Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hasan Ahmed is active.

Publication


Featured researches published by Hasan Ahmed.


Nature | 2012

Increased HIV-1 vaccine efficacy against viruses with genetic signatures in Env V2

Morgane Rolland; Paul T. Edlefsen; Brendan B. Larsen; Sodsai Tovanabutra; Eric Sanders-Buell; Tomer Hertz; Allan C. deCamp; Chris Carrico; Sergey Menis; Craig A. Magaret; Hasan Ahmed; Michal Juraska; Lennie Chen; Philip Konopa; Snehal Nariya; Julia N. Stoddard; Kim Wong; Haishuang Zhao; Wenjie Deng; Brandon Maust; Meera Bose; Shana Howell; A Bates; Michelle Lazzaro; Annemarie O'Sullivan; Esther Lei; Andrea Bradfield; Grace Ibitamuno; Vatcharain Assawadarachai; Robert J. O'Connell

The RV144 trial demonstrated 31% vaccine efficacy at preventing human immunodeficiency virus (HIV)-1 infection. Antibodies against the HIV-1 envelope variable loops 1 and 2 (Env V1 and V2) correlated inversely with infection risk. We proposed that vaccine-induced immune responses against V1/V2 would have a selective effect against, or sieve, HIV-1 breakthrough viruses. A total of 936 HIV-1 genome sequences from 44 vaccine and 66 placebo recipients were examined. We show that vaccine-induced immune responses were associated with two signatures in V2 at amino acid positions 169 and 181. Vaccine efficacy against viruses matching the vaccine at position 169 was 48% (confidence interval 18% to 66%; P = 0.0036), whereas vaccine efficacy against viruses mismatching the vaccine at position 181 was 78% (confidence interval 35% to 93%; P = 0.0028). Residue 169 is in a cationic glycosylated region recognized by broadly neutralizing and RV144-derived antibodies. The predicted distance between the two signature sites (21 ± 7 Å) and their match/mismatch dichotomy indicate that multiple factors may be involved in the protection observed in RV144. Genetic signatures of RV144 vaccination in V2 complement the finding of an association between high V1/V2-binding antibodies and reduced risk of HIV-1 acquisition, and provide evidence that vaccine-induced V2 responses plausibly had a role in the partial protection conferred by the RV144 regimen.


The Journal of Infectious Diseases | 2012

Magnitude and Breadth of the Neutralizing Antibody Response in the RV144 and Vax003 HIV-1 Vaccine Efficacy Trials

David C. Montefiori; Chitraporn Karnasuta; Ying Huang; Hasan Ahmed; Peter B. Gilbert; Mark S. de Souza; Robert McLinden; Sodsai Tovanabutra; Agnes Laurence-Chenine; Eric Sanders-Buell; M. Anthony Moody; Mattia Bonsignori; Christina Ochsenbauer; John C. Kappes; Haili Tang; Kelli M. Greene; Hongmei Gao; Celia C. LaBranche; Charla Andrews; Victoria R. Polonis; Supachai Rerks-Ngarm; Punnee Pitisuttithum; Sorachai Nitayaphan; Jaranit Kaewkungwal; Steve Self; Phillip W. Berman; Donald P. Francis; Faruk Sinangil; Carter Lee; Jim Tartaglia

Background. A recombinant canarypox vector expressing human immunodeficiency virus type 1 (HIV-1) Gag, Pro, and membrane-linked gp120 (vCP1521), combined with a bivalent gp120 protein boost (AIDSVAX B/E), provided modest protection against HIV-1 infection in a community-based population in Thailand (RV144 trial). No protection was observed in Thai injection drug users who received AIDSVAX B/E alone (Vax003 trial). We compared the neutralizing antibody response in these 2 trials. Methods. Neutralization was assessed with tier 1 and tier 2 strains of virus in TZM-bl and A3R5 cells. Results. Neutralization of several tier 1 viruses was detected in both RV144 and Vax003. Peak titers were higher in Vax003 and waned rapidly in both trials. The response in RV144 was targeted in part to V3 of gp120.vCP1521 priming plus 2 boosts with gp120 protein was superior to 2 gp120 protein inoculations alone, confirming a priming effect for vCP1521. Sporadic weak neutralization of tier 2 viruses was detected only in Vax003 and A3R5 cells. Conclusion. The results suggest either that weak neutralizing antibody responses can be partially protective against HIV-1 in low-risk heterosexual populations or that the modest efficacy seen in RV144 was mediated by other immune responses, either alone or in combination with neutralizing antibodies.


PLOS Computational Biology | 2015

Comprehensive Sieve Analysis of Breakthrough HIV-1 Sequences in the RV144 Vaccine Efficacy Trial

Paul T. Edlefsen; Morgane Rolland; Tomer Hertz; Sodsai Tovanabutra; Andrew J. Gartland; Allan C. deCamp; Craig A. Magaret; Hasan Ahmed; Raphael Gottardo; Michal Juraska; Connor O. McCoy; Brendan B. Larsen; Eric Sanders-Buell; Chris Carrico; Sergey Menis; Meera Bose; Rv Sequencing Team; Miguel A. Arroyo; Robert J. O’Connell; Sorachai Nitayaphan; Punnee Pitisuttithum; Jaranit Kaewkungwal; Supachai Rerks-Ngarm; Merlin L. Robb; Tatsiana Kirys; Ivelin S. Georgiev; Peter D. Kwong; Konrad Scheffler; Sergei L. Kosakovsky Pond; Jonathan M. Carlson

The RV144 clinical trial showed the partial efficacy of a vaccine regimen with an estimated vaccine efficacy (VE) of 31% for protecting low-risk Thai volunteers against acquisition of HIV-1. The impact of vaccine-induced immune responses can be investigated through sieve analysis of HIV-1 breakthrough infections (infected vaccine and placebo recipients). A V1/V2-targeted comparison of the genomes of HIV-1 breakthrough viruses identified two V2 amino acid sites that differed between the vaccine and placebo groups. Here we extended the V1/V2 analysis to the entire HIV-1 genome using an array of methods based on individual sites, k-mers and genes/proteins. We identified 56 amino acid sites or “signatures” and 119 k-mers that differed between the vaccine and placebo groups. Of those, 19 sites and 38 k-mers were located in the regions comprising the RV144 vaccine (Env-gp120, Gag, and Pro). The nine signature sites in Env-gp120 were significantly enriched for known antibody-associated sites (p = 0.0021). In particular, site 317 in the third variable loop (V3) overlapped with a hotspot of antibody recognition, and sites 369 and 424 were linked to CD4 binding site neutralization. The identified signature sites significantly covaried with other sites across the genome (mean = 32.1) more than did non-signature sites (mean = 0.9) (p < 0.0001), suggesting functional and/or structural relevance of the signature sites. Since signature sites were not preferentially restricted to the vaccine immunogens and because most of the associations were insignificant following correction for multiple testing, we predict that few of the genetic differences are strongly linked to the RV144 vaccine-induced immune pressure. In addition to presenting results of the first complete-genome analysis of the breakthrough infections in the RV144 trial, this work describes a set of statistical methods and tools applicable to analysis of breakthrough infection genomes in general vaccine efficacy trials for diverse pathogens.


Journal of Virology | 2014

Analysis of HLA A*02 Association with Vaccine Efficacy in the RV144 HIV-1 Vaccine Trial

Andrew J. Gartland; Sue Li; John McNevin; Georgia D. Tomaras; Raphael Gottardo; Holly Janes; Youyi Fong; Daryl Morris; Daniel E. Geraghty; Gustavo H. Kijak; Paul T. Edlefsen; Nicole Frahm; Brendan B. Larsen; Sodsai Tovanabutra; Eric Sanders-Buell; Allan C. deCamp; Craig A. Magaret; Hasan Ahmed; Jodie P. Goodridge; Lennie Chen; Philip Konopa; Snehal Nariya; Julia N. Stoddard; Kim Wong; Hong Zhao; Wenjie Deng; Brandon Maust; Meera Bose; Shana Howell; A Bates

ABSTRACT The RV144 HIV-1 vaccine trial demonstrated partial efficacy of 31% against HIV-1 infection. Studies into possible correlates of protection found that antibodies specific to the V1 and V2 (V1/V2) region of envelope correlated inversely with infection risk and that viruses isolated from trial participants contained genetic signatures of vaccine-induced pressure in the V1/V2 region. We explored the hypothesis that the genetic signatures in V1 and V2 could be partly attributed to selection by vaccine-primed T cells. We performed a T-cell-based sieve analysis of breakthrough viruses in the RV144 trial and found evidence of predicted HLA binding escape that was greater in vaccine versus placebo recipients. The predicted escape depended on class I HLA A*02- and A*11-restricted epitopes in the MN strain rgp120 vaccine immunogen. Though we hypothesized that this was indicative of postacquisition selection pressure, we also found that vaccine efficacy (VE) was greater in A*02-positive (A*02+) participants than in A*02− participants (VE = 54% versus 3%, P = 0.05). Vaccine efficacy against viruses with a lysine residue at site 169, important to antibody binding and implicated in vaccine-induced immune pressure, was also greater in A*02+ participants (VE = 74% versus 15%, P = 0.02). Additionally, a reanalysis of vaccine-induced immune responses that focused on those that were shown to correlate with infection risk suggested that the humoral responses may have differed in A*02+ participants. These exploratory and hypothesis-generating analyses indicate there may be an association between a class I HLA allele and vaccine efficacy, highlighting the importance of considering HLA alleles and host immune genetics in HIV vaccine trials. IMPORTANCE The RV144 trial was the first to show efficacy against HIV-1 infection. Subsequently, much effort has been directed toward understanding the mechanisms of protection. Here, we conducted a T-cell-based sieve analysis, which compared the genetic sequences of viruses isolated from infected vaccine and placebo recipients. Though we hypothesized that the observed sieve effect indicated postacquisition T-cell selection, we also found that vaccine efficacy was greater for participants who expressed HLA A*02, an allele implicated in the sieve analysis. Though HLA alleles have been associated with disease progression and viral load in HIV-1 infection, these data are the first to suggest the association of a class I HLA allele and vaccine efficacy. While these statistical analyses do not provide mechanistic evidence of protection in RV144, they generate testable hypotheses for the HIV vaccine community and they highlight the importance of assessing the impact of host immune genetics in vaccine-induced immunity and protection. (This study has been registered at ClinicalTrials.gov under registration no. NCT00223080.)


PLOS Pathogens | 2013

HIV-1 Vaccine-Induced T-Cell Reponses Cluster in Epitope Hotspots that Differ from Those Induced in Natural Infection with HIV-1

Tomer Hertz; Hasan Ahmed; David P. Friedrich; Danilo R. Casimiro; Steven G. Self; Lawrence Corey; M. Juliana McElrath; Susan Buchbinder; Helen Horton; Nicole Frahm; Michael N. Robertson; Barney S. Graham; Peter B. Gilbert

Several recent large clinical trials evaluated HIV vaccine candidates that were based on recombinant adenovirus serotype 5 (rAd-5) vectors expressing HIV-derived antigens. These vaccines primarily elicited T-cell responses, which are known to be critical for controlling HIV infection. In the current study, we present a meta-analysis of epitope mapping data from 177 participants in three clinical trials that tested two different HIV vaccines: MRKAd-5 HIV and VRC-HIVAD014-00VP. We characterized the population-level epitope responses in these trials by generating population-based epitope maps, and also designed such maps using a large cohort of 372 naturally infected individuals. We used these maps to address several questions: (1) Are vaccine-induced responses randomly distributed across vaccine inserts, or do they cluster into immunodominant epitope hotspots? (2) Are the immunodominance patterns observed for these two vaccines in three vaccine trials different from one another? (3) Do vaccine-induced hotspots overlap with epitope hotspots induced by chronic natural infection with HIV-1? (4) Do immunodominant hotspots target evolutionarily conserved regions of the HIV genome? (5) Can epitope prediction methods be used to identify these hotspots? We found that vaccine responses clustered into epitope hotspots in all three vaccine trials and some of these hotspots were not observed in chronic natural infection. We also found significant differences between the immunodominance patterns generated in each trial, even comparing two trials that tested the same vaccine in different populations. Some of the vaccine-induced immunodominant hotspots were located in highly variable regions of the HIV genome, and this was more evident for the MRKAd-5 HIV vaccine. Finally, we found that epitope prediction methods can partially predict the location of vaccine-induced epitope hotspots. Our findings have implications for vaccine design and suggest a framework by which different vaccine candidates can be compared in early phases of evaluation.


The Journal of Infectious Diseases | 2016

Vaccination With Heterologous HIV-1 Envelope Sequences and Heterologous Adenovirus Vectors Increases T-Cell Responses to Conserved Regions: HVTN 083

Stephen R. Walsh; Zoe Moodie; Andrew J. Fiore-Gartland; Cecilia Morgan; Marissa B. Wilck; Scott M. Hammer; Susan Buchbinder; Spyros A. Kalams; Paul A. Goepfert; Mark J. Mulligan; Michael C. Keefer; Lindsey R. Baden; Edith Swann; Shannon Grant; Hasan Ahmed; Fusheng Li; Tomer Hertz; Steven G. Self; David P. Friedrich; Nicole Frahm; Hua-Xin Liao; David C. Montefiori; Georgia D. Tomaras; M. Juliana McElrath; John Hural; Barney S. Graham; Xia Jin

BACKGROUND Increasing the breadth of human immunodeficiency virus type 1 (HIV-1) vaccine-elicited immune responses or targeting conserved regions may improve coverage of circulating strains. HIV Vaccine Trials Network 083 tested whether cellular immune responses with these features are induced by prime-boost strategies, using heterologous vectors, heterologous inserts, or a combination of both. METHODS A total of 180 participants were randomly assigned to receive combinations of adenovirus vectors (Ad5 or Ad35) and HIV-1 envelope (Env) gene inserts (clade A or B) in a prime-boost regimen. RESULTS T-cell responses to heterologous and homologous insert regimens targeted a similar number of epitopes (ratio of means, 1.0; 95% confidence interval [CI], .6-1.6; P = .91), but heterologous insert regimens induced significantly more epitopes that were shared between EnvA and EnvB than homologous insert regimens (ratio of means, 2.7; 95% CI, 1.2-5.7; P = .01). Participants in the heterologous versus homologous insert groups had T-cell responses that targeted epitopes with greater evolutionary conservation (mean entropy [±SD], 0.32 ± 0.1 bits; P = .003), and epitopes recognized by responders provided higher coverage (49%; P = .035). Heterologous vector regimens had higher numbers of total, EnvA, and EnvB epitopes than homologous vector regimens (P = .02, .044, and .045, respectively). CONCLUSIONS These data demonstrate that vaccination with heterologous insert prime boosting increased T-cell responses to shared epitopes, while heterologous vector prime boosting increased the number of T-cell epitopes recognized. CLINICAL TRIALS REGISTRATION NCT01095224.


PLOS ONE | 2017

Sieve analysis of breakthrough HIV-1 sequences in HVTN 505 identifies vaccine pressure targeting the CD4 binding site of Env-gp120

Allan C. deCamp; Morgane Rolland; Paul T. Edlefsen; Eric Sanders-Buell; Breana Hall; Craig A. Magaret; Andrew J. Fiore-Gartland; Michal Juraska; Lindsay N. Carpp; Shelly Karuna; Meera Bose; Steven LePore; Shana Miller; Annemarie O'Sullivan; Kultida Poltavee; Hongjun Bai; Kalpana Dommaraju; Hong Zhao; Kim Wong; Lennie Chen; Hasan Ahmed; Derrick Goodman; Matthew Zirui Tay; Raphael Gottardo; Richard A. Koup; Robert T. Bailer; John R. Mascola; Barney S. Graham; Mario Roederer; Robert J. O’Connell

Although the HVTN 505 DNA/recombinant adenovirus type 5 vector HIV-1 vaccine trial showed no overall efficacy, analysis of breakthrough HIV-1 sequences in participants can help determine whether vaccine-induced immune responses impacted viruses that caused infection. We analyzed 480 HIV-1 genomes sampled from 27 vaccine and 20 placebo recipients and found that intra-host HIV-1 diversity was significantly lower in vaccine recipients (P ≤ 0.04, Q-values ≤ 0.09) in Gag, Pol, Vif and envelope glycoprotein gp120 (Env-gp120). Furthermore, Env-gp120 sequences from vaccine recipients were significantly more distant from the subtype B vaccine insert than sequences from placebo recipients (P = 0.01, Q-value = 0.12). These vaccine effects were associated with signatures mapping to CD4 binding site and CD4-induced monoclonal antibody footprints. These results suggest either (i) no vaccine efficacy to block acquisition of any viral genotype but vaccine-accelerated Env evolution post-acquisition; or (ii) vaccine efficacy against HIV-1s with Env sequences closest to the vaccine insert combined with increased acquisition due to other factors, potentially including the vaccine vector.


Vaccine | 2016

A study of vaccine-induced immune pressure on breakthrough infections in the Phambili phase 2b HIV-1 vaccine efficacy trial.

Tomer Hertz; M.G. Logan; Morgane Rolland; Craig A. Magaret; Cecilia Rademeyer; Andrew J. Fiore-Gartland; Paul T. Edlefsen; Allan C. deCamp; Hasan Ahmed; Nobubelo Ngandu; Brendan B. Larsen; Nicole Frahm; Jinny C. Marais; R. Thebus; Daniel E. Geraghty; John Hural; Larry Corey; James G. Kublin; Glenda Gray; M.J. McElrath; James I. Mullins; Peter B. Gilbert; Carolyn Williamson

INTRODUCTION The Merck Adenovirus-5 Gag/Pol/Nef HIV-1 subtype-B vaccine evaluated in predominately subtype B epidemic regions (Step Study), while not preventing infection, exerted vaccine-induced immune pressure on HIV-1 breakthrough infections. Here we investigated if the same vaccine exerted immune pressure when tested in the Phambili Phase 2b study in a subtype C epidemic. MATERIALS AND METHODS A sieve analysis, which compares breakthrough viruses from placebo and vaccine arms, was performed on 277 near full-length genomes generated from 23 vaccine and 20 placebo recipients. Vaccine coverage was estimated by computing the percentage of 9-mers that were exact matches to the vaccine insert. RESULTS There was significantly greater protein distances from the vaccine immunogen sequence in Gag (p=0.045) and Nef (p=0.021) in viruses infecting vaccine recipients compared to placebo recipients. Twenty-seven putative sites of vaccine-induced pressure were identified (p<0.05) in Gag (n=10), Pol (n=7) and Nef (n=10), although they did not remain significant after adjustment for multiple comparisons. We found the epitope sieve effect in Step was driven by HLA A∗02:01; an allele which was found in low frequency in Phambili participants compared to Step participants. Furthermore, the coverage of the vaccine against subtype C Phambili viruses was 31%, 46% and 14% for Gag, Pol and Nef, respectively, compared to subtype B Step virus coverage of 56%, 61% and 26%, respectively. DISCUSSION This study presents evidence of sieve effects in Gag and Nef; however could not confirm effects on specific amino acid sites. We propose that this weaker signal of vaccine immune pressure detected in the Phambili study compared to the Step study may have been influenced by differences in host genetics (HLA allele frequency) and reduced impact of vaccine-induced immune responses due to mismatch between the viral subtype in the vaccine and infecting subtypes.


Retrovirology | 2012

T-cell based sieve analysis ties HLA A*02 to vaccine efficacy and IgA-C1 immune correlate in RV144 Thai trial

Tomer Hertz; Andrew J. Gartland; Holly Janes; Sue Li; Youyi Fong; Georgia D. Tomaras; Daryl Morris; Daniel E. Geraghty; Gustavo H. Kijak; Paul T. Edlefsen; Morgane Rolland; Brendan B. Larsen; Sodsai Tovanabutra; Eric Sanders-Buell; Allan C. deCamp; Craig A. Magaret; Hasan Ahmed; Snehal Nariya; Kim Wong; Hong Zhao; Wenjie Deng; Brandon Maust; Meera Bose; Shana Howell; Michelle Lazzaro; A Bates; Esther Lei; Andrea Bradfield; Grace Ibitamuno; Vatcharain Assawadarachai

T-cell based sieve analysis ties HLA A*02 to vaccine efficacy and IgA-C1 immune correlate in RV144 Thai trial T Hertz, A Gartland, H Janes, S Li, Y Fong, GD Tomaras, D Morris, D Geraghty, GH Kijak, PT Edlefsen, M Rolland, BB Larsen, S Tovanabutra, E Sanders-Buell, AC DeCamp, CA Magaret, H Ahmed, S Nariya, K Wong, H Zhao, W Deng, BS Maust, M Bose, S Howell, M Lazzaro, A Bates, E Lei, A Bradfield, G Ibitamuno, V Assawadarachai, RJ O’Connel, MS deSouza, S Nitayaphan, S Rerks-Ngarm, ML Robb, MJ McElrath, BF Haynes, NL Michael, PB Gilbert, JI Mullins, JH Kim


Retrovirology | 2012

Rapid development of cross-clade neutralizing antibody responses after clade B gp120/gp140 protein priming and clade c gp140 protein boosting

Paul Spearman; Georgia D. Tomaras; David C. Montefiori; Yunda Huang; Hasan Ahmed; Marnie Elizaga; John Hural; Julie McElrath; L. Ouedraogo; Michael Pensiero; C. Butler; Spyros A. Kalams; Edgar Turner Overton; Susan W. Barnett

Background Immunization with heterologous Env protein immunogens following an immunologic rest period has the potential to generate cross-clade neutralizing antibody responses. We identified individuals who had received a clade B Env protein with MF59 4-17 years earlier, most in combination with a DNA or ALVAC prime, and administered a clade C protein boost in an open label phase 1 trial. Methods

Collaboration


Dive into the Hasan Ahmed's collaboration.

Top Co-Authors

Avatar

Allan C. deCamp

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Craig A. Magaret

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Eric Sanders-Buell

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Paul T. Edlefsen

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Tomer Hertz

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Meera Bose

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Morgane Rolland

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Sodsai Tovanabutra

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge