and control replication after repeated low-dose heterologous SIV challenge. J Virol :10-.

Significant Protection against High-Dose Simian Immunodeficiency Virus Challenge Conferred by a New Prime-Boost Vaccine Regimen

Significant Protection against High-Dose Simian Immunodeficiency Virus Challenge Conferred by a New Prime-Boost Vaccine Regimen JOURNAL OF VIROLOGY, June 2011, p. 5764 5772 Vol. 85, No. 12 0022-538X/11/$12.00 doi:10.1128/jvi.00342-11 Copyright 2011, American Society for Microbiology. All Rights Reserved. Significant Protection

More information

NIH Public Access Author Manuscript Nature. Author manuscript; available in PMC 2009 July 1.

NIH Public Access Author Manuscript Nature. Author manuscript; available in PMC 2009 July 1. NIH Public Access Author Manuscript Published in final edited form as: Nature. 2009 January 1; 457(7225): 87 91. doi:10.1038/nature07469. Immune Control of an SIV Challenge by a T Cell-Based Vaccine in

More information

Low-Dose Mucosal Simian Immunodeficiency Virus Infection Restricts Early Replication Kinetics and Transmitted Virus Variants in Rhesus Monkeys

Low-Dose Mucosal Simian Immunodeficiency Virus Infection Restricts Early Replication Kinetics and Transmitted Virus Variants in Rhesus Monkeys JOURNAL OF VIROLOGY, Oct. 2010, p. 10406 10412 Vol. 84, No. 19 0022-538X/10/$12.00 doi:10.1128/jvi.01155-10 Copyright 2010, American Society for Microbiology. All Rights Reserved. Low-Dose Mucosal Simian

More information

Immunization with Single-Cycle SIV Significantly Reduces Viral Loads After an Intravenous Challenge with SIV mac 239

Immunization with Single-Cycle SIV Significantly Reduces Viral Loads After an Intravenous Challenge with SIV mac 239 Immunization with Single-Cycle SIV Significantly Reduces Viral Loads After an Intravenous Challenge with SIV mac 239 Bin Jia 1, Sharon K. Ng 1, M. Quinn DeGottardi 1, Michael Piatak Jr. 2, Eloísa Yuste

More information

Immunization with single-cycle SIV significantly reduces viral loads after an intravenous challenge with SIV(mac)239

Immunization with single-cycle SIV significantly reduces viral loads after an intravenous challenge with SIV(mac)239 University of Massachusetts Medical School escholarship@umms Preventive and Behavioral Medicine Publications and Presentations Preventive and Behavioral Medicine 1-23-2009 Immunization with single-cycle

More information

Should There be Further Efficacy Testing of T-T cell Based Vaccines that do not Induce Broadly Neutralizing Antibodies?

Should There be Further Efficacy Testing of T-T cell Based Vaccines that do not Induce Broadly Neutralizing Antibodies? Dale and Betty Bumpers Vaccine Research Center National Institute of Allergy and Infectious Diseases National Institutes of Health Department of Health and Human Services Should There be Further Efficacy

More information

Received 26 September 2003/Accepted 22 December 2003

Received 26 September 2003/Accepted 22 December 2003 JOURNAL OF VIROLOGY, Apr. 2004, p. 3930 3940 Vol. 78, No. 8 0022-538X/04/$08.00 0 DOI: 10.1128/JVI.78.8.3930 3940.2004 Copyright 2004, American Society for Microbiology. All Rights Reserved. Highly Effective

More information

Recombinant Baculovirus Derived HIV-1 Virus-Like Particles Elicit Potent Neutralizing Antibody Responses

Recombinant Baculovirus Derived HIV-1 Virus-Like Particles Elicit Potent Neutralizing Antibody Responses Recombinant Baculovirus Derived HIV-1 Virus-Like Particles Elicit Potent Neutralizing Antibody Responses Weimin Liu University of Alabama at Birmingham Introduction and Rationale Virus-like particles (VLPs)

More information

Understanding HIV. Transmitted/Founder Viruses. Brandon Keele SAIC-Frederick National Cancer Institute

Understanding HIV. Transmitted/Founder Viruses. Brandon Keele SAIC-Frederick National Cancer Institute Understanding HIV Transmission Utilizing Transmitted/Founder Viruses Brandon Keele SAIC-Frederick National Cancer Institute AIDS Vaccine 2011 15 September 2011 Overview Several years ago, the CHAVI sought

More information

GOVX-B11: A Clade B HIV Vaccine for the Developed World

GOVX-B11: A Clade B HIV Vaccine for the Developed World GeoVax Labs, Inc. 19 Lake Park Drive Suite 3 Atlanta, GA 3 (678) 384-72 GOVX-B11: A Clade B HIV Vaccine for the Developed World Executive summary: GOVX-B11 is a Clade B HIV vaccine targeted for use in

More information

Is an HIV Vaccine Possible?

Is an HIV Vaccine Possible? 304 BJID 2009; 13 (August) Is an HIV Vaccine Possible? Nancy A. Wilson 1,2 and David I. Watkins 1,2 1 Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison; 2 Wisconsin National

More information

DNA and Protein Vaccination Confers Protection Upon Mucosal Challenge with Heterologous SIVsmE660 (OA 10.04)

DNA and Protein Vaccination Confers Protection Upon Mucosal Challenge with Heterologous SIVsmE660 (OA 10.04) DNA and Protein Vaccination Confers Protection Upon Mucosal Challenge with Heterologous SIVsmE660 (OA 10.04) Rashmi Jalah Human Retrovirus Pathogenesis Section, Vaccine Branch, CCR, National Cancer Institute

More information

HIV and Challenges of Vaccine Development

HIV and Challenges of Vaccine Development Dale and Betty Bumpers Vaccine Research Center National Institute of Allergy and Infectious Diseases National Institutes of Health HIV and Challenges of Vaccine Development Richard A. Koup, MD INTEREST

More information

Received 22 April 2009/Accepted 19 June 2009

Received 22 April 2009/Accepted 19 June 2009 JOURNAL OF VIROLOGY, Sept. 2009, p. 9584 9590 Vol. 83, No. 18 0022-538X/09/$08.00 0 doi:10.1128/jvi.00821-09 Copyright 2009, American Society for Microbiology. All Rights Reserved. Protective Efficacy

More information

Novel Heterologous Prime-Boost Vaccine Strategies for HIV. Dan Barouch April 18, 2012

Novel Heterologous Prime-Boost Vaccine Strategies for HIV. Dan Barouch April 18, 2012 Novel Heterologous Prime-Boost Vaccine Strategies for HIV Dan Barouch April 18, 2012 Desired Features of a Next Generation HIV-1 Vaccine Candidate The RV1 study suggests that an HIV-1 vaccine is possible

More information

Current Strategies in HIV-1 Vaccine Development Using Replication-Defective Adenovirus as a Case Study

Current Strategies in HIV-1 Vaccine Development Using Replication-Defective Adenovirus as a Case Study Note: I have added some clarifying comments to the slides -- please click on Comments under View to see them. Current Strategies in HIV-1 Vaccine Development Using Replication-Defective Adenovirus as a

More information

Why are validated immunogenicity assays important for HIV vaccine development?

Why are validated immunogenicity assays important for HIV vaccine development? Why are validated immunogenicity assays important for HIV vaccine development? There is a need to compare immunogenicity of products in the pipeline, when similar or different in class when developed by

More information

Are we targeting the right HIV determinants?

Are we targeting the right HIV determinants? QuickTime et un décompresseur TIFF (non compressé) sont requis pour visionner cette image. AIDS Vaccine 2009 October 22 nd 2009 - Paris Are we targeting the right HIV determinants? Françoise BARRÉ-SINOUSSI

More information

Strategies for an HIV vaccine

Strategies for an HIV vaccine Strategies for an HIV vaccine Norman L. Letvin J Clin Invest. 2002;110(1):15-27. https://doi.org/10.1172/jci15985. Perspective The development of an HIV vaccine poses an unprecedented challenge to the

More information

Supporting Information

Supporting Information Supporting Information Sui et al..7/pnas.997 Pre-CLP CM9 LA9 SL Tat# Pol Vif % Tetramer + CD + CD + Vac+IL- +IL- Vac Fig. S. Frequencies of six different CD + CD + Mamu-A*-tetramer + cells were measured

More information

NIH Public Access Author Manuscript Nat Med. Author manuscript; available in PMC 2010 September 1.

NIH Public Access Author Manuscript Nat Med. Author manuscript; available in PMC 2010 September 1. NIH Public Access Author Manuscript Published in final edited form as: Nat Med. 2010 March ; 16(3): 319 323. doi:10.1038/nm.2089. Mosaic HIV-1 Vaccines Expand the Breadth and Depth of Cellular Immune Responses

More information

Supplementary Figure 1. ALVAC-protein vaccines and macaque immunization. (A) Maximum likelihood

Supplementary Figure 1. ALVAC-protein vaccines and macaque immunization. (A) Maximum likelihood Supplementary Figure 1. ALVAC-protein vaccines and macaque immunization. (A) Maximum likelihood tree illustrating CRF01_AE gp120 protein sequence relationships between 107 Envs sampled in the RV144 trial

More information

Immunogenicity of ALVAC HIV (vcp1521) and AIDSVAX B/E Prime Boost Vaccination in RV144, Thai Phase III HIV Vaccine Trial

Immunogenicity of ALVAC HIV (vcp1521) and AIDSVAX B/E Prime Boost Vaccination in RV144, Thai Phase III HIV Vaccine Trial Immunogenicity of ALVAC HIV (vcp1521) and AIDSVAX B/E Prime Boost Vaccination in RV144, Thai Phase III HIV Vaccine Trial M. de Souza, R. Trichavaroj, A. Schuetz, W. Chuenarom, Y. Phuang ngern, S. Jongrakthaitae,

More information

Department of Surgery, Duke University, Durham, North Carolina; and 7 Southern Research Institute, Frederick, Maryland

Department of Surgery, Duke University, Durham, North Carolina; and 7 Southern Research Institute, Frederick, Maryland MAJOR ARTICLE Highly Attenuated Rabies Virus Based Vaccine Vectors Expressing Simian-Human Immunodeficiency Virus 89.6P Env and Simian Immunodeficiency Virus mac239 Gag Are Safe in Rhesus Macaques and

More information

ALVAC -HIV and AIDSVAX B/E Prime-Boost HIV-1 Preventive Vaccine Regimen. Results of the Thai HIV Vaccine Trial, RV144

ALVAC -HIV and AIDSVAX B/E Prime-Boost HIV-1 Preventive Vaccine Regimen. Results of the Thai HIV Vaccine Trial, RV144 ALVAC -HIV and AIDSVAX B/E Prime-Boost HIV-1 Preventive Vaccine Regimen Results of the Thai HIV Vaccine Trial, RV144 SupachaiRerks-Ngarm, PunneePittisutthithum, SorachaiNitayaphan, JaranitKaewkungwal,

More information

Figure S1. Schematic presentation of genomic replication of idsiv after transfection and infection. After transfection of idsiv plasmid DNA into 293T

Figure S1. Schematic presentation of genomic replication of idsiv after transfection and infection. After transfection of idsiv plasmid DNA into 293T Figure S1. Schematic presentation of genomic replication of idsiv after transfection and infection. After transfection of idsiv plasmid DNA into 293T cells, the RNA genomes with all modifications are generated

More information

DEBATE ON HIV ENVELOPE AS A T CELL IMMUNOGEN HAS BEEN GAG-GED

DEBATE ON HIV ENVELOPE AS A T CELL IMMUNOGEN HAS BEEN GAG-GED DEBATE ON HIV ENVELOPE AS A T CELL IMMUNOGEN HAS BEEN GAG-GED Viv Peut Kent Laboratory, University of Melbourne, Australia WHY ENVELOPE? Env subject to both humoral and cellular immune responses Perhaps

More information

Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques

Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques SUPPORTING INFORMATION FOR: Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques Lia Vassena, 1,2 Huiyi Miao, 1 Raffaello Cimbro,

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION ` SUPPLEMENTAL FIGURES doi:10.1038/nature10003 Supplemental Figure 1: RhCMV/SIV vectors establish and indefinitely maintain high frequency SIV-specific T cell responses in diverse tissues: The figure shows

More information

Immunogens and Antigen Processing: Report from a Global HIV Vaccine Enterprise Working Group

Immunogens and Antigen Processing: Report from a Global HIV Vaccine Enterprise Working Group report Immunogens and Antigen Processing: Report from a Global HIV Vaccine Enterprise Working Group John Mascola, C Richter King & Ralph Steinman on behalf of a Working Group convened by the Global HIV

More information

HIV Anti-HIV Neutralizing Antibodies

HIV Anti-HIV Neutralizing Antibodies ,**/ The Japanese Society for AIDS Research The Journal of AIDS Research : HIV HIV Anti-HIV Neutralizing Antibodies * Junji SHIBATA and Shuzo MATSUSHITA * Division of Clinical Retrovirology and Infectious

More information

Antibody-Dependent Cell-Mediated Cytotoxicity in Simian Immunodeficiency Virus-Infected Rhesus Monkeys

Antibody-Dependent Cell-Mediated Cytotoxicity in Simian Immunodeficiency Virus-Infected Rhesus Monkeys JOURNAL OF VIROLOGY, July 2011, p. 6906 6912 Vol. 85, No. 14 0022-538X/11/$12.00 doi:10.1128/jvi.00326-11 Copyright 2011, American Society for Microbiology. All Rights Reserved. Antibody-Dependent Cell-Mediated

More information

Combinatorial Vaccines for AIDS and other Infectious Diseases

Combinatorial Vaccines for AIDS and other Infectious Diseases Dale and Betty Bumpers Vaccine Research Center National Institute of Allergy and Infectious Diseases National Institutes of Health Department of Health and Human Services Combinatorial Vaccines for AIDS

More information

Antibody Dependent Cellular Cytotxic activity: Past and Future. Guido Ferrari, M.D. Duke University Medical Center

Antibody Dependent Cellular Cytotxic activity: Past and Future. Guido Ferrari, M.D. Duke University Medical Center Antibody Dependent Cellular Cytotxic activity: Past and Future Guido Ferrari, M.D. Duke University Medical Center Mechanism of Antibody Dependent Cellular Cytotoxicity (ADCC) ADCC Effector Cells (NK, monocytes/macrophages,

More information

Replicating measles-shiv vaccine induces long term preservation of central memory CD4 cells in the gut of macaques challenged with SHIV89.

Replicating measles-shiv vaccine induces long term preservation of central memory CD4 cells in the gut of macaques challenged with SHIV89. Replicating measles-shiv vaccine induces long term preservation of central memory CD4 cells in the gut of macaques challenged with SHIV89.6P Frédéric Tangy Viral Genomics and Vaccination Laboratory Measles

More information

Received 29 August 2002/Accepted 3 December 2002

Received 29 August 2002/Accepted 3 December 2002 JOURNAL OF VIROLOGY, Mar. 2003, p. 3099 3118 Vol. 77, No. 5 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.5.3099 3118.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Simian-Human

More information

JVI Accepts, published online ahead of print on 7 March 2007 J. Virol. doi: /jvi

JVI Accepts, published online ahead of print on 7 March 2007 J. Virol. doi: /jvi JVI Accepts, published online ahead of print on 7 March 2007 J. Virol. doi:10.1128/jvi.02763-06 Copyright 2007, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved.

More information

Efficacy of Multivalent Adenovirus-Based Vaccine against Simian Immunodeficiency Virus Challenge

Efficacy of Multivalent Adenovirus-Based Vaccine against Simian Immunodeficiency Virus Challenge JOURNAL OF VIROLOGY, Mar. 2010, p. 2996 3003 Vol. 84, No. 6 0022-538X/10/$12.00 doi:10.1128/jvi.00969-09 Copyright 2010, American Society for Microbiology. All Rights Reserved. Efficacy of Multivalent

More information

Magnitude and Diversity of Cytotoxic-T-Lymphocyte Responses Elicited by Multiepitope DNA Vaccination in Rhesus Monkeys

Magnitude and Diversity of Cytotoxic-T-Lymphocyte Responses Elicited by Multiepitope DNA Vaccination in Rhesus Monkeys JOURNAL OF VIROLOGY, Sept. 2003, p. 10113 10118 Vol. 77, No. 18 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.18.10113 10118.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Magnitude

More information

Development of a Universal T Cell Vaccine. Tomáš Hanke Weatherall Institute of Molecular Medicine University of Oxford United Kingdom

Development of a Universal T Cell Vaccine. Tomáš Hanke Weatherall Institute of Molecular Medicine University of Oxford United Kingdom Development of a Universal T Cell Vaccine Tomáš Hanke Weatherall Institute of Molecular Medicine University of Oxford United Kingdom Development of HIV-1 vaccines Induction of cell-mediated responses Immunogens

More information

It has been 25 years since HIV-1 was identified as the causative

It has been 25 years since HIV-1 was identified as the causative Vol 4j2 October 8jdoi:.38/nature732 Challenges in the development of an HIV-1 vaccine Dan H. Barouch 1 The development of a safe and effective human immunodeficiency virus (HIV)-1 vaccine is a critically

More information

staining and flow cytometry

staining and flow cytometry Detection of influenza virus-specific T cell responses by intracellular by cytokine intracellular staining cytokine staining and flow cytometry Detection of influenza virus-specific T cell responses and

More information

Progress on new vaccine strategies against chronic viral infections

Progress on new vaccine strategies against chronic viral infections Progress on new vaccine strategies against chronic viral infections Jay A. Berzofsky,, Masaki Terabe, Igor M. Belyakov J Clin Invest. 2004;114(4):450-462. https://doi.org/10.1172/jci22674. Review Among

More information

JOURNAL OF VIROLOGY, Oct. 1999, p Vol. 73, No. 10. Copyright 1999, American Society for Microbiology. All Rights Reserved.

JOURNAL OF VIROLOGY, Oct. 1999, p Vol. 73, No. 10. Copyright 1999, American Society for Microbiology. All Rights Reserved. JOURNAL OF VIROLOGY, Oct. 1999, p. 8201 8215 Vol. 73, No. 10 0022-538X/99/$04.00 0 Copyright 1999, American Society for Microbiology. All Rights Reserved. Role of Immune Responses against the Envelope

More information

A global approach to HIV-1 vaccine development

A global approach to HIV-1 vaccine development A global approach to HIV-1 vaccine development The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters. Citation Published Version Accessed

More information

Progress in HIV Vaccine Development Magdalena Sobieszczyk, MD, MPH. Division of Infectious Diseases Columbia University Medical Center

Progress in HIV Vaccine Development Magdalena Sobieszczyk, MD, MPH. Division of Infectious Diseases Columbia University Medical Center Progress in HIV Vaccine Development Magdalena Sobieszczyk, MD, MPH Division of Infectious Diseases Columbia University Medical Center 1 Outline A short history of HIV vaccine design and development Describe

More information

Received 14 April 2005/Accepted 7 August 2005

Received 14 April 2005/Accepted 7 August 2005 JOURNAL OF VIROLOGY, Dec. 2005, p. 15547 15555 Vol. 79, No. 24 0022-538X/05/$08.00 0 doi:10.1128/jvi.79.24.15547 15555.2005 Copyright 2005, American Society for Microbiology. All Rights Reserved. Attenuation

More information

EMERGING ISSUES IN THE HUMORAL IMMUNE RESPONSE TO HIV. (Summary of the recommendations from an Enterprise Working Group)

EMERGING ISSUES IN THE HUMORAL IMMUNE RESPONSE TO HIV. (Summary of the recommendations from an Enterprise Working Group) AIDS Vaccine 07, Seattle, August 20-23, 2007 EMERGING ISSUES IN THE HUMORAL IMMUNE RESPONSE TO HIV (Summary of the recommendations from an Enterprise Working Group) The Working Group Reston, Virginia,

More information

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART BIOE 301 LECTURE 10 MITALI BANERJEE A VACCINE FOR HIV HIV HAART Visit wikipedia.org and learn the mechanism of action of the five classes of antiretroviral drugs. (1) Reverse transcriptase inhibitors (RTIs)

More information

HIV 101: Fundamentals of HIV Infection

HIV 101: Fundamentals of HIV Infection HIV 101: Fundamentals of HIV Infection David H. Spach, MD Professor of Medicine University of Washington Seattle, Washington Learning Objectives After attending this presentation, learners will be able

More information

Received 17 March 2003/Accepted 16 July 2003

Received 17 March 2003/Accepted 16 July 2003 JOURNAL OF VIROLOGY, Nov. 2003, p. 11563 11577 Vol. 77, No. 21 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.21.11563 11577.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Multigene

More information

HIV 1 Preventive Vaccine Regimen. Community based Trial in Thailand V 144. for the MOPH TAVEG Collaboration

HIV 1 Preventive Vaccine Regimen. Community based Trial in Thailand V 144. for the MOPH TAVEG Collaboration V ALVAC HIV and AIDSVAX B/E Prime Boost HIV 1 Preventive Vaccine Regimen Final Results of the Phase III Community based Trial in Thailand Supachai Rerks Ngarm, Punnee Pittisutthithum, Sorachai Nitayaphan,

More information

The humoral immune responses to IBV proteins.

The humoral immune responses to IBV proteins. The humoral immune responses to IBV proteins. E. Dan Heller and Rosa Meir The Hebrew University of Jerusalem, Israel COST FA1207 meeting WG2 + WG3, Budapest, Jan. 2015 1 IBV encodes four major structural

More information

Mosaic vaccines elicit CD8 + T lymphocyte responses that confer enhanced immune coverage of diverse HIV strains in monkeys

Mosaic vaccines elicit CD8 + T lymphocyte responses that confer enhanced immune coverage of diverse HIV strains in monkeys Mosaic vaccines elicit CD + T lymphocyte responses that fer enhanced immune coverage of diverse HIV strains in monkeys 21 Nature America, Inc. All rights reserved. Sampa Santra 1, Hua-Xin Liao 2, Ruijin

More information

Establishment and Targeting of the Viral Reservoir in Rhesus Monkeys

Establishment and Targeting of the Viral Reservoir in Rhesus Monkeys Establishment and Targeting of the Viral Reservoir in Rhesus Monkeys Dan H. Barouch, M.D., Ph.D. Center for Virology and Vaccine Research Beth Israel Deaconess Medical Center Ragon Institute of MGH, MIT,

More information

"Risk behaviour and time as covariates for effi cacy of the HIV vaccine regimen ALVAC-HIV (vcp1521) and AIDSVAX B/E: a posthoc

Risk behaviour and time as covariates for effi cacy of the HIV vaccine regimen ALVAC-HIV (vcp1521) and AIDSVAX B/E: a posthoc University of Nebraska - Lincoln DigitalCommons@University of Nebraska - Lincoln US Army Research U.S. Department of Defense 212 Risk behaviour and time as covariates for effi cacy of the HIV vaccine regimen

More information

HIV cure: current status and implications for the future

HIV cure: current status and implications for the future HIV cure: current status and implications for the future Carolyn Williamson, PhD Head of Medical Virology, Faculty Health Sciences, University of Cape Town CAPRISA Research Associate, Centre of Excellence

More information

Received 19 September 2007/Accepted 21 November 2007

Received 19 September 2007/Accepted 21 November 2007 JOURNAL OF VIROLOGY, Feb. 2008, p. 1723 1738 Vol. 82, No. 4 0022-538X/08/$08.00 0 doi:10.1128/jvi.02084-07 Copyright 2008, American Society for Microbiology. All Rights Reserved. Patterns of CD8 Immunodominance

More information

An Effective AIDS Vaccine Based on Live Attenuated Vesicular Stomatitis Virus Recombinants

An Effective AIDS Vaccine Based on Live Attenuated Vesicular Stomatitis Virus Recombinants Cell, Vol. 106, 539 549, September 7, 2001, Copyright 2001 by Cell Press An Effective AIDS Vaccine Based on Live Attenuated Vesicular Stomatitis Virus Recombinants Nina F. Rose, 1 Preston A. Marx, 2,3

More information

X/01/$ DOI: /JVI Copyright 2001, American Society for Microbiology. All Rights Reserved.

X/01/$ DOI: /JVI Copyright 2001, American Society for Microbiology. All Rights Reserved. JOURNAL OF VIROLOGY, Apr. 2001, p. 3753 3765 Vol. 75, No. 8 0022-538X/01/$04.00 0 DOI: 10.1128/JVI.75.8.3753 3765.2001 Copyright 2001, American Society for Microbiology. All Rights Reserved. Route of Simian

More information

Zheng, BJ; Du, LY; Zhao, GY; Lin, YP; Sui, HY; Chan, C; Ma, S; Guan, Y; Yuen, KY. Citation Hong Kong Medical Journal, 2008, v. 14 suppl. 4, p.

Zheng, BJ; Du, LY; Zhao, GY; Lin, YP; Sui, HY; Chan, C; Ma, S; Guan, Y; Yuen, KY. Citation Hong Kong Medical Journal, 2008, v. 14 suppl. 4, p. Title Studies of SARS virus vaccines Author(s) Zheng, BJ; Du, LY; Zhao, GY; Lin, YP; Sui, HY; Chan, C; Ma, S; Guan, Y; Yuen, KY Citation Hong Kong Medical Journal, 2008, v. 14 suppl. 4, p. 39-43 Issued

More information

A centralized gene-based HIV-1 vaccine elicits broad cross-clade cellular immune responses in rhesus monkeys

A centralized gene-based HIV-1 vaccine elicits broad cross-clade cellular immune responses in rhesus monkeys A centralized gene-based HIV-1 vaccine elicits broad cross-clade cellular immune responses in rhesus monkeys Sampa Santra, Bette T. Korber, Mark Muldoon, Dan H. Barouch, Gary J. Nabel, Feng Gao, Beatrice

More information

NIH Public Access Author Manuscript Nat Rev Microbiol. Author manuscript; available in PMC 2014 November 19.

NIH Public Access Author Manuscript Nat Rev Microbiol. Author manuscript; available in PMC 2014 November 19. NIH Public Access Author Manuscript Published in final edited form as: Nat Rev Microbiol. 2014 November ; 12(11): 765 771. doi:10.1038/nrmicro3360. Novel vaccine vectors for HIV-1 Dan H. Barouch and Center

More information

On an individual level. Time since infection. NEJM, April HIV-1 evolution in response to immune selection pressures

On an individual level. Time since infection. NEJM, April HIV-1 evolution in response to immune selection pressures HIV-1 evolution in response to immune selection pressures BISC 441 guest lecture Zabrina Brumme, Ph.D. Assistant Professor, Faculty of Health Sciences Simon Fraser University http://www3.niaid.nih.gov/topics/hivaids/understanding/biology/structure.htm

More information

ADCC Develops Over Time during Persistent Infection with Live-Attenuated SIV and Is Associated with Complete Protection against SIV(mac)251 Challenge

ADCC Develops Over Time during Persistent Infection with Live-Attenuated SIV and Is Associated with Complete Protection against SIV(mac)251 Challenge University of Massachusetts Medical School escholarship@umms Preventive and Behavioral Medicine Publications and Presentations Preventive and Behavioral Medicine 8-2012 ADCC Develops Over Time during Persistent

More information

HIV/AIDS: vaccines and alternate strategies for treatment and prevention

HIV/AIDS: vaccines and alternate strategies for treatment and prevention Ann. N.Y. Acad. Sci. ISSN 0077-8923 ANNALS OF THE NEW YORK ACADEMY OF SCIENCES Online Meeting Report HIV/AIDS: vaccines and alternate strategies for treatment and prevention Yegor Voronin 1 and Sanjay

More information

HIV-1 Vaccine Development: Recent Advances in the CTL Platform Spotty Business ACCEPTED

HIV-1 Vaccine Development: Recent Advances in the CTL Platform Spotty Business ACCEPTED JVI Accepts, published online ahead of print on 7 November 2007 J. Virol. doi:10.1128/jvi.01634-07 Copyright 2007, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved.

More information

Received 16 February 2004/Accepted 5 June 2004

Received 16 February 2004/Accepted 5 June 2004 JOURNAL OF VIROLOGY, Oct. 2004, p. 11434 11438 Vol. 78, No. 20 0022-538X/04/$08.00 0 DOI: 10.1128/JVI.78.20.11434 11438.2004 Copyright 2004, American Society for Microbiology. All Rights Reserved. Heterologous

More information

Efficacy Results from the Step Study

Efficacy Results from the Step Study Efficacy Results from the Step Study (Merck V520 Protocol 023/HVTN 502) A Phase II Test-of-Concept Trial of the MRKAd5 HIV-1 Gag/Pol/Nef Trivalent Vaccine CROI February 5, 2008 Boston, MA Rationale for

More information

Rapid perforin upregulation directly ex vivo by CD8 + T cells is a defining characteristic of HIV elite controllers

Rapid perforin upregulation directly ex vivo by CD8 + T cells is a defining characteristic of HIV elite controllers Rapid perforin upregulation directly ex vivo by CD8 + T cells is a defining characteristic of HIV elite controllers Adam R. Hersperger Department of Microbiology University of Pennsylvania Evidence for

More information

Supporting Online Material for

Supporting Online Material for www.sciencemag.org/cgi/content/full/1175194/dc1 Supporting Online Material for A Vital Role for Interleukin-21 in the Control of a Chronic Viral Infection John S. Yi, Ming Du, Allan J. Zajac* *To whom

More information

Therapeutic DNA Vaccine Induces Broad T Cell Responses in the Gut and Sustained Protection from Viral Rebound and AIDS in SIV-Infected Rhesus Macaques

Therapeutic DNA Vaccine Induces Broad T Cell Responses in the Gut and Sustained Protection from Viral Rebound and AIDS in SIV-Infected Rhesus Macaques Therapeutic DNA Vaccine Induces Broad T Cell Responses in the Gut and Sustained Protection from Viral Rebound and AIDS in SIV-Infected Rhesus Macaques Deborah Heydenburg Fuller 1,2,3 * a, Premeela Rajakumar

More information

Updated information and services can be found at:

Updated information and services can be found at: REFERENCES CONTENT ALERTS Neutralizing Antibodies Elicited by Immunization of Monkeys with DNA Plasmids and Recombinant Adenoviral Vectors Expressing Human Immunodeficiency Virus Type 1 Proteins John R.

More information

Received 9 August 2004/Accepted 26 October 2004

Received 9 August 2004/Accepted 26 October 2004 JOURNAL OF VIROLOGY, Mar. 2005, p. 3358 3369 Vol. 79, No. 6 0022-538X/05/$08.00 0 doi:10.1128/jvi.79.6.3358 3369.2005 Copyright 2005, American Society for Microbiology. All Rights Reserved. Protection

More information

HIV-1 p24 ELISA Pair Set Cat#: orb54951 (ELISA Manual)

HIV-1 p24 ELISA Pair Set Cat#: orb54951 (ELISA Manual) HIV-1 p24 ELISA Pair Set Cat#: orb54951 (ELISA Manual) BACKGROUND Human Immunodeficiency Virus ( HIV ) can be divided into two major types, HIV type 1 (HIV-1) and HIV type 2 (HIV-2). HIV-1 is related to

More information

Vaccine-Induced T Cells Control Reversion of AIDS Virus Immune Escape Mutants

Vaccine-Induced T Cells Control Reversion of AIDS Virus Immune Escape Mutants JOURNAL OF VIROLOGY, Apr. 2007, p. 4137 4144 Vol. 81, No. 8 0022-538X/07/$08.00 0 doi:10.1128/jvi.02193-06 Copyright 2007, American Society for Microbiology. All Rights Reserved. Vaccine-Induced T Cells

More information

CD40L-Adjuvanted DNA/MVA SIV Vaccine Enhances Protection. Against Neutralization Resistant Mucosal SIV Infection

CD40L-Adjuvanted DNA/MVA SIV Vaccine Enhances Protection. Against Neutralization Resistant Mucosal SIV Infection JVI Accepted Manuscript Posted Online 4 February 2015 J. Virol. doi:10.1128/jvi.03527-14 Copyright 2015, American Society for Microbiology. All Rights Reserved. 1 2 CD40L-Adjuvanted DNA/MVA SIV Vaccine

More information

HIV-1 vaccine-induced immunity in the test-of-concept Step Study: a case cohort analysis

HIV-1 vaccine-induced immunity in the test-of-concept Step Study: a case cohort analysis HIV-1 vaccine-induced immunity in the test-of-concept Step Study: a case cohort analysis M Juliana McElrath, Stephen C De Rosa, Zoe Moodie, Sheri Dubey, Lisa Kierstead, Holly Janes, Olivier D Defawe, Donald

More information

The effect of adenovirus-specific antibodies on adenoviral vector induced, transgene product specific T cell responses

The effect of adenovirus-specific antibodies on adenoviral vector induced, transgene product specific T cell responses Article The effect of adenovirus-specific antibodies on adenoviral vector induced, transgene product specific T cell responses Juliana C. Small,*,,1 Larissa H. Haut,*,1 Ang Bian,* and Hildegund C. J. Ertl*,2

More information

Received 30 December 2006/Accepted 25 February 2007

Received 30 December 2006/Accepted 25 February 2007 JOURNAL OF VIROLOGY, May 2007, p. 5202 5211 Vol. 81, No. 10 0022-538X/07/$08.00 0 doi:10.1128/jvi.02881-06 Copyright 2007, American Society for Microbiology. All Rights Reserved. Long-Term Control of Simian

More information

Modulation of DNA Vaccine-Elicited CD8 T-Lymphocyte Epitope Immunodominance Hierarchies

Modulation of DNA Vaccine-Elicited CD8 T-Lymphocyte Epitope Immunodominance Hierarchies JOURNAL OF VIROLOGY, Dec. 2006, p. 11991 11997 Vol. 80, No. 24 0022-538X/06/$08.00 0 doi:10.1128/jvi.01348-06 Copyright 2006, American Society for Microbiology. All Rights Reserved. Modulation of DNA Vaccine-Elicited

More information

Supplementary information. MARCH8 inhibits HIV-1 infection by reducing virion incorporation of envelope glycoproteins

Supplementary information. MARCH8 inhibits HIV-1 infection by reducing virion incorporation of envelope glycoproteins Supplementary information inhibits HIV-1 infection by reducing virion incorporation of envelope glycoproteins Takuya Tada, Yanzhao Zhang, Takayoshi Koyama, Minoru Tobiume, Yasuko Tsunetsugu-Yokota, Shoji

More information

Heterologous Envelope Immunogens Contribute to AIDS Vaccine Protection in Rhesus Monkeys

Heterologous Envelope Immunogens Contribute to AIDS Vaccine Protection in Rhesus Monkeys JOURNAL OF VIROLOGY, July 2004, p. 7490 7497 Vol. 78, No. 14 0022-538X/04/$08.00 0 DOI: 10.1128/JVI.78.14.7490 7497.2004 Copyright 2004, American Society for Microbiology. All Rights Reserved. Heterologous

More information

Professor Andrew McMichael

Professor Andrew McMichael BHIVA AUTUMN CONFERENCE 2011 Including CHIVA Parallel Sessions Professor Andrew McMichael University of Oxford 17 18 November 2011, Queen Elizabeth II Conference Centre, London BHIVA AUTUMN CONFERENCE

More information

Supporting Information

Supporting Information Supporting Information Hatziioannou et al. 10.1073/pnas.0812587106 SI Text Viral Constructs and Viral Stock Generation. To generate the HIV-1 constructs used throughout these studies, the env gene in an

More information

Supplemental Materials and Methods Plasmids and viruses Quantitative Reverse Transcription PCR Generation of molecular standard for quantitative PCR

Supplemental Materials and Methods Plasmids and viruses Quantitative Reverse Transcription PCR Generation of molecular standard for quantitative PCR Supplemental Materials and Methods Plasmids and viruses To generate pseudotyped viruses, the previously described recombinant plasmids pnl4-3-δnef-gfp or pnl4-3-δ6-drgfp and a vector expressing HIV-1 X4

More information

VIROLOGY. Engineering Viral Genomes: Retrovirus Vectors

VIROLOGY. Engineering Viral Genomes: Retrovirus Vectors VIROLOGY Engineering Viral Genomes: Retrovirus Vectors Viral vectors Retrovirus replicative cycle Most mammalian retroviruses use trna PRO, trna Lys3, trna Lys1,2 The partially unfolded trna is annealed

More information

JAK3 inhibitor administration in vivo in chronically SIV Infected Rhesus Macaques

JAK3 inhibitor administration in vivo in chronically SIV Infected Rhesus Macaques JAK3 inhibitor administration in vivo in chronically SIV Infected Rhesus Macaques preferentially depletes NK Cells Ladawan Kowawisatsut 1 Kovit Pattanapanyasat 1 Aftab A. Ansari 2 1 Department of Immunology

More information

NIH Public Access Author Manuscript J Acquir Immune Defic Syndr. Author manuscript; available in PMC 2013 September 01.

NIH Public Access Author Manuscript J Acquir Immune Defic Syndr. Author manuscript; available in PMC 2013 September 01. NIH Public Access Author Manuscript Published in final edited form as: J Acquir Immune Defic Syndr. 2012 September 1; 61(1): 19 22. doi:10.1097/qai.0b013e318264460f. Evaluation of HIV-1 Ambiguous Nucleotide

More information

T cell Vaccine Strategies for HIV, the Virus. With a Thousand Faces

T cell Vaccine Strategies for HIV, the Virus. With a Thousand Faces JVI Accepts, published online ahead of print on 13 May 2009 J. Virol. doi:10.1128/jvi.00114-09 Copyright 2009, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved.

More information

Fostering Clinical Development for HIV-1 Vaccine

Fostering Clinical Development for HIV-1 Vaccine W Fostering Clinical Development for HIV-1 Vaccine Ravimiarenduse alane seminar 9. oktoobril Tallinnas Mart Ustav, PhD CSO, SVP 1 FIT Biotech Founded in 1995 Operations in Tampere, Finland and Tartu, Estonia

More information

Macaques vaccinated with live-attenuated SIV control replication of heterologous virus

Macaques vaccinated with live-attenuated SIV control replication of heterologous virus ARTICLE Macaques vaccinated with live-attenuated SIV control replication of heterologous virus Matthew R. Reynolds, 1 Andrea M. Weiler, 1 Kim L. Weisgrau, 1 Shari M. Piaskowski, 1 Jessica R. Furlott, 1

More information

Innate and Cellular Immunology Control of Infection by Cell-mediated Immunity

Innate and Cellular Immunology Control of Infection by Cell-mediated Immunity Innate & adaptive Immunity Innate and Cellular Immunology Control of Infection by Cell-mediated Immunity Helen Horton PhD Seattle Biomedical Research Institute Depts of Global Health & Medicine, UW Cellular

More information

A Path to an HIV Vaccine: GSID Consortium Activities. Faruk Sinangil, PhD 4th Annual CAVD Meeting Miami, FL December 1-4, 2009

A Path to an HIV Vaccine: GSID Consortium Activities. Faruk Sinangil, PhD 4th Annual CAVD Meeting Miami, FL December 1-4, 2009 A Path to an HIV Vaccine: GSID Consortium Activities Faruk Sinangil, PhD 4th Annual CAVD Meeting Miami, FL December 1-4, 2009 Project Goals Acquire and disseminate information that will contribute to the

More information

Commercially available HLA Class II tetramers (Beckman Coulter) conjugated to

Commercially available HLA Class II tetramers (Beckman Coulter) conjugated to Class II tetramer staining Commercially available HLA Class II tetramers (Beckman Coulter) conjugated to PE were combined with dominant HIV epitopes (DRB1*0101-DRFYKTLRAEQASQEV, DRB1*0301- PEKEVLVWKFDSRLAFHH,

More information

Higher priming doses enhance HIV-specific humoral but not cellular. responses in a randomized, double-blind phase Ib clinical trial of

Higher priming doses enhance HIV-specific humoral but not cellular. responses in a randomized, double-blind phase Ib clinical trial of Higher priming doses enhance HIV-specific humoral but not cellular responses in a randomized, double-blind phase Ib clinical trial of preventive HIV-1 vaccines Pierre-Alexandre Bart a,b, Yunda Huang c,

More information

Comparative Efficacy of Subtype AE Simian-Human Immunodeficiency Virus Priming and Boosting Vaccines in Pigtail Macaques

Comparative Efficacy of Subtype AE Simian-Human Immunodeficiency Virus Priming and Boosting Vaccines in Pigtail Macaques JOURNAL OF VIROLOGY, Jan. 2007, p. 292 300 Vol. 81, No. 1 0022-538X/07/$08.00 0 doi:10.1128/jvi.01727-06 Copyright 2007, American Society for Microbiology. All Rights Reserved. Comparative Efficacy of

More information

The global spread of the human immunodeficiency virus (HIV)

The global spread of the human immunodeficiency virus (HIV) Immunological and Virological Analyses of Rhesus Macaques Immunized with Chimpanzee Adenoviruses Expressing the Simian Immunodeficiency Virus Gag/Tat Fusion Protein and Challenged Intrarectally with Repeated

More information

An AIDS vaccine: Why is it so difficult?

An AIDS vaccine: Why is it so difficult? An AIDS vaccine: Why is it so difficult? Jaap Goudsmit, MD, PhD Professor of Poverty-related Communicable Diseases, AMC Chairman of the Board CPCD CSO, Crucell Holland BV An AIDS vaccine: Why is it so

More information