Skip to main content
Journal of Virology logoLink to Journal of Virology
. 2013 Mar;87(6):3526–3537. doi: 10.1128/JVI.02686-12

Antibodies to gp120 and PD-1 Expression on Virus-Specific CD8+ T Cells in Protection from Simian AIDS

Monica Vaccari a, Rabih Halwani b, L Jean Patterson c, Adriano Boasso d, Jennifer Beal c, Elzbieta Tryniszewska a,e, Anna Hryniewicz a, David Venzon g, Elias K Haddad f, Mohamed El-Far b,*, Margherita Rosati h, George N Pavlakis h, Barbara K Felber i, Saleh Al-Muhsen b, Marjorie Robert-Guroff c, Rafick-Pierre Sekaly f, Genoveffa Franchini a,
PMCID: PMC3592123  PMID: 23325679

Abstract

We compared the relative efficacies against simian immunodeficiency virus (SIV) challenge of three vaccine regimens that elicited similar frequencies of SIV-specific CD4+ and CD8+ T-cell responses but differed in the level of antibody responses to the gp120 envelope protein. All macaques were primed with DNA plasmids expressing SIV gag, pol, env, and Retanef genes and were boosted with recombinant modified vaccinia Ankara virus (MVA) expressing the same genes, either once (1 × MVA) or twice (2 × MVA), or were boosted once with MVA followed by a single boost with replication-competent adenovirus (Ad) type 5 host range mutant (Ad5 h) expressing SIV gag and nef genes but not Retanef or env (1 × MVA/Ad5). While two of the vaccine regimens (1 × MVA and 1 × MVA/Ad5) protected from high levels of SIV replication only during the acute phase of infection, the 2 × MVA regimen, with the highest anti-SIV gp120 titers, protected during the acute phase and transiently during the chronic phase of infection. Mamu-A*01 macaques of this third group exhibited persistent Gag CD8+CM9+ effector memory T cells with low expression of surface Programmed death-1 (PD-1) receptor and high levels of expression of genes associated with major histocompatibility complex class I (MHC-I) and MHC-II antigen. The fact that control of SIV replication was associated with both high titers of antibodies to the SIV envelope protein and durable effector SIV-specific CD8+ T cells suggests the hypothesis that the presence of antibodies at the time of challenge may increase innate immune recruiting activity by enhancing antigen uptake and may result in improvement of the quality and potency of secondary SIV-specific CD8+ T-cell responses.

INTRODUCTION

The human immunodeficiency virus type 1 (HIV-1) vaccine platforms brought to the clinic so far have induced either T-cell responses alone, nonneutralizing antibodies to primary HIV isolates, or both (14).

Recombinant poxvirus vector-based HIV vaccine candidates are among the most intensively studied live vectors (5). Numerous studies have demonstrated their ability to induce balanced CD4+ and CD8+ T-cell responses as well as mucosal immune responses against heterologous antigens. Live recombinant poxvirus vectors such as modified vaccinia Ankara virus (MVA) (611), and genetically attenuated NYVAC or ALVAC (8, 12, 13) have been evaluated in combination with DNA immunization in preclinical studies in macaques (5). This approach has generated high-frequency CD4+ and CD8+ T-cell responses (8, 10, 11, 1416) and high titers of nonneutralizing antibodies when combined with viral recombinant proteins (13; our unpublished data). In the simian immunodeficiency virus (SIV) macaque model, the combination of DNA and these viral vector-based vaccine modalities has induced the expression and mobilization of proinflammatory cytokines and reduced by 1 to 2 log the viral load in plasma during primary and chronic infection, depending on the virus used in the challenges (613, 1519; Vaccari et al., unpublished). Protection from high viral replication afforded by these modalities has been correlated not only with vaccine-induced central memory (CM) CD8+ T-cell responses but also with CD4+ T-cell responses (14). In addition, a direct role of vaccine-induced CD8+ and CD4+ T-cell responses in the control of SIV replication has been demonstrated by depleting CD4+ T cells during vaccination and by depleting CD8+ T cells in vaccinated animals before challenge exposure (11, 20). The protection afforded by poxvirus-based SIV vaccines in the preclinical setting using a high-dose challenge modality is, however, not long lasting, and eventually viral replication returns to the levels observed in control animals (14).

Recombinant adenoviruses (Ad) represent a second group of prominent vaccine vectors. Replication-defective Ad vectors have been the most extensively studied (21), but replication-competent Ad vectors are also highly promising, especially as they target multiple mucosal sites regardless of immunization route (22). In the chimpanzee model, replicating Ad-HIV recombinants in combination with envelope protein boosts have elicited potent, long-lasting neutralizing antibodies (23) and protection against homologous and heterologous HIV challenges (24). In the rhesus macaque model, similar prime/boost approaches using replicating Ad type 5 host range mutant (Ad5 h) viruses carrying HIV and/or SIV genes have elicited strong humoral and cellular systemic and mucosal immune responses and sterilizing protection (25) or strongly reduced viremia (2629) following high-dose intrarectal or intravenous simian/human immunodeficiency virus (SHIV) or SIVmac251 challenges, as well as delayed acquisition following repeated low-dose SIVmac251 intrarectal challenges (30).

Nevertheless, in the phase 2b efficacy trial (termed Step), replication-defective Ad5-HIV vaccines expressing Gag, Pol, and Nef failed to prevent HIV acquisition. Moreover, an early vaccine-enhanced risk of HIV acquisition, seen predominantly in uncircumcised men with preexisting Ad5 antibody, was recently confirmed, although the risk waned over time (31). Though the results of this clinical trial were discouraging, a “sieve effect” was confirmed, showing that vaccine-elicited T-cell responses exerted selective pressure on the HIV-infecting founder virus population, although they were not potent enough to prevent infection or decrease viral loads significantly (32). The absence of the envelope gene in this vaccine approach may also account for the lack of protection. The recent results of the RV144 vaccine trial in Thailand suggest that vaccine-elicited antibodies, together with T-cell responses to the HIV-1 envelope, confer some degree of protection against HIV infection (4).

Here, we evaluated the ability of a vaccination approach that induces a combination of T-cell and antibody responses to protect against a high-dose SIVmac251 challenge. We wished to test (i) whether vaccination with DNA followed by recombinant modified vaccinia Ankara virus twice (2 × MVA) could induce T-cell immune responses quantitatively different from those obtained with DNA followed by 1 × MVA or 1 × MVA/Ad5; (ii) whether the omission of the second boost with MVA (1 × MVA) or the absence of the envelope in the Ad5 boost (1 × MVA/Ad5) affected the antibody response to the envelope protein and protection; (iii) whether boosting with replication-competent Ad could provide a higher frequency of long-lasting virus-specific CD8+ and CD4+ T-cell responses in systemic compartments; and (iv) which combination of DNA and live vector-based vaccines would best protect against a high dose of the pathogenic SIVmac251. Our results support the concept that vaccine-induced antibodies to the envelope protein are important in protection and may contribute to the maintenance of effector CD8+ T-cell responses in infected animals.

MATERIALS AND METHODS

Animals, treatments, and SIVmac251 challenge.

All animals used in this study were colony-bred rhesus macaques (Macaca mulatta), obtained from Covance Research Products (Alice, TX). The animals were housed and handled in accordance with the standards of the Association for the Assessment and Accreditation of Laboratory Animal Care International. The care and use of the animals were in compliance with all relevant institutional (NIH) guidelines.

A total of 27 macaques were enrolled in the study. Twenty-one macaques were divided into three groups of seven animals each (Fig. 1). The 1 × MVA group included two Mamu-A*01+ animals, while each of the remaining groups had three. The 21 macaques were immunized at weeks 0 and 4 with intramuscular (3-mg) and intradermal (1-mg) inoculations of DNA-SIV Retanef (8) and individual plasmid DNAs encoding codon-optimized SIV gag, pol, and env genes (33), all given at different sites as previously described (8, 12, 34). All animals were boosted at week 26 with 108 PFU of recombinant MVA expressing SIVmac251 env and gag-pol genes and SIVmac239 Retanef genes delivered intramuscularly at two sites for each animal. Seven animals received no further immunization (1 × MVA group), and seven animals received a second boost with the two MVA recombinants at week 53 (2 × MVA group). The remaining seven macaques were vaccinated with 5 × 108 PFU each of two replication-competent Ad5 h recombinants expressing SIV239gag (35) and SIV239nefΔ1–14 (36) by the intranasal route at week 53 (1 × MVA/Ad5 group). Six control macaques were mock vaccinated with the equivalent amount of nonrecombinant MVA (3 animals, 1 Mamu-A*01+) or Ad5 h (3 animals, 1 Mamu-A*01+) at weeks 26 and 53. All macaques were challenged intrarectally with a high dose (1:10 stock dilution) of pathogenic SIVmac251 21 or 26 weeks after the last immunization (Fig. 1A).

Fig 1.

Fig 1

Clinical and virological outcome of vaccinations. (A) Schematic representation of the immunization regimens. The arrows indicate times of immunizations and of challenge exposure to SIVmac251, performed by the intrarectal route at week 74 in all animals and at week 52 in the 1 × MVA group. (B, C, D, E) Plasma virus levels are presented for each animal. *, identified Mamu-A*01+ macaques. (F, G, H) Comparison of mean virus load in plasma in macaques from the different groups.

Binding antibody assay.

To detect anti-SIVmac251-binding antibodies, serial dilutions of plasma were incubated with SIVmac251 lysate spiked with native purified gp120 or p27 Gag protein of SIVmac251 bound to microtiter enzyme-linked immunosorbent assay plates (19). Endpoint titers were defined as the reciprocal of the highest serum dilution that gave an optical absorbency at 450 nm at least 2 standard deviations greater than average values obtained with negative-control serum.

Preparation of lymphocytes from blood and tissues.

Mononuclear cells from blood and lymph nodes (LNs) were isolated by density gradient centrifugation on Ficoll and resuspended in RPMI 1640 medium (Gibco BRL, Gaithersburg, MD) containing 10% fetal bovine serum (R-10).

Measurement of viral RNA.

SIVmac251 in plasma was quantified by nucleic acid sequence-based amplification as previously described (23). Total RNA was extracted from isolated cells using the guanidium thiocyanate-phenol-chloroform method, modified for TRIzol (Invitrogen, Carlsbad, CA). RNA (1 μg) was reverse transcribed into first-strand cDNA using random hexanucleotide primers, oligo(dT), and Moloney murine leukemia virus reverse transcriptase (Promega, Madison, WI). cDNA quantification was performed by real-time PCR, conducted with the ABI Prism 7900HT (Applied Biosystems, Foster City, CA). All reactions were performed using a SYBR green PCR mix (Qiagen, Valencia, CA) according to the following thermal profile: denaturation at 95°C for 15 s, annealing at 60°C for 15 s, and extension at 72°C for 15 s (data collection was performed during the extension step). Primer sequences were as follows: GAPDH forward, 5-GTCTGGAAAAACCTGCCAAG-3, and reverse, 5-ACCTGGTGCTCAGTGTAGCC-3; SIVgag forward, 5-GCAGAGGAGGAAATTACCCAGTAC-3, and reverse, 5-CAATTTTACCCAGGCATTTAATGTT-3.

Lymphocyte proliferation assays.

Peripheral blood mononuclear cells (PBMC) were incubated with antigens (SIVp27 Gag or gp120 envelope proteins) at a concentration of 5 μg/ml for 5 days at 37°C in 96-well plates. Concanavalin-A (ConA; Sigma-Aldrich) at a concentration of 2 μg/ml was used as polyclonal stimulator, while cultures without ConA or Gag or Envelope proteins were used to determine the background proliferative response. For the in vitro Programmed death-1 (PD-1) and its natural ligand PD-L1 blockade experiments, blocking antibody specific to PD-L1 (eBiosciences) was added to cell cultures at a concentration of 10 μg/ml. A standard surface-staining protocol was followed for CD4+ and CD8+ T cells using anti-human CD4-PE (BD-Pharmingen) and CD8-phycoerythrin (PE)-Texas Red (ECD) (Cedarlane) monoclonal antibodies (MAbs). Between 250,000 and 1 × 106 events were acquired for each condition. Data were analyzed using DIVA software (BD Biosciences). Alternatively, PBMC were cultured at 105 cells per well in triplicate for 3 days in the absence or presence of native high-performance liquid chromatography-purified SIV p27 Gag or gp120 Env protein (Advanced BioScience Laboratories, Kensington, MD) or with ConA as a positive control. The cells were then pulsed overnight with 1 μCi of [3H]thymidine before harvest. Proliferation is expressed as the stimulation index, calculated as the fold increase in thymidine incorporation into cellular DNA of stimulated versus unstimulated (medium only) cells.

Intracellular staining and enzyme-linked immunospot (ELISpot) assays.

SIV-specific CD4+ and CD8+ T-cell responses were detected using pools of 15-meric peptides overlapping by 11 amino acids covering entire Gag, Env, and Pol proteins of SIVmac239. Cells (1 × 106) in RPMI 1640 medium (containing 10% human serum and antibiotics) were incubated in the absence or the presence of a specific peptide pool at 1 μg/ml of each peptide for 1 h or in the presence of the superantigen staphylococcal enterotoxin B (Sigma, St. Louis, MO) at a 1-mg/ml final concentration as a positive control. The costimulatory MAbs CD28 and CD49d (0.5 μg/ml; BD Pharmingen, San Diego, CA) were added to all the samples to maximize the detection of T cells with higher activation thresholds. The positive control was treated with staphylococcal enterotoxin B, brefeldin A (GolgiPlug; BD Biosciences) at a final concentration of 10 μg/ml was added, and the cells were incubated for an additional 5 h. The cells were washed, stained for the surface antigens with anti-human CD3alt epsilon (clone SP34; BD Pharmingen) and anti-human CD8beta Abs (clone 2ST8.5H7; Beckman Coulter), permeabilized by incubation in Cytofix/Cytoperm solution (BD Biosciences), and stained with anti-human interleukin-2 (IL-2)-fluorescein isothiocyanate (FITC) (clone MQ1-17H12) and anti-gamma interferon (IFN-γ)-allophycocyanin (APC) MAbs (clone B27; BD Pharmingen). The results were calculated as the total number of cytokine-positive cells with background subtracted.

T-cell ELISpot assay was performed using Mabtech ELISpot plus for Monkey IFN-γ kits in triplicate on freshly purified PBMC. Cells (3 × 105 cells/well) were used with or without stimulation with the SIVmac251 Gag or Env overlapping peptides as described previously (11). ConA was used as a positive control (final concentration, 1 μg/ml). The cells were then incubated at 37°C in 5% CO2 atmosphere for 24 h and washed. The plates were assayed for IFN-γ-producing cells per the manufacturer's protocol. The spot-forming cells (SFC) were counted on an ELISpot reader. The results are expressed as number of spots per 106 cells after subtracting the number of spots in unstimulated wells.

Flow cytometry and isolation of Tetramer+ and memory T-cell subpopulations.

Mononuclear cells from blood were separated and stained as previously described (11).

The frequency of CCR5-positive CD4+ T-cells was assessed by staining mononuclear cells with anti-human CD3 antibody (clone SP34), anti-human CD4 antibody (clone L200), and anti-human CCR5 antibody (clone 3A9; BD Biosciences). SIV-specific CD8+ T cells staining was performed with anti-human CD8β antibody (clone 2ST8.5H7; Beckman Coulter), anti-human CD28 antibody (clone CD28.2; Pharmingen, San Diego, CA), anti-human CD95 antibody (clone DX2; BD Pharmingen), and Gag181-189 CM9 (p11C; CTPYDINQM)-Mamu-A*01 tetrameric complexes (Beckman Coulter) for 30 min at room temperature.

Lymph nodes isolated from the macaques at week 24 following challenge were labeled as follows. Cells isolated from Mamu-A*01+ animals were labeled with CD8 (CD8-PE-Texas Red [ECD]; BD), CM9 (Tet-PE; BD), and 7-AAD (PE-Cy5; BD). Cells isolated from non-Mamu-A*01+ animals were labeled with CD4 (clone L200-PerCP-Cy5.5; BD Biosciences), CD8 (CD8-PE-Texas Red [ECD]; Cedarlane), CD95 (clone DX2-PE-Cy5; BD Biosciences), CD28 (clone CD28.2-Pacific Blue; custom made, BD Biosciences), CCR7 (clone 3D12-PE-Cy7; BD Biosciences), 7-AAD (PE-Cy5; BD Biosciences). For Mamu-A*01+ animals, cells were sorted for CD8+CM9+ cells. For non-Mamu-A*01+ animals, cells were sorted into CM and effector memory (EM) T cells. Sorting was performed using a flow cytometer (FACSAria; BD Biosciences). The purity of the CM and EM subpopulations ranged from 96 to 99%. Cells were sorted directly into RLT buffer and followed by RNA extraction. All procedures were done at 4°C to minimize any changes in cell phenotype or gene expression.

In vitro infection of human DCs with MVA and Ad5.

PBMC, isolated from 5 cytomegalovirus (CMV)-negative subjects, were infected in vitro with MVA or nonreplicating Ad5, and PD-L1 levels of expression on dendritic cells (DCs) were monitored following infection. A noninfected group (Mock) was also monitored. Briefly, 3 × 106 cells were infected, or not, with either MVA (gpe) or Ad5 (gag) at a multiplicity of infection (MOI) of 10 and then incubated at 37°C for 24 h. Cells were then labeled with anti-CD3-Alexa 700, anti-CD19-Alexa 700, anti-CD14-Alexa 700 (BD Biosciences), anti-CD16-Alexa 700 (Biolegend), anti-HLADR-APC-Cy7, anti-CD11c-APC, anti-CD123-PE, anti-CD86-PE-cy5, and anti-PD-L1:PE-Cy7 (BD Biosciences) and analyzed with a BD LSRII flow cytometer. Myeloid DCs (mDCs) were identified as CD3, CD19, CD16, CD14, HLADR+, CD11c+, and CD123; and plasmacytoid dendritic cells (pDCs) as CD3, CD19, CD16, CD14, HLADR+, CD11, and CD123+. The level of activation, and hence infection, of DCs was monitored by measuring CD86 expression (n = 5 for the three groups, MVA, Adv, and Mock).

RNA isolation, amplification, and microarray hybridization.

CM9+ cells within LN of Ad5 h and MVA-vaccinated macaques were sorted 24 weeks following challenge using a BD FACSARIA I. Sample RNA was extracted using an RNA extraction kit (Qiagen) and amplified using an RNA kit (MessageAmp; Ambion) according to the manufacturer's instructions. Quantification was performed using a spectrophotometer (NanoDrop Technologies), and RNA quality was assessed using the Experion automated electrophoresis system (Bio-Rad). Total RNA was then amplified and labeled using the Illumina TotalPrep RNA amplification kit, which is based on the Eberwine amplification protocol. This protocol involves an initial cDNA synthesis step followed by in vitro transcription for cRNA synthesis. The biotinylated cRNA was hybridized onto Illumina Human RefSeq-8 BeadChips at 58°C for 20 h and quantified using an Illumina BeadStation 500GX scanner and Illumina BeadStudio v3 software. We optimized the use of the Illumina microarray human RefSeq-8 BeadChips for the analysis of macaque gene expression.

Microarray data preprocessing.

Illumina probe results were exported from BeadStudio as raw data and screened for quality. Samples failing chip visual inspection and control examination were removed. Gene expression data were analyzed using Bioconductor, an open-source software library for the analysis of genomic data based on R, a language and environment for statistical computing and graphics (www.r-project.org). Genes were then filtered by intensity and by variance filters to allow a reduction in the number of tests and a corresponding increase in power of the differential gene expression analysis (37). The resulting matrix showing filtered genes as rows and samples as columns was log2 transformed and used as input for linear modeling using Bioconductor's limma package. P values from the resulting comparison were adjusted for multiple testing. This method controls the false-discovery rate, which was set to 0.05 in this analysis. Data were then analyzed using gene set enrichment analysis (GSEA) and ingenuity pathway analysis. GSEA is a computational method that determines whether a previously defined set of genes shows statistically significant, concordant differences between two biological states (e.g., phenotypes). GSEA was performed using the C2 pathways collection from the Molecular Signatures Database (MSigDB), which is a collection of gene sets for use with GSEA software. Microarrays were scanned at 16 bits using the ScanArray Express (Packard Instrument Co.). The microarrays were then screened for quality, first by visual inspection of the array with flagging of poor-quality spots and then with automated scripts that scanned the quantified output files and measured overall density distribution on each channel and the number of flagged spots. Lowess normalization was then applied on the scanned chips.

RESULTS

Vaccination with 2 × MVA, but not 1 × MVA or 1 × MVA/Ad5, is associated with durable control of viral replication.

All vaccinated and control macaques were exposed intrarectally to a single high dose of SIVmac251 as previously described (10, 11, 38) and outlined in Fig. 1A. All animals became infected. Plasma SIV RNA copies were measured from the time of exposure up to 24 weeks postchallenge (Fig. 1B to E). A peak of viral replication was observed at 2 weeks from exposure in all animals, as previously observed with this SIVmac251 challenge stock (10, 11, 38). All vaccinated groups experienced significantly lower levels in plasma at week 2 during primary infection than the mock-vaccinated animals (for all groups, P = 0.0051; Fig. 1B to H). The mean virus levels over weeks 2 to 16 postinfection were significantly lower than in the control group in the 2 × MVA group only (P = 0.0070, by the Wilcoxon rank-sum test) (Fig. 1G). No significant differences were observed in mean virus levels in plasma among the 2 × MVA, 1 × MVA/Ad5, and 1 × MVA groups at any time point analyzed (data not shown). The frequency of CCR5+CD4+ T cells was assessed in mononuclear cells isolated from blood, peripheral lymph nodes, rectal mucosal pinch biopsy specimens, and bronchoalveolar lavage fluids collected at week 24 postinfection from 5, 6, and 4 animals in the 2 × MVA and 1 × MVA/Ad5 groups (data not shown). Despite the lack of significant difference in virus levels in plasma among the groups of vaccinated animals, there was a significantly higher percentage of CCR5+CD4+ T cells in the rectal mucosa of the animals in the 2 × MVA group than in the controls (P = 0.042), while no differences were found for the 1 × MVA/Ad5 group (data not shown). The frequency of CD4+ T cells in the 2 × MVA group was also significantly higher in the bronchoalveolar lavage fluids than in the 1 × MVA/Ad5 group and the controls (P = 0.017 and 0.007, respectively, by the Wilcoxon rank sum test) (data not shown). These data suggest a better regeneration of CD4+ T cells at mucosal sites in animals vaccinated with the 2 × MVA vaccine.

The 2 × MVA vaccine induced significantly higher anti-gp120 titers than 1 × MVA or 1 × MVA/Ad5 regimens but equivalent frequencies of T-cell responses.

Serum binding antibodies to the Gag and Envelope proteins were measured by enzyme-linked immunosorbent assay (ELISA). At 3 to 4 weeks after the last immunization, antibody titers to p27 Gag did not differ significantly among the vaccinated groups (Fig. 2A). In contrast, we found significantly higher antibody titers to gp120 in the 2 × MVA-immunized group (P = 0.044 across the three groups (P = 0.029 between 2 × MVA and 1 × MVA/Ad5), whereas the difference only approached statistical significance between the 2 × MVA and 1 × MVA groups (P = 0.07; Fig. 2B). The titers of binding antibodies to SIV gp120, however, did not correlate significantly with the virus levels in plasma (Fig. 2C).

Fig 2.

Fig 2

Antibody titers and kinetics of CD8+CM9+ T-cell response in the vaccinated macaques. Endpoint antibody titers to p27 Gag (A) and gp120 (B) in the sera of the immunized monkeys. (C) Correlative analysis between the titers of the gp120-binding antibodies in all the vaccinated animals and plasma viral loads at week 6 postinfection. (D to G) Frequency and kinetics of CM9+CD8+ T cells in blood of the macaques during immunization and following challenge exposure to SIVmac251. (H to K) Relative frequency of CD95+CD28 (effector memory [EM] in solid black bars) and CD95+CD28+ (central memory [CM] in white bars) CM9+CD8+ T cells in blood of vaccinated and control macaques following challenge exposure to SIVmac251. The data are shown as means for each group, calculated using the individual frequency of EM or CM on the total Gag-CM9+ memory population.

Prior to the final boost with either MVA-SIV or Ad5-SIV at week 53 (Fig. 1A), the DNA/MVA regimens elicited equivalent T-cell responses, as shown by measurement of T-cell proliferation to gp120 and Gag in the 2 × MVA group and 1 × MVA/Ad5 group on freshly collected blood 1 week following the week 26 immunization and up to the time of the last boost (see Fig. S1A in the supplemental material). Similarly, the ability of PBMC to produce IFN-γ in an ELISpot assay or of CD4+ or CD8+ T cells to produce IFN-γ and IL-2 in an intracellular cytokine staining (ICS) assay following in vitro stimulation with overlapping Gag, Env, Nef, and Pol peptides did not differ between the 2 × MVA and 1 × MVA/Ad groups (see Fig. S1B to D in the supplemental material). None of these T-cell responses correlated significantly with protection from high virus levels in plasma (data not shown).

Sustained expansion of Gag tetramer CM9+CD8+ T effector cells in 2 × MVA-vaccinated macaques.

We studied the frequency of CD8+CM9+ Gag-specific T cells in the blood of the Mamu-A*01+ animals included in each group and found that the overall frequency of the CM9+CD8+ T cells did not differ significantly in the three vaccinated groups after the first MVA boost (week 26), as expected (Fig. 2D to F). The administration of either an additional MVA or an additional Ad5 vaccine did not further increase these responses (Fig. 2E and F). Following challenge exposure, the overall frequency of CM9+ Gag-specific tetramer cells (area under the curve) did not differ among the three vaccinated groups. However, the CD8+CM9+ population that was still expanding at week 16 or 18 postchallenge in the 1 × MVA and 2 × MVA groups (Fig. 2D and E) had already begun to contract in the 1 × MVA-Ad5 group (Fig. 2F). As expected, the kinetics of expansion of CD8+CM9+ T cells was delayed in the mock-vaccinated group and was already contracted by week 16 (Fig. 2G).

The kinetics of expansion of CD8+CM9+ T cells in the blood suggested either differences in trafficking to tissues or that the vaccine regimens, in addition to eliciting different levels of antibody responses to gp120 (Fig. 2B), may have elicited CD8+CM9+ T cells with a different proliferative potential. To investigate this hypothesis, we analyzed the memory phenotype of the CM9+CD8+ T-cell population in the blood of the vaccinated animals. We found a marginally significant larger and more-sustained expansion of effector memory (EM; CD28CD95+) CM9+ T cells in macaques immunized with the 2 × MVA than in macaques immunized with the 1 × MVA/Ad5 regimen vaccine (P = 0.051, by repeated measures analysis of variance at all postchallenge time points tested) (Fig. 2I and J). The 1 × MVA/Ad5 group macaques showed a more balanced response of both EM and central memory (CM; CD28+CD95+) CM9+ T cells (Fig. 2H). Thus, boosting with MVA or adenovirus did not induce immune responses that could be differentiated by the frequency of IFN-γ or by the overall frequency of total tetramer+ CD8+ T cells in Mamu-A*01+ macaques; rather, the data suggested a possible difference in the ability of antigen-specific cells to proliferate and expand following challenge exposure.

Differential PD-1 expression on antigen-specific cells from vaccinated macaques.

To investigate the mechanism behind this observation, we analyzed Gag CM9+ CD8+ T cells for the expression of Programmed death-1 (PD-1), a critical mediator of virus-specific CD8+ T-cell exhaustion (39), in the six Mamu-A*01+ macaques included in the 2 × MVA and the 1 × MVA/Ad5 groups.

Gag-CM9+ T cells were collected from the Mamu-A*01+ animals in 2 × MVA and 1 × MVA/Ad-5 groups at week 74 (before the challenge) and at weeks 7 and 24 following challenge exposure. Cells were labeled with carboxyfluorescein diacetate succinimidyl ester (CFSE) and stimulated with a pool of Gag peptides. The level of expression of PD-1 on nonproliferating CFSEhi CM9+ cells from macaque 906 in the 2 × MVA group and macaque 221 in the 1 × MVA/Ad5 group is shown in Fig. 3A. The levels of expression of PD-1 in both proliferating (CFSElo) and nonproliferating (CFSEhi) CM9 tetramer+ T cells before (week 74, day 0) and after (weeks 7 and 24) challenge exposure to SIVmac251 (Fig. 3B to D) were lower in macaques immunized with 2 × MVA than in macaques immunized with 1 × MVA/Ad5 (Fig. 3B to D) when the mean fluorescence intensity (MFI) values for PD-1 were compared at all time points (P = 0.003 for CFSEhi and P = 0.05 for CFSElo; Fig. 3E). PD-1 levels on CFSEhi and CFSElo cells from mock-vaccinated macaques were comparable to those of 1 × MVA/Ad5-vaccinated macaques (data not shown). Importantly, in vitro treatment with blocking antibody for PD-L1 (40, 41), the natural ligand for PD-1, significantly restored proliferation in Gag-specific CD8+ (Fig. 3F) and CD4+ (Fig. 3G) T cells isolated from lymph nodes of Mamu-A*01 and non-Mamu-A*01 macaques (4 from the 2 × MVA group, 3 from the 1 × MVA/Ad5 group, and 4 from the control group) at week 24 postinfection. These results confirm that the interaction of PD-L1 with PD-1 negatively regulates T-cell proliferation.

Fig 3.

Fig 3

PD-1 expression on CD8+CM9+ T-cell response in the vaccinated macaques. (A) CFSE gating strategy and mean fluorescence intensity (MFI) of PD-1 expression on nonproliferating CFSEhi CM9+CD8+ T cells from representative macaques. (B to D) PD-1 level of expression on CFSElo or CFSEhi CM9+ CD8+ T cells from animals from the 2 × MVA and 1 × MVA/Ad5 groups were analyzed at week 74 (before challenge) and at weeks 7 and 24 after challenge exposure to SIVmac251. (E) PD-1 expression levels on low and high proliferating CD8+CM9+ T cells in the two groups of vaccinated animals at all time points. The plot includes all the data points, and each dot represents one macaque. (F and G) Frequency of Gag-specific T cells 5 days following stimulation with Gag peptides in the presence (or absence) of anti-PD-L1 blocking antibody; CD8+ T cells (F) and CD4+ T cells (G) are gated on CFSElow cells. (H to J) PD-L1 expression on human mDCs (H) and on pDCs (J) and CD86 expression on human mDCs (I) following in vitro infection with Ad5 or MVA.

Next, we investigated the ability of the vaccine vectors used in this study to increase the expression of PD-L1 on dendritic cells. Infection of in vitro-matured human myeloid dendritic cells (mDC) by nonreplicating Ad5 significantly enhanced the level of PD-L1 expression compared to MVA-infected mDCs (Fig. 3H), and the CD86 maturation marker was significantly increased by infection with MVA but not Ad5 (Fig. 3I). No difference in PD-L1 expression was observed following exposure of pDC to either MVA or Ad5 (Fig. 3J).

SIV-specific CD8+ T cells elicited by the 1 × MVA/Ad5 vaccine exhibit a more proapoptotic gene expression profile than do those elicited by the 2 × MVA regimen.

To better define and confirm the qualitative differences in the SIV responses induced by these two vaccine regimens, we determined the gene expression profile of SIV-specific CM9+ CD8+ T cells. We obtained total RNA from CD8+CM9+ cells sorted from lymph nodes collected from 2 Mamu-A*01+ animals in the 1 × MVA/Ad5 group and 3 animals in the 2 × MVA group at week 24 postinfection and analyzed it by microarray (Table 1). The expression of genes involved in survival and apoptosis was analyzed using both the gene set enrichment analysis (GSEA) and the ingenuity pathway analysis. The significantly upregulated and downregulated pathways are presented in Fig. 4 and in Fig. S2 in the supplemental material. Figure 4 (upper panel) illustrates 8 significant pathways (P values < 0.05), 6 upregulated in 1 × MVA/Ad5 CM9+ cells and 2 upregulated in 2 × MVA CM9+ cells. Within those pathways, we have selected the most up- and downregulated genes; 27 were upregulated in 1 × MVA/Ad5 CM9+ cells, and 8 were upregulated in 2 × MVA CM9+ cells. The genes within the 6 upregulated pathways in the 1 × MVA/Ad5 group were involved mostly in apoptosis, cellular anergy and inactivation, or transcriptional suppression as indicated in Fig. 4 (upper panel) and Table 1. Of particular importance are genes like PPP2R5C, ATF2, ATF4, PTEN, MAP3K4, MAP3K7, MAPK1, NIN, RAP1, and YWHAZ, which are known to have proapoptotic activity and to be implicated in the negative control of cell growth and division (4244). On the other hand, genes within pathways upregulated in 2 × MVA such as TAPBP, HLA-DQB1, HLA-DMB, CIITA, IRAK1, IGBP1, and CCND2 are involved in major histocompatibility complex (MHC) formation and presentation, indicating immune activation as well as cell persistence (4547). The upregulation of genes involved in MHC class II maturation and peptide presentation were also observed independently when we analyzed the data using the ingenuity approach (Fig. 4, lower panel). Collectively, the data from two independent analysts suggest that the CM9+ cells in the 1 × MVA/Ad5-immunized animals could be more prone to apoptosis than those in the 2 × MVA-immunized animals.

Table 1.

Definition of pathways and genes differentially expressed in Adv versus MVA Tet+ cells

Pathway or gene name Function or description
Pathways
    HSA04010_MAPK_SIGNALING_PATHWAY Genes involved in MAPK signaling pathway
    HSA04540_GAP_JUNCTION Genes involved in gap junction
    M_2.9_ERK_TRANSACTIVATION_CYTOSKELETAL_MAPK_JNK Progressively downregulated through 12 h following treatment of WS1 human skin fibroblasts with UVC at a high dose
    UVC_HIGH_D4_DN Downregulated at any time point following treatment of XPB/CS fibroblasts with 3 J/m2 UVC
    UVC_XPCS_ALL_DN Downregulated at 4 h following treatment of XPB/CS fibroblasts with 3 J/m2 UVC (genes involved in apoptosis)
    UVC_XPCS_4HR_DN Upregulated in mouse hematopoietic stem cells
    HSC_HSCANDPROGENITORS_FETAL Genes involved in antigen processing and presentation
    HSA04612_ANTIGEN_PROCESSING_AND_PRESENTATION Genes involved in MAPK signaling pathway
Genes
    Upregulated in Tet+ MVA compared to Tet+ Adv
        TAPBP Required for MHC formation
        CIITA Involved in transcription of HLA class II and important for antigen presentation
        HLA-DQB1 Involved in transcription of HLA class II and important for antigen presentation
        HLA-DMB Helps to load HLA class II
        IRAK1
        IGBP1 B-cell activation
        PSMC5 Important for MHC-I processing
        CCND2 Important for cell division
    Uppregulated in Tet+ Adv compared to Tet+ MVA
        MAP3K4 Apoptosis
        JUN Apoptosis
        ATF4 Apoptosis
        ATF2 Apoptosis
        RAP1A Plays a role in apoptosis
        DUSP5 Apoptosis
        TAOK1 Apoptosis
        PTEN Tumor suppressor
        ITPR1 Apoptotic gene
        TUBB2B
        PRKACB Apoptosis
        PRKCB1 Apoptotic gene
        SOS1 Differential role
        UBE2G1 Ubiquitination
        POGZ Gene suppressor
        ARID5B Gene repression
        PPP2R5C Negative control of cell division
        TRIM33 Gene repression
        SPEN Gene transcription repression
        CDYL Oncogene repression
        CNOT4 Transcription repression
        CBLB Anti-inflammatory

Fig 4.

Fig 4

Heat map representation of gene expression within CM9+ CD8+ T cells at 24 weeks following challenge. (Upper panel) Genes from representative pathways analyzed with GSEA and displayed as a heat map to demonstrate differential expression between Adv CM9+ and MVA CM9+ cells. Each pathway comprising selected top upregulated genes is represented by a different color. The false color expression in log2 scale is depicted on the right side of the figure. The heat map shows the expression level of each gene (red, upregulated; blue, downregulated). Eight significant pathways are shown (P values < 0.05) (6 upregulated in Ad5 CM9+ samples and 2 upregulated in MVA CM9+ cells), as is a subset of 30 most up- and downregulated genes (22 upregulated in Ad5 CM9+ samples and 8 upregulated in MVA CM9+ samples). (Lower panel) Genes upregulated and downregulated in antigen presentation pathways.

To determine if the differences observed in proapoptotic gene expression among the Mamu-A*01+ macaques could explain the greater level of sustained viral control observed in all the 2 × MVA-vaccinated animals (Fig. 1G) but not in all the 1 × MVA/Ad5-vaccinated macaques (Fig. 1H), we examined viral loads in just the Mamu-A*01+ animals (see Fig. S1E in the supplemental material). No difference in viremia control was seen between groups (Fig. S1E). Moreover, 1 × MVA-Ad5-immunized macaques that exhibited proapoptotic gene expression (Fig. 4, upper panel) were not correlated with viremia control. Macaque 907 exhibited a high persistent viral load, whereas macaque 221 controlled viremia. Similarly, among the 2 × MVA-vaccinated macaques that displayed profiles indicative of immune activation and cell persistence, only macaque 902 controlled viremia, whereas macaques 906 and 314 exhibited high viral loads (see Fig. S1E in the supplemental material). Thus, more-complex interactions must be involved in the control of viremia in these macaques, as discussed below.

DISCUSSION

Heterologous prime-boost vaccination has been shown to enhance immune responses and control viral replication (48). MVA and nonreplicating Ad5 vectors have the capability to infect immature dendritic cells, but they may differently influence the ability of dendritic cells to present antigens (4951). In the case of nonreplicating Ad5, dendritic cell activation has been demonstrated to depend on the recognition of a heparin-sensitive receptor recognized by the Ad5 fiber, the Shaft (52), suggesting that the same mechanisms of activation of dendritic cells may also be used by the replicating Ad5 h used in our study. We reasoned that because the DNA/MVA platform induces a high frequency of CD4+ T-cell responses and Ad5 induces potent CD8+ T-cell responses (8, 33, 4951), the combination of a DNA prime followed by boosts with the two vectors could increase immune responses to Gag and enhance protection (53). In this study, we demonstrated that boosting DNA vaccines with two consecutive doses of MVA that express the Envelope protein (2 × MVA) elicited a significantly higher binding antibody response to the SIV envelope protein and conferred more-durable protection against SIVmac251 replication than boosting with 1 × MVA followed by an Ad5 h vector that did not express the Envelope protein (1 × MVA/Ad5). This result was expected, since repeated inoculation with MVA vectors expressing the envelope protein boosts antibody responses (7).

T-cell frequency and functionality, measured by ELISpot following in vitro stimulation with SIV Gag or by proliferative responses to Gag and Env, were comparable between the two groups before and after the second boosts with either MVA or Ad5 h. However, following challenge exposure and infection, there was a clear difference in the kinetics of expansion and durability of CM9+ T memory cells in blood of the vaccinated Mamu-A*01+ macaque groups. While SIV-specific cells within the 1 × MVA/Ad5 group peaked at 2 weeks following challenge exposure to SIVmac251, those of the 2 × MVA group were still expanding and did not peak until week 16 following challenge exposure. The memory CM9+ T cells appeared to decrease faster in the 1 × MVA-Ad5 group than in the 2 × MVA or the 1 × MVA group.

The 2 × MVA vaccination regimen induced a significantly lower level of PD-1 expression on SIV-specific cells when all the time points were taken together (Fig. 2E). But most striking were the distinctly different gene expression profiles associated with apoptosis seen between the SIV-specific T cells in the 2 × MVA compared to the 1 × MVA/Ad5 macaques that carried the Mamu-A*01 allele. The decreased activation of several proapoptotic pathways in the 2 × MVA animals suggested a reasonable explanation for the different rate of expansion and contraction of Gag-specific effector cells in this group.

PD-1 is believed to be a major regulator of apoptosis (39) that can impact the frequency of HIV-specific CD8+ T cells and could be manipulated to improve CD8+ T-cell survival and function (28, 54, 55). Upregulation of PD-1 expression on T cells and PD-L1 expression on DCs (28, 55) could be the consequence of SIVmac251 continuous replication in the infected animals, since at these time points, all animals were viremic, although the 2 × MVA animals controlled viremia better than did the control macaques. However, because the low level of PD-1 on SIV-specific cells was also observed before exposure to SIVmac251 (prechallenge time point week 74 [day 0]; Fig. 4, lower panel) and persisted after infection in the 1 × MVA/Ad5 group, we believe that differential PD-1 expression may be due to immunogenic differences between Ad5 and MVA. Indeed, our data using the nonreplicating Ad5 demonstrated the elicitation of higher expression of PD-L1 on human myeloid dendritic cells in vitro than MVA.

PD-L1 has been shown to interact with B7-1 costimulatory molecule to inhibit T-cell responses. Extensive studies have shown that a PD-1/PD-L1 blockade restores exhausted T cells during chronic viral infections and in tumors (28, 55). Song et al. evaluated the effects of soluble PD-1 (sPD-1) as a blockade of PD-1 and PD-L1 on vaccine-elicited antigen-specific T-cell responses in mice (56). Coadministration of sPD-1 with an Ad-based vaccine could increase antigen-specific CD8+ T-cell responses, indicating a vaccine type-independent adjuvant effect of sPD-1. This coadministration of sPD-1 DNA augmented T-cell proliferation and reduced T-cell apoptosis through upregulation of Bcl-xL expression during T-cell activation.

Gene array analysis revealed the activation of proapoptotic pathways within SIV-specific T cells of the 1 × MVA/Ad5 group 24 weeks following challenge. This is in accordance with the lower frequency of the CM9+CD8+ T cells at the specified time points in the 1 × MVA/Ad5 group. This apoptotic state could be triggered by PD-1-mediated apoptosis and cell contraction (41, 57). Genes within the 5 pathways upregulated in the Ad5-vaccinated group were mostly genes involved in apoptosis, cellular anergy, and inactivation or were transcription suppressor genes. PPP2R5C is implicated in the negative control of cell growth and division (54, 57). ATF2 and ATF4 have proapoptotic activity and are involved in the upregulation of Fas and FasL (56). PTEN is known to inhibit AKT phosphorylation, involved in phosphorylation of FOXO3a in different cell models (58) as well as it negatively regulates the activation of NF-κB and Iκ kinase β (IKKβ) (59). In fact, both PTEN and DUSP5, also upregulated in the Ad5 group, are upstream of FOXO3a and are believed to inhibit its phosphorylation, leading to a reduction in cell survival. Other genes that are also upregulated and are part of the mitogen-activated protein kinase (MAPK) or extracellular signal-regulated kinase (ERK) pathway are MAP3K4 and MAP3K7 (both activate the JNK MAPK pathways in response to cellular stress). MAPK1, NIN, RAP1, and YWHAZ are all involved in many vital cellular processes such as metabolism, protein trafficking, signal transduction, apoptosis, and cell cycle regulation. Moreover, we have preliminary results indicating that the PD-1 inhibitory effect is mediated through dephosphorylation of FOXO3a, which in turn upregulates expression of proapoptotic FOXO3a target genes such as Fas ligand and Bim genes (R. P. Sekaly, unpublished data). Therefore, the high level of expression of PD-1 on SIV-specific CD8+ T cells in the 1 × MVA/Ad5 group may drive the upregulation of the above genes, leading to cellular anergy and death.

Nevertheless, the lack of correlation of viremia control with the proapoptotic gene expression profiles does not allow us to conclude that this mechanism contributed to the lesser protection during the chronic phase in the 1 × MVA/Ad5 group. Microarray analysis was conducted only on 2 Mamu-A*01 macaques in this group, and more extensive studies may still be needed to prove this hypothesis. However, other factors may have offset the impact of any enhanced apoptosis or exhaustion. We note that the 1 × MVA/Ad5-immunized macaques exhibited a balanced T-cell EM and CM response (Fig. 2I). Both EM (60) and CM (61) T cells have been shown to be important for vaccine-elicited protection. CM T cells are important for replenishing memory T-cell populations. It is possible that gene expression profiles of the 1 × MVA/Ad5-vaccinated macaques reflect in part the restraint needed to prevent uncontrolled CM T-cell expansion.

An alternative explanation for the enhanced protection of the 2 × MVA group was suggested by the gene expression profiles. The pathways upregulated in the 2 × MVA group included the antigen processing and presentation pathway as well as the hematopoietic stem cells (HSC) and progenitors pathway, all involved in MHC-I and -II maturation and peptide presentation. TAPBP is involved in the association of MHC-I with transporter associated with antigen processing (TAP), which is required for the assembly of MHC-I with peptide (peptide loading). Both HLA-DQB1 and HLA-DMB belong to the HLA class II beta chain paralogues and play a central role in the immune system by presenting peptides derived from extracellular proteins (62). CIITA is essential for transcriptional activity of the HLA class II promoter (46). Upregulated genes within the HSC and progenitor pathway (Fig. 4, upper panel) are involved in cell survival and persistence. IRAK1 binds to the IL-1 type I receptor following IL-1 engagement, triggering intracellular signaling cascades leading to transcriptional upregulation and mRNA stabilization. IGBP1 is involved in the proliferation and differentiation of B cells (22), while CCND2 is essential for the control of the cell cycle (45). This upregulation of genes involved in MHC-II maturation and peptide presentation was also scored when the gene data were analyzed using the ingenuity approach (Fig. 4, lower panel). In fact, the MHC-II presentation pathway was one of three gene expression pathways that were significantly differentiated using that approach (see Fig. S2 in the supplemental material). The higher titers of binding antibodies to gp120 elicited by vaccination in the 2 × MVA group, which per se do not correlate with better control of plasma virus levels, may rather have enhanced antigen uptake through complex formation and stimulation of the Fcγ receptor, thus influencing the quality of the secondary T-cell response to SIVmac251 (63, 64). The microarray data that demonstrate upregulation of genes involved in MHC-I and II antigen presentation in the 3 Mamu-A*01 macaques of the 2 × MVA group support this hypothesis (Fig. 4, lower panel), although further studies on larger numbers of animals will be necessary to confirm it.

Poxvirus-Ad vector combinations are being increasingly explored in HIV vaccine design, and dissecting immune correlates of protection is difficult. In both mice (65) and macaques (66), the order of heterologous vector administration influences the memory profile of virus-specific T cells. As suggested here, an envelope protein boost, in addition to providing potentially protective antibodies, may also impact T-cell responses. It will be important to elucidate whether this potential mechanism influences vaccine protective efficacy.

Supplementary Material

Supplemental material

ACKNOWLEDGMENTS

This research was supported by the Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research.

We thank Gene Shearer and Richard S. Blumberg for helpful discussions, Nancy Miller for the gift of the SIVmac251 challenge stock, and Teresa Habina for editorial assistance.

Footnotes

Published ahead of print 16 January 2013

Supplemental material for this article may be found at http://dx.doi.org/10.1128/JVI.02686-12.

REFERENCES

  • 1. Buchbinder SP, Mehrotra DV, Duerr A, Fitzgerald DW, Mogg R, Li D, Gilbert PB, Lama JR, Marmor M, Del RC, McElrath MJ, Casimiro DR, Gottesdiener KM, Chodakewitz JA, Corey L, Robertson MN. 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 372:1881–1893 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Edupuganti S, Weber D, Poole C. 2004. Cytotoxic T-lymphocyte responses to canarypox vector-based HIV vaccines in HIV-seronegative individuals: a meta-analysis of published studies. HIV Clin. Trials 5:259–268 [DOI] [PubMed] [Google Scholar]
  • 3. Flynn NM, Forthal DN, Harro CD, Judson FN, Mayer KH, Para MF. 2005. Placebo-controlled phase 3 trial of a recombinant glycoprotein 120 vaccine to prevent HIV-1 infection. J. Infect. Dis. 191:654–665 [DOI] [PubMed] [Google Scholar]
  • 4. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, Kaewkungwal J, Chiu J, Paris R, Premsri N, Namwat C, SMde Adams E, Benenson M, Gurunathan S, Tartaglia J, McNeil JG, Francis DP, Stablein D, Birx DL, Chunsuttiwat S, Khamboonruang C, Thongcharoen P, Robb ML, Michael NL, Kunasol P, Kim JH. 2009. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N. Engl. J. Med. 361:2209–2220 [DOI] [PubMed] [Google Scholar]
  • 5. Franchini G, Gurunathan S, Baglyos L, Plotkin S, Tartaglia J. 2004. Poxvirus-based vaccine candidates for HIV: two decades of experience with special emphasis on canarypox vectors. Expert Rev. Vaccines 3(Suppl 1):S75–S88 [DOI] [PubMed] [Google Scholar]
  • 6. Allen TM, Vogel TU, Fuller DH, Mothe BR, Steffen S, Boyson JE, Shipley T, Fuller J, Hanke T, Sette A, Altman JD, Moss B, McMichael AJ, Watkins DI. 2000. Induction of AIDS virus-specific CTL activity in fresh, unstimulated peripheral blood lymphocytes from rhesus macaques vaccinated with a DNA prime/modified vaccinia virus Ankara boost regimen. J. Immunol. 164:4968–4978 [DOI] [PubMed] [Google Scholar]
  • 7. Amara RR, Villinger F, Altman JD, Lydy SL, O'Neil SP, Staprans SI, Montefiori DC, Xu Y, Herndon JG, Wyatt LS, Candido MA, Kozyr NL, Earl PL, Smith JM, Ma HL, Grimm BD, Hulsey ML, Miller J, McClure HM, McNicholl JM, Moss B, Robinson HL. 2001. Control of a mucosal challenge and prevention of AIDS by a multiprotein DNA/MVA vaccine. Science 292:69–74 [DOI] [PubMed] [Google Scholar]
  • 8. Hel Z, Tsai WP, Tryniszewska E, Nacsa J, Markham PD, Lewis MG, Pavlakis GN, Felber BK, Tartaglia J, Franchini G. 2006. Improved vaccine protection from simian AIDS by the addition of nonstructural simian immunodeficiency virus genes. J. Immunol. 176:85–96 [DOI] [PubMed] [Google Scholar]
  • 9. Sutter G, Moss B. 1992. Nonreplicating vaccinia vector efficiently expresses recombinant genes. Proc. Natl. Acad. Sci. U. S. A. 89:10847–10851 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Vaccari M, Boasso A, Ma ZM, Cecchinato V, Venzon D, Doster MN, Tsai WP, Shearer GM, Fuchs D, Felber BK, Pavlakis GN, Miller CJ, Franchini G. 2008. CD4+ T-cell loss and delayed expression of modulators of immune responses at mucosal sites of vaccinated macaques following SIV(mac251) infection. Mucosal Immunol. 1:497–507 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Vaccari M, Mattapallil J, Song K, Tsai WP, Hryniewicz A, Venzon D, Zanetti M, Reimann KA, Roederer M, Franchini G. 2008. Reduced protection from simian immunodeficiency virus SIVmac251 infection afforded by memory CD8+ T cells induced by vaccination during CD4+ T-cell deficiency. J. Virol. 82:9629–9638 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Hel Z, Johnson JM, Tryniszewska E, Tsai WP, Harrod R, Fullen J, Tartaglia J, Franchini G. 2002. A novel chimeric Rev, Tat, and Nef (Retanef) antigen as a component of an SIV/HIV vaccine. Vaccine 20:3171–3186 [DOI] [PubMed] [Google Scholar]
  • 13. Vaccari M, Keele BF, Bosinger SE, Doster MN, Zhong-Min Ma Pollara J, Hryniewicz A, Ferrari G, Yongjun G, Forthal DN, Venzon D, Fenizia C, Morgan T, Montefiori DC, Lifson J, Miller C, Silvestri G, Rosati M, Felber BK, Pavlakis G, Tartaglia J, Franchini G. 16 January 2013. Protection afforded by an HIV vaccine candidate in macaques dependent on the dose of SIVmac251 challenge exposure. J. Virol. doi:10.1128/JVI.02863-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Hel Z, Nacsa J, Tryniszewska E, Tsai WP, Parks RW, Montefiori DC, Felber BK, Tartaglia J, Pavlakis GN, Franchini G. 2002. Containment of simian immunodeficiency virus infection in vaccinated macaques: correlation with the magnitude of virus-specific pre- and postchallenge CD4+ and CD8+ T cell responses. J. Immunol. 169:4778–4787 [DOI] [PubMed] [Google Scholar]
  • 15. Manrique M, Micewicz E, Kozlowski PA, Wang SW, Aurora D, Wilson RL, Ghebremichael M, Mazzara G, Montefiori D, Carville A, Mansfield KG, Aldovini A. 2008. DNA-MVA vaccine protection after X4 SHIV challenge in macaques correlates with day-of-challenge antiviral CD4+ cell-mediated immunity levels and postchallenge preservation of CD4+ T cell memory. AIDS Res. Hum. Retroviruses 24:505–519 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Paris RM, Kim JH, Robb ML, Michael NL. 2010. Prime-boost immunization with poxvirus or adenovirus vectors as a strategy to develop a protective vaccine for HIV-1. Expert Rev. Vaccines 9:1055–1069 [DOI] [PubMed] [Google Scholar]
  • 17. Casimiro DR, Wang F, Schleif WA, Liang X, Zhang ZQ, Tobery TW, Davies ME, McDermott AB, O'Connor DH, Fridman A, Bagchi A, Tussey LG, Bett AJ, Finnefrock AC, Fu TM, Tang A, Wilson KA, Chen M, Perry HC, Heidecker GJ, Freed DC, Carella A, Punt KS, Sykes KJ, Huang L, Ausensi VI, Bachinsky M, Sadasivan-Nair U, Watkins DI, Emini EA, Shiver JW. 2005. Attenuation of simian immunodeficiency virus SIVmac239 infection by prophylactic immunization with DNA and recombinant adenoviral vaccine vectors expressing Gag. J. Virol. 79:15547–15555 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Patterson LJ, Kuate S, Daltabuit-Test M, Li Q, Xiao P, McKinnon K, DiPasquale J, Cristillo A, Venzon D, Haase A, Robert-Guroff M. 2012. Replicating adenovirus-simian immunodeficiency virus (SIV) vectors efficiently prime SIV-specific systemic and mucosal immune responses by targeting myeloid dendritic cells and persisting in rectal macrophages, regardless of immunization route. Clin. Vaccine Immunol. 19:629–637 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Patterson LJ, Malkevitch N, Venzon D, Pinczewski J, Gomez-Roman VR, Wang L, Kalyanaraman VS, Markham PD, Robey FA, Robert-Guroff M. 2004. Protection against mucosal simian immunodeficiency virus SIV(mac251) challenge by using replicating adenovirus-SIV multigene vaccine priming and subunit boosting. J. Virol. 78:2212–2221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Van Rompay KK, Abel K, Lawson JR, Singh RP, Schmidt KA, Evans T, Earl P, Harvey D, Franchini G, Tartaglia J, Montefiori D, Hattangadi S, Moss B, Marthas ML. 2005. Attenuated poxvirus-based simian immunodeficiency virus (SIV) vaccines given in infancy partially protect infant and juvenile macaques against repeated oral challenge with virulent SIV. J. Acquir. Immune Defic. Syndr. 38:124–134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Barouch DH. 2010. Novel adenovirus vector-based vaccines for HIV-1. Curr. Opin. HIV AIDS 5:386–390 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Inui S, Sanjo H, Maeda K, Yamamoto H, Miyamoto E, Sakaguchi N. 1998. Ig receptor binding protein 1 (alpha4) is associated with a rapamycin-sensitive signal transduction in lymphocytes through direct binding to the catalytic subunit of protein phosphatase 2A. Blood 92:539–546 [PubMed] [Google Scholar]
  • 23. Romano JW, Williams KG, Shurtliff RN, Ginocchio C, Kaplan M. 1997. NASBA technology: isothermal RNA amplification in qualitative and quantitative diagnostics. Immunol. Invest. 26:15–28 [DOI] [PubMed] [Google Scholar]
  • 24. Lubeck MD, Natuk R, Myagkikh M, Kalyan N, Aldrich K, Sinangil F, Alipanah S, Murthy SC, Chanda PK, Nigida SM, Jr, Markham PD, Zolla-Pazner S, Steimer K, Wade M, Reitz MS, Jr, Arthur LO, Mizutani S, Davis A, Hung PP, Gallo RC, Eichberg J, Robert-Guroff M. 1997. Long-term protection of chimpanzees against high-dose HIV-1 challenge induced by immunization. Nat. Med. 3:651–658 [DOI] [PubMed] [Google Scholar]
  • 25. Bogers WM, Davis D, Baak I, Kan E, Hofman S, Sun Y, Mortier D, Lian Y, Oostermeijer H, Fagrouch Z, Dubbes R, van der Maas M, Mooij P, Koopman G, Verschoor E, Langedijk JP, Zhao J, Brocca-Cofano E, Robert-Guroff M, Srivastava I, Barnett S, Heeney JL. 2008. Systemic neutralizing antibodies induced by long interval mucosally primed systemically boosted immunization correlate with protection from mucosal SHIV challenge. Virology 382:217–225 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Demberg T, Florese RH, Heath MJ, Larsen K, Kalisz I, Kalyanaraman VS, Lee EM, Pal R, Venzon D, Grant R, Patterson LJ, Korioth-Schmitz B, Buzby A, Dombagoda D, Montefiori DC, Letvin NL, Cafaro A, Ensoli B, Robert-Guroff M. 2007. A replication-competent adenovirus-human immunodeficiency virus (Ad-HIV) tat and Ad-HIV env priming/Tat and envelope protein boosting regimen elicits enhanced protective efficacy against simian/human immunodeficiency virus SHIV89.6P challenge in rhesus macaques. J. Virol. 81:3414–3427 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Hidajat R, Xiao P, Zhou Q, Venzon D, Summers LE, Kalyanaraman VS, Montefiori DC, Robert-Guroff M. 2009. Correlation of vaccine-elicited systemic and mucosal nonneutralizing antibody activities with reduced acute viremia following intrarectal simian immunodeficiency virus SIVmac251 challenge of rhesus macaques. J. Virol. 83:791–801 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Petrovas C, Casazza JP, Brenchley JM, Price DA, Gostick E, Adams WC, Precopio ML, Schacker T, Roederer M, Douek DC, Koup RA. 2006. PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection. J. Exp. Med. 203:2281–2292 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Xiao P, Zhao J, Patterson LJ, Brocca-Cofano E, Venzon D, Kozlowski PA, Hidajat R, Demberg T, Robert-Guroff M. 2010. Multiple vaccine-elicited nonneutralizing antienvelope antibody activities contribute to protective efficacy by reducing both acute and chronic viremia following simian/human immunodeficiency virus SHIV89.6P challenge in rhesus macaques. J. Virol. 84:7161–7173 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Xiao P, Patterson LJ, Kuate S, Brocca-Cofano E, Thomas MA, Venzon D, Zhao J, DiPasquale J, Fenizia C, Lee EM, Kalisz I, Kalyanaraman VS, Pal R, Montefiori D, Keele BF, Robert-Guroff M. 2012. Replicating adenovirus-SIV recombinant priming and envelope protein boosting elicits localized, mucosal IgA immunity in rhesus macaques correlated with delayed acquisition following a repeated low-dose rectal SIVmac251 challenge. J. Virol. 86:4644–4657 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Duerr A, Huang Y, Buchbinder S, Coombs RW, Sanchez J, Del RC, Casapia M, Santiago S, Gilbert P, Corey L, Robertson MN. 2012. Extended follow-up confirms early vaccine-enhanced risk of HIV acquisition and demonstrates waning effect over time among participants in a randomized trial of recombinant adenovirus HIV vaccine (Step Study). J. Infect. Dis. 206:258–266 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Rolland M, Tovanabutra S, Decamp AC, Frahm N, Gilbert PB, Sanders-Buell E, Heath L, Magaret CA, Bose M, Bradfield A, O'Sullivan A, Crossler J, Jones T, Nau M, Wong K, Zhao H, Raugi DN, Sorensen S, Stoddard JN, Maust BS, Deng W, Hural J, Dubey S, Michael NL, Shiver J, Corey L, Li F, Self SG, Kim J, Buchbinder S, Casimiro DR, Robertson MN, Duerr A, McElrath MJ, McCutchan FE, Mullins JI. 2011. Genetic impact of vaccination on breakthrough HIV-1 sequences from the STEP trial. Nat. Med. 17:366–371 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. von Gegerfelt AS, Rosati M, Alicea C, Valentin A, Roth P, Bear J, Franchini G, Albert PS, Bischofberger N, Boyer JD, Weiner DB, Markham P, Israel ZR, Eldridge JH, Pavlakis GN, Felber BK. 2007. Long-lasting decrease of viremia in macaques chronically infected with simian immunodeficiency SIVmac251after therapeutic DNA immunization. J. Virol. 81:1972–1979 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Hel Z, Venzon D, Poudyal M, Tsai WP, Giuliani L, Woodward R, Chougnet C, Shearer G, Altman JD, Watkins D, Bischofberger N, Abimiku A, Markham P, Tartaglia J, Franchini G. 2000. Viremia control following antiretroviral treatment and therapeutic immunization during primary SIV251 infection of macaques. Nat. Med. 6:1140–1146 [DOI] [PubMed] [Google Scholar]
  • 35. Zhao J, Lou Y, Pinczewski J, Malkevitch N, Aldrich K, Kalyanaraman VS, Venzon D, Peng B, Patterson LJ, Edghill-Smith Y, Woodward R, Pavlakis GN, Robert-Guroff M. 2003. Boosting of SIV-specific immune responses in rhesus macaques by repeated administration of Ad5hr-SIVenv/rev and Ad5hr-SIVgag recombinants. Vaccine 21:4022–4035 [DOI] [PubMed] [Google Scholar]
  • 36. Patterson LJ, Malkevitch N, Pinczewski J, Venzon D, Lou Y, Peng B, Munch C, Leonard M, Richardson E, Aldrich K, Kalyanaraman VS, Pavlakis GN, Robert-Guroff M. 2003. Potent, persistent induction and modulation of cellular immune responses in rhesus macaques primed with Ad5hr-simian immunodeficiency virus (SIV) env/rev, gag, and/or nef vaccines and boosted with SIV gp120. J. Virol. 77:8607–8620 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Hackstadt AJ, Hess AM. 2009. Filtering for increased power for microarray data analysis. BMC Bioinform. 10:11 doi:10.1186/1471-2105-10-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Gomez-Roman VR, Patterson LJ, Venzon D, Liewehr D, Aldrich K, Florese R, Robert-Guroff M. 2005. Vaccine-elicited antibodies mediate antibody-dependent cellular cytotoxicity correlated with significantly reduced acute viremia in rhesus macaques challenged with SIVmac251. J. Immunol. 174:2185–2189 [DOI] [PubMed] [Google Scholar]
  • 39. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, Horton HF, Fouser L, Carter L, Ling V, Bowman MR, Carreno BM, Collins M, Wood CR, Honjo T. 2000. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 192:1027–1034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, Ahmed R. 2006. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439:682–687 [DOI] [PubMed] [Google Scholar]
  • 41. Blackburn SD, Shin H, Freeman GJ, Wherry EJ. 2008. Selective expansion of a subset of exhausted CD8 T cells by alphaPD-L1 blockade. Proc. Natl. Acad. Sci. U. S. A. 105:15016–15021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Chen KC, Chiou YL, Chang LS. 2009. JNK1/c-Jun and p38 alpha MAPK/ATF-2 pathways are responsible for upregulation of Fas/FasL in human chronic myeloid leukemia K562 cells upon exposure to Taiwan cobra phospholipase A2. J. Cell. Biochem. 108:612–620 [DOI] [PubMed] [Google Scholar]
  • 43. Dong H, Zhu G, Tamada K, Chen L. 1999. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat. Med. 5:1365–1369 [DOI] [PubMed] [Google Scholar]
  • 44. Herr I, Posovszky C, Di Marzio LD, Cifone MG, Boehler T, Debatin KM. 2000. Autoamplification of apoptosis following ligation of CD95-L, TRAIL and TNF-alpha. Oncogene 19:4255–4262 [DOI] [PubMed] [Google Scholar]
  • 45. Callahan MJ, Nagymanyoki Z, Bonome T, Johnson ME, Litkouhi B, Sullivan EH, Hirsch MS, Matulonis UA, Liu J, Birrer MJ, Berkowitz RS, Mok SC. 2008. Increased HLA-DMB expression in the tumor epithelium is associated with increased CTL infiltration and improved prognosis in advanced-stage serous ovarian cancer. Clin. Cancer Res. 14:7667–7673 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Dong G, Wearsch PA, Peaper DR, Cresswell P, Reinisch KM. 2009. Insights into MHC class I peptide loading from the structure of the tapasin-ERp57 thiol oxidoreductase heterodimer. Immunity 30:21–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Riley JL, Westerheide SD, Price JA, Brown JA, Boss JM. 1995. Activation of class II MHC genes requires both the X box region and the class II transactivator (CIITA). Immunity 2:533–543 [DOI] [PubMed] [Google Scholar]
  • 48. Schneider J, Gilbert SC, Hannan CM, Degano P, Prieur E, Sheu EG, Plebanski M, Hill AV. 1999. Induction of CD8+ T cells using heterologous prime-boost immunisation strategies. Immunol. Rev. 170:29–38 [DOI] [PubMed] [Google Scholar]
  • 49. Guerra S, Najera JL, Gonzalez JM, Lopez-Fernandez LA, Climent N, Gatell JM, Gallart T, Esteban M. 2007. Distinct gene expression profiling after infection of immature human monocyte-derived dendritic cells by the attenuated poxvirus vectors MVA and NYVAC. J. Virol. 81:8707–8721 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Liu L, Chavan R, Feinberg MB. 2008. Dendritic cells are preferentially targeted among hematolymphocytes by modified vaccinia virus Ankara and play a key role in the induction of virus-specific T cell responses in vivo. BMC Immunol. 9:15 doi:10.1186/1471-2172-9-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Lore K, Adams WC, Havenga MJ, Precopio ML, Holterman L, Goudsmit J, Koup RA. 2007. Myeloid and plasmacytoid dendritic cells are susceptible to recombinant adenovirus vectors and stimulate polyfunctional memory T cell responses. J. Immunol. 179:1721–1729 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Cheng C, Gall JG, Kong WP, Sheets RL, Gomez PL, King CR, Nabel GJ. 2007. Mechanism of ad5 vaccine immunity and toxicity: fiber shaft targeting of dendritic cells. PLoS Pathog. 3:e25 doi:10.1371/journal.ppat.0030025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Gilbert SC, Schneider J, Hannan CM, Hu JT, Plebanski M, Sinden R, Hill AV. 2002. Enhanced CD8 T cell immunogenicity and protective efficacy in a mouse malaria model using a recombinant adenoviral vaccine in heterologous prime-boost immunisation regimes. Vaccine 20:1039–1045 [DOI] [PubMed] [Google Scholar]
  • 54. Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, Mackey EW, Miller JD, Leslie AJ, Depierres C, Mncube Z, Duraiswamy J, Zhu B, Eichbaum Q, Altfeld M, Wherry EJ, Coovadia HM, Goulder PJ, Klenerman P, Ahmed R, Freeman GJ, Walker BD. 2006. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 443:350–354 [DOI] [PubMed] [Google Scholar]
  • 55. Petrovas C, Price DA, Mattapallil J, Ambrozak DR, Geldmacher C, Cecchinato V, Vaccari M, Tryniszewska E, Gostick E, Roederer M, Douek DC, Morgan SH, Davis SJ, Franchini G, Koup RA. 2007. SIV-specific CD8+ T cells express high levels of PD1 and cytokines but have impaired proliferative capacity in acute and chronic SIVmac251 infection. Blood 110:928–936 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Song MY, Park SH, Nam HJ, Choi DH, Sung YC. 2011. Enhancement of vaccine-induced primary and memory CD8(+) T-cell responses by soluble PD-1. J. Immunother. 34:297–306 [DOI] [PubMed] [Google Scholar]
  • 57. Trautmann L, Janbazian L, Chomont N, Said EA, Wang G, Gimmig S, Bessette B, Boulassel MR, Delwart E, Sepulveda H, Balderas RS, Routy JP, Haddad EK, Sekaly RP. 2006. Upregulation of PD-1 expression on HIV-specific CD8 + T cells leads to reversible immune dysfunction. Nat. Med. 12:1198–1202 [DOI] [PubMed] [Google Scholar]
  • 58. Cantley LC, Neel BG. 1999. New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway. Proc. Natl. Acad. Sci. U. S. A. 96:4240–4245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Koul D, Takada Y, Shen R, Aggarwal BB, Yung WK. 2006. PTEN enhances TNF-induced apoptosis through modulation of nuclear factor-kappaB signaling pathway in human glioma cells. Biochem. Biophys. Res. Commun. 350:463–471 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Hansen SG, Ford JC, Lewis MS, Ventura AB, Hughes CM, Coyne-Johnson L, Whizin N, Oswald K, Shoemaker R, Swanson T, Legasse AW, Chiuchiolo MJ, Parks CL, Axthelm MK, Nelson JA, Jarvis MA, Piatak M, Jr, Lifson JD, Picker LJ. 2011. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature 473:523–527 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Vaccari M, Trindade CJ, Venzon D, Zanetti M, Franchini G. 2005. Vaccine-induced CD8+ central memory T cells in protection from simian AIDS. J. Immunol. 175:3502–3507 [DOI] [PubMed] [Google Scholar]
  • 62. van Grevenynghe J, Procopio FA, He Z, Chomont N, Riou C, Zhang Y, Gimmig S, Boucher G, Wilkinson P, Shi Y, Yassine-Diab B, Said EA, Trautmann L, El Far M, Balderas RS, Boulassel MR, Routy JP, Haddad EK, Sekaly RP. 2008. Transcription factor FOXO3a controls the persistence of memory CD4(+) T cells during HIV infection. Nat. Med. 14:266–274 [DOI] [PubMed] [Google Scholar]
  • 63. Nimmerjahn F, Ravetch JV. 2007. Fc-receptors as regulators of immunity. Adv. Immunol. 96:179–204 [DOI] [PubMed] [Google Scholar]
  • 64. Qiao SW, Kobayashi K, Johansen FE, Sollid LM, Andersen JT, Milford E, Roopenian DC, Lencer WI, Blumberg RS. 2008. Dependence of antibody-mediated presentation of antigen on FcRn. Proc. Natl. Acad. Sci. U. S. A. 105:9337–9342 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Pillai VK, Kannanganat S, Penaloza-Macmaster P, Chennareddi L, Robinson HL, Blackwell J, Amara RR. 2011. Different patterns of expansion, contraction and memory differentiation of HIV-1 Gag-specific CD8 T cells elicited by adenovirus type 5 and modified vaccinia Ankara vaccines. Vaccine 29:5399–5406 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Barouch DH, Liu J, Li H, Maxfield LF, Abbink P, Lynch DM, Iampietro MJ, SanMiguel A, Seaman MS, Ferrari G, Forthal DN, Ourmanov I, Hirsch VM, Carville A, Mansfield KG, Stablein D, Pau MG, Schuitemaker H, Sadoff JC, Billings EA, Rao M, Robb ML, Kim JH, Marovich MA, Goudsmit J, Michael NL. 2012. Vaccine protection against acquisition of neutralization-resistant SIV challenges in rhesus monkeys. Nature 482:89–93 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental material

Articles from Journal of Virology are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES