Received 18 April 2006/Accepted 9 July 2006

Size: px
Start display at page:

Download "Received 18 April 2006/Accepted 9 July 2006"

Transcription

1 JOURNAL OF VIROLOGY, Oct. 2006, p Vol. 80, No X/06/$ doi: /jvi Copyright 2006, American Society for Microbiology. All Rights Reserved. Induction of Human Immunodeficiency Virus Type 1 (HIV-1)-Specific T-Cell Responses in HIV Vaccine Trial Participants Who Subsequently Acquire HIV-1 Infection Helen Horton, 1,2 Colin Havenar-Daughton, 3 Deborah Lee, 1 Erin Moore, 1 Jianhong Cao, 1 John McNevin, 1 Thomas Andrus, 3 Haiying Zhu, 3 Abbe Rubin, 4 Tuofu Zhu, 3 Connie Celum, 2 and M. Juliana McElrath 1,2,3 * Program in Infectious Diseases 1 and Statistical Center for HIV/AIDS Research and Prevention, 4 Fred Hutchinson Cancer Research Center, Seattle, Washington, and Departments of Medicine 2 and Laboratory Medicine, 3 University of Washington School of Medicine, Seattle, Washington Received 18 April 2006/Accepted 9 July 2006 Candidate human immunodeficiency virus type 1 (HIV-1) vaccines designed to elicit T-cell immunity in HIV-1-uninfected persons are under investigation in phase I to III clinical trials. Little is known about how these vaccines impact the immunologic response postinfection in persons who break through despite vaccination. Here, we describe the first comprehensive characterization of HIV-specific T-cell immunity in vaccine study participants following breakthrough HIV-1 infection in comparison to 16 nonvaccinated subjects with primary HIV-1 infection. Whereas none of the 16 breakthrough infections possessed vaccine-induced HIV-1- specific T-cell responses preinfection, 85% of vaccinees and 86% of nonvaccinees with primary HIV-1 infection developed HIV-specific T-cell responses postinfection. Breakthrough subjects T cells recognized 43 unique HIV-1 T-cell epitopes, of which 8 are newly described, and 25% were present in the vaccine. The frequencies of gamma interferon (IFN- )-secreting cells recognizing epitopes within gene products that were and were not encoded by the vaccine were not different (P 0.64), which suggests that responses were not anamnestic. Epitopes within Nef and Gag proteins were the most commonly recognized in both vaccinated and nonvaccinated infected subjects. One individual controlled viral replication without antiretroviral therapy and, notably, mounted a novel HIV-specific HLA-C14-restricted Gag LYNTVATL-specific T-cell response. Longitudinally, HIV-specific T cells in this individual were able to secrete IFN- and tumor necrosis factor alpha, as well as proliferate and degranulate in response to their cognate antigenic peptides up to 5 years postinfection. In conclusion, a vaccinee s ability to mount an HIV-specific T-cell response postinfection is not compromised by previous immunization, since the CD8 T-cell responses postinfection are similar to those seen in vaccinenaïve individuals. Finding an individual who is controlling infection highlights the importance of comprehensive studies of breakthrough infections in vaccine trials to determine whether host genetics/immune responses and/or viral characteristics are responsible for controlling viral replication. The human immunodeficiency virus (HIV) pandemic is of staggering proportions, with nearly 40 million individuals infected (1). This has created an urgent need to develop a safe, efficacious vaccine as the single prevention strategy to benefit the largest number of persons. In the absence of established correlates of protection, most experts predict that both humoral and cellular immunity will be required for sustained immunity against HIV type 1 (HIV-1). Successful vaccines against other viral infections rely on induction of protective levels of antiviral antibodies (10). Likewise, the first generation of HIV vaccine candidates, notably, recombinant monomeric envelope protein subunit vaccines, was designed to elicit antibody responses. Studies of individuals who participated in these trials but subsequently became infected with HIV (breakthrough infections), therefore, have focused on characterization of their HIV-specific neutralizing antibody responses prior to infection and on the ability of these antibodies * Corresponding author. Mailing address: Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N D3-100, Seattle, WA Phone: (206) Fax: (206) jmcelrat@fhcrc.org. to neutralize viruses infecting vaccine recipients (5, 12, 17, 23). Unfortunately, these studies revealed that nearly all vaccineinduced antibodies by this approach lack potency and breadth of reactivity against transmitting strains (discussed in reference 8) and ultimately fail to provide HIV protection. While new strategies for the design of vaccines which induce effective anti-hiv-l antibodies are still being identified, the emphasis has shifted to the design of vaccines that will induce antiviral CD8 T-cell immunity. Although HIV-specific CD8 T cells alone are unlikely to prevent HIV infection, they may have an important impact in rapidly controlling viral replication following infection. A large effort has centered upon the development of recombinant attenuated poxviruses as a safe approach in humans to induce HIV-1-specific T-cell immunity. Recombinant canarypox HIV vaccines, the most extensively tested to date, induce CD8 T-cell responses at various frequencies (9 to 70%) depending upon the construct tested and the method of detection (4, 11, 15, 18). Here we investigate the characteristics of the T cells present during early infection in individuals previously enrolled in HIV vaccine trials evaluating recombinant canarypox candidate vaccines, contrasting their responses with those of nonvaccinated 9779

2 9780 HORTON ET AL. J. VIROL. subjects who have recently acquired HIV-1 infection. The vaccine constructs contained full-length gag (HIV-1 LAI) and env gp120 (HIV-1 MN) coding regions, with some constructs also containing parts of pol and nef. Several individuals also received a recombinant gp120 SF-2 protein boost (Chiron) or a recombinant gp120 MN protein boost (Vaxgen Inc.). Vaccinees with breakthrough infections described in this report took part in phase I/II HIV vaccine trials where the net cumulative cytotoxic T-lymphocyte (CTL) response rates were between 33 and 50%, as assessed by the chromium release assay (4, 18). We determined whether any of the T-cell responses detected during early infection in these individuals represented anamnestic responses induced by immunization prior to infection. Furthermore, we characterized longitudinally the function of HIV-specific T cells postinfection in the only volunteer controlling infection in the absence of antiretroviral therapy (ART). These are the first comprehensive data describing HIV-specific T-cell responses pre- and postinfection in participants who became HIV-infected in HIV vaccine trials of vaccine candidates designed to elicit CD8 T-cell immunity. MATERIALS AND METHODS Study population. Volunteers enrolled in the National Institutes of Health (NIH)-sponsored HIV Vaccine Trials Network protocol 402 were evaluated in this study. Each individual had participated in phase I or II trials examining safety and immunogenicity of recombinant canarypox vectors (Sanofi Pasteur) with or without a recombinant gp120 protein vaccine boost (Vaxgen Inc., Chiron Corp.). Results from these studies and the description of the candidate vaccines have been previously published (3, 4, 11, 18). Each study participant became infected as a direct result of high-risk behavior. The institutional review boards at each site approved the trials, and each subject gave informed consent for participation in the studies. The time of infection was estimated using an algorithm based on the participant s last negative and first positive HIV-1 Western blot assay or plasma RNA reverse transcriptase PCR, acute retroviral symptoms, and exposure history (17). When participants did not report exposure or seroconversion symptoms, the date was assigned as the mid-point between the last negative and first positive serological or DNA test. Plasma HIV-1 RNA was determined by quantitative reverse transcription-pcr assay (Roche Molecular Systems, Branchburg, N.J.) (14), which has a sensitivity of 50 copies/ml. Peripheral blood CD4 and CD8 T-cell counts were determined by flow cytometry with consensus methods (14) and expressed as cells per microliter. HLA typing was performed at the Puget Sound Blood Center by sequence-specific primer PCR as previously described (7). The comparison population consisted of 16 patients with newly diagnosed HIV-1 infection who were followed longitudinally at the University of Washington Primary Infection Clinic (9). Primary infection was documented by signs and symptoms consistent with an acute retroviral syndrome (26, 27). The duration of infection in these subjects was designated from the date of onset of acute symptoms. Peptides. Peptides for the gamma interferon (IFN- ) ELISpot were either synthesized by Fred Hutchinson Cancer Research Center Shared Resources Facility (Seattle, WA) or supplied by the NIH AIDS Research and Reference Reagent Program (Bethesda, MD). Peptides were 15 amino acids in length, overlapping by 11 amino acids, and were based on the proteins encoded by the HxB2 clade B HIV sequence, except for Env (based on MN) and Nef (based on BRU). The IFN- ELISpot assays were performed by stimulating peripheral blood mononuclear cells (PBMC) with pools of 50 peptides (each peptide at a final concentration of 1 g/ml). Experiments were performed to define the 15-mer recognized within a peptide pool using peptide matrices and, whenever possible, the 8- to 11-mer optimal peptide as previously described (25). IFN- ELISpot. The ELISpot assay methodology has been described in detail elsewhere (25). In brief, cryopreserved PBMC were thawed, washed, and rested overnight in R10 (RPMI 1640 [GibcoBRL, New York] containing 10% fetal calf serum [Gemini Bioproducts, California], 2 mm L-glutamine [GibcoBRL], 100 U/ml penicillin G, 100 g/ml streptomycin sulfate) before use in ELISpot assays. Cells were resuspended in R10 and aliquoted at to cells/well. Peptide antigens were added at a final concentration of 1 to 2 g/ml, R10 alone provided a negative control, and 1 g/ml phytohemagglutinin (PHA-P) served as a positive control. Due to limited numbers of cells, experimental and control testing was performed in duplicate only. Spots were counted using the CTL Analyzer and software (Cellular Technology Ltd., Cleveland, OH). For functional avidity measurements, a standard IFN- ELISpot was performed with the indicated peptides at twofold dilutions ranging from 2 g/ml to g/ml. The regression curve was drawn with the sigmoidal fit tool in the Origin 6.0 software. CFSE labeling and polychromatic flow cytometry. PBMC were labeled with 1.2 M carboxyl fluorescein succinimidyl ester (CFSE; Molecular Probes, Inc., Eugene, Oreg.) for 8 min in the dark at 37 C, and free CFSE was quenched with 100% 4 C fetal calf serum for 1 min. Labeled cells were washed before in vitro culture with 2 g/ml of HIV-1 peptides for 5 days. As a positive control, the cells were stimulated with 30 ng/ml anti-cd3 and 1 g/ml anti-cd28 monoclonal antibodies. On day 5 following CFSE labeling, cells were washed in R10 and rested overnight. The next day cells were washed and stimulated for 6 h in the presence of brefeldin A (10 g/ml) with either staphylococcal enterotoxin B (1 g/ml; Sigma) as a positive control, HIV-1 peptides (1 g/ml each peptide/ sample), or no peptide as a negative control. CD107a-PECy5 was added during stimulation. Intracellular staining was performed with standard techniques (BD CFC protocol), using previously titrated antibody reagents. GolgiStop (BD) was used in addition to brefeldin A in panels with CD107a. CD3-allophycocyanin-Cy7, CD8-peridinin chlorophyll a-cy5.5, IFN- -phycoerythrin-cy7, interleukin-2 (IL-2) phycoerythrin, tumor necrosis factor alpha (TNF- ) allophycocyanin, and CD107a-phycoerythrin-Cy5 monoclonal antibodies were supplied by BD. Data acquisition was performed on an LSRII flow cytometer (BD), collecting 100,000 to 200,000 lymphocyte-gated or 10,000 to 40,000 CD8 events per sample, and analyzed with Flowjo software (Tree Star Inc., Ashland, OR). Positive responses for the ex vivo cytokine secretion assay were designated when the percentage of bright cytokine-positive CD8 T cells was twice that of the negative control and at least 0.05% (background subtracted). Statistical analysis. The two-sided exact Wilcoxon rank-sum test was used to assess differences in the breadth and magnitude of HIV-1-specific T-cell responses between vaccine-primed and vaccine-naïve individuals during early infection. All statistical computations were performed using Jmp (version 5.0; SAS Institute) or SAS (version 8.2; SAS Institute) software. RESULTS Study participants. Sixteen HIV-1 vaccine trial participants between the ages of 22 and 43 years (median, 32 years) were diagnosed with HIV-1 infection after study enrollment; 13 were vaccine recipients, and 3 were placebo recipients (Table 1). The vaccinees received a recombinant canarypox candidate vaccine, with or without a recombinant gp120 protein boost. Ten of the 13 completed their vaccination series (a total of four to five immunizations) before acquiring HIV-1 infection between November 1996 and May 2002 as a result of high-risk behavior. The predominant risk factor for HIV-1 acquisition was men having high-risk sexual contact with men (MSM). Two participants were women reporting low-risk sexual activities at enrollment of the vaccine trial. The median length of time between last vaccination and estimated time of infection was 249 days (Table 1; see Materials and Methods for the algorithm used to estimate time of infection). Median plasma HIV-1 RNA and CD4 counts in these individuals after HIV-1 infection (sampled on assay date) (Table 2) were not statistically significant between vaccinees (4.78 log 10 copies/ml; 552 cells/mm 3 ) and placebo recipients (4.27 log 10 copies/ml; 657 cells/mm 3 )(P 0.4 and 0.1, respectively; Wilcoxon rank sum test). A recent publication provides further details of the subjects clinical courses (20). Characterization of HIV-specific T-cell responses early in infection in previous vaccine recipients. HIV-1-specific T-cell responses were measured by IFN- ELISpot as close to the estimated date of infection as possible. Of note, volunteers were typically followed every 3 to 6 months after completion of

3 VOL. 80, 2006 T-CELL RESPONSES IN HIV VACCINE TRIAL PARTICIPANTS 9781 TABLE 1. Demographic profile and treatment of study population Participant Treatment Immunogen a prior to infection No. of vaccinations date immunizations according to the study protocol, which did not permit us to necessarily capture events during acute infection. HIV-specific T cells were defined as recognizing 15-mers, unless there were sufficient cells to define the optimal 8- to 11-mer epitopic response. All assays were performed on cryopreserved PBMC isolated at times when the volunteers were not receiving ART, with the exception of 402FIE and 014A Mean viability was 85% (median, 87%; range, 64 to 100%), and mean PHA response was 9,477 spot-forming cells (SFC)/10 6 PBMC (median, 9,940 SFC/10 6 PBMC; range, 587 SFC/10 6 PBMC to too numerous to count). The peptides recognized by PBMC from each individual are summarized in Table 2, and their positions throughout the encoded gene products are shown in Fig. 1A. We detected postinfection HIV-specific T-cell responses in all but two participants by IFN- ELISpot. One of these individuals (014A-011) was receiving ART and had an undetectable viral load, which may account for the lack of responses detected in this individual. PBMC from the other individual were viable (88% viability), but the PHA response was weak (587 SFC/10 6 PBMC), which suggests that there were fewer T cells than would be expected. The number of HIV peptides recognized by IFN- -secreting CD8 T cells ranged from 1 to 5 (median, 4) among the placebos and from 0 to 9 (median, 3) among the vaccine recipients. Of the 11 vaccinees who mounted detectable T-cell responses, 9 possessed T cells that recognized HIV-1 epitopes that were present in the vaccines they received, but 10 of these individuals also possessed HIV- 1-specific T cells recognizing epitopes that were not present in the vaccines (Table 2). The magnitude of the HIV-specific T-cell responses in the vaccinees ranged from 70 to 5,310 SFC/10 6 PBMC and was not correlated with plasma HIV-1 Interval between last treatment and HIV Gender Race acquisition (days) b Age c (yr) Risk factor d 014A-003 Placebo ALVAC/saline 4 67 Male Other 30 MSM 014A-006 Placebo ALVAC/saline 3 e 34 Male White 25 MSM 402FIE Placebo ALVAC/saline Male White 31 MSM 014A-001 Vaccine vcp205/gp Female Other 33 Low risk 014A-004 Vaccine vcp Male White 35 MSM 014A-005 Vaccine vcp205/gp Male White 43 MSM 014A-008 Vaccine vcp Male Other 41 IDU 014A-009 Vaccine vcp Male White 25 MSM 014A-011 Vaccine vcp205 3 e 13 Male White 42 MSM 40287N Vaccine vcp125/gp ,176 Female Black 22 Low risk 402I03 Vaccine vcp65/gp Male White 32 MSM 402I15 Vaccine vcp205/gp Male White 34 MSM Vaccine vcp205/gp ,589 Male White 38 MSM 2x0226 Vaccine vcp205/gp Male Other 31 MSM Vaccine vcp Male White 27 MSM Vaccine vcp1452/gp120 1 e 14 Male Black 27 Bisexual a Recombinant canarypox vectors are as follows: vcp125 expresses HIV-1 MN gp160 (Pasteur Merieux/Connaught); vcp65 expresses rabies virus glycoprotein (Pasteur Merieux/Connaught); vcp205 expresses HIV-1 envelope gp120 (strain MN) linked to the transmembrane portion of HIV-1 gp41 (strain LAI) and the HIV-1 LAI genes encoding the entire Gag protein and a portion of the Pol sequence, sufficient to evoke the protease portion (Pasteur Merieux/Connaught); vcp1452 expresses HIV-1 envelope gp120 (strain MN) linked to the transmembrane portion of HIV-1 gp41 (strain LAI), the HIV-1 LAI gene encoding the entire Gag protein, a portion of the pol gene encoding the protease, and a synthetic polynucleotide encompassing several known human CTL epitopes from the nef and pol gene products. It also contains sequences encoding the E3L and K3L vaccinia virus proteins into the C6 site (vcp1452) (Virogenetics Corp., Troy, NY, and Sanofi Pasteur, S.A., Marcy L Etoile, France). b See Materials and Methods for the algorithm used for estimating time of HIV-1 infection. c Age at enrollment of parent vaccine protocol. d MSM, men having sex with men; IDU, injection drug user. All volunteers reported HIV high-risk behavior prior to enrollment, except for those designated. e Participant did not complete vaccination regimen. RNA viral load (r 0.357, P 0.26) measured at the same time point. It was not possible to assess the correlation of T-cell response magnitude and viral load in the placebo group due to the small sample size (n 3). Forty-four unique HIV peptides were recognized in the 16 participants (13 vaccine and 3 placebo recipients), and of these, 8 have not been described previously (shown in Table 2). CD8 (not CD4 ) T cells mediated all responses when subsets were examined by IFN- intracellular staining using flow cytometry (data not shown). Nef and Gag proteins were the most commonly recognized among HIV-1 proteins by T cells in these individuals (Fig. 1A). Six of 16 individuals recognized at least one Nef epitope and 6 unique Nef epitopes were identified, whereas 11 of 16 individuals recognized at least 1 Gag epitope, with 15 unique epitopes identified. The frequency of IFN- -secreting cells was not greater for epitopes within the vaccine than for those outside the vaccine (P 0.64; Wilcoxon rank sum test). There was insufficient power to determine whether any single epitope or HLA allele was overrepresented in this cohort (see Table 2 for epitopes and HLA typing). Comparison of early T-cell responses detected in vaccinees to those in vaccine-naïve individuals. HIV-specific T-cell responses identified in the 13 vaccinated individuals were compared to responses seen during early infection in 16 study participants enrolled in a longitudinal primary infection cohort (9) who had never received an HIV-1 vaccine. These individuals were selected based on availability of cryopreserved PBMC from leukapheresis performed during primary infection. They were comparable in age, ethnicity, gender, risk behavior, and date of infection to the individuals in our study (data not shown). Of the 35 unique peptides identified in the vaccine recipients (compared to the acutely infected cohort),

4 9782 HORTON ET AL. J. VIROL. TABLE 2. HIV-1 epitopes recognized by study participant CD8 T cells before and after HIV-1 infection Participant identifier Vaccine treatment Class I HLA type Days postinfection Viral load RNA (copies/ml) HIV-1 epitope i IFN- SFC/ 10 6 PBMC 014A-003 Placebo A*0101, A*2902; B*1501, B* ,525 Gag TSTLQEQIGW A-006 Placebo A*0101; B*0801, B* ,749,235 Env IVELLGRRGWEVLKY 20 1,790 Env RQGLERALL 20 2,835 Env ERYLKDQQL 20 2,350 Nef FLKEKGGL Nef AAVDLSHFL 20 2, FIE a Placebo A3, A32; B38, B ,069 Gag RLRPGGKKK Gag VDRFYKTLRAEQASQ Env MHEDIISLW Vpr GLGQHIYETYGDTWA e A-001 vcp205/gp120 A*0202, A*8001; B*1801, B* NA b Gag QAISPRTLNAW Env RIRQGLERA Nef YPLTFGWCY A-004 vcp205 A*0201, A*1101; B*4002, B* Pol TAFTIPSI Gag CTERQANFL Gag SLYNTVATL h Nef AVDLSHFLK A-005 vcp205/gp120 A*2501, A*3002; B*0702, B* ,775 Gag QAISPRTLNAW Gag ETINEEAAEW Env TENFNMWKNNMVEQM Env IPRRIRQGL Nef FPVTPQVPLR Tat CCFHCQVC Pol QKQGQGQWTYQIYQE Pol KIQNFRVYY Vpr EAVRHFPRIWLHGLG A-008 vcp205 A*0201, A*0301; B*4501, B* ,483 Gag RLRPGGKKK A-009 vcp205 A1, A2; B8, B ,413 Gag EIYKRWII Env MHEDIISLW Env RRGWEVLKY Env RQGLERALL Nef IHSQRRQDILDLWIY A-011 vcp205 A*0201; B*31012, B* c None 40287N vcp125/gp120 A1, A33; B8, B ,392 Gag GGKKKYKL Gag EIYKRWII Vpr EAVRHFPRIWLHGLG Vpu EYRKILRQRKIDRLI I03 f vcp65/gp120 A3, A33; B15 (63), B ,109 Gag IRLRPGGKK 20 3,960 Gag KRWIILGLNK 20 5,310 Gag GLNKIVRMY Gag MMQRGNFRNQRKIVK e 20 2,510 Env SFNCGGEFF Env LQRAGRAILHIPTRI I15 vcp205 A1, A10 (26); B8, B17 (57) 92 41,742 d Gag QMVHQAISPRTLNAW e Rev DEELIRTVRLIKLLY e Nef FLKEKGGL vcp205/gp160 A2, A2; B62, B ,069 Gag PPIPVGDIY Vif DAKLVITTY x0226 vcp205/gp120 NA b 30 NA b Vif DAKLVITTY 20 2,943 Env PIPIHYCAPAGFAIL vcp1452 A*2501, A*6601; B*0702, B* ,509 d None g vcp1452/gp120 A*3001, A*3002; B*4201/02, B*4403/26/30 Before After 91 32,511 d Pol YPGIKVRQL ND j 490 Pol KIQNFRVYY 365 Pol DDTVLEEMSLPGRWK 215 a Some responses mapped in this individual at 143 dpi (viral load of 400 and participant on ART). b NA, data not available. c On ART at time of mapping. d No viral load was obtained on the visit date of sample collection. Thus, the closest viral load reported for 402I15 was 3.3 months post-sample date; for , it was 0.4 months post-sample date; for , it was 0.7 months post-sample date. e By matrix only. The individual peptide was not tested due to insufficient cells. f Responses to Vif and Vpr peptide pools were not tested in this individual. Pol pools 9 and 10 were also positive, but an exact peptide(s) was not identified due to insufficient cells. g Only a weak PHA response (587 SFC/10 6 PBMC) was detected at this time point. h See later figures and Discussion. i The vaccine contained the epitopes shown in bold. j ND, not determined.

5 VOL. 80, 2006 T-CELL RESPONSES IN HIV VACCINE TRIAL PARTICIPANTS 9783 Downloaded from FIG. 1. A. Peptide sequences and amino acid locations within each protein of HIV peptides recognized by CD8 T cells during early infection in individuals who have participated in previous canarypox trials of potential HIV vaccines. B. Comparison of epitopes recognized by HIV-specific T cells early in infection in vaccine-naïve individuals from the primary infection cohort (blue bars) with HIV peptides recognized by T cells from vaccinated individuals (red bars) or placebos (green bars). Open boxes indicate newly defined epitopes. 12 corresponded to epitopes identified in the primary infection group (9), 16 were epitopes previously described in chronic infection (1, 16), and 7 are newly described (Fig. 1B). Only two of the seven newly described epitopes were contained in the vaccines that the subjects received. Between the two populations of relatively small sample size, no statistical differences were found in the number of epitopes (P 0.13) or in the number of HIV-1 proteins (P 0.18) recognized by T cells. Similarly, no significant difference was found between the frequencies of cells expressing IFN- in vaccinees versus placebo recipients (P 0.13; Wilcoxon rank sum test), suggesting that the responses observed in vaccinees were unlikely to be anamnestic. Characterization of preseroconversion HIV-specific T-cell responses. Cryopreserved PBMC were assayed preinfection, at the closest time before infection as possible, to determine whether any of the responses seen in early infection were anamnestic responses induced by immunization prior to infection. Mean viability was 87% (median, 87%; range, 72 to 97%), and mean PHA response was 4,548 SFC/10 6 PBMC (median, 1,942 SFC/10 6 PBMC; range, 245 to 19,585 SFC/10 6 PBMC). Only one individual (014A-004) possessed measurable preseroconversion HIV-1-specific T-cell responses after four immunizations with vcp205 (Table 2). Interestingly, this individual never received ART and is the only subject among these vaccine trial study participants with breakthrough infection who is controlling viral replication in the absence of ART (Fig. 2A). Three of the four HIV-specific responses identified in this individual postseroconversion were present before seroconversion, albeit at very low frequencies (below 150 SFC/10 6 PBMC). These T-cell responses were boosted after seroconversion (Fig. 2B), suggesting that they could represent memory responses that were induced upon immunization. However, a Nef AVDLSHFLK-specific T-cell response was detected prior on July 3, 2018 by guest

6 9784 HORTON ET AL. J. VIROL. FIG. 2. A. Longitudinal plasma viral load (RNA copies/ml) and CD4 counts (cells/ l) in individual B. HIV-specific T-cell responses, measured by IFN- ELISpot, before and after seroconversion in to seroconversion, despite the fact that Nef was not present in the vaccine the subject received. T cells specific for this epitope were restricted by HLA-A*11, and their presence preseroconversion was confirmed by major histocompatibility complex (MHC)-peptide tetramer binding (0.51% of CD3 CD8 T cells [data not shown]). We looked for the presence of proviral DNA at the last immunization date to determine whether this individual was already infected when the HIV-specific T cells were first detected. Using limiting-dilution PCR as previously described (29), we confirmed that this individual was indeed infected at a very low level ( copies per million PBMC), despite being HIV-1 seronegative by Western blotting and negative by standard plasma viral RNA assays. No provirus was detected using this method 8 months prior to the last immunization date, although the number of cells that were available for assay was limited (T. Zhu et al., unpublished data). IFN- -secreting T cells specific for the well-characterized HLA-A2-restricted SLYNTVATL (SL9) peptide were detected in volunteer 014A-004 by the IFN- ELISpot assay (Table 2). PBMC from this individual obtained at postseroconversion time points, however, failed to bind the HLA-A2/ SL9 peptide tetramer. Of note, T cells from these same time points had substantial reactivity to the SL9 peptide by IFN- ELISpot (Fig. 2B and data not shown). Further characterization of the optimal epitope recognized by these T cells revealed it to be an 8-mer, LYNTVATL (LL8) (Fig. 3A), which is a single amino acid short of the A2-restricted SL9 epitope. The newly defined LL8 epitope was restricted by HLA-C14 (Fig. FIG. 3. A. Characterization of the optimal epitope, LL8, recognized by T cells from B. Identification of HLA-C14 as the restricting MHC molecule for LL8-specific T cells in C. Peptide titrations of the Gag-LL8 epitope. Standard IFN- ELISpot was performed with the indicated peptides at twofold dilutions ranging from 2 g/ml to g/ml. The regression curve was drawn with the sigmoidal fit tool in the Origin 6.0 software. Data shown are background subtracted. 3B), not previously shown to restrict HIV-1 epitopes, and T cells recognizing this epitope were of high avidity (50% effective concentration, 0.26 nm) (Fig. 3C). In this subject who was controlling HIV-1 infection, we examined additional antiviral functions of the epitope-specific CD8 T cells defined at day 46 postinfection and at three additional time points over 5 years after infection (Fig. 4). Similar quantities of CD8 T cells secreted TNF- and IFN- when pulsed with their cognate peptide (Gag p17 LYNTV ATL, Gag p2p7p1p6 CTERQANFL, RT TAFTIPSI, and Nef AVDLSHFLK), whereas IL-2-secreting cells were either absent or just at the threshold level of detection by the ex vivo

7 VOL. 80, 2006 T-CELL RESPONSES IN HIV VACCINE TRIAL PARTICIPANTS 9785 FIG. 4. A. Gating scheme for ex vivo intracellular cytokine staining assay. B. Representative example of ex vivo intracellular cytokine and CD107a expression data. PBMC were stimulated for 6 h with no antigen, LL8 peptide, or staphylococcal enterotoxin B (SEB). Data are gated on CD3 CD8 live lymphocytes. C. Postinfection cytokine secretion and degranulation (as measured by CD107a expression) profiles of epitopespecific T-cell responses assessed longitudinally in Background-subtracted percent secretion of IFN-, TNF-, and IL-2 and percent expression of CD107a in CD8 T cells are shown for each epitope at 409, 751, and 1,838 dpi. Only CD8 T-cell responses that were 2 times the background and at least 0.05% are reported. D. Representative example of CFSE proliferation data. Each row shows data from cultures stimulated for 5 days with no antigen or with either LL8 peptide or anti-cd3/anti-cd28 monoclonal antibody. Cultures were split into three aliquots and restimulated for 6 h with no antigen, LL8 peptide, or SEB before staining as for the ex vivo intracellular cytokine staining assay. Data are gated on CD3 CD8 live lymphocytes. E. Postinfection proliferative ability (measured as percentage of CD8 T cells that are CFSE lo ) of epitopespecific T cells measured longitudinally in The percentage of proliferating cells that secrete IFN-, TNF-, or IL-2 and the percentage that express CD107a in response to cognate peptide are also shown.

8 9786 HORTON ET AL. J. VIROL. assay over the course of infection. At all time points, the majority of this subject s HIV-specific T cells were able to degranulate as measured by CD107a expression and proliferate as measured by CFSE lo staining (Fig. 4E). Of note, ex vivo IL-2 expression of epitope-specific CD8 T cells did not correlate with their ability to proliferate over 5 days of peptide stimulation. Most notably, the relative magnitudes of IFN- secreting CD8 T cells did change over time, with the numbers of those recognizing LL8, AK9, and TI8 diminishing while the CL9-specific cells increased, as observed prior to seroconversion. DISCUSSION The new generations of candidate HIV vaccines currently being evaluated in clinical trials aim primarily to induce HIVspecific CD8 T-cell-mediated immunity. Specific T cells can only recognize HIV once the virus has infected cells or through cross-presentation (22). These vaccines are, therefore, unlikely to prevent infection itself but may aid in the efficient elimination of virus-infected cells, thus empowering the individual to control the virus. Although a recent study detailed responses from a single vaccinated individual pre- and postinfection (6), this study details the first thorough evaluation of pre- and postinfection HIV-specific T-cell responses in 16 participants in phase I/II canarypox prime-boost vaccine trials, 13 of whom received active immunogen. We found no significant evidence of anamnestic responses to the vaccine epitopes versus the virus with which they were infected; however, since these vaccines have low immunogenicity and therefore have not moved forward into HIV Vaccine Trials Network (HVTN) phase IIb trials, this is perhaps not surprising. All of the individuals who received active immunogen were nonresponsive to the immunization regimen by our detection system, and their inability to mount a vaccine response may explain why no anamnestic response was observed. We identified one participant who controlled his infection without antiretroviral therapy for 5 years. This control could be related to his HLA type, and this finding highlights the importance of comprehensive study of breakthrough infections in vaccine trials to determine whether it is the host genetics, characteristics of the initial transmitted virus, and/or immune responses which are controlling viral replication. Eleven of the 13 individuals who received the vaccine had taken part in trials of canarypox prime-boost regimens where the net cumulative CTL response rates were between 33 and 50%, as assessed by the chromium release assay (4, 18). The other two vaccine recipients had taken part in a phase II trial where the net cumulative CTL response rate, as assessed by IFN- ELISpot, was only 9%. Sera from 9 of the 16 study participants had been assessed for neutralizing antibodies during the vaccination series, and of these, 6 had detectable antibody titers against the vaccine strain (neutralization titers ranged from 12 to 1,941 [data not shown]). No vaccine-induced HIV-specific CTL were detected using the ELISpot assay (20). It is possible that vaccine-induced responses were present at frequencies that were too low to detect by this assay, but the fact that the mean frequency of responses recognizing epitopes within the vaccine was no greater than that outside the vaccine suggests that there were no recall responses present prior to infection. Since none of the individuals in this study possessed detectable CTL prior to infection, it is interesting to speculate that the lack of infection in individuals with CTL responses is due to their HIV-specific vaccine-induced T-cell responses. However, the HIV-1 seroincidence rate among vaccine recipients was comparable to that in the HIVNET Vaccine Preparedness Study (1.38 per 100 person-years) (20, 28); due to the small sample sizes and differences in immunogens, reliable estimates of efficacy for these canarypox vaccines in reducing HIV-1 acquisition cannot be derived. That the postinfection T-cell responses were similar in specificity, breadth, and magnitude between the acute infection group and the vaccine recipients indicates that an individual s ability to raise HIV-specific T-cell immunity during a viral challenge is not affected by having seen HIV in the context of vaccination. However, this may be because the vaccines in this study did not induce strong responses. It remains to be determined whether infection-induced responses will be disrupted by more immunogenic vaccines. Of note, Nef and Gag were recognized most frequently in the vaccinated individuals, which is in agreement with recent data from other primary infection cohorts, as is the fact that all proteins were targeted by HIV-specific T cells (1, 9). Differences between the specific responses observed in our primary infection group and those described in this study may be explained by the average time point for PBMC collection, which was 113 days postinfection (dpi) in our study compared to 30 dpi for the acute infection cohort. Furthermore, the numbers of individuals characterized for T-cell responses at the epitopic level against all HIV proteins during acute infection (1, 2, 9) are far more limited than for those characterized during chronic infection. This, together with the diversity of MHC haplotypes among individuals, means that the database of the complete repertoire of epitopes recognized during primary infection is not sufficiently robust to make distinctions between epitopes recognized during acute versus chronic infection at this time. Of the 44 epitopes recognized, 31 have been previously defined as restricted by 21 different HLA molecules. The number of epitopes/alleles was too small to be able to determine if any single epitope/hla allele was overrepresented in these breakthrough infections. 014A-004 is the only individual who is controlling viral replication in the absence of ART. He never reported signs and symptoms consistent with an acute retroviral syndrome and has, to date, maintained extraordinarily low viral loads after infection. He possessed no neutralizing antibody preseroconversion (reciprocal titer giving 90% inhibition was 12 at day 46 before infection) and low levels soon after seroconversion (reciprocal titer giving 90% inhibition ranged from 107 to 117 between 38 and 87 days after infection). This individual has been tested for a number of genes that might explain his ability to control viral replication. His MHC haplotype is HLA-A2/ A11/B51/B61/C2/C14. Interestingly, HLA-C14 has been associated with nonprogression (19), and this individual has a new HLA-C14-restricted response that has not been previously described. This response is of high avidity, which may explain his ability to control viral replication, since high-avidity CTL responses have been shown to be more protective than lowavidity responses (13). The LYNTVATL (LL8), HLA-C14- restricted epitope is a single amino acid short of SLYNTVATL (SL9), which has been described as a very common HLA-A2-

9 VOL. 80, 2006 T-CELL RESPONSES IN HIV VACCINE TRIAL PARTICIPANTS 9787 restricted epitope recognized by T cells from chronically infected individuals. SL9 is not normally seen during acute infection (16) and, indeed, although this individual possesses HLA-A2, he does not recognize SL9 during acute infection. Not only is the LL8 epitope novel, but also, to our knowledge, this is the first description of any HLA-C14-restricted HIV epitope. Since HLA-A2 is present at a very high frequency (25%) in the Caucasian population, it is interesting to speculate that some LL8/C14-restricted responses may have been erroneously attributed to SL9/A2 if these persons possess both HLA-A2 and HLA-C14. Notably, HIV-specific T cells present in this individual were able to proliferate at all time points measured. Maintenance of proliferative ability in HIV-specific CD8 T cells has been shown to be associated with nonprogressive disease (24). Other than the presence of HLA-C14, this individual does not possess other known receptor polymorphisms or mutations in CCR5, CCR2, SDF-1, or DC-SIGN that would explain his ability to control viral replication. He is heterozygous for the CCR5 promoter P1 haplotype (21). It is possible that his ability to control viral replication and his delay in time to seroconversion were related to vaccine-induced immune responses, although there are insufficient immunologic data at present to substantiate this. Alternatively, this individual may have been infected with a less fit viral strain. Further studies are under way to examine this possibility. Immunological and virological data from another vaccinated individual (202-T07) who received the same vaccine as 014A- 004 but also became HIV infected have recently been reported (6). This individual possessed an HLA-B27-restricted response induced by vaccination. The authors inferred that he also possessed at least two other Gag-specific T-cell responses induced by vaccination, although there were insufficient cells to characterize these at the epitopic level. The authors postulated that this individual may have progressed to disease more quickly after vaccination than would be predicted for an HLA-B27 individual. 014A-004 differs in a few respects from 202-T07 in that he possessed T-cell responses to Gag, Pol, and Nef prior to seroconversion, whereas 202-T07 possessed only Gag-specific responses. In addition, since 014A-004 was actually infected at a low level, it is likely that antigen was more persistent in this individual than in 202-T07. This raises the intriguing possibility that low-level persistence of antigen may be important for inducing immune responses that are capable of controlling viremia. In conclusion, these data provide the first comprehensive analysis of the HIV-specific T-cell response in infected individuals who have previously been inoculated with vaccines designed to induce CD8 T-cell immunity against HIV. Of note, we show that an individual s ability to mount an HIVspecific T-cell immune response is not compromised by HIV vaccination. Although the canarypox prime-boost vaccine strategies, as tested in the initial phase I/II trials, did not progress into HVTN efficacy trials, new vaccines that induce higher frequencies of CD8 CTL are currently being evaluated. It will be important to perform similar studies as those described here on responses from breakthrough individuals to determine whether vaccine-induced HIV-specific T-cell responses will aid in control of HIV infection. ACKNOWLEDGMENTS We thank the study volunteers for their commitment and the clinical staff at the primary infection clinic and the collaborating HVTUs for their dedication. In addition, we appreciate the collaboration of numerous investigators from the HVTN, NIH Division of AIDS, Sanofi Pasteur, Vaxgen, Inc., and Chiron Corporation. This work was supported by grant numbers U01 AI 4674, U01 AI 46725, P01 AI , RO1 AI 49109, RO1 AI45402, RO1 AI 55336, and M01-RR from the National Institutes of Health. REFERENCES 1. Addo, M. M., X. G. Yu, A. Rathod, D. Cohen, R. L. Eldridge, D. Strick, M. N. Johnston, C. Corcoran, A. G. Wurcel, C. A. Fitzpatrick, M. E. Feeney, W. R. Rodriguez, N. Basgoz, R. Draenert, D. R. Stone, C. Brander, P. J. R. Goulder, E. S. Rosenberg, M. Altfeld, and B. D. Walker Comprehensive epitope analysis of human immunodeficiency virus type 1 (HIV-1)-specific T-cell responses directed against the entire expressed HIV-1 genome demonstrate broadly directed responses, but no correlation to viral load. J. Virol. 77: Altfeld, M., E. S. Rosenberg, R. Shankarappa, J. S. Mukherjee, F. M. Hecht, R. L. Eldridge, M. M. Addo, S. H. Poon, M. N. Phillips, G. K. Robbins, P. E. Sax, S. Boswell, J. O. Kahn, C. Brander, P. J. Goulder, J. A. Levy, J. I. Mullins, and B. D. Walker Cellular immune responses and viral diversity in individuals treated during acute and early HIV-1 infection. J. Exp. Med. 193: Belshe, R. B., G. J. Gorse, M. J. Mulligan, T. G. Evans, M. C. Keefer, J. L. Excler, A. M. Duliege, J. Tartaglia, W. I. Cox, J. McNamara, K. L. Hwang, A. Bradney, D. Montefiori, K. J. Weinhold, et al Induction of immune responses to HIV-1 by canarypox virus (ALVAC) HIV-1 and gp120 SF-2 recombinant vaccines in uninfected volunteers. AIDS 12: Belshe, R. B., C. Stevens, G. J. Gorse, S. Buchbinder, K. Weinhold, H. Sheppard, D. Stablein, S. Self, J. McNamara, S. Frey, J. Flores, J. L. Excler, M. Klein, R. E. Habib, A. M. Duliege, C. Harro, L. Corey, M. Keefer, M. Mulligan, P. Wright, C. Celum, F. Judson, K. Mayer, D. McKirnan, M. Marmor, and G. Woody Safety and immunogenicity of a canarypoxvectored human immunodeficiency virus type 1 vaccine with or without gp120: a phase 2 study in higher- and lower-risk volunteers. J. Infect. Dis. 183: Berman, P. W., A. M. Gray, T. Wrin, J. C. Vennari, D. J. Eastman, G. R. Nakamura, D. P. Francis, G. Gorse, and D. H. Schwartz Genetic and immunologic characterization of viruses infecting MN-rgp120-vaccinated volunteers. J. Infect. Dis. 176: Betts, M. R., B. Exley, D. A. Price, A. Bansal, Z. T. Camacho, V. Teaberry, S. M. West, D. R. Ambrozak, G. Tomaras, M. Roederer, J. M. Kilby, J. Tartaglia, R. Belshe, F. Gao, D. C. Douek, K. J. Weinhold, R. A. Koup, P. Goepfert, and G. Ferrari Characterization of functional and phenotypic changes in anti-gag vaccine-induced T cell responses and their role in protection after HIV-1 infection. Proc. Natl. Acad. Sci. USA 102: Bunce, M., G. C. Fanning, and K. I. Welsh Comprehensive, serologically equivalent DNA typing for HLA-B by PCR using sequence-specific primers (PCR-SSP). Tissue Antigens 45: Burton, D. R., and J. P. Moore Why do we not have an HIV vaccine and how can we make one? Nat. Med. 4: Cao, J., J. McNevin, S. Holte, L. Fink, L. Corey, and M. J. McElrath Comprehensive analysis of human immunodeficiency virus type 1 (HIV-1)- specific gamma interferon-secreting CD8 T cells in primary HIV-1 infection. J. Virol. 77: Cassidy, W. M., B. Watson, V. A. Ioli, K. Williams, S. Bird, and D. J. West A randomized trial of alternative two- and three-dose hepatitis B vaccination regimens in adolescents: antibody responses, safety, and immunologic memory. Pediatrics 107: Clements-Mann, M. L., K. Weinhold, T. J. Matthews, B. S. Graham, G. J. Gorse, M. C. Keefer, M. J. McElrath, R. H. Hsieh, J. Mestecky, S. Zolla- Pazner, J. Mascola, D. Schwartz, R. Siliciano, L. Corey, P. F. Wright, R. Belshe, R. Dolin, S. Jackson, S. Xu, P. Fast, M. C. Walker, D. Stablein, J. L. Excler, J. Tartaglia, E. Paoletti, et al Immune responses to human immunodeficiency virus (HIV) type 1 induced by canarypox expressing HIV- 1MN gp120, HIV-1SF2 recombinant gp120, or both vaccines in seronegative adults. J. Infect. Dis. 177: Connor, R. I., B. T. Korber, B. S. Graham, B. H. Hahn, D. D. Ho, B. D. Walker, A. U. Neumann, S. H. Vermund, J. Mestecky, S. Jackson, E. Fenamore, Y. Cao, F. Gao, S. Kalams, K. J. Kunstman, D. McDonald, N. McWilliams, A. Trkola, J. P. Moore, and S. M. Wolinsky Immunological and virological analyses of persons infected by human immunodeficiency virus type 1 while participating in trials of recombinant gp120 subunit vaccines. J. Virol. 72: Derby, M., M. Alexander-Miller, R. Tse, and J. Berzofsky High-avidity CTL exploit two complementary mechanisms to provide better protection against viral infection than low-avidity CTL. J. Immunol. 166: Dewar, R. L., H. C. Highbarger, M. D. Sarmiento, J. A. Todd, M. B.

10 9788 HORTON ET AL. J. VIROL. Vasudevachari, R. T. Davey, Jr., J. A. Kovacs, N. P. Salzman, H. C. Lane, and M. S. Urdea Application of branched DNA signal amplification to monitor human immunodeficiency virus type 1 burden in human plasma. J. Infect. Dis. 170: Evans, T. G Evaluation of canarypox-induced CD8 responses following immunization by measuring the effector population IFN- production. Immunol. Lett. 77: Goulder, P. J., M. A. Altfeld, E. S. Rosenberg, T. Nguyen, Y. Tang, R. L. Eldridge, M. M. Addo, S. He, J. S. Mukherjee, M. N. Phillips, M. Bunce, S. A. Kalams, R. P. Sekaly, B. D. Walker, and C. Brander Substantial differences in specificity of HIV-specific cytotoxic T cells in acute and chronic HIV infection. J. Exp. Med. 193: Graham, B. S., M. J. McElrath, R. I. Connor, D. H. Schwartz, G. J. Gorse, M. C. Keefer, M. J. Mulligan, T. J. Matthews, S. M. Wolinsky, D. C. Montefiori, S. H. Vermund, J. S. Lambert, L. Corey, R. B. Belshe, R. Dolin, P. F. Wright, B. T. Korber, M. C. Wolff, P. E. Fast, et al Analysis of intercurrent human immunodeficiency virus type 1 infections in phase I and II trials of candidate AIDS vaccines. J. Infect. Dis. 177: Gupta, K., M. Hudgens, L. Corey, M. J. McElrath, K. Weinhold, D. C. Montefiori, G. J. Gorse, S. E. Frey, M. C. Keefer, T. G. Evans, R. Dolin, D. H. Schwartz, C. Harro, B. Graham, P. W. Spearman, M. Mulligan, and P. Goepfert Safety and immunogenicity of a high-titered canarypox vaccine in combination with rgp120 in a diverse population of HIV-1-uninfected adults: AIDS Vaccine Evaluation Group protocol 022A. J. Acquir. Immune Defic. Syndr. 29: Hendel, H., S. Caillat-Zucman, H. Lebuanec, M. Carrington, S. J. O Brien, J. M. Andrieu, F. Schachter, D. Zagury, J. Rappaport, C. Winkler, G. W. Nelson, and J.-F. Zagury New class I and II HLA alleles strongly associated with opposite patterns of progression to AIDS. J. Immunol. 162: Lee, D., B. S. Graham, Y. Chiu, P. B. Gilbert, M. J. McElrath, R. B. Belshe, S. P. Buchbinder, H. W. Sheppard, B. A. Koblin, K. H. Mayer, M. C. Keefer, M. J. Mulligan, and C. L. Celum Breakthrough infections during phase I-II prime-boost HIV-1 vaccine trials with canarypox vectors (ALVAC) and recombinant gp120 or gp160 boost. J. Infect. Dis. 190: Liu, H., Y. Hwangbo, S. Holte, J. Lee, C. Wang, N. Kaupp, H. Zhu, C. Celum, L. Corey, M. J. McElrath, and T. Zhu Analysis of genetic polymorphisms in CCR5, CCR2, stromal cell-derived factor-1, RANTES, and dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin in seronegative individuals repeatedly exposed to HIV-1. J. Infect. Dis. 190: Maranon, C., J. F. Desoutter, G. Hoeffel, W. Cohen, D. Hanau, and A. Hosmalin Dendritic cells cross-present HIV antigens from live as well as apoptotic infected CD4 T lymphocytes. Proc. Natl. Acad. Sci. USA 101: McElrath, M. J., L. Corey, P. D. Greenberg, T. J. Matthews, D. C. Montefiori, L. Rowen, L. Hood, and J. I. Mullins Human immunodeficiency virus type 1 infection despite prior immunization with a recombinant envelope vaccine regimen. Proc. Natl. Acad. Sci. USA 93: Migueles, S. A., A. C. Laborico, W. L. Shupert, M. S. Sabbaghian, R. Rabin, C. W. Hallahan, D. Van Baarle, S. Kostense, F. Miedema, M. McLaughlin, L. Ehler, J. Metcalf, S. Liu, and M. Connors HIV-specific CD8 T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nat. Immunol. 3: Russell, N., M. Hudgens, R. Ha, C. Havenar-Daughton, and M. J. McElrath Moving to human immunodeficiency type 1 vaccine efficacy trials: defining T cell responses as potential correlates of immunity. J. Infect. Dis. 187: Schacker, T Primary HIV infection. Early diagnosis and treatment are critical to outcome. Postgrad. Med. 102: , Schacker, T., A. C. Collier, J. Hughes, T. Shea, and L. Corey Clinical and epidemiologic features of primary HIV infection. Ann. Intern. Med. 125: Seage, G. R., III, S. E. Holte, D. Metzger, B. A. Koblin, M. Gross, C. Celum, M. Marmor, G. Woody, K. H. Mayer, C. Stevens, F. N. Judson, D. McKirnan, A. Sheon, S. Self, and S. P. Buchbinder Are US populations appropriate for trials of human immunodeficiency virus vaccine? The HIVNET Vaccine Preparedness study. Am. J. Epidemiol. 153: Zhu, T., L. Corey, Y. Hwangbo, J. M. Lee, G. H. Learn, J. I. Mullins, and M. J. McElrath Persistence of extraordinarily low levels of genetically homogeneous human immunodeficiency virus type 1 in exposed seronegative individuals. J. Virol. 77: Downloaded from on July 3, 2018 by guest

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

Superior Control of HIV-1 Replication by CD8+ T Cells Targeting Conserved Epitopes: Implications for HIV Vaccine Design

Superior Control of HIV-1 Replication by CD8+ T Cells Targeting Conserved Epitopes: Implications for HIV Vaccine Design Superior Control of HIV-1 Replication by CD8+ T Cells Targeting Conserved Epitopes: Implications for HIV Vaccine Design Pratima Kunwar 1,7, Natalie Hawkins 2, Warren L. Dinges 1,3, Yi Liu 5, Erin E. Gabriel

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

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

Received 15 November 2000; revised 6 February 2001; electronically published

Received 15 November 2000; revised 6 February 2001; electronically published 1343 Safety and Immunogenicity of a Canarypox-Vectored Human Immunodeficiency Virus Type 1 Vaccine with or without gp120: A Phase 2 Study in Higher- and Lower-Risk Volunteers Robert B. Belshe, 1 Cladd

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

HIV-1 acute infection: evidence for selection?

HIV-1 acute infection: evidence for selection? HIV-1 acute infection: evidence for selection? ROLLAND Morgane University of Washington Cohort & data S6 S5 T4 S4 T2 S2 T1 S1 S7 T3 DPS (days post symptoms) 3 (Fiebig I) 7 (Fiebig I) 13 (Fiebig V) 14 (Fiebig

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

How HIV Causes Disease Prof. Bruce D. Walker

How HIV Causes Disease Prof. Bruce D. Walker How HIV Causes Disease Howard Hughes Medical Institute Massachusetts General Hospital Harvard Medical School 1 The global AIDS crisis 60 million infections 20 million deaths 2 3 The screen versions of

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

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

RAISON D ETRE OF THE IMMUNE SYSTEM:

RAISON D ETRE OF THE IMMUNE SYSTEM: RAISON D ETRE OF THE IMMUNE SYSTEM: To Distinguish Self from Non-Self Thereby Protecting Us From Our Hostile Environment. Innate Immunity Acquired Immunity Innate immunity: (Antigen nonspecific) defense

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

Supplementary Information Titles

Supplementary Information Titles Supplementary Information Titles Journal: Nature Medicine Article Title: Corresponding Author: Supplementary Item & Number Supplementary Figure 1 Supplementary Figure 2 Supplementary Figure 3 Supplementary

More information

Selection on the Human Immunodeficiency Virus Type 1 Proteome following Primary Infection

Selection on the Human Immunodeficiency Virus Type 1 Proteome following Primary Infection JOURNAL OF VIROLOGY, Oct. 2006, p. 9519 9529 Vol. 80, No. 19 0022-538X/06/$08.00 0 doi:10.1128/jvi.00575-06 Copyright 2006, American Society for Microbiology. All Rights Reserved. Selection on the Human

More information

Fayth K. Yoshimura, Ph.D. September 7, of 7 HIV - BASIC PROPERTIES

Fayth K. Yoshimura, Ph.D. September 7, of 7 HIV - BASIC PROPERTIES 1 of 7 I. Viral Origin. A. Retrovirus - animal lentiviruses. HIV - BASIC PROPERTIES 1. HIV is a member of the Retrovirus family and more specifically it is a member of the Lentivirus genus of this family.

More information

NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections

NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections Amy Chung Dr. Ivan Stratov Prof. Stephen Kent ADCC process consists of Target cell QuickTime and a TIFF (Uncompressed) FcγR decompressor

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

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

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

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

RAISON D ETRE OF THE IMMUNE SYSTEM:

RAISON D ETRE OF THE IMMUNE SYSTEM: RAISON D ETRE OF THE IMMUNE SYSTEM: To Distinguish Self from Non-Self Thereby Protecting Us From Our Hostile Environment. Innate Immunity Adaptive Immunity Innate immunity: (Antigen - nonspecific) defense

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

Potential cross reactions between HIV 1 specific T cells and the microbiome. Andrew McMichael Suzanne Campion

Potential cross reactions between HIV 1 specific T cells and the microbiome. Andrew McMichael Suzanne Campion Potential cross reactions between HIV 1 specific T cells and the microbiome Andrew McMichael Suzanne Campion Role of the Microbiome? T cell (and B cell) immune responses to HIV and Vaccines are influenced

More information

Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection

Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection Anne Piantadosi 1,2[, Bhavna Chohan 1,2[, Vrasha Chohan 3, R. Scott McClelland 3,4,5, Julie Overbaugh 1,2* 1 Division of Human

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

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

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

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

HVTN P5 Vaccine Trials

HVTN P5 Vaccine Trials HVTN P5 Vaccine Trials Erica Andersen-Nissen, PhD Director, Cape Town HVTN Immunology Laboratory Considerations for a Pan-African HIV Vaccine Development Agenda Kigali, Rwanda 16-17 March 2015 HVTN Mission

More information

Micropathology Ltd. University of Warwick Science Park, Venture Centre, Sir William Lyons Road, Coventry CV4 7EZ

Micropathology Ltd. University of Warwick Science Park, Venture Centre, Sir William Lyons Road, Coventry CV4 7EZ www.micropathology.com info@micropathology.com Micropathology Ltd Tel 24hrs: +44 (0) 24-76 323222 Fax / Ans: +44 (0) 24-76 - 323333 University of Warwick Science Park, Venture Centre, Sir William Lyons

More information

Ex vivo Human Antigen-specific T Cell Proliferation and Degranulation Willemijn Hobo 1, Wieger Norde 1 and Harry Dolstra 2*

Ex vivo Human Antigen-specific T Cell Proliferation and Degranulation Willemijn Hobo 1, Wieger Norde 1 and Harry Dolstra 2* Ex vivo Human Antigen-specific T Cell Proliferation and Degranulation Willemijn Hobo 1, Wieger Norde 1 and Harry Dolstra 2* 1 Department of Laboratory Medicine - Laboratory of Hematology, Radboud University

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

HIV Vaccine. 15 September 2016 นพ.นคร เปรมศร ผ อ านวยการส าน กงาน โครงการศ กษาว คซ นเอดส ทดลอง

HIV Vaccine. 15 September 2016 นพ.นคร เปรมศร ผ อ านวยการส าน กงาน โครงการศ กษาว คซ นเอดส ทดลอง HIV Vaccine 15 September 2016 นพ.นคร เปรมศร ผ อ านวยการส าน กงาน โครงการศ กษาว คซ นเอดส ทดลอง GLOBAL STATISTICS 2015 17 million people were accessing antiretroviral therapy 36.7 million people globally

More information

Supplementary Figure 1. Enhanced detection of CTLA-4 on the surface of HIV-specific

Supplementary Figure 1. Enhanced detection of CTLA-4 on the surface of HIV-specific SUPPLEMENTARY FIGURE LEGEND Supplementary Figure 1. Enhanced detection of CTLA-4 on the surface of HIV-specific CD4 + T cells correlates with intracellular CTLA-4 levels. (a) Comparative CTLA-4 levels

More information

Human Immunodeficiency Virus

Human Immunodeficiency Virus Human Immunodeficiency Virus Virion Genome Genes and proteins Viruses and hosts Diseases Distinctive characteristics Viruses and hosts Lentivirus from Latin lentis (slow), for slow progression of disease

More information

New Preventive Technology: Providing New Options to Stop the Spread of HIV/AIDS. Dublin, Ireland. June 24, AIDS Vaccines.

New Preventive Technology: Providing New Options to Stop the Spread of HIV/AIDS. Dublin, Ireland. June 24, AIDS Vaccines. New Preventive Technology: Providing New Options to Stop the Spread of HIV/AIDS Dublin, Ireland June 24, 2004 AIDS Vaccines - An R&D Briefing This paper has been prepared by the International AIDS Vaccine

More information

Activation of NK Cells by ADCC Antibodies and HIV Disease Progression

Activation of NK Cells by ADCC Antibodies and HIV Disease Progression RAPID COMMUNICATION Activation of NK Cells by ADCC Antibodies and HIV Disease Progression Amy W. Chung, BSc, PhD,* Marjon Navis, PhD,* Gamze Isitman, BSc,* Leia Wren, BSc,* Julie Silvers, RN, Janaki Amin,

More information

Supplementary Fig. 1: Ex vivo tetramer enrichment with anti-c-myc beads

Supplementary Fig. 1: Ex vivo tetramer enrichment with anti-c-myc beads Supplementary Fig. 1: Ex vivo tetramer enrichment with anti-c-myc beads Representative example of comparative ex vivo tetramer enrichment performed in three independent experiments with either conventional

More information

Under the Radar Screen: How Bugs Trick Our Immune Defenses

Under the Radar Screen: How Bugs Trick Our Immune Defenses Under the Radar Screen: How Bugs Trick Our Immune Defenses Session 7: Cytokines Marie-Eve Paquet and Gijsbert Grotenbreg Whitehead Institute for Biomedical Research HHV-8 Discovered in the 1980 s at the

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

HIV acute infections and elite controllers- what can we learn?

HIV acute infections and elite controllers- what can we learn? HIV acute infections and elite controllers- what can we learn? Thumbi Ndung u, BVM, PhD KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH) and HIV Pathogenesis Programme (HPP), Doris Duke

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

Primary Human Immunodeficiency Virus Type 1 (HIV-1) Infection during HIV-1 Gag Vaccination

Primary Human Immunodeficiency Virus Type 1 (HIV-1) Infection during HIV-1 Gag Vaccination JOURNAL OF VIROLOGY, Mar. 2008, p. 2784 2791 Vol. 82, No. 6 0022-538X/08/$08.00 0 doi:10.1128/jvi.01720-07 Copyright 2008, American Society for Microbiology. All Rights Reserved. Primary Human Immunodeficiency

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

Conserved epitopes on HIV-1, FIV and SIV p24 proteins are recognized by HIV-1 infected subjects

Conserved epitopes on HIV-1, FIV and SIV p24 proteins are recognized by HIV-1 infected subjects Human Vaccines & Immunotherapeutics ISSN: 2164-5515 (Print) 2164-554X (Online) Journal homepage: http://www.tandfonline.com/loi/khvi20 Conserved epitopes on HIV-1, FIV and SIV p24 proteins are recognized

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

Challenges of Integrating Mucosal Immune Assays into HIV Vaccine Trials KAVI

Challenges of Integrating Mucosal Immune Assays into HIV Vaccine Trials KAVI Challenges of Integrating Mucosal Immune Assays into HIV Vaccine Trials Omu Anzala, MBChB, PhD KAVI University of Nairobi Outline Why mucosal immunology Experience from Kenya Challenges The way forward

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

HIV INFECTION: An Overview

HIV INFECTION: An Overview HIV INFECTION: An Overview UNIVERSITY OF PAPUA NEW GUINEA SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY & MOLECULAR BIOLOGY PBL MBBS II SEMINAR VJ

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

Estimating primate effector T cell responses to DNA vaccination

Estimating primate effector T cell responses to DNA vaccination Estimating primate effector T cell responses to DNA vaccination Oct 22 nd, 21 Devon J. Shedlock, PhD th Vaccine Renaissance Conference Estimating vaccineinduced effector T cell responses Vaccineinduced

More information

HIV-1 Dual Infection and Neurocognitive Impairment

HIV-1 Dual Infection and Neurocognitive Impairment HIV-1 Dual Infection and Neurocognitive Impairment Gabriel Wagner, MD Assistant Professor of Medicine Infectious Diseases & Global Public Health UC San Diego HIV-Associated End Organ Damage Antiretroviral

More information

CD4 + T-cell-inducing HIV vaccines may have an impact on viral control

CD4 + T-cell-inducing HIV vaccines may have an impact on viral control CD4 + T-cell-inducing HIV vaccines may have an impact on viral control Clues from cytokines produced by CD4 + T-cells from HIVinfected patients and vaccinated seronegative volunteers Eva Van Braeckel 1,

More information

Fully Differentiated HIV-1 Specific CD8+ T Effector Cells are More Frequently Detectable in Controlled than in Progressive HIV-1 Infection

Fully Differentiated HIV-1 Specific CD8+ T Effector Cells are More Frequently Detectable in Controlled than in Progressive HIV-1 Infection Fully Differentiated HIV-1 Specific CD8+ T Effector Cells are More Frequently Detectable in Controlled than in Progressive HIV-1 Infection The Harvard community has made this article openly available.

More information

What to Measure, How to Measure It

What to Measure, How to Measure It Dale and Betty Bumpers Vaccine Research Center National Institute of Allergy and Infectious Diseases National Institutes of Health Monitoring Memory T-cells: What to Measure, How to Measure It Pratip K.

More information

Perforin Expression Directly Ex Vivo by HIV-Specific CD8+ T-Cells Is. Is a Correlate of HIV Elite Control

Perforin Expression Directly Ex Vivo by HIV-Specific CD8+ T-Cells Is. Is a Correlate of HIV Elite Control Perforin Expression Directly Ex Vivo by HIV-Specific CD8+ T-Cells Is a Correlate of HIV Elite Control The Harvard community has made this article openly available. Please share how this access benefits

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

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

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

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

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

HVTN Laboratory Program: Immunogenicity and Research Assays

HVTN Laboratory Program: Immunogenicity and Research Assays HVTN Laboratory Program: Immunogenicity and Research Assays Erica Andersen-Nissen, PhD Director, Cape Town HVTN Immunology Laboratory Considerations for a Pan-African HIV Vaccine Development Agenda Kigali,

More information

Biomedical Engineering for Global Health. Lecture 10 HIV/AIDS vaccine development

Biomedical Engineering for Global Health. Lecture 10 HIV/AIDS vaccine development Biomedical Engineering for Global Health Lecture 10 HIV/AIDS vaccine development Review of lecture 9 How do vaccines work? Types ofvaccines: Review of lecture 9 Are vaccines effective? -Edward Jenner s

More information

An HIV-1 clade C DNA prime, NYVAC boost vaccine regimen induces reliable, polyfunctional, and long-lasting T cell responses

An HIV-1 clade C DNA prime, NYVAC boost vaccine regimen induces reliable, polyfunctional, and long-lasting T cell responses ARTICLE An HIV-1 clade C DNA prime, NYVAC boost vaccine regimen induces reliable, polyfunctional, and long-lasting T cell responses The Journal of Experimental Medicine CORRESPONDENCE Giuseppe Pantaleo:

More information

Genetic and Immunologic Heterogeneity among Persons Who Control HIV Infection in the Absence of Therapy

Genetic and Immunologic Heterogeneity among Persons Who Control HIV Infection in the Absence of Therapy MAJOR ARTICLE Genetic and Immunologic Heterogeneity among Persons Who Control HIV Infection in the Absence of Therapy Florencia Pereyra, 1,2 Marylyn M. Addo, 1 Daniel E. Kaufmann, 1 Yang Liu, 5 Toshiyuki

More information

Immunodeficiency. (2 of 2)

Immunodeficiency. (2 of 2) Immunodeficiency (2 of 2) Acquired (secondary) immunodeficiencies More common Many causes such as therapy, cancer, sarcoidosis, malnutrition, infection & renal disease The most common of which is therapy-related

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, Mar. 2001, p. 2142 2153 Vol. 75, No. 5 0022-538X/01/$04.00 0 DOI: 10.1128/JVI.75.5.2142 2153.2001 Copyright 2001, American Society for Microbiology. All Rights Reserved. Mature Dendritic

More information

Identification and Characterization of CD4 T cells actively transcribing HIV RNA in Peripheral Blood

Identification and Characterization of CD4 T cells actively transcribing HIV RNA in Peripheral Blood Dale and Betty Bumpers Vaccine Research Center National Institute of Allergy and Infectious Diseases National Institutes of Health Identification and Characterization of CD4 T cells actively transcribing

More information

Therapeutic Immunization with Autologous DC Pulsed with Autologous Inactivated HIV-1 Infected Apoptotic Cells

Therapeutic Immunization with Autologous DC Pulsed with Autologous Inactivated HIV-1 Infected Apoptotic Cells Therapeutic Immunization with Autologous DC Pulsed with Autologous Inactivated HIV-1 Infected Apoptotic Cells Sharon A. Riddler, MD, MPH University of Pittsburgh May 2008 Slide 1 HIV and DC Vaccines During

More information

HIV Immunopathogenesis. Modeling the Immune System May 2, 2007

HIV Immunopathogenesis. Modeling the Immune System May 2, 2007 HIV Immunopathogenesis Modeling the Immune System May 2, 2007 Question 1 : Explain how HIV infects the host Zafer Iscan Yuanjian Wang Zufferey Abhishek Garg How does HIV infect the host? HIV infection

More information

HIV-1 Subtypes: An Overview. Anna Maria Geretti Royal Free Hospital

HIV-1 Subtypes: An Overview. Anna Maria Geretti Royal Free Hospital HIV-1 Subtypes: An Overview Anna Maria Geretti Royal Free Hospital Group M Subtypes A (1, 2, 3) B C D F (1, 2) G H J K Mechanisms of HIV-1 genetic diversification Point mutations RT error rate: ~1 per

More information

Toinform rational design of prophylactic vaccines and

Toinform rational design of prophylactic vaccines and Kinetics of Expansion of Epitope-Specific T Cell Responses during Primary HIV-1 Infection 1 Emma L. Turnbull, 2 * MaiLee Wong,* Shuyi Wang, Xiping Wei, Nicola A. Jones,* Karen E. Conrod,* Diana Aldam,

More information

Citation for published version (APA): Von Eije, K. J. (2009). RNAi based gene therapy for HIV-1, from bench to bedside

Citation for published version (APA): Von Eije, K. J. (2009). RNAi based gene therapy for HIV-1, from bench to bedside UvA-DARE (Digital Academic Repository) RNAi based gene therapy for HIV-1, from bench to bedside Von Eije, K.J. Link to publication Citation for published version (APA): Von Eije, K. J. (2009). RNAi based

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

Therapeutic strategies for immune reconstitution in acquired immunodeficiency syndrome

Therapeutic strategies for immune reconstitution in acquired immunodeficiency syndrome 30 1, 1, 2, 3 1. ( ), 201508; 2., 200040; 3., 200032 : ( AIDS) ( HIV) 20 90,,,,,, AIDS, CD4 + T ( CTL), HIV, : ; ; Therapeutic strategies for immune reconstitution in acquired immunodeficiency syndrome

More information

Safety and Immunogenicity of an HIV Subtype B and E Prime-Boost Vaccine Combination in HIV-Negative Thai Adults

Safety and Immunogenicity of an HIV Subtype B and E Prime-Boost Vaccine Combination in HIV-Negative Thai Adults BRIEF REPORT Safety and Immunogenicity of an HIV Subtype B and E Prime-Boost Vaccine Combination in HIV-Negative Thai Adults Sorachai Nitayaphan, 1 Punnee Pitisuttithum, 3 Chitraporn Karnasuta, 2 Chirapa

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

JOURNAL OF VIROLOGY, May 2000, p Vol. 74, No. 9. Copyright 2000, American Society for Microbiology. All Rights Reserved.

JOURNAL OF VIROLOGY, May 2000, p Vol. 74, No. 9. Copyright 2000, American Society for Microbiology. All Rights Reserved. JOURNAL OF VIROLOGY, May 2000, p. 4127 4138 Vol. 74, No. 9 0022-538X/00/$04.00 0 Copyright 2000, American Society for Microbiology. All Rights Reserved. Anti-Human Immunodeficiency Virus Type 1 (HIV-1)

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

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

Progressive Telomere Shortening of Epstein-Barr Virus Specific Memory T Cells during HIV Infection: Contributor to Exhaustion?

Progressive Telomere Shortening of Epstein-Barr Virus Specific Memory T Cells during HIV Infection: Contributor to Exhaustion? BRIEF REPORT Progressive Telomere Shortening of Epstein-Barr Virus Specific Memory T Cells during HIV Infection: Contributor to Exhaustion? Debbie van Baarle, 1 Nening M. Nanlohy, 1 Sigrid Otto, 1 Fiona

More information

Cytotoxicity assays. Rory D. de Vries, PhD 1. Viroscience lab, Erasmus MC, Rotterdam, the Netherlands

Cytotoxicity assays. Rory D. de Vries, PhD 1. Viroscience lab, Erasmus MC, Rotterdam, the Netherlands Cytotoxicity assays Rory D. de Vries, PhD 1 1 Viroscience lab, Erasmus MC, Rotterdam, the Netherlands Anti-influenza immunity Humoral / CD4+ / CD8+ / NK? Function of CTL Elimination of virus-infected cells?

More information

The Kitchen Sink. Glenda Gray AIDS vaccine at the cross roads: how to adapt to a new preven9on age? Monday 7 th October, 2013, Barcelona

The Kitchen Sink. Glenda Gray AIDS vaccine at the cross roads: how to adapt to a new preven9on age? Monday 7 th October, 2013, Barcelona The Kitchen Sink Glenda Gray AIDS vaccine at the cross roads: how to adapt to a new preven9on age? Monday 7 th October, 2013, Barcelona The only important slide of my presentation. All efficacy trial in

More information

Madhav V. Dhodapkar, Joseph Krasovsky, Ralph M. Steinman, and Nina Bhardwaj

Madhav V. Dhodapkar, Joseph Krasovsky, Ralph M. Steinman, and Nina Bhardwaj Mature dendritic cells boost functionally superior CD8 + T-cell in humans without foreign helper epitopes Rapid PUBLICATION Madhav V. Dhodapkar, Joseph Krasovsky, Ralph M. Steinman, and Nina Bhardwaj Laboratory

More information

Viral Persistence Alters CD8 T-Cell Immunodominance and Tissue Distribution and Results in Distinct Stages of Functional Impairment

Viral Persistence Alters CD8 T-Cell Immunodominance and Tissue Distribution and Results in Distinct Stages of Functional Impairment JOURNAL OF VIROLOGY, Apr. 2003, p. 4911 4927 Vol. 77, No. 8 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.8.4911 4927.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Viral Persistence

More information

Defining kinetic properties of HIV-specific CD8 + T-cell responses in acute infection

Defining kinetic properties of HIV-specific CD8 + T-cell responses in acute infection Defining kinetic properties of HIV-specific CD8 + T-cell responses in acute infection Yiding Yang 1 and Vitaly V. Ganusov 1,2,3 1 Department of Microbiology, University of Tennessee, Knoxville, TN 37996,

More information

Alternate Antibody-Based Therapeutic Strategies To Purge the HIV Cell Reservoir

Alternate Antibody-Based Therapeutic Strategies To Purge the HIV Cell Reservoir Alternate Antibody-Based Therapeutic Strategies To Purge the HIV Cell Reservoir Giuseppe Pantaleo, M.D. Professor of Medicine Head, Division of Immunology and Allergy Executive Director, Swiss Vaccine

More information

Review Article. AIDS vaccine: Present status and future challenges

Review Article. AIDS vaccine: Present status and future challenges Review Article AIDS vaccine: Present status and future challenges P. K. Nigam, Manjula Kerketta Department of Dermatology and Venereology, Pt. JNM Medical College and associated Dr. BRAM Hospital, Raipur,

More information

MID 36. Cell. HIV Life Cycle. HIV Diagnosis and Pathogenesis. HIV-1 Virion HIV Entry. Life Cycle of HIV HIV Entry. Scott M. Hammer, M.D.

MID 36. Cell. HIV Life Cycle. HIV Diagnosis and Pathogenesis. HIV-1 Virion HIV Entry. Life Cycle of HIV HIV Entry. Scott M. Hammer, M.D. Life Cycle Diagnosis and Pathogenesis Scott M. Hammer, M.D. -1 Virion Entry Life Cycle of Entry -1 virion -1 Virus virion envelope Cell membrane receptor RELEASE OF PROGENY VIRUS REVERSE Co- TRANSCRIPTION

More information

CD27 and CD57 Expression Reveals Atypical Differentiation of Human Immunodeficiency Virus Type 1-Specific Memory CD8 T Cells

CD27 and CD57 Expression Reveals Atypical Differentiation of Human Immunodeficiency Virus Type 1-Specific Memory CD8 T Cells CLINICAL AND VACCINE IMMUNOLOGY, Jan. 2007, p. 74 80 Vol. 14, No. 1 1556-6811/07/$08.00 0 doi:10.1128/cvi.00250-06 Copyright 2007, American Society for Microbiology. All Rights Reserved. CD27 and CD57

More information

5. Over the last ten years, the proportion of HIV-infected persons who are women has: a. Increased b. Decreased c. Remained about the same 1

5. Over the last ten years, the proportion of HIV-infected persons who are women has: a. Increased b. Decreased c. Remained about the same 1 Epidemiology 227 April 24, 2009 MID-TERM EXAMINATION Select the best answer for the multiple choice questions. There are 60 questions and 9 pages on the examination. Each question will count one point.

More information

Early Induction and Maintenance of Env-Specific T-Helper Cells following Human Immunodeficiency Virus Type 1 Infection

Early Induction and Maintenance of Env-Specific T-Helper Cells following Human Immunodeficiency Virus Type 1 Infection JOURNAL OF VIROLOGY, Feb. 2003, p. 2663 2674 Vol. 77, No. 4 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.4.2663 2674.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Early Induction

More information

HIV Vaccine. Sunee Sirivichayakul, Ph.D. Faculty of Medicine Chulalongkorn University. August 22, 2014

HIV Vaccine. Sunee Sirivichayakul, Ph.D. Faculty of Medicine Chulalongkorn University. August 22, 2014 HIV Vaccine Sunee Sirivichayakul, Ph.D. Faculty of Medicine Chulalongkorn University August 22, 2014 Immunity Natural immunity Active natural immunity e.g., infection Passive natural immunity e.g., trans-placental

More information

HIV & AIDS: Overview

HIV & AIDS: Overview HIV & AIDS: Overview UNIVERSITY OF PAPUA NEW GUINEA SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY & MOLECULAR BIOLOGY PBL SEMINAR VJ TEMPLE 1 What

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

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

Cellular Immunity in Aging and HIV: Correlates of Protection. Immune Senescence

Cellular Immunity in Aging and HIV: Correlates of Protection. Immune Senescence Cellular Immunity in Aging and HIV: Correlates of Protection Janet E. McElhaney, MD Professor of Medicine Allan M. McGavin Chair in Research Geriatrics University of British Columbia Vancouver, BC and

More information

IL-12 and GM-CSF in DNA/MVA Immunizations against HIV-1 CRF12_BF Nef Induced T-Cell Responses With an Enhanced Magnitude, Breadth and Quality

IL-12 and GM-CSF in DNA/MVA Immunizations against HIV-1 CRF12_BF Nef Induced T-Cell Responses With an Enhanced Magnitude, Breadth and Quality IL-12 and GM-CSF in DNA/MVA Immunizations against HIV-1 CRF12_BF Nef Induced T-Cell Responses With an Enhanced Magnitude, Breadth and Quality Ana María Rodríguez, María Fernanda Pascutti., Cynthia Maeto.,

More information

Micro 301 HIV/AIDS. Since its discovery 31 years ago 12/3/ Acquired Immunodeficiency Syndrome (AIDS) has killed >32 million people

Micro 301 HIV/AIDS. Since its discovery 31 years ago 12/3/ Acquired Immunodeficiency Syndrome (AIDS) has killed >32 million people Micro 301 HIV/AIDS Shiu-Lok Hu hus@uw.edu December 3, 2012 Since its discovery 31 years ago Acquired Immunodeficiency Syndrome (AIDS) has killed >32 million people In 2011 34.0 million [31.4 35.9 million]

More information