Network


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

Hotspot


Dive into the research topics where Stephen C. De Rosa is active.

Publication


Featured researches published by Stephen C. De Rosa.


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

BACKGROUNDnIn 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.nnnMETHODSnIn 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.nnnRESULTSnOf 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.nnnCONCLUSIONSnThis 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.


The Journal of Infectious Diseases | 2013

Safety and Comparative Immunogenicity of an HIV-1 DNA Vaccine in Combination with Plasmid Interleukin 12 and Impact of Intramuscular Electroporation for Delivery

Spyros A. Kalams; Scott Parker; Marnie Elizaga; Barbara Metch; Srilatha Edupuganti; John Hural; Stephen C. De Rosa; Donald K. Carter; Kyle Rybczyk; Ian Frank; Jonathan D. Fuchs; Beryl A. Koblin; Denny H. Kim; Patrice Joseph; Michael C. Keefer; Lindsey R. Baden; John H. Eldridge; Jean D. Boyer; Adam Sherwat; Massimo Cardinali; Mary Allen; Michael Pensiero; Christopher Collett Butler; Amir S. Khan; Jian Yan; Niranjan Y. Sardesai; James G. Kublin; David B. Weiner

BACKGROUNDnDNA vaccines have been very poorly immunogenic in humans but have been an effective priming modality in prime-boost regimens. Methods to increase the immunogenicity of DNA vaccines are needed.nnnMETHODSnHIV Vaccine Trials Network (HVTN) studies 070 and 080 were multicenter, randomized, clinical trials. The human immunodeficiency virus type 1 (HIV-1) PENNVAX®-B DNA vaccine (PV) is a mixture of 3 expression plasmids encoding HIV-1 Clade B Env, Gag, and Pol. The interleukin 12 (IL-12) DNA plasmid expresses human IL-12 proteins p35 and p40. Study subjects were healthy HIV-1-uninfected adults 18-50 years old. Four intramuscular vaccinations were given in HVTN 070, and 3 intramuscular vaccinations were followed by electroporation in HVTN 080. Cellular immune responses were measured by intracellular cytokine staining after stimulation with HIV-1 peptide pools.nnnRESULTSnVaccination was safe and well tolerated. Administration of PV plus IL-12 with electroporation had a significant dose-sparing effect and provided immunogenicity superior to that observed in the trial without electroporation, despite fewer vaccinations. A total of 71.4% of individuals vaccinated with PV plus IL-12 plasmid with electroporation developed either a CD4(+) or CD8(+) T-cell response after the second vaccination, and 88.9% developed a CD4(+) or CD8(+) T-cell response after the third vaccination.nnnCONCLUSIONSnUse of electroporation after PV administration provided superior immunogenicity than delivery without electroporation. This study illustrates the power of combined DNA approaches to generate impressive immune responses in humans.


PLOS ONE | 2011

A Phase IIA Randomized Clinical Trial of a Multiclade HIV-1 DNA Prime Followed by a Multiclade rAd5 HIV-1 Vaccine Boost in Healthy Adults (HVTN204)

Gavin J. Churchyard; Cecilia Morgan; Elizabeth Adams; John Hural; Barney S. Graham; Zoe Moodie; Doug Grove; Glenda Gray; Linda-Gail Bekker; M. Juliana McElrath; Georgia D. Tomaras; Paul A. Goepfert; Spyros A. Kalams; Lindsey R. Baden; Michelle Lally; Raphael Dolin; William A. Blattner; Artur Kalichman; J. Peter Figueroa; Jean W. Pape; Mauro Schechter; Olivier D. Defawe; Stephen C. De Rosa; David C. Montefiori; Gary J. Nabel; Lawrence Corey; Michael C. Keefer

Background The safety and immunogenicity of a vaccine regimen consisting of a 6-plasmid HIV-1 DNA prime (envA, envB, envC, gagB, polB, nefB) boosted by a recombinant adenovirus serotype-5 (rAd5) HIV-1 with matching inserts was evaluated in HIV-seronegative participants from South Africa, United States, Latin America and the Caribbean. Methods 480 participants were evenly randomized to receive either: DNA (4 mg IM by Biojector) at 0, 1 and 2 months, followed by rAd5 (1010 PU IM by needle/syringe) at 6 months; or placebo. Participants were monitored for reactogenicity and adverse events throughout the 12-month study. Peak and duration of HIV-specific humoral and cellular immune responses were evaluated after the prime and boost. Results The vaccine was well tolerated and safe. T-cell responses, detected by interferon-γ (IFN-γ) ELISpot to global potential T-cell epitopes (PTEs) were observed in 70.8% (136/192) of vaccine recipients overall, most frequently to Gag (54.7%) and to Env (54.2%). In U.S. vaccine recipients T-cell responses were less frequent in Ad5 sero-positive versus sero-negative vaccine recipients (62.5% versus 85.7% respectively, pu200a=u200a0.035). The frequency of HIV-specific CD4+ and CD8+ T-cell responses detected by intracellular cytokine staining were similar (41.8% and 47.2% respectively) and most secreted ≥2 cytokines. The vaccine induced a high frequency (83.7%–94.6%) of binding antibody responses to consensus Group M, and Clades A, B and C gp140 Env oligomers. Antibody responses to Gag were elicited in 46% of vaccine recipients. Conclusion The vaccine regimen was well-tolerated and induced polyfunctional CD4+ and CD8+ T-cells and multi-clade anti-Env binding antibodies. Trial Registration: ClinicalTrials.gov NCT00125970


Proceedings of the National Academy of Sciences of the United States of America | 2012

Merck Ad5/HIV induces broad innate immune activation that predicts CD8⁺ T-cell responses but is attenuated by preexisting Ad5 immunity.

Erica Andersen-Nissen; Eric R. Peterson; Alicia Sato; M. Kristina Hamilton; Joleen Borgerding; Akshay T. Krishnamurty; Joanne T. Chang; Devin J. Adams; Tiffany R. Hensley; Alexander I. Salter; Cecilia Morgan; Ann Duerr; Stephen C. De Rosa; Alan Aderem; M. Juliana McElrath

To better understand how innate immune responses to vaccination can lead to lasting protective immunity, we used a systems approach to define immune signatures in humans over 1 wk following MRKAd5/HIV vaccination that predicted subsequent HIV-specific T-cell responses. Within 24 h, striking increases in peripheral blood mononuclear cell gene expression associated with inflammation, IFN response, and myeloid cell trafficking occurred, and lymphocyte-specific transcripts decreased. These alterations were corroborated by marked serum inflammatory cytokine elevations and egress of circulating lymphocytes. Responses of vaccinees with preexisting adenovirus serotype 5 (Ad5) neutralizing antibodies were strongly attenuated, suggesting that enhanced HIV acquisition in Ad5-seropositive subgroups in the Step Study may relate to the lack of appropriate innate activation rather than to increased systemic immune activation. Importantly, patterns of chemoattractant cytokine responses at 24 h and alterations in 209 peripheral blood mononuclear cell transcripts at 72 h were predictive of subsequent induction and magnitude of HIV-specific CD8+ T-cell responses. This systems approach provides a framework to compare innate responses induced by vectors, as shown here by contrasting the more rapid, robust response to MRKAd5/HIV with that to yellow fever vaccine. When applied iteratively, the findings may permit selection of HIV vaccine candidates eliciting innate immune response profiles more likely to drive HIV protective immunity.


Proceedings of the National Academy of Sciences of the United States of America | 2005

Characterization of subsets of CD4+ memory T cells reveals early branched pathways of T cell differentiation in humans

Kaimei Song; Ronald L. Rabin; Brenna J. Hill; Stephen C. De Rosa; Stephen P. Perfetto; Hongwei H. Zhang; John F. Foley; Jeffrey S. Reiner; Jie Liu; Joseph J. Mattapallil; Mario Roederer; Joshua M. Farber

The pathways for differentiation of human CD4+ T cells into functionally distinct subsets of memory cells in vivo are unknown. The identification of these subsets and pathways has clear implications for the design of vaccines and immune-targeted therapies. Here, we show that populations of apparently naïve CD4+ T cells express the chemokine receptors CXCR3 or CCR4 and demonstrate patterns of gene expression and functional responses characteristic of memory cells. The proliferation history and T cell receptor repertoire of these chemokine-receptor+ cells suggest that they are very early memory CD4+ T cells that have “rested down” before acquiring the phenotypes described for “central” or “effector” memory T cells. In addition, the chemokine-receptor+ “naïve” populations contain Th1 and Th2 cells, respectively, demonstrating that Th1/Th2 differentiation can occur very early in vivo in the absence of markers conventionally associated with memory cells. We localized ligands for CXCR3 and CCR4 to separate foci in T cell zones of tonsil, suggesting that the chemokine-receptor+ subsets may be recruited and contribute to segregated, polarized microenvironments within lymphoid organs. Importantly, our data suggest that CD4+ T cells do not differentiate according to a simple schema from naïve → CD45RO+ noneffector/central memory → effector/effector memory cells. Rather, developmental pathways branch early on to yield effector/memory populations that are highly heterogeneous and multifunctional and have the potential to become stable resting cells.


JAMA | 2016

Safety and Immunogenicity of Novel Adenovirus Type 26– and Modified Vaccinia Ankara–Vectored Ebola Vaccines: A Randomized Clinical Trial

Iain D. Milligan; Malick Gibani; Richard P. Sewell; Elizabeth A. Clutterbuck; Danielle Campbell; Emma Plested; Elizabeth Nuthall; Merryn Voysey; Laura Silva-Reyes; M. Juliana McElrath; Stephen C. De Rosa; Nicole Frahm; Kristen W. Cohen; Georgi Shukarev; Nicola Orzabal; Wilbert van Duijnhoven; Carla Truyers; Nora Bachmayer; Daniel Splinter; Nathaly Samy; Maria Grazia Pau; Hanneke Schuitemaker; Kerstin Luhn; Benoit Callendret; Johan Van Hoof; Macaya Douoguih; Katie Ewer; Brian Angus; Andrew J. Pollard; Matthew D. Snape

IMPORTANCEnDeveloping effective vaccines against Ebola virus is a global priority.nnnOBJECTIVEnTo evaluate an adenovirus type 26 vector vaccine encoding Ebola glycoprotein (Ad26.ZEBOV) and a modified vaccinia Ankara vector vaccine, encoding glycoproteins from Ebola virus, Sudan virus, Marburg virus, and Tai Forest virus nucleoprotein (MVA-BN-Filo).nnnDESIGN, SETTING, AND PARTICIPANTSnSingle-center, randomized, placebo-controlled, observer-blind, phase 1 trial performed in Oxford, United Kingdom, enrolling healthy 18- to 50-year-olds from December 2014; 8-month follow-up was completed October 2015.nnnINTERVENTIONSnParticipants were randomized into 4 groups, within which they were simultaneously randomized 5:1 to receive study vaccines or placebo. Those receiving active vaccines were primed with Ad26.ZEBOV (5u2009×u200910(10) viral particles) or MVA-BN-Filo (1u2009×u200910(8) median tissue culture infective dose) and boosted with the alternative vaccine 28 or 56 days later. A fifth, open-label group received Ad26.ZEBOV boosted by MVA-BN-Filo 14 days later.nnnMAIN OUTCOMES AND MEASURESnThe primary outcomes were safety and tolerability. All adverse events were recorded until 21 days after each immunization; serious adverse events were recorded throughout the trial. Secondary outcomes were humoral and cellular immune responses to immunization, as assessed by enzyme-linked immunosorbent assay and enzyme-linked immunospot performed at baseline and from 7 days after each immunization until 8 months after priming immunizations.nnnRESULTSnAmong 87 study participants (median age, 38.5 years; 66.7% female), 72 were randomized into 4 groups of 18, and 15 were included in the open-label group. Four participants did not receive a booster dose; 67 of 75 study vaccine recipients were followed up at 8 months. No vaccine-related serious adverse events occurred. No participant became febrile after MVA-BN-Filo, compared with 3 of 60 participants (5%; 95% CI, 1%-14%) receiving Ad26.ZEBOV in the randomized groups. In the open-label group, 4 of 15 Ad26.ZEBOV recipients (27%; 95% CI, 8%-55%) experienced fever. In the randomized groups, 28 of 29 Ad26.ZEBOV recipients (97%; 95% CI, 82%- 99.9%) and 7 of 30 MVA-BN-Filo recipients (23%; 95% CI, 10%-42%) had detectable Ebola glycoprotein-specific IgG 28 days after primary immunization. All vaccine recipients had specific IgG detectable 21 days postboost and at 8-month follow-up. Within randomized groups, at 7 days postboost, at least 86% of vaccine recipients showed Ebola-specific T-cell responses.nnnCONCLUSIONS AND RELEVANCEnIn this phase 1 study of healthy volunteers, immunization with Ad26.ZEBOV or MVA-BN-Filo did not result in any vaccine-related serious adverse events. An immune response was observed after primary immunization with Ad26.ZEBOV; boosting by MVA-BN-Filo resulted in sustained elevation of specific immunity. These vaccines are being further assessed in phase 2 and 3 studies.nnnTRIAL REGISTRATIONnclinicaltrials.gov Identifier: NCT02313077.


The Journal of Infectious Diseases | 2011

A Trimeric, V2-Deleted HIV-1 Envelope Glycoprotein Vaccine Elicits Potent Neutralizing Antibodies but Limited Breadth of Neutralization in Human Volunteers

Paul Spearman; Michelle Lally; Marnie Elizaga; David C. Montefiori; Georgia D. Tomaras; M. Juliana McElrath; John Hural; Stephen C. De Rosa; Alicia Sato; Yunda Huang; Sharon E. Frey; Paul A. Sato; John Donnelly; Susan W. Barnett; Lawrence Corey

BACKGROUNDnA key missing element in the development of a successful human immunodeficiency virus (HIV) vaccine is an immunogen that can generate broadly cross-neutralizing antibodies against primary isolates of the virus.nnnMETHODSnThis phase 1 clinical trial employed a DNA prime and subunit envelope protein boost in an attempt to generate cellular and humoral immune responses that might be desirable in a protective HIV vaccine. Priming was performed via intramuscular injection with gag and env DNA adsorbed to polylactide coglycolide microspheres, followed by boosting with a recombinant trimeric envelope (Env) glycoprotein delivered in MF59 adjuvant.nnnRESULTSnThe DNA prime and protein boost were generally safe and well-tolerated. Env-specific CD4(+) cellular responses were generated that were predominantly detected after Env protein boosting. Neutralizing antibody responses against the homologous SF162 viral isolate were remarkably strong and were present in the majority of vaccine recipients, including a strong response against CD4-induced epitopes on gp120. Despite the promising potency of this vaccine approach, neutralization breadth against heterologous tier 2 strains of HIV-1 was minimal.nnnCONCLUSIONSnPotent neutralization against neutralization-sensitive strains of HIV is achievable in humans through a DNA prime, recombinant oligomeric Env protein boost regimen. Eliciting substantial breadth of neutralization remains an elusive goal.nnnCLINICAL TRIALS REGISTRATIONnNCT00073216.


Journal of Immunology | 2011

HIV-DNA Priming Alters T Cell Responses to HIV-Adenovirus Vaccine Even When Responses to DNA Are Undetectable

Stephen C. De Rosa; Evan P. Thomas; John Bui; Yunda Huang; Allan C. deCamp; Cecilia Morgan; Spyros A. Kalams; Georgia D. Tomaras; Rama Akondy; Rafi Ahmed; Chuen Yen Lau; Barney S. Graham; Gary J. Nabel; M. Juliana McElrath

Many candidate HIV vaccines are designed to primarily elicit T cell responses. Although repeated immunization with the same vaccine boosts Ab responses, the benefit for T cell responses is ill defined. We compared two immunization regimens that include the same recombinant adenoviral serotype 5 (rAd5) boost. Repeated homologous rAd5 immunization fails to increase T cell responses, but increases gp140 Ab responses 10-fold. DNA prime, as compared with rAd5 prime, directs long-term memory CD8+ T cells toward a terminally differentiated effector memory phenotype with cytotoxic potential. Based on the kinetics of activated cells measured directly ex vivo, the DNA vaccination primes for both CD4+ and CD8+ T cells, despite the lack of detection of the latter until after the boost. These results suggest that heterologous prime-boost combinations have distinct immunological advantages over homologous prime-boosts and suggest that the effect of DNA on subsequent boosting may not be easily detectable directly after the DNA vaccination.


Vaccine | 2011

A phase I trial of preventive HIV vaccination with heterologous poxviral-vectors containing matching HIV-1 inserts in healthy HIV-uninfected subjects

Michael C. Keefer; Sharon E. Frey; Marnie Elizaga; Barbara Metch; Stephen C. De Rosa; Paulo Feijó Barroso; Georgia D. Tomaras; Massimo Cardinali; Paul A. Goepfert; Artur Kalichman; Valerie Philippon; M. Juliana McElrath; Xia Jin; Guido Ferrari; Olivier D. Defawe; Gail P. Mazzara; David C. Montefiori; Michael Pensiero; Dennis Panicali; Lawrence Corey

We evaluated replication-defective poxvirus vectors (modified vaccinia Ankara [MVA] and fowlpox [FPV]) in a homologous and heterologous vector prime-boost vaccination regimen containing matching HIV inserts (MVA-HIV and FPV-HIV) given at months 0, 1, 3, 5 and 7 in 150 healthy HIV-negative vaccinia-naïve participants. FPV-HIV alone was poorly immunogenic, while the high dose (10(9)pfu/2 ml) of MVA-HIV alone elicited maximal responses after two injections: CD4+ and CD8+ T-cell responses in 26/55 (47.3%) and 5/60 (8.3%) of participants, respectively, and IFN-γ ELISpot responses in 28/62 (45.2%). The infrequent CD8+ T-cell responses following MVA-HIV priming were boosted only by the heterologous (FPV-HIV) construct in 14/27 (51.9%) of participants post 4th vaccination. Alternatively, HIV envelope-specific binding antibodies were demonstrated in approximately two-thirds of recipients of the homologous boosting regimen, but in less than 20% of subjects after the heterologous vector boost. Thus, a heterologous poxvirus vector prime-boost regimen can induce HIV-specific CD8+ T-cell and CD4+ T-cell responses, which may be an important feature of an optimal regimen for preventive HIV vaccination.


Cytometry Part A | 2008

Nine-color flow cytometry for accurate measurement of T cell subsets and cytokine responses. Part II: Panel performance across different instrument platforms.

Bridget McLaughlin; Nicole Baumgarth; Martin Bigos; Mario Roederer; Stephen C. De Rosa; John D. Altman; Douglas F. Nixon; Janet Ottinger; Judy Li; Laurel Beckett; Barbara L. Shacklett; Thomas G. Evans; David M. Asmuth

Cellular immune responses elicited by vaccination are complex and require polychromatic analysis to accurately characterize the phenotype and function of rare, responding cells. Technical challenges and a lack of instrument standardization between research sites have limited the application of polychromatic cytometry in multicenter clinical trials. Two previously developed six‐color T cell subset immunophenotyping reagent panels deliberately designed to accommodate three additional low frequency functional measurements were compared for their reproducibility of staining across three different flow cytometers. We repeatedly measured similar T cell subset frequencies between the two reagent panels and across the three different cytometers. Spectral overlap reduced sensitivity in two of the three open measurement channels (PE [IL‐2] and APC [IFNγ]) for one reagent combination, particularly in subsets with low cytokine expression. There was no significant interassay variation for measurements across instrument platforms. Careful panel design will identify reagent combinations that minimize spectral spillover into channels reserved for cytokine measurement and comparable results can be achieved using different cytometers, however, it is important to establish standardized quality control procedures for each instrument to minimize variation between cytometers.

Collaboration


Dive into the Stephen C. De Rosa's collaboration.

Top Co-Authors

Avatar

M. Juliana McElrath

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cecilia Morgan

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John Hural

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Lawrence Corey

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nicole Frahm

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Allan C. deCamp

Fred Hutchinson Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge