Network


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

Hotspot


Dive into the research topics where Stephen Whitney is active.

Publication


Featured researches published by Stephen Whitney.


Journal of Virology | 2013

Antibodies with High Avidity to the gp120 Envelope Protein in Protection from Simian Immunodeficiency Virus SIVmac251 Acquisition in an Immunization Regimen That Mimics the RV-144 Thai Trial

Poonam Pegu; Monica Vaccari; Shari N. Gordon; Brandon F. Keele; Melvin N. Doster; Yongjun Guan; Guido Ferrari; Ranajit Pal; Maria Grazia Ferrari; Stephen Whitney; Lauren Hudacik; Erik Billings; Mangala Rao; David C. Montefiori; Georgia D. Tomaras; S. Munir Alam; Claudio Fenizia; Jeffrey D. Lifson; Donald Stablein; Jim Tartaglia; Nelson L. Michael; Jerome H. Kim; David Venzon; Genoveffa Franchini

ABSTRACT The recombinant canarypox vector, ALVAC-HIV, together with human immunodeficiency virus (HIV) gp120 envelope glycoprotein, has protected 31.2% of Thai individuals from HIV acquisition in the RV144 HIV vaccine trial. This outcome was unexpected, given the limited ability of the vaccine components to induce CD8+ T-cell responses or broadly neutralizing antibodies. We vaccinated macaques with an immunization regimen intended to mimic the RV144 trial and exposed them intrarectally to a dose of the simian immunodeficiency virus SIVmac251 that transmits few virus variants, similar to HIV transmission to humans. Vaccination induced anti-envelope antibodies in all vaccinees and CD4+ and CD8+ T-cell responses. Three of the 11 macaques vaccinated with ALVAC-SIV/gp120 were protected from SIVmac251 acquisition, but the result was not significant. The remaining vaccinees were infected and progressed to disease. The magnitudes of vaccine-induced SIVmac251-specific T-cell responses and binding antibodies were not significantly different between protected and infected animals. However, sera from protected animals had higher avidity antibodies to gp120, recognized the variable envelope regions V1/V2, and reduced SIVmac251 infectivity in cells that express high levels of α4β7 integrins, suggesting a functional role of antibodies to V2. The current results emphasize the utility of determining the titer of repeated mucosal challenge in the preclinical evaluation of HIV vaccines.


Nature Medicine | 2016

Adjuvant-dependent innate and adaptive immune signatures of risk of SIVmac251 acquisition.

Monica Vaccari; Shari N. Gordon; Slim Fourati; Luca Schifanella; Namal P.M. Liyanage; Mark J. Cameron; Brandon F. Keele; Xiaoying Shen; Georgia D. Tomaras; Erik Billings; Mangala Rao; Amy W. Chung; Karen G. Dowell; Chris Bailey-Kellogg; Eric P. Brown; Margaret E. Ackerman; Diego A. Vargas-Inchaustegui; Stephen Whitney; Melvin N. Doster; Nicolo Binello; Poonam Pegu; David C. Montefiori; Kathryn E. Foulds; David S. Quinn; Mitzi Donaldson; Frank Liang; Karin Loré; Mario Roederer; Richard A. Koup; Adrian B. McDermott

A recombinant vaccine containing Aventis Pasteurs canarypox vector (ALVAC)–HIV and gp120 alum decreased the risk of HIV acquisition in the RV144 vaccine trial. The substitution of alum with the more immunogenic MF59 adjuvant is under consideration for the next efficacy human trial. We found here that an ALVAC–simian immunodeficiency virus (SIV) and gp120 alum (ALVAC–SIV + gp120) equivalent vaccine, but not an ALVAC–SIV + gp120 MF59 vaccine, was efficacious in delaying the onset of SIVmac251 in rhesus macaques, despite the higher immunogenicity of the latter adjuvant. Vaccine efficacy was associated with alum-induced, but not with MF59-induced, envelope (Env)-dependent mucosal innate lymphoid cells (ILCs) that produce interleukin (IL)-17, as well as with mucosal IgG to the gp120 variable region 2 (V2) and the expression of 12 genes, ten of which are part of the RAS pathway. The association between RAS activation and vaccine efficacy was also observed in an independent efficacious SIV-vaccine approach. Whether RAS activation, mucosal ILCs and antibodies to V2 are also important hallmarks of HIV-vaccine efficacy in humans will require further studies.


Vaccine | 1993

Studies on reactogenicity and immunogenicity of attenuated bivalent cold recombinant influenza type A (CRA) and inactivated trivalent influenza virus (TI) vaccines in infants and young children

Pedro A. Piedra; W. Paul Glezen; Innocent N. Mbawuike; William C. Gruber; Barbara D. Baxter; F.James Boland; Richard W. Byrd; Lawrence L. Fan; Jessica K. Lewis; Linda J. Rhodes; Stephen Whitney; Larry H. Taber

Fifty-two infants seronegative to or without prior infection with influenza type A viruses were enrolled in a study to evaluate reactogenicity and immunogenicity of three bivalent cold recombinant type A (CRA) and two trivalent inactivated influenza (TI) vaccines. Controls consisted of infants receiving normal saline by nose drops (Pli.n.) or intramuscularly (Pli.m.). CRA and TI vaccines were monitored for local and systemic reactions after vaccination. Serum specimens obtained prior to and 6 weeks postvaccination were analysed for neutralizing antibody to influenza H1N1 and H3N2 viruses. CRA vaccines and Pli.n. recipients had similar numbers of acute respiratory infections and comparable rates of illnesses during the trial. Significantly fewer CRA vaccinees without an intercurrent viral infection had fever (0/16 versus 4/10, p = 0.04) and cough (4/16 versus 9/10, p = 0.002) than CRA vaccinees with a confirmed intercurrent viral infection. Recipients of TI vaccine and Pli.m. did not develop reactions at the injection site. For each of the CRA vaccines tested, a dominant CRA virus was identified. The dominant CRA viruses were isolated from a greater number of infants or for a longer duration than the non-dominant CRA viruses. All 14 non-dominant CRA viruses were recovered from infants within the first week after vaccination; 24 of 77 dominant CRA viruses were recovered more than 7 days after vaccination. The immunogenicity of CRA vaccines was not affected by a confirmed intercurrent viral infection or low titres of influenza-specific antibody.(ABSTRACT TRUNCATED AT 250 WORDS)


Journal of Medical Primatology | 2005

Polyvalent DNA prime and envelope protein boost HIV-1 vaccine elicits humoral and cellular responses and controls plasma viremia in rhesus macaques following rectal challenge with an R5 SHIV isolate

Ranajit Pal; Shixia Wang; V. S. Kalyanaraman; B.C. Nair; Stephen Whitney; Timothy Keen; Lindsey Hocker; Lauren Hudacik; Nicolas Rose; Anthony D. Cristillo; Innocent Mboudjeka; Siyuan Shen; Te-Hui Wu-Chou; David C. Montefiori; John R. Mascola; Shan Lu; Phillip D. Markham

Abstract:  Immunization of macaques with multivalent DNA encoding gp120 genes from HIV‐1 subtypes A, B, C and E and a gag gene followed by boosting with homologous gp120 proteins elicited strong anti‐gp120 antibodies capable of neutralizing homologous and to a lesser degree heterologous HIV‐1 isolates. Both Env‐ and Gag‐specific cell mediated immune (CMI) responses were detected in the immunized animals. Following rectal challenge with an SHIV isolate encoding HIV‐1Ba‐Lenv, plasma viremia in the infected immunized animals was significantly lower than that observed in the naïve animals. Further, one of six immunized animals was completely protected whereas all six naïve animals were infected. These results demonstrate that a vaccine based on priming with a polyvalent DNA vaccine from multiple HIV‐1 subtypes followed by boosting with homologous Env proteins elicits anti‐HIV‐1 immune responses capable of controlling rectal transmission of SHIVBa‐L.


Virology | 2011

Rapid SIV Env-specific mucosal and serum antibody induction augments cellular immunity in protecting immunized, elite- controller macaques against high dose heterologous SIV challenge

L. Jean Patterson; Mara Daltabuit-Test; Peng Xiao; Jun Zhao; William K. Hu; Ulrike Wille-Reece; Egidio Brocca-Cofano; V. S. Kalyanaraman; Irene Kalisz; Stephen Whitney; Eun Mi Lee; Ranajit Pal; David C. Montefiori; Satya Dandekar; Robert A. Seder; Mario Roederer; Roger W. Wiseman; Vanessa M. Hirsch; Marjorie Robert-Guroff

Three Indian rhesus macaques, Ad-SIV primed/protein boosted and exposed twice to high-dose mucosal SIV(mac251) challenges, exhibited elite control of viremia over 6.5 years. They were negative for host factors associated with control of SIV infection. After a third intrarectal challenge with SIV(smE660), all controlled viremia, with one (macaque #5) maintaining undetectable viremia in blood. Acquisition was not blocked, but virus was contained in the jejunum and draining lymph nodes. Polyfunctional memory T cell responses and high-titered neutralizing and non-neutralizing serum and mucosal antibodies were present before and maintained post-challenge. The level of protection seen for animal #5 was predicted from analyses of gene transcription in jejunum 2 weeks post-challenge. Macaques #7 and #9, exhibiting lower pre-challenge cellular and humoral immunity, partially controlled the SIV(smE660) challenge. Initial vaccine-induced control by macaque #5 extended to the SIV(smE660) challenge due to multiple immune mechanisms that were boosted and augmented by cryptic SIV exposure.


Vaccine | 2011

Vaccine induced antibodies to the first variable loop of human immunodeficiency virus type 1 gp120, mediate antibody-dependent virus inhibition in macaques.

Izabela Bialuk; Stephen Whitney; Vibeke Andresen; Ruth H. Florese; Janos Nacsa; Valentina Cecchinato; Valerio W. Valeri; Jean-Michel Heraud; Shari N. Gordon; Robyn Washington Parks; David C. Montefiori; David Venzon; Thorsten Demberg; Marjorie Robert Guroff; Gary Landucci; Donald N. Forthal; Genoveffa Franchini

The role of antibodies directed against the hyper variable envelope region V1 of human immunodeficiency virus type 1 (HIV-1), has not been thoroughly studied. We show that a vaccine able to elicit strain-specific non-neutralizing antibodies to this region of gp120 is associated with control of highly pathogenic chimeric SHIV(89.6P) replication in rhesus macaques. The vaccinated animal that had the highest titers of antibodies to the amino terminus portion of V1, prior to challenge, had secondary antibody responses that mediated cell killing by antibody-dependent cellular cytotoxicity (ADCC), as early as 2 weeks after infection and inhibited viral replication by antibody-dependent cell-mediated virus inhibition (ADCVI), by 4 weeks after infection. There was a significant inverse correlation between virus level and binding antibody titers to the envelope protein, (R=-0.83, p=0.015), and ADCVI (R=-0.84 p=0.044). Genotyping of plasma virus demonstrated in vivo selection of three SHIV(89.6P) variants with changes in potential N-linked glycosylation sites in V1. We found a significant inverse correlation between virus levels and titers of antibodies that mediated ADCVI against all the identified V1 virus variants. A significant inverse correlation was also found between neutralizing antibody titers to SHIV(89.6) and virus levels (R=-0.72 p=0.0050). However, passive inoculation of purified immunoglobulin from animal M316, the macaque that best controlled virus, to a naïve macaque, resulted in a low serum neutralizing antibodies and low ADCVI activity that failed to protect from SHIV(89.6P) challenge. Collectively, while our data suggest that anti-envelope antibodies with neutralizing and non-neutralizing Fc(R-dependent activities may be important in the control of SHIV replication, they also demonstrate that low levels of these antibodies alone are not sufficient to protect from infection.


Journal of Virology | 2015

Comparable Antigenicity and Immunogenicity of Oligomeric Forms of a Novel, Acute HIV-1 Subtype C gp145 Envelope for Use in Preclinical and Clinical Vaccine Research.

Lindsay Wieczorek; Shelly J. Krebs; Vaniambadi Kalyanaraman; Stephen Whitney; Sodsai Tovanabutra; Carlos G. Moscoso; Eric Sanders-Buell; Constance Williams; Bonnie M. Slike; Sebastian Molnar; Vincent Dussupt; S. Munir Alam; Agnès Laurence Chenine; Tina Tong; Edgar L. Hill; Hua-Xin Liao; Michael Hoelscher; Leonard Maboko; Susan Zolla-Pazner; Barton F. Haynes; Michael Pensiero; Francine E. McCutchan; Shawyon Malek-Salehi; R. Holland Cheng; Merlin L. Robb; Thomas C. VanCott; Nelson L. Michael; Mary Marovich; Carl R. Alving; Gary R. Matyas

ABSTRACT Eliciting broadly reactive functional antibodies remains a challenge in human immunodeficiency virus type 1 (HIV-1) vaccine development that is complicated by variations in envelope (Env) subtype and structure. The majority of new global HIV-1 infections are subtype C, and novel antigenic properties have been described for subtype C Env proteins. Thus, an HIV-1 subtype C Env protein (CO6980v0c22) from an infected person in the acute phase (Fiebig stage I/II) was developed as a research reagent and candidate immunogen. The gp145 envelope is a novel immunogen with a fully intact membrane-proximal external region (MPER), extended by a polylysine tail. Soluble gp145 was enriched for trimers that yielded the expected “fan blade” motifs when visualized by cryoelectron microscopy. CO6980v0c22 gp145 reacts with the 4E10, PG9, PG16, and VRC01 HIV-1 neutralizing monoclonal antibodies (MAbs), as well as the V1/V2-specific PGT121, 697, 2158, and 2297 MAbs. Different gp145 oligomers were tested for immunogenicity in rabbits, and purified dimers, trimers, and larger multimers elicited similar levels of cross-subtype binding and neutralizing antibodies to tier 1 and some tier 2 viruses. Immunized rabbit sera did not neutralize the highly resistant CO6980v0c22 pseudovirus but did inhibit the homologous infectious molecular clone in a peripheral blood mononuclear cell (PBMC) assay. This Env is currently in good manufacturing practice (GMP) production to be made available for use as a clinical research tool and further evaluation as a candidate vaccine. IMPORTANCE At present, the product pipeline for HIV vaccines is insufficient and is limited by inadequate capacity to produce large quantities of vaccine to standards required for human clinical trials. Such products are required to evaluate critical questions of vaccine formulation, route, dosing, and schedule, as well as to establish vaccine efficacy. The gp145 Env protein presented in this study forms physical trimers, binds to many of the well-characterized broad neutralizing MAbs that target conserved Env epitopes, and induce cross-subtype neutralizing antibodies as measured in both cell line and primary cell assays. This subtype C Env gp145 protein is currently undergoing good manufacturing practice production for use as a reagent for preclinical studies and for human clinical research. This product will serve as a reagent for comparative studies and may represent a next-generation candidate HIV immunogen.


Journal of Biological Chemistry | 2015

Antigenicity and Immunogenicity of a Trimeric Envelope Protein from an Indian Clade C HIV-1 Isolate

Rangasamy Sneha Priya; Menon Veena; Irene Kalisz; Stephen Whitney; Dhopeshwarkar Priyanka; Celia C. LaBranche; Mullapudi Sri Teja; David C. Montefiori; Ranajit Pal; Sundarasamy Mahalingam; V. S. Kalyanaraman

Background: The RV144 trial has established the importance of studying diverse HIV-1 envelopes for immunogen design. Results: HIV-1 93IN101 gp145 exposes conserved regions of envelope protein and induces broadly neutralizing antibodies. Conclusion: Characterization of trimeric gp145 from an Indian isolate demonstrates its potential use as a multiclade candidate HIV-1 vaccine to combat AIDS. Significance: Findings encourage studies on various Indian clade C envelope proteins. Human immunodeficiency virus type 1 (HIV-1) isolates from India mainly belong to clade C and are quite distinct from clade C isolates from Africa in terms of their phylogenetic makeup, serotype, and sensitivity to known human broadly neutralizing monoclonal antibodies. Because many of these properties are associated with the envelope proteins of HIV-1, it is of interest to study the envelope proteins of Indian clade C isolates as part of the ongoing efforts to develop a vaccine against HIV-1. To this end, we purified trimeric uncleaved gp145 of a CCR5 tropic Indian clade C HIV-1 (93IN101) from the conditioned medium of 293 cells. The purified protein was shown to be properly folded with stable structure by circular dichroism. Conformational integrity was further demonstrated by its high affinity binding to soluble CD4, CD4 binding site antibodies such as b12 and VRC01, quaternary epitope-specific antibody PG9, and CD4-induced epitope-specific antibody 17b. Sera from rabbits immunized with gp145 elicited high titer antibodies to various domains of gp120 and neutralized a broad spectrum of clade B and clade C HIV-1 isolates. Similar to other clade B and clade C envelope immunogens, most of the Tier 1 neutralizing activity could be absorbed with the V3-specific peptide. Subsequent boosting of these rabbits with a clade B HIV-1 Bal gp145 resulted in an expanded breadth of neutralization of HIV-1 isolates. The present study strongly supports the inclusion of envelopes from Indian isolates in a future mixture of HIV-1 vaccines.


PLOS ONE | 2014

Nedd4-mediated increase in HIV-1 Gag and Env proteins and immunity following DNA-vaccination of BALB/c mice.

Brad Lewis; Stephen Whitney; Lauren Hudacik; Lindsey Galmin; Maria Cecilia Huaman; Anthony D. Cristillo

The late assembly domain of many viruses is critical for budding. Within these domains, encoded in viral structural proteins, are the conserved motifs PTAP, PPxY and YPxL. These sequences are the key determinants for association of viral proteins with intracellular molecules such as Tsg101, Nedd4 and AIP1/ALIX. While roles for Tsg101 and AIP1/ALIX in HIV-1 budding have been well established, less is known about the role of Nedd4. Recent studies, however, have identified a function for Nedd4-like protein in HIV-1 release. In this study, we investigated post-transcriptional changes of Nedd4 following SHIVSF162P3 infection of rhesus macaques, its role on HIV-1 p24 and gp120 levels in vitro and its potential as an immune modulator in HIV vaccination of BALB/c mice. Increased Nedd4 protein levels were noted in both CD4+ and CD8+ T cells following SHIVSF162P3-infection of naïve macaques. Transient co-transfection studies in 293 cells with HXB2 and Nedd4 demonstrated a Nedd4-mediated increase in p24 and gp120 levels. This increase was found to be dependent on the Ca2+/calmodulin-regulated phospholipid binding C2 domain and not ubiquitin ligase activity or HIV LTR activity. Co-transfection of Nedd4 with plasmid DNA expressing Gag or Env was further shown to augment both intracellular and extracellular Gag or Env proteins. To assess the potential of Nedd4 as an immune modulator, BALB/c mice were immunized intramuscularly with plasmid DNA encoding HIV gag, env and Nedd4. Nedd4 co-administration was found to increase serum anti-p24 but not anti-gp120 antibodies. Nedd4 co-injection was found to have no affect on Gag- or Env-specific IFNγ but had a trend of increased Gag-specific IL-6, IL-17A and TNFα that was not seen following Env stimulation. Based on our initial findings, Nedd4-mediated changes in HIV protein levels and its potential use in HIV-1 vaccine development warrants further investigation.


Retrovirology | 2012

Characterization in rabbits & nonhuman primates of the neutralizing antibody response elicited by DNA & protein vaccination with SIVmac251 & SIVsmE660

Vp Menon; Stephen Whitney; J Francis; L Ajayi; Nikolai Petrovsky; David C. Montefiori; Ranajit Pal; K Vaniambadi

Methods We evaluated immunogenicity of DNA and protein vaccines encoding SIVmac251 and SIVsmE660 gp145 in rabbits and rhesus macaques. DNA vaccines encoding wild type gp145 or mutated gp160 truncated at Q708 were used. Trimeric wildtype gp145 proteins, stably expressed and purified from 293T cells, were used with Advax delta inulin adjuvant to boost after DNA immunization. Macaques were electroporated with wild type DNA of both isolates followed by adjuvanted homologous protein boosts. Rabbits received DNA vaccine alone, delivered by electroporation. Neutralization assays were performed in TZM-bl cells with SIVmac251 and SIVsmE660 isolates that are partially resistant to neutralization.

Collaboration


Dive into the Stephen Whitney's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

V. S. Kalyanaraman

North Shore University Hospital

View shared research outputs
Top Co-Authors

Avatar

Mangala Rao

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Shari N. Gordon

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Melvin N. Doster

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Poonam Pegu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Shan Lu

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Shixia Wang

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge