Network


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

Hotspot


Dive into the research topics where J. Robert Putnak is active.

Publication


Featured researches published by J. Robert Putnak.


Vaccine | 2010

Development of a recombinant tetravalent dengue virus vaccine: immunogenicity and efficacy studies in mice and monkeys.

David E. Clements; Beth-Ann Coller; Michael M. Lieberman; Steven A. Ogata; Gordon Wang; Kent E. Harada; J. Robert Putnak; John M. Ivy; Michael McDonell; Gary Bignami; Iain Peters; Julia Leung; Carolyn Weeks-Levy; Eileen Nakano; Tom Humphreys

Truncated recombinant dengue virus envelope protein subunits (80E) are efficiently expressed using the Drosophila Schneider-2 (S2) cell expression system. Binding of conformationally sensitive antibodies as well as X-ray crystal structural studies indicate that the recombinant 80E subunits are properly folded native-like proteins. Combining the 80E subunits from each of the four dengue serotypes with ISCOMATRIX adjuvant, an adjuvant selected from a set of adjuvants tested for maximal and long lasting immune responses, results in high titer virus neutralizing antibody responses. Immunization of mice with a mixture of all four 80E subunits and ISCOMATRIX adjuvant resulted in potent virus neutralizing antibody responses to each of the four serotypes. The responses to the components of the tetravalent mixture were equivalent to the responses to each of the subunits administered individually. In an effort to evaluate the potential protective efficacy of the Drosophila expressed 80E, the dengue serotype 2 (DEN2-80E) subunit was tested in both the mouse and monkey challenge models. In both models protection against viral challenge was achieved with low doses of antigen in the vaccine formulation. In non-human primates, low doses of the tetravalent formulation induced good virus neutralizing antibody titers to all four serotypes and protection against challenge with the two dengue virus serotypes tested. In contrast to previous reports, where subunit vaccine candidates have generally failed to induce potent, protective responses, native-like soluble 80E proteins expressed in the Drosophila S2 cells and administered with appropriate adjuvants are highly immunogenic and capable of eliciting protective responses in both mice and monkeys. These results support the development of a dengue virus tetravalent vaccine based on the four 80E subunits produced in the Drosophila S2 cell expression system.


Human Vaccines | 2009

Phase 2 clinical trial of three formulations of tetravalent live-attenuated dengue vaccine in flavivirus-naïve adults

Wellington Sun; Dennis Cunningham; Steven S. Wasserman; Judith Perry; J. Robert Putnak; Kenneth H. Eckels; David W. Vaughn; Stephen J. Thomas; Niranjan Kanesa-thasan; Bruce L. Innis; Robert Edelman

Sixteen dose formulations of our live-attenuated tetravalent dengue virus vaccines (TDV) were previously evaluated for safety and immunogenicity. Two of the sixteen candidate TDV formulations (Formulations 13 and 14) were selected for further evaluation. A new TDV formulation, Formulation 17, using a higher primary dog kidney (PDK) cell passage Dengue-1 virus (DENV-1) and a lower PDK cell passage DENV-4, was developed to optimize the neutralizing antibody response. All three formulations consist of combinations of 10exp3-5 pfu/dose of the four dengue vaccine virus serotypes. This double-blind, randomized trial in 71 healthy adult subjects evaluated vaccine safety, reactogenicity and immunogenicity. TDV’s were given subcutaneously in the deltoid on Day 0 and 180 (6 months). Subjects were seen in clinic on Study Days 0, 10, 28, 180, 190 and 208 and filled out daily symptom diaries for 21 days after each vaccination. Formulation 13 was the most reactogenic, while both Formulations 14 and 17 were similar in reported reactions. Seventy-five percent, 31% and 31% of subjects were viremic on Day 10 after primary vaccination with Formulations 13, 14 and 17 respectively. Viremia was not detected in any subject following the second dose of vaccine. The immunogenicity endpoint was neutralizing antibody titer one month after the second vaccination. Thirty-six percent, 40% and 63% of vaccinated subjects developed tetravalent neutralizing antibodies after two doses of Formulations 13, 14 and 17, respectively. Formulation 17 was selected for further clinical evaluation based on this study.


American Journal of Tropical Medicine and Hygiene | 2008

Safety and Immunogenicity of a Tetravalent Live-attenuated Dengue Vaccine in Flavivirus Naive Children

Sriluck Simasathien; Stephen J. Thomas; Veerachai Watanaveeradej; Ananda Nisalak; Célia Barberousse; Bruce L. Innis; Wellington Sun; J. Robert Putnak; Kenneth H. Eckels; Yanee Hutagalung; Robert V. Gibbons; Chunlin Zhang; Rafael De La Barrera; Richard G. Jarman; Wipa Chawachalasai; Mammen P. Mammen

A Phase I/II observer-blind, randomized, controlled trial evaluated the safety and immunogenicity of a dengue virus (DENV) vaccine candidate in healthy Thai infants (aged 12-15 months) without measurable pre-vaccination neutralizing antibodies to DENV and Japanese encephalitis virus. Fifty-one subjects received two doses of either DENV (N = 34; four received 1/10th dose) or control vaccine (N = 17; dose 1, live varicella; dose 2, Haemophilus influenzae type b). After each vaccine dose, adverse events (AEs) were solicited for 21 days, and non-serious AEs were solicited for 30 days; serious AEs (SAEs) were recorded throughout the study. Laboratory safety assessments were performed at 10 and 30 days; neutralizing antibodies were measured at 30 days. The DENV vaccine was well-tolerated without any related SAEs. After the second dose, 85.7% of full-dose DENV vaccinees developed at least trivalent and 53.6% developed tetravalent neutralizing antibodies ≥ 1:10 to DENV (control group = 0%). This vaccine candidate, therefore, warrants continued development in this age group (NCT00322049; clinicaltrials.gov).


Vaccine | 2001

A purified inactivated Japanese encephalitis virus vaccine made in vero cells

Ashok K. Srivastava; J. Robert Putnak; Sung H. Lee; Sun P. Hong; Sang B. Moon; David A. Barvir; Bangti Zhao; Russell A. Olson; Soo-Ok Kim; Wangdon Yoo; Andrew C. Towle; David W. Vaughn; Bruce L. Innis; Kenneth H. Eckels

A second generation, purified, inactivated vaccine (PIV) against Japanese encephalitis (JE) virus was produced and tested in mice where it was found to be highly immunogenic and protective. The JE-PIV was made from an attenuated strain of JE virus propagated in certified Vero cells, purified, and inactivated with formalin. Its manufacture followed current GMP guidelines for the production of biologicals. The manufacturing process was efficient in generating a high yield of virus, essentially free of contaminating host cell proteins and nucleic acids. The PIV was formulated with aluminum hydroxide and administered to mice by subcutaneous inoculation. Vaccinated animals developed high-titered JE virus neutralizing antibodies in a dose dependent fashion after two injections. The vaccine protected mice against morbidity and mortality after challenge with live, virulent, JE virus. Compared with the existing licensed mouse brain-derived vaccine, JE-Vax, the Vero cell-derived JE-PIV was more immunogenic and as effective as preventing encephalitis in mice. The JE-PIV is currently being tested for safety and immunogenicity in volunteers.


The Journal of Infectious Diseases | 2006

Protection of Rhesus Monkeys against Dengue Virus Challenge after Tetravalent Live Attenuated Dengue Virus Vaccination

Wellington Sun; Ananda Nisalak; Montip Gettayacamin; Kenneth H. Eckels; J. Robert Putnak; David W. Vaughn; Bruce L. Innis; Stephen J. Thomas; Timothy P. Endy

Rhesus monkeys develop viremia after dengue virus (DENV) inoculation and have been used as an animal model to study DENV infection and DENV vaccine candidates. We evaluated, in this model, the protective efficacy of a live attenuated tetravalent DENV vaccine (TDV) candidate against parenteral challenge with parental near-wild-type DENV strains. Twenty monkeys were vaccinated with TDV at 0 and 1 month, and 20 unvaccinated monkeys served as controls. Vaccinated animals and their controls were inoculated with 10(3)-10(4) pfu of challenge virus 4.5 months after the second vaccination. Primary vaccination resulted in 95%, 100%, 70%, and 15% seroconversion to DENV serotypes 1, 2, 3, and 4 (DENV-1, -2, -3, and -4), respectively. After the second vaccination, the seropositivity rates were 100%, 100%, 90%, and 70%, respectively. Vaccination with TDV resulted in complete protection against viremia from DENV-2 challenge and in 80%, 80%, and 50% protection against challenge with DENV-1, -3, and -4, respectively. Our results suggest that the TDV can elicit protective immunity against all 4 DENV serotypes. Interference among the 4 vaccine viruses may have resulted in decreased antibody responses to DENV-3 and -4, which would require reformulation or dose optimization to minimize this interference during testing of the vaccine in humans.


The Journal of Infectious Diseases | 2013

Experimental Dengue Virus Challenge of Human Subjects Previously Vaccinated With Live Attenuated Tetravalent Dengue Vaccines

Wellington Sun; Kenneth H. Eckels; J. Robert Putnak; Arthur Lyons; Stephen J. Thomas; David W. Vaughn; Robert V. Gibbons; Stefan Fernandez; Vicky Gunther; Mammen P. Mammen; John D. Statler; Bruce L. Innis

BACKGROUND Protection against dengue requires immunity against all 4 serotypes of dengue virus (DENV). Experimental challenge may be useful in evaluating vaccine-induced immunity. METHODS Ten subjects previously vaccinated with a live attenuated tetravalent dengue vaccine (TDV) and 4 DENV-naive control subjects were challenged by subcutaneous inoculation of either 10(3) plaque-forming units (PFU) of DENV-1 or 10(5) PFU of DENV-3. Two additional subjects who did not develop DENV-3 neutralizing antibody (NAb) from TDV were revaccinated with 10(4) PFU of live attenuated DENV-3 vaccine to evaluate memory response. RESULTS All 5 TDV recipients were protected against DENV-1 challenge. Of the 5 TDV recipients challenged with DENV-3, 2 were protected. All DENV-3-challenge subjects who developed viremia also developed elevated liver enzyme levels, and 2 had values that were >10 times greater than normal. Of the 2 subjects revaccinated with DENV-3 vaccine, 1 showed a secondary response to DENV-2, while neither showed such response to DENV-3. All 4 control subjects developed dengue fever from challenge. Protection was associated with presence of NAb, although 1 subject was protected despite a lack of measurable NAb at the time of DENV-1 challenge. CONCLUSIONS Vaccination with TDV induced variable protection against subcutaneous challenge. DENV-3 experimental challenge was associated with transient but marked elevations of transaminases.


Virology | 1988

Functional and antigenic domains of the dengue-2 virus nonstructural glycoprotein NS-1

J. Robert Putnak; Peter C. Charles; R. Padmanabhan; Koji Irie; Charles H. Hoke; Donald S. Burke

The gene coding for the nonstructural glycoprotein of dengue-2 virus was cloned, sequenced, and expressed in Escherichia coli. There was about 70% conservation at the amino acid level with dengue serotypes 1 and 4 suggesting an important common function for this protein. Conserved hydrophobic domains were found both before the amino-terminus and at the carboxy-terminus, consistent with transmembrane roles. Evidence for at least partial translocation of NS-1 through the inner membrane of E. coli was found. Also conserved were two signals for N-linked glycosylation located near the middle of NS-1. Various regions of NS-1 were tested for antigenicity with mouse and rabbit polyclonal and mouse monoclonal antibodies. The mouse polyclonal antibodies, made against a crude dengue-infected mouse brain immunogen, reacted most strongly with N-terminal regions of NS-1, whereas, the rabbit antiserum, made against purified NS-1 protein, reacted strongest with C-terminal regions. These findings suggest that immunogen presentation or species differences could be important. Although most of the monoclonals appeared to be unreactive in Western blots with expressed NS-1 proteins, two appeared to react strongly; the region from amino acid (a.a.) 273 to a.a. 346 was required for antibody binding. This region, located adjacent to the two conserved C-terminal hydrophobic domains, is highly charged and contains 5 of the 10 conserved cysteine residues of NS-1.


PLOS Pathogens | 2017

Impact of prior flavivirus immunity on Zika virus infection in rhesus macaques

Michael K. McCracken; Gregory D. Gromowski; Heather Friberg; Xiaoxu Lin; Peter Abbink; Rafael De La Barrera; Kenneth H. Eckles; Lindsey S Garver; Michael Boyd; David Jetton; Dan H. Barouch; Matthew C. Wise; Bridget S. Lewis; Jeffrey R. Currier; Kayvon Modjarrad; Mark Milazzo; Michelle Liu; Anna B. Mullins; J. Robert Putnak; Nelson L. Michael; Richard G. Jarman; Stephen J. Thomas

Studies have demonstrated cross-reactivity of anti-dengue virus (DENV) antibodies in human sera against Zika virus (ZIKV), promoting increased ZIKV infection in vitro. However, the correlation between in vitro and in vivo findings is not well characterized. Thus, we evaluated the impact of heterotypic flavivirus immunity on ZIKV titers in biofluids of rhesus macaques. Animals previously infected (≥420 days) with DENV2, DENV4, or yellow fever virus were compared to flavivirus-naïve animals following infection with a Brazilian ZIKV strain. Sera from DENV-immune macaques demonstrated cross-reactivity with ZIKV by antibody-binding and neutralization assays prior to ZIKV infection, and promoted increased ZIKV infection in cell culture assays. Despite these findings, no significant differences between flavivirus-naïve and immune animals were observed in viral titers, neutralizing antibody levels, or immune cell kinetics following ZIKV infection. These results indicate that prior infection with heterologous flaviviruses neither conferred protection nor increased observed ZIKV titers in this non-human primate ZIKV infection model.


American Journal of Tropical Medicine and Hygiene | 2015

An Adjuvanted, Tetravalent Dengue Virus-Purified Inactivated Vaccine Candidate Induces Long-Lasting and Protective Antibody Responses Against Dengue Challenge in Rhesus Macaques

Stefan Fernandez; Stephen J. Thomas; Rafael De La Barrera; Rawiwan Imerbsin; Richard G. Jarman; Benoı̂t Baras; Jean-François Toussaint; Sally Mossman; Bruce L. Innis; Alexander C. Schmidt; Marie-Pierre Malice; Pascale Festraets; Lucile Warter; J. Robert Putnak; Kenneth H. Eckels

The immunogenicity and protective efficacy of a candidate tetravalent dengue virus purified inactivated vaccine (TDENV PIV) formulated with alum or an Adjuvant System (AS01, AS03 tested at three different dose levels, or AS04) was evaluated in a 0, 1-month vaccination schedule in rhesus macaques. One month after dose 2, all adjuvanted formulations elicited robust and persisting neutralizing antibody titers against all four dengue virus serotypes. Most of the formulations tested prevented viremia after challenge, with the dengue serotype 1 and 2 virus strains administered at 40 and 32 weeks post-dose 2, respectively. This study shows that inactivated dengue vaccines, when formulated with alum or an Adjuvant System, are candidates for further development.


Vaccines#R##N#New Approaches to Immunological Problems | 1992

New Approaches to Flavivirus Vaccine Development

Jacob J. Schlesinger; J. Robert Putnak; Kenneth H. Eckels

Publisher Summary This chapter discusses new approaches to flavivirus vaccine development. Flaviviruses are small, enveloped single-positive strand RNA viruses of the newly established family, Flaviviridae. Genetically and serologically related to each other, the 68 known flaviviruses include a large number of arthropod-borne agents of human disease, the most important of which, in terms of prevalence and pathogenicity, are the mosquito-borne yellow fever, dengue, and Japanese encephalitis viruses, and the far eastern and western subtypes of tick-borne encephalitis virus. The chapter presents recent advances in the molecular biology of flaviviruses that will offer novel alternatives to further flavivirus vaccine development. The capacity of medically important flaviviruses to cause lethal encephalitis in mice has been invaluable in the early assessment of candidate vaccine preparations. However, inadequacy of this model as a reliable measure of vaccine attenuation or protective immunogenicity in humans, where the pathogenesis of flaviviral disease may be vastly different, cannot be too strongly emphasized. In the case of dengue where infection of monkeys is clinically unapparent, the absence of a suitable experimental model of human infection presents a formidable barrier to the understanding of the pathogenesis of this disease and to the development of a vaccine against it.

Collaboration


Dive into the J. Robert Putnak's collaboration.

Top Co-Authors

Avatar

Kenneth H. Eckels

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stephen J. Thomas

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar

Wellington Sun

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Rafael De La Barrera

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Niranjan Kanesa-thasan

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Richard G. Jarman

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Charles H. Hoke

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Arthur Lyons

Walter Reed Army Institute of Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge