Network


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

Hotspot


Dive into the research topics where James B. Whitney is active.

Publication


Featured researches published by James B. Whitney.


Nature | 2013

Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys

Dan H. Barouch; James B. Whitney; Brian Moldt; Florian Klein; Thiago Y. Oliveira; Jinyan Liu; Kathryn E. Stephenson; Hui-Wen Chang; Karthik Shekhar; Sanjana Gupta; Joseph P. Nkolola; Michael S. Seaman; Kaitlin M. Smith; Erica N. Borducchi; Crystal Cabral; Jeffrey Y. Smith; Stephen Blackmore; Srisowmya Sanisetty; James R. Perry; Matthew Beck; Mark G. Lewis; William Rinaldi; Arup K. Chakraborty; Pascal Poignard; Michel C. Nussenzweig; Dennis R. Burton

Human immunodeficiency virus type 1 (HIV-1)-specific monoclonal antibodies with extraordinary potency and breadth have recently been described. In humanized mice, combinations of monoclonal antibodies have been shown to suppress viraemia, but the therapeutic potential of these monoclonal antibodies has not yet been evaluated in primates with an intact immune system. Here we show that administration of a cocktail of HIV-1-specific monoclonal antibodies, as well as the single glycan-dependent monoclonal antibody PGT121, resulted in a rapid and precipitous decline of plasma viraemia to undetectable levels in rhesus monkeys chronically infected with the pathogenic simian–human immunodeficiency virus SHIV-SF162P3. A single monoclonal antibody infusion afforded up to a 3.1 log decline of plasma viral RNA in 7 days and also reduced proviral DNA in peripheral blood, gastrointestinal mucosa and lymph nodes without the development of viral resistance. Moreover, after monoclonal antibody administration, host Gag-specific T-lymphocyte responses showed improved functionality. Virus rebounded in most animals after a median of 56 days when serum monoclonal antibody titres had declined to undetectable levels, although, notably, a subset of animals maintained long-term virological control in the absence of further monoclonal antibody infusions. These data demonstrate a profound therapeutic effect of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys as well as an impact on host immune responses. Our findings strongly encourage the investigation of monoclonal antibody therapy for HIV-1 in humans.


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.


Science Translational Medicine | 2011

Immune and Genetic Correlates of Vaccine Protection Against Mucosal Infection by SIV in Monkeys.

Norman L. Letvin; Srinivas S. Rao; David C. Montefiori; Michael S. Seaman; Yue Sun; So-Yon Lim; Wendy W. Yeh; Mohammed Asmal; Rebecca Gelman; Ling Shen; James B. Whitney; Cathal Seoighe; Miguel Lacerda; Sheila M. Keating; Philip J. Norris; Michael G. Hudgens; Peter B. Gilbert; Adam P. Buzby; Linh Mach; Jinrong Zhang; Harikrishnan Balachandran; George M. Shaw; Stephen D. Schmidt; John Paul Todd; Alan Dodson; John R. Mascola; Gary J. Nabel

A vaccine protecting monkeys against mucosal infection by simian immunodeficiency virus sheds light on immune and genetic correlates of protection. Unraveling Immune Correlates of Vaccine Protection Developing an effective vaccine against HIV-1, the virus that causes AIDS, has been a huge challenge that has stymied AIDS researchers for several decades. A key problem for HIV vaccine trials has been the lack of immune correlates that indicate which antibody and T cell responses in the vaccinees correlate directly with a protective effect. The only HIV vaccine trial to date that has shown a protective effect is the RV144 trial carried out in Thailand between 2003 and 2006, with the final results reported in 2009. In this trial of 16,400 Thai volunteers, those vaccinated with a prime-boost HIV vaccine showed a reduction in the rate of infection by HIV-1 of 31% compared to volunteers given a placebo. The protective effect was seen for up to 3 years after the initial vaccination, but the immune correlates of protection by this vaccine are still not known. In an effort to learn more about possible immune correlates of HIV vaccine protection, Letvin and colleagues used a prime/boost vaccine regimen in monkeys that was similar to that used in the RV144 trial. Monkeys were vaccinated with a plasmid DNA prime/recombinant adenovirus serotype 5 (rAd5) boost vaccine regimen and then were challenged with intrarectal doses of one of two isolates of the simian immunodeficiency virus (SIV) every week for 12 weeks. Although the vaccine had no impact on acquisition of the SIVmac251 isolate (which is tough for the monkey immune system to neutralize), the vaccine provided a 50% reduction in infection with the SIVsmE660 isolate (which more readily undergoes neutralization). The authors then examined a variety of immune responses in the protected vaccinated monkeys including cellular, antibody, and innate immune responses; they also examined whether protective host alleles were present in the protected animals. They found that low levels of neutralizing antibodies and a CD4+ T cell response against the HIV envelope (Env) protein correlated with the protective effect. In addition, monkeys that expressed two TRIM5 alleles that help to restrict SIV replication in host cells were protected by the vaccine, whereas monkeys expressing one TRIM5 allele that is permissive for SIV replication were not. This study begins to unravel the immune and genetic correlates of protection in nonhuman primates and highlights the need to scrutinize these types of correlates in future trials of HIV vaccines in human volunteers. The RV144 vaccine trial in Thailand demonstrated that an HIV vaccine could prevent infection in humans and highlights the importance of understanding protective immunity against HIV. We used a nonhuman primate model to define immune and genetic mechanisms of protection against mucosal infection by the simian immunodeficiency virus (SIV). A plasmid DNA prime/recombinant adenovirus serotype 5 (rAd5) boost vaccine regimen was evaluated for its ability to protect monkeys from infection by SIVmac251 or SIVsmE660 isolates after repeat intrarectal challenges. Although this prime-boost vaccine regimen failed to protect against SIVmac251 infection, 50% of vaccinated monkeys were protected from infection with SIVsmE660. Among SIVsmE660-infected animals, there was about a one-log reduction in peak plasma virus RNA in monkeys expressing the major histocompatibility complex class I allele Mamu-A*01, implicating cytotoxic T lymphocytes in the control of SIV replication once infection is established. Among Mamu-A*01–negative monkeys challenged with SIVsmE660, no CD8+ T cell response or innate immune response was associated with protection against virus acquisition. However, low levels of neutralizing antibodies and an envelope-specific CD4+ T cell response were associated with vaccine protection in these monkeys. Moreover, monkeys that expressed two TRIM5 alleles that restrict SIV replication were more likely to be protected from infection than monkeys that expressed at least one permissive TRIM5 allele. This study begins to elucidate the mechanisms of vaccine protection against immunodeficiency viruses and highlights the need to analyze these immune and genetic correlates of protection in future trials of HIV vaccine strategies.


Science | 2015

Protective Efficacy of Adenovirus/Protein Vaccines Against SIV Challenges in Rhesus Monkeys

Dan H. Barouch; Galit Alter; Thomas A. Broge; Caitlyn Linde; Margaret E. Ackerman; Eric P. Brown; Erica N. Borducchi; Kaitlin M. Smith; Joseph P. Nkolola; Jinyan Liu; Jennifer Shields; Lily Parenteau; James B. Whitney; Peter Abbink; David Ng’ang’a; Michael S. Seaman; Christy L. Lavine; James R. Perry; Wenjun Li; Arnaud D. Colantonio; Mark G. Lewis; Bing Chen; Holger Wenschuh; Ulf Reimer; Michael Piatak; Jeffrey D. Lifson; Scott A. Handley; Herbert W. Virgin; Marguerite Koutsoukos; Clarisse Lorin

To defeat SIV, add a protein boost Despite 30 years of effort, no HIV-1 vaccine exists. Barouch et al. evaluated one promising strategy in rhesus macaques, a preclinical model commonly used to test potential HIV-1 vaccine candidates. They immunized monkeys with adenovirus-36 vectors engineered to express SIV (simian immunodeficiency virus) genes and then boosted them with a recombinant gp120 envelope glycoprotein (Env) from SIV. This regimen afforded greater protection than a strategy that instead used a viral vector–based boost. A parallel trial using a SHIV (simian/human immunodeficiency virus)–based vaccine and challenge model produced similar results. Whether this particular approach will be equally successful in humans remains to be tested. Science, this issue p. 320 A viral vector–recombinant envelope glycoprotein–based HIV-1 vaccine strategy protected 50% of monkeys from infection. Preclinical studies of viral vector–based HIV-1 vaccine candidates have previously shown partial protection against neutralization-resistant virus challenges in rhesus monkeys. In this study, we evaluated the protective efficacy of adenovirus serotype 26 (Ad26) vector priming followed by purified envelope (Env) glycoprotein boosting. Rhesus monkeys primed with Ad26 vectors expressing SIVsmE543 Env, Gag, and Pol and boosted with AS01B-adjuvanted SIVmac32H Env gp140 demonstrated complete protection in 50% of vaccinated animals against a series of repeated, heterologous, intrarectal SIVmac251 challenges that infected all controls. Protective efficacy correlated with the functionality of Env-specific antibody responses. Comparable protection was also observed with a similar Ad/Env vaccine against repeated, heterologous, intrarectal SHIV-SF162P3 challenges. These data demonstrate robust protection by Ad/Env vaccines against acquisition of neutralization-resistant virus challenges in rhesus monkeys.


Journal of Virology | 2006

Molecularly Cloned SHIV-1157ipd3N4: a Highly Replication- Competent, Mucosally Transmissible R5 Simian-Human Immunodeficiency Virus Encoding HIV Clade C env

Ruijiang Song; Agnès-Laurence Chenine; Robert A. Rasmussen; C. R. Ruprecht; S. Mirshahidi; Ricky D. Grisson; Weidong Xu; James B. Whitney; L. M. Goins; Helena Ong; Pei-Lin Li; E. Shai-Kobiler; T. Wang; C. M. McCann; Hong Zhang; Charles Wood; C. Kankasa; W. E. Secor; Harold M. McClure; E. Strobert; James G. Else; Ruth M. Ruprecht

ABSTRACT Human immunodeficiency virus type 1 (HIV-1) clade C causes >50% of all HIV infections worldwide, and an estimated 90% of all transmissions occur mucosally with R5 strains. A pathogenic R5 simian-human immunodeficiency virus (SHIV) encoding HIV clade C env is highly desirable to evaluate candidate AIDS vaccines in nonhuman primates. To this end, we generated SHIV-1157i, a molecular clone from a Zambian infant isolate that carries HIV clade C env. SHIV-1157i was adapted by serial passage in five monkeys, three of which developed peripheral CD4+ T-cell depletion. After the first inoculated monkey developed AIDS at week 137 postinoculation, transfer of its infected blood to a naïve animal induced memory T-cell depletion and thrombocytopenia within 3 months in the recipient. In parallel, genomic DNA from the blood donor was amplified to generate the late proviral clone SHIV-1157ipd3. To increase the replicative capacity of SHIV-1157ipd3, an extra NF-κB binding site was engineered into its 3′ long terminal repeat, giving rise to SHIV-1157ipd3N4. This virus was exclusively R5 tropic and replicated more potently in rhesus peripheral blood mononuclear cells than SHIV-1157ipd3 in the presence of tumor necrosis factor alpha. Rhesus macaques of Indian and Chinese origin were next inoculated intrarectally with SHIV-1157ipd3N4; this virus replicated vigorously in both sets of monkeys. We conclude that SHIV-1157ipd3N4 is a highly replication-competent, mucosally transmissible R5 SHIV that represents a valuable tool to test candidate AIDS vaccines targeting HIV-1 clade C Env.


PLOS Pathogens | 2010

TRIM5α Modulates Immunodeficiency Virus Control in Rhesus Monkeys

So-Yon Lim; Thomas Rogers; Tiffany Chan; James B. Whitney; Jonghwa Kim; Joseph Sodroski; Norman L. Letvin

The cytoplasmic TRIM5α proteins of certain mammalian lineages efficiently recognize the incoming capsids of particular retroviruses and potently restrict infection in a species-specific manner. Successful retroviruses have evolved capsids that are less efficiently recognized by the TRIM5α proteins of the natural hosts. To address whether TRIM5α contributes to the outcome of retroviral infection in a susceptible host species, we investigated the impact of TRIM5 polymorphisms in rhesus monkeys on the course of a simian immunodeficiency virus (SIV) infection. Full-length TRIM5α cDNAs were derived from each of 79 outbred monkeys and sequenced. Associations were explored between the expression of particular TRIM5 alleles and both the permissiveness of cells to SIV infection in vitro and clinical sequelae of SIV infection in vivo. Natural variation in the TRIM5α B30.2(SPRY) domain influenced the efficiency of SIVmac capsid binding and the in vitro susceptibility of cells from the monkeys to SIVmac infection. We also show the importance in vivo of the interaction of SIVmac with different allelic forms of TRIM5, demonstrating that particular alleles are associated with as much as 1.3 median log difference in set-point viral loads in SIVmac-infected rhesus monkeys. Moreover, these allelic forms of TRIM5 were associated with the extent of loss of central memory (CM) CD4+ T cells and the rate of progression to AIDS in the infected monkeys. These findings demonstrate a central role for TRIM5α in limiting the replication of an immunodeficiency virus infection in a primate host.


Nature Medicine | 2016

Zika viral dynamics and shedding in rhesus and cynomolgus macaques

Christa E. Osuna; So-Yon Lim; Claire Deleage; Bryan D. Griffin; Derek R. Stein; Lukas T. Schroeder; Robert W. Omange; Katharine Best; Ma Luo; Peter Hraber; Hanne Andersen-Elyard; Erwing Fabian Cardozo Ojeda; Scott Huang; Dana L. Vanlandingham; Stephen Higgs; Alan S. Perelson; Jacob D. Estes; David Safronetz; Mark G. Lewis; James B. Whitney

Infection with Zika virus has been associated with serious neurological complications and fetal abnormalities. However, the dynamics of viral infection, replication and shedding are poorly understood. Here we show that both rhesus and cynomolgus macaques are highly susceptible to infection by lineages of Zika virus that are closely related to, or are currently circulating in, the Americas. After subcutaneous viral inoculation, viral RNA was detected in blood plasma as early as 1 d after infection. Viral RNA was also detected in saliva, urine, cerebrospinal fluid (CSF) and semen, but transiently in vaginal secretions. Although viral RNA during primary infection was cleared from blood plasma and urine within 10 d, viral RNA was detectable in saliva and seminal fluids until the end of the study, 3 weeks after the resolution of viremia in the blood. The control of primary Zika virus infection in the blood was correlated with rapid innate and adaptive immune responses. We also identified Zika RNA in tissues, including the brain and male and female reproductive tissues, during early and late stages of infection. Re-infection of six animals 45 d after primary infection with a heterologous strain resulted in complete protection, which suggests that primary Zika virus infection elicits protective immunity. Early invasion of Zika virus into the nervous system of healthy animals and the extent and duration of shedding in saliva and semen underscore possible concern for additional neurologic complications and nonarthropod-mediated transmission in humans.


Current Opinion in Infectious Diseases | 2004

Live attenuated HIV vaccines: pitfalls and prospects.

James B. Whitney; Ruth M. Ruprecht

Purpose of review When simian immunodeficiency virus (SIV) deleted in the nef gene caused no disease in macaques and provided protection against wild-type SIV challenge, hopes were high that the removal of nef would convert a pathogenic immunodeficiency virus into a live attenuated vaccine. We seek to highlight recent studies focused on several major issues regarding live attenuated AIDS viruses as vaccine candidates: (1) safety, (2) efficacy, (3) the correlates of immune protection, and (4) the molecular determinants for lentiviral virulence or attenuation. Recent findings Nef-deletion mutants have retained virulence; compared with wild-type SIV, disease progression was slowed but not abrogated. After long-term observation, all adult macaques given SIVmac239Δ3 exhibited immune dysfunction; over 50% had T-cell depletion, and 18% developed AIDS. Vaccine efficacy has been disappointing, with limited or no cross-protection and no protection against homologous virus challenge years after initial vaccination. To date, the correlates of protective immunity have defied precise definition; no dominant mechanism has yet emerged. Data from passive serum transfer and CD8+ T-cell depletion studies have raised the possibility that alternate mechanism of protection may be operative. Due to relentless viral replication and continuous selective pressure, initially benign viruses can generate virulent progeny with unpredictable genotypes. Summary Neither safety nor efficacy of the current live attenuated primate immunodeficiency virus vaccines has withstood the test of time. However, such viruses are invaluable tools to address two key questions: (1) what are the correlates of protection, and (2) what are the molecular determinants of viral immunopathogenesis?


Journal of Virology | 2003

A Structurally Disordered Region at the C Terminus of Capsid Plays Essential Roles in Multimerization and Membrane Binding of the Gag Protein of Human Immunodeficiency Virus Type 1

Chen Liang; Jing Hu; James B. Whitney; Lawrence Kleiman; Mark A. Wainberg

ABSTRACT Crystal structures of human immunodeficiency virus type 1 (HIV-1) capsid protein (CA) reveal that the last 11 C-terminal amino acids are disordered. This disordered region contains a glycine-rich sequence 353-GVGGP-357 (numbering refers to the initiation methionine of Gag) that is highly conserved within the Gag proteins of HIV-1, HIV-2, and simian immunodeficiency virus, which suggests the importance of this sequence in virus replication. In the present study, we demonstrate that changing any individual residue within this short region in the context of the full-length HIV-1 genome virtually abolishes production of extracellular virus particles, in either the presence or absence of viral protease activity. This severe defect in virus particle production results from impaired Gag multimerization, as well as from decreased Gag association with the cellular membranes, as demonstrated by the results of gradient sedimentation and membrane flotation centrifugation assays. These findings are further supported by the diffuse distribution pattern of the mutant Gag within the cytoplasm, as opposed to the punctate distribution of the wild-type Gag on the plasma membrane. On the basis of these results, we propose that the disordered feature of amino acid stretch 353-GVGGP-357 in the CA crystal forms may have allowed Gag to adopt multiple conformations and that such structural flexibility is needed by Gag in order to construct geometrically complex particles.


PLOS Pathogens | 2016

A Subset of Latency-Reversing Agents Expose HIV-Infected Resting CD4+ T-Cells to Recognition by Cytotoxic T-Lymphocytes.

R. Brad Jones; Stefanie Mueller; Rachel O’Connor; Katherine Rimpel; Derek D. Sloan; Dan Karel; Hing C. Wong; Emily K. Jeng; Allison S. Thomas; James B. Whitney; So-Yon Lim; Colin Kovacs; Erika Benko; Sara Karandish; Szu-han Huang; Maria J. Buzon; Mathias Lichterfeld; Alivelu Irrinki; Jeffrey P. Murry; Angela Tsai; Helen Yu; Romas Geleziunas; Alicja Trocha; Mario A. Ostrowski; Darrell J. Irvine; Bruce D. Walker

Resting CD4+ T-cells harboring inducible HIV proviruses are a critical reservoir in antiretroviral therapy (ART)-treated subjects. These cells express little to no viral protein, and thus neither die by viral cytopathic effects, nor are efficiently cleared by immune effectors. Elimination of this reservoir is theoretically possible by combining latency-reversing agents (LRAs) with immune effectors, such as CD8+ T-cells. However, the relative efficacy of different LRAs in sensitizing latently-infected cells for recognition by HIV-specific CD8+ T-cells has not been determined. To address this, we developed an assay that utilizes HIV-specific CD8+ T-cell clones as biosensors for HIV antigen expression. By testing multiple CD8+ T-cell clones against a primary cell model of HIV latency, we identified several single agents that primed latently-infected cells for CD8+ T-cell recognition, including IL-2, IL-15, two IL-15 superagonists (IL-15SA and ALT-803), prostratin, and the TLR-2 ligand Pam3CSK4. In contrast, we did not observe CD8+ T-cell recognition of target cells following treatment with histone deacetylase inhibitors or with hexamethylene bisacetamide (HMBA). In further experiments we demonstrate that a clinically achievable concentration of the IL-15 superagonist ‘ALT-803’, an agent presently in clinical trials for solid and hematological tumors, primes the natural ex vivo reservoir for CD8+ T-cell recognition. Thus, our results establish a novel experimental approach for comparative evaluation of LRAs, and highlight ALT-803 as an LRA with the potential to synergize with CD8+ T-cells in HIV eradication strategies.

Collaboration


Dive into the James B. Whitney's collaboration.

Top Co-Authors

Avatar

So-Yon Lim

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Norman L. Letvin

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michael S. Seaman

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peter Hraber

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Dan H. Barouch

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christa E. Osuna

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

John R. Mascola

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Srinivas S. Rao

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mark G. Lewis

Southern Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge