Network


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

Hotspot


Dive into the research topics where Nelson L. Michael is active.

Publication


Featured researches published by Nelson L. Michael.


The New England Journal of Medicine | 2009

Vaccination with ALVAC and AIDSVAX to Prevent HIV-1 Infection in Thailand

Supachai Rerks-Ngarm; Punnee Pitisuttithum; Sorachai Nitayaphan; Jaranit Kaewkungwal; Joseph Chiu; Robert Paris; Nakorn Premsri; Chawetsan Namwat; Mark S. de Souza; Elizabeth Adams; Michael Benenson; Sanjay Gurunathan; Jim Tartaglia; John G. McNeil; Donald P. Francis; Donald Stablein; Deborah L. Birx; Supamit Chunsuttiwat; Chirasak Khamboonruang; Thongcharoen P; Merlin L. Robb; Nelson L. Michael; Prayura Kunasol; Jerome H. Kim

BACKGROUND The development of a safe and effective vaccine against the human immunodeficiency virus type 1 (HIV-1) is critical to pandemic control. METHODS In a community-based, randomized, multicenter, double-blind, placebo-controlled efficacy trial, we evaluated four priming injections of a recombinant canarypox vector vaccine (ALVAC-HIV [vCP1521]) plus two booster injections of a recombinant glycoprotein 120 subunit vaccine (AIDSVAX B/E). The vaccine and placebo injections were administered to 16,402 healthy men and women between the ages of 18 and 30 years in Rayong and Chon Buri provinces in Thailand. The volunteers, primarily at heterosexual risk for HIV infection, were monitored for the coprimary end points: HIV-1 infection and early HIV-1 viremia, at the end of the 6-month vaccination series and every 6 months thereafter for 3 years. RESULTS In the intention-to-treat analysis involving 16,402 subjects, there was a trend toward the prevention of HIV-1 infection among the vaccine recipients, with a vaccine efficacy of 26.4% (95% confidence interval [CI], -4.0 to 47.9; P=0.08). In the per-protocol analysis involving 12,542 subjects, the vaccine efficacy was 26.2% (95% CI, -13.3 to 51.9; P=0.16). In the modified intention-to-treat analysis involving 16,395 subjects (with the exclusion of 7 subjects who were found to have had HIV-1 infection at baseline), the vaccine efficacy was 31.2% (95% CI, 1.1 to 52.1; P=0.04). Vaccination did not affect the degree of viremia or the CD4+ T-cell count in subjects in whom HIV-1 infection was subsequently diagnosed. CONCLUSIONS This ALVAC-HIV and AIDSVAX B/E vaccine regimen may reduce the risk of HIV infection in a community-based population with largely heterosexual risk. Vaccination did not affect the viral load or CD4+ count in subjects with HIV infection. Although the results show only a modest benefit, they offer insight for future research. (ClinicalTrials.gov number, NCT00223080.)


The New England Journal of Medicine | 2012

Immune-Correlates Analysis of an HIV-1 Vaccine Efficacy Trial

Barton F. Haynes; Peter B. Gilbert; M. Juliana McElrath; Susan Zolla-Pazner; Georgia D. Tomaras; S. Munir Alam; David T. Evans; David C. Montefiori; Chitraporn Karnasuta; Ruengpueng Sutthent; Hua-Xin Liao; Anthony L. DeVico; George K. Lewis; Constance Williams; Abraham Pinter; Youyi Fong; Holly Janes; Allan C. deCamp; Yunda Huang; Mangala Rao; Erik Billings; Nicos Karasavvas; Merlin L. Robb; Viseth Ngauy; Mark S. de Souza; Robert Paris; Guido Ferrari; Robert T. Bailer; Kelly A. Soderberg; Charla Andrews

BACKGROUND In the RV144 trial, the estimated efficacy of a vaccine regimen against human immunodeficiency virus type 1 (HIV-1) was 31.2%. We performed a case-control analysis to identify antibody and cellular immune correlates of infection risk. METHODS In pilot studies conducted with RV144 blood samples, 17 antibody or cellular assays met prespecified criteria, of which 6 were chosen for primary analysis to determine the roles of T-cell, IgG antibody, and IgA antibody responses in the modulation of infection risk. Assays were performed on samples from 41 vaccinees who became infected and 205 uninfected vaccinees, obtained 2 weeks after final immunization, to evaluate whether immune-response variables predicted HIV-1 infection through 42 months of follow-up. RESULTS Of six primary variables, two correlated significantly with infection risk: the binding of IgG antibodies to variable regions 1 and 2 (V1V2) of HIV-1 envelope proteins (Env) correlated inversely with the rate of HIV-1 infection (estimated odds ratio, 0.57 per 1-SD increase; P=0.02; q=0.08), and the binding of plasma IgA antibodies to Env correlated directly with the rate of infection (estimated odds ratio, 1.54 per 1-SD increase; P=0.03; q=0.08). Neither low levels of V1V2 antibodies nor high levels of Env-specific IgA antibodies were associated with higher rates of infection than were found in the placebo group. Secondary analyses suggested that Env-specific IgA antibodies may mitigate the effects of potentially protective antibodies. CONCLUSIONS This immune-correlates study generated the hypotheses that V1V2 antibodies may have contributed to protection against HIV-1 infection, whereas high levels of Env-specific IgA antibodies may have mitigated the effects of protective antibodies. Vaccines that are designed to induce higher levels of V1V2 antibodies and lower levels of Env-specific IgA antibodies than are induced by the RV144 vaccine may have improved efficacy against HIV-1 infection.


Nature | 2012

Vaccine protection against acquisition of neutralization-resistant SIV challenges in rhesus monkeys

Dan H. Barouch; Jinyan Liu; Hualin Li; Lori F. Maxfield; Peter Abbink; Diana M. Lynch; M. Justin Iampietro; Adam SanMiguel; Michael S. Seaman; Guido Ferrari; Donald N. Forthal; Ilnour Ourmanov; Vanessa M. Hirsch; Angela Carville; Keith G. Mansfield; Donald Stablein; Maria G. Pau; Hanneke Schuitemaker; Jerald C. Sadoff; Erik Billings; Mangala Rao; Merlin L. Robb; Jerome H. Kim; Mary Marovich; Jaap Goudsmit; Nelson L. Michael

Preclinical studies of human immunodeficiency virus type 1 (HIV-1) vaccine candidates have typically shown post-infection virological control, but protection against acquisition of infection has previously only been reported against neutralization-sensitive virus challenges. Here we demonstrate vaccine protection against acquisition of fully heterologous, neutralization-resistant simian immunodeficiency virus (SIV) challenges in rhesus monkeys. Adenovirus/poxvirus and adenovirus/adenovirus-vector-based vaccines expressing SIVSME543 Gag, Pol and Env antigens resulted in an 80% or greater reduction in the per-exposure probability of infection against repetitive, intrarectal SIVMAC251 challenges in rhesus monkeys. Protection against acquisition of infection showed distinct immunological correlates compared with post-infection virological control and required the inclusion of Env in the vaccine regimen. These data demonstrate the proof-of-concept that optimized HIV-1 vaccine candidates can block acquisition of stringent, heterologous, neutralization-resistant virus challenges in rhesus monkeys.


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.


Nature | 2014

Rapid seeding of the viral reservoir prior to SIV viraemia in rhesus monkeys

James B. Whitney; Alison L. Hill; Srisowmya Sanisetty; Pablo Penaloza-MacMaster; Jinyan Liu; Mayuri Shetty; Lily Parenteau; Crystal Cabral; Jennifer Shields; Stephen Blackmore; Jeffrey Y. Smith; Amanda L. Brinkman; Lauren Peter; Sheeba Mathew; Kaitlin M. Smith; Erica N. Borducchi; Daniel I. S. Rosenbloom; Mark G. Lewis; Jillian Hattersley; Bei Li; Joseph Hesselgesser; Romas Geleziunas; Merlin L. Robb; Jerome H. Kim; Nelson L. Michael; Dan H. Barouch

The viral reservoir represents a critical challenge for human immunodeficiency virus type 1 (HIV-1) eradication strategies. However, it remains unclear when and where the viral reservoir is seeded during acute infection and the extent to which it is susceptible to early antiretroviral therapy (ART). Here we show that the viral reservoir is seeded rapidly after mucosal simian immunodeficiency virus (SIV) infection of rhesus monkeys and before systemic viraemia. We initiated suppressive ART in groups of monkeys on days 3, 7, 10 and 14 after intrarectal SIVMAC251 infection. Treatment with ART on day 3 blocked the emergence of viral RNA and proviral DNA in peripheral blood and also substantially reduced levels of proviral DNA in lymph nodes and gastrointestinal mucosa as compared with treatment at later time points. In addition, treatment on day 3 abrogated the induction of SIV-specific humoral and cellular immune responses. Nevertheless, after discontinuation of ART following 24 weeks of fully suppressive therapy, virus rebounded in all animals, although the monkeys that were treated on day 3 exhibited a delayed viral rebound as compared with those treated on days 7, 10 and 14. The time to viral rebound correlated with total viraemia during acute infection and with proviral DNA at the time of ART discontinuation. These data demonstrate that the viral reservoir is seeded rapidly after intrarectal SIV infection of rhesus monkeys, during the ‘eclipse’ phase, and before detectable viraemia. This strikingly early seeding of the refractory viral reservoir raises important new challenges for HIV-1 eradication strategies.


Journal of Virology | 2012

Antibody-Dependent Cellular Cytotoxicity-Mediating Antibodies from an HIV-1 Vaccine Efficacy Trial Target Multiple Epitopes and Preferentially Use the VH1 Gene Family

Mattia Bonsignori; Justin Pollara; M. Anthony Moody; Michael D. Alpert; Xi Chen; Kwan-Ki Hwang; Peter B. Gilbert; Ying Huang; Thaddeus C. Gurley; Daniel M. Kozink; Dawn J. Marshall; John F. Whitesides; Chun-Yen Tsao; Jaranit Kaewkungwal; Sorachai Nitayaphan; Punnee Pitisuttithum; Supachai Rerks-Ngarm; Jerome H. Kim; Nelson L. Michael; Georgia D. Tomaras; David C. Montefiori; George K. Lewis; Anthony L. DeVico; David T. Evans; Guido Ferrari; Hua-Xin Liao; Barton F. Haynes

ABSTRACT The ALVAC-HIV/AIDSVAX-B/E RV144 vaccine trial showed an estimated efficacy of 31%. RV144 secondary immune correlate analysis demonstrated that the combination of low plasma anti-HIV-1 Env IgA antibodies and high levels of antibody-dependent cellular cytotoxicity (ADCC) inversely correlate with infection risk. One hypothesis is that the observed protection in RV144 is partially due to ADCC-mediating antibodies. We found that the majority (73 to 90%) of a representative group of vaccinees displayed plasma ADCC activity, usually (96.2%) blocked by competition with the C1 region-specific A32 Fab fragment. Using memory B-cell cultures and antigen-specific B-cell sorting, we isolated 23 ADCC-mediating nonclonally related antibodies from 6 vaccine recipients. These antibodies targeted A32-blockable conformational epitopes (n = 19), a non-A32-blockable conformational epitope (n = 1), and the gp120 Env variable loops (n = 3). Fourteen antibodies mediated cross-clade target cell killing. ADCC-mediating antibodies displayed modest levels of V-heavy (VH) chain somatic mutation (0.5 to 1.5%) and also displayed a disproportionate usage of VH1 family genes (74%), a phenomenon recently described for CD4-binding site broadly neutralizing antibodies (bNAbs). Maximal ADCC activity of VH1 antibodies correlated with mutation frequency. The polyclonality and low mutation frequency of these VH1 antibodies reveal fundamental differences in the regulation and maturation of these ADCC-mediating responses compared to VH1 bNAbs.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Vaccine-induced plasma IgA specific for the C1 region of the HIV-1 envelope blocks binding and effector function of IgG

Georgia D. Tomaras; Guido Ferrari; Xiaoying Shen; S. Munir Alam; Hua-Xin Liao; Justin Pollara; Mattia Bonsignori; M. Anthony Moody; Youyi Fong; Xi Chen; Brigid Poling; Cindo O. Nicholson; Ruijun Zhang; Xiaozhi Lu; Robert Parks; Jaranit Kaewkungwal; Sorachai Nitayaphan; Punnee Pitisuttithum; Supachai Rerks-Ngarm; Peter B. Gilbert; Jerome H. Kim; Nelson L. Michael; David C. Montefiori; Barton F. Haynes

Analysis of correlates of risk of infection in the RV144 HIV-1 vaccine efficacy trial demonstrated that plasma IgG against the HIV-1 envelope (Env) variable region 1 and 2 inversely correlated with risk, whereas HIV-1 Env-specific plasma IgA responses directly correlated with risk. In the secondary analysis, antibody-dependent cellular cytotoxicity (ADCC) was another inverse correlate of risk, but only in the presence of low plasma IgA Env-specific antibodies. Thus, we investigated the hypothesis that IgA could attenuate the protective effect of IgG responses through competition for the same Env binding sites. We report that Env-specific plasma IgA/IgG ratios are higher in infected than in uninfected vaccine recipients in RV144. Moreover, Env-specific IgA antibodies from RV144 vaccinees blocked the binding of ADCC-mediating mAb to HIV-1 Env glycoprotein 120 (gp120). An Env-specific monomeric IgA mAb isolated from an RV144 vaccinee also inhibited the ability of natural killer cells to kill HIV-1–infected CD4+ T cells coated with RV144-induced IgG antibodies. We show that monomeric Env-specific IgA, as part of postvaccination polyclonal antibody response, may modulate vaccine-induced immunity by diminishing ADCC effector function.


Nature | 2016

Vaccine protection against Zika virus from Brazil

Rafael A. Larocca; Peter Abbink; Jean Pierre Schatzmann Peron; Paolo Marinho de Andrade Zanotto; M. Justin Iampietro; Alexander Badamchi-Zadeh; Michael Boyd; David Ng’ang’a; Marinela Kirilova; Ramya Nityanandam; Noe B. Mercado; Zhenfeng Li; Edward T. Moseley; Christine A. Bricault; Erica N. Borducchi; Patricia B. Giglio; David Jetton; George H. Neubauer; Joseph P. Nkolola; Lori F. Maxfield; Rafael De La Barrera; Richard G. Jarman; Kenneth H. Eckels; Nelson L. Michael; Stephen J. Thomas; Dan H. Barouch

Zika virus (ZIKV) is a flavivirus that is responsible for the current epidemic in Brazil and the Americas. ZIKV has been causally associated with fetal microcephaly, intrauterine growth restriction, and other birth defects in both humans and mice. The rapid development of a safe and effective ZIKV vaccine is a global health priority, but very little is currently known about ZIKV immunology and mechanisms of immune protection. Here we show that a single immunization with a plasmid DNA vaccine or a purified inactivated virus vaccine provides complete protection in susceptible mice against challenge with a strain of ZIKV involved in the outbreak in northeast Brazil. This ZIKV strain has recently been shown to cross the placenta and to induce fetal microcephaly and other congenital malformations in mice. We produced DNA vaccines expressing ZIKV pre-membrane and envelope (prM-Env), as well as a series of deletion mutants. The prM-Env DNA vaccine, but not the deletion mutants, afforded complete protection against ZIKV, as measured by absence of detectable viraemia following challenge, and protective efficacy correlated with Env-specific antibody titers. Adoptive transfer of purified IgG from vaccinated mice conferred passive protection, and depletion of CD4 and CD8 T lymphocytes in vaccinated mice did not abrogate this protection. These data demonstrate that protection against ZIKV challenge can be achieved by single-shot subunit and inactivated virus vaccines in mice and that Env-specific antibody titers represent key immunologic correlates of protection. Our findings suggest that the development of a ZIKV vaccine for humans is likely to be achievable.


Science | 2016

Protective efficacy of multiple vaccine platforms against Zika virus challenge in rhesus monkeys

Peter Abbink; Rafael A. Larocca; Rafael De La Barrera; Christine A. Bricault; Edward T. Moseley; Michael Boyd; Marinela Kirilova; Zhenfeng Li; David Ng’ang’a; Ovini Nanayakkara; Ramya Nityanandam; Noe B. Mercado; Erica N. Borducchi; Arshi Agarwal; Amanda L. Brinkman; Crystal Cabral; Abishek Chandrashekar; Patricia B. Giglio; David Jetton; Jessica Jimenez; Benjamin C. Lee; Shanell Mojta; Katherine Molloy; Mayuri Shetty; George H. Neubauer; Kathryn E. Stephenson; Jean Pierre Schatzmann Peron; Paolo Marinho de Andrade Zanotto; Johnathan Misamore; Brad Finneyfrock

Zika virus (ZIKV) is responsible for a major ongoing epidemic in the Americas and has been causally associated with fetal microcephaly. The development of a safe and effective ZIKV vaccine is therefore an urgent global health priority. Here we demonstrate that three different vaccine platforms protect against ZIKV challenge in rhesus monkeys. A purified inactivated virus vaccine induced ZIKV-specific neutralizing antibodies and completely protected monkeys against ZIKV strains from both Brazil and Puerto Rico. Purified immunoglobulin from vaccinated monkeys also conferred passive protection in adoptive transfer studies. A plasmid DNA vaccine and a single-shot recombinant rhesus adenovirus serotype 52 vector vaccine, both expressing ZIKV premembrane and envelope, also elicited neutralizing antibodies and completely protected monkeys against ZIKV challenge. These data support the rapid clinical development of ZIKV vaccines for humans.


Science Translational Medicine | 2014

Vaccine-Induced Env V1–V2 IgG3 Correlates with Lower HIV-1 Infection Risk and Declines Soon After Vaccination

Nicole L. Yates; Hua-Xin Liao; Youyi Fong; Allan C. deCamp; Nathan Vandergrift; William T. Williams; S. Munir Alam; Guido Ferrari; Zhi-Yong Yang; Kelly E. Seaton; Phillip W. Berman; Michael D. Alpert; David T. Evans; Robert J. O’Connell; Donald P. Francis; Faruk Sinangil; Carter Lee; Sorachai Nitayaphan; Supachai Rerks-Ngarm; Jaranit Kaewkungwal; Punnee Pitisuttithum; James Tartaglia; Abraham Pinter; Susan Zolla-Pazner; Peter B. Gilbert; Gary J. Nabel; Nelson L. Michael; Jerome H. Kim; David C. Montefiori; Barton F. Haynes

A V1-V2 IgG3 response to HIV correlates with a decreased risk of HIV-1 infection and is one vaccine-induced humoral response that is higher in a clinical trial showing HIV-1 vaccine efficacy compared to a trial showing nonefficacy. Env IgG3 Takes Center Stage Only one HIV-1 vaccine trial (RV144), to date, has demonstrated some level of vaccine efficacy. IgG antibodies to the V1-V2 region of the HIV-1 envelope correlated with decreased HIV-1 risk. However, a previous vaccine trial (VAX003) also induced these types of antibodies but failed to demonstrate efficacy, thus raising the question about whether the quality of the V1-V2 IgG response and the context of other immune responses were important. Yates et al. report that these two trials did induce a qualitatively distinct antibody subclass response, with more V1V2 IgG3 responses and correlations with antiviral function induced by the partially efficacious RV144 vaccine regimen compared to the VAX003 vaccine regimen that lacks efficacy. The authors then demonstrated that these specific IgG3 antibodies correlated with a decreased risk of infection in a placebo-controlled, blinded study of RV144 vaccinees with and without subsequent HIV-1 infection. Vaccine-induced HIV-1 antibody subclass profiles, specifically Env IgG3, should be evaluated in future HIV-1 vaccine efficacy trials to further refine immune correlates of protection. HIV-1–specific immunoglobulin G (IgG) subclass antibodies bind to distinct cellular Fc receptors. Antibodies of the same epitope specificity but of a different subclass therefore can have different antibody effector functions. The study of IgG subclass profiles between different vaccine regimens used in clinical trials with divergent efficacy outcomes can provide information on the quality of the vaccine-induced B cell response. We show that HIV-1–specific IgG3 distinguished two HIV-1 vaccine efficacy studies (RV144 and VAX003 clinical trials) and correlated with decreased risk of HIV-1 infection in a blinded follow-up case-control study with the RV144 vaccine. HIV-1–specific IgG3 responses were not long-lived, which was consistent with the waning efficacy of the RV144 vaccine. These data suggest that specific vaccine-induced HIV-1 IgG3 should be tested in future studies of immune correlates in HIV-1 vaccine efficacy trials.

Collaboration


Dive into the Nelson L. Michael's collaboration.

Top Co-Authors

Avatar

Merlin L. Robb

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Jerome H. Kim

International Vaccine Institute

View shared research outputs
Top Co-Authors

Avatar

Supachai Rerks-Ngarm

Thailand Ministry of Public Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sodsai Tovanabutra

Henry M. Jackson Foundation for the Advancement of Military Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric Sanders-Buell

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Mary Marovich

Walter Reed Army Institute of Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge