Network


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

Hotspot


Dive into the research topics where Jean D. Boyer is active.

Publication


Featured researches published by Jean D. Boyer.


The Journal of Infectious Diseases | 1998

First Human Trial of a DNA-Based Vaccine for Treatment of Human Immunodeficiency Virus Type 1 Infection: Safety and Host Response

Rob Roy MacGregor; Jean D. Boyer; Kenneth E. Ugen; Kim Lacy; Stephen J. Gluckman; Mark L. Bagarazzi; Michael A. Chattergoon; Yaela Baine; Terry J. Higgins; Richard B. Ciccarelli; Leslie R. Coney; Richard S. Ginsberg; David B. Weiner

A DNA-based vaccine containing human immunodeficiency virus type 1 (HIV-1) env and rev genes was tested for safety and host immune response in 15 asymptomatic HIV-infected patients who were not using antiviral drugs and who had CD4+ lymphocyte counts of > or = 500 per microliter of blood. Successive groups received three doses of vaccine (30, 100, or 300 microg) at 10-week intervals in a dose-escalation trial. Vaccine administration induced no local or systemic reactions, and no laboratory abnormalities were detected. Specifically, no patient developed anti-DNA antibody or muscle enzyme elevations. No consistent change occurred in CD4 or CD8 lymphocyte counts or in plasma HIV concentration. Antibody against gp120 increased in individual patients in the 100- and 300-/microg groups. Some increases were noted in cytotoxic T lymphocyte activity against gp160-bearing targets and in lymphocyte proliferative activity. The safety and potential immunogenicity of an HIV-directed DNA-based vaccine was demonstrated, a finding that should encourage further studies.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Gene transfer in humans using a conditionally replicating lentiviral vector

Bruce L. Levine; Laurent Humeau; Jean D. Boyer; Rob-Roy MacGregor; Tessio Rebello; Xiaobin Lu; Gwendolyn K. Binder; Vladimir Slepushkin; Franck Lemiale; John R. Mascola; Frederic D. Bushman; Boro Dropulic; Carl H. June

We report findings from a clinical evaluation of lentiviral vectors in a phase I open-label nonrandomized clinical trial for HIV. This trial evaluated the safety of a conditionally replicating HIV-1-derived vector expressing an antisense gene against the HIV envelope. Five subjects with chronic HIV infection who had failed to respond to at least two antiviral regimens were enrolled. A single i.v. infusion of gene-modified autologous CD4 T cells was well tolerated in all patients. Viral loads were stable, and one subject exhibited a sustained decrease in viral load. CD4 counts remained steady or increased in four subjects, and sustained gene transfer was observed. Self-limiting mobilization of the vector was observed in four of five patients. There is no evidence for insertional mutagenesis after 21–36 months of observation. Immune function improved in four subjects. Lentiviral vectors appear promising for gene transfer to humans.


European Journal of Immunology | 1998

Modulation of amplitude and direction of in vivo immune responses by co-administration of cytokine gene expression cassettes with DNA immunogens

Jong J. Kim; Neil N. Trivedi; Liesl K. Nottingham; Lake Morrison; Anthony Tsai; Yin Hu; Sundarasamy Mahalingam; Kesen Dang; Lois Ahn; Nicole K. Doyle; Darren M. Wilson; Michael A. Chattergoon; Ara A. Chalian; Jean D. Boyer; Michael G. Agadjanyan; David B. Weiner

Immunization with nucleic acids has been shown to induce both antigen‐specific cellular and humoral immune responses in vivo. We hypothesize that immunization with DNA could be enhanced by directing specific immune responses induced by the vaccine based on the differential correlates of protection known for a particular pathogen. Recently we and others reported that specific immune responses generated by DNA vaccine could be modulated by co‐delivery of gene expression cassettes encoding for IL‐12, granulocyte‐macrophage colony‐stimulating factor and the co‐stimulatory molecule CD86. To further engineer the immune response in vivo, we investigated the induction and regulation of immune responses following the co‐delivery of pro‐inflammatory cytokine (IL‐1α, TNF‐α, and TNF‐β), Th1 cytokine (IL‐2, IL‐12, IL‐15, and IL‐18), and Th2 cytokine (IL‐4, IL‐5 and IL‐10) genes. We observed enhancement of antigen‐specific humoral response with the co‐delivery of Th2 cytokine genes IL‐4, IL‐5, and IL‐10 as well as those of IL‐2 and IL‐18. A dramatic increase in antigen‐specific T helper cell proliferation was seen with IL‐2 and TNF‐α gene co‐injections. In addition, we observed a significant enhancement of the cytotoxic response with the co‐administration of TNF‐α and IL‐15 genes with HIV‐1 DNA immunogens. These increases in CTL response were both MHC class I restricted and CD8+ T cell dependent. Together with earlier reports on the utility of co‐immunizing using immunologically important molecules together with DNA immunogens, we demonstrate the potential of this strategy as an important tool for the development of more rationally designed vaccines.


Journal of Clinical Investigation | 1998

CD8 positive T cells influence antigen-specific immune responses through the expression of chemokines.

Jong J. Kim; Liesl K. Nottingham; Jeong I. Sin; Anthony Tsai; Lake Morrison; Jim Oh; Kesen Dang; Yin Hu; Ken Kazahaya; Mosi Bennett; Tzvete Dentchev; Darren M. Wilson; Ara A. Chalian; Jean D. Boyer; Michael G. Agadjanyan; David B. Weiner

The potential roles of CD8(+) T-cell-induced chemokines in the expansion of immune responses were examined using DNA immunogen constructs as model antigens. We coimmunized cDNA expression cassettes encoding the alpha-chemokines IL-8 and SDF-1alpha and the beta-chemokines MIP-1alpha, RANTES, and MCP-1 along with DNA immunogens and analyzed the resulting antigen-specific immune responses. In a manner more similar to the traditional immune modulatory role of CD4(+) T cells via the expression of Th1 or Th2 cytokines, CD8(+) T cells appeared to play an important role in immune expansion and effector function by producing chemokines. For instance, IL-8 was a strong inducer of CD4(+) T cells, indicated by strong T helper proliferative responses as well as an enhancement of antibody responses. MIP-1alpha had a dramatic effect on antibody responses and modulated the shift of immune responses to a Th2-type response. RANTES coimmunization enhanced the levels of antigen-specific Th1 and cytotoxic T lymphocyte (CTL) responses. Among the chemokines examined, MCP-1 was the most potent activator of CD8(+) CTL activity. The enhanced CTL results are supported by the increased expression of Th1 cytokines IFN-gamma and TNF-alpha and the reduction of IgG1/IgG2a ratio. Our results support that CD8(+) T cells may expand both humoral and cellular responses in vivo through the elaboration of specific chemokines at the peripheral site of infection during the effector stage of the immune response.


The FASEB Journal | 1998

Modulating the immune response to genetic immunization

Adam D. Cohen; Jean D. Boyer; David B. Weiner

Genetic immunization, also known as DNA or polynucleotide immunization, is a novel strategy for vaccine development in which plasmid DNA encoding either individual or a collection of antigens is directly administered to a host. Such immunization leads to host expression of the delivered foreign gene, resulting in the induction of a specific immune response against the in vivo produced antigen. DNA immunization has been shown to induce protective immune responses in several infectious disease and cancer experimental model systems. Furthermore, DNA vaccines have recently entered the clinic for analysis as both prophylactic and therapeutic agents. Although the mechanisms of immunity to DNA have not yet been fully elucidated, it has become apparent that the immune response achieved by DNA vaccination is quite malleable, and can be manipulated by altering the conditions under which the vaccine is administered. Either through changing the method or location of immunization, altering the number of immunostimulatory sequences in the plasmid, altering the immunization regimen, or coadministering genes for cytokines or costimulatory molecules, one can modulate both the magnitude and orientation of the subsequent immune response. Through maximization of this feature of DNA immunization, we will likely be able to design vaccines and immunotherapeutic agents that are tailored to the correlates of protection for a particular disease, resulting in a new generation of more focused and effective immune stimulating agents.—Cohen, A. D., Boyer, J. D., Weiner, D. B. Modulating the immune response to genetic immunization. FASEB J. 12, 1611–1626 (1998)


Journal of Immunology | 2005

Coimmunization with an Optimized IL-15 Plasmid Results in Enhanced Function and Longevity of CD8 T Cells That Are Partially Independent of CD4 T Cell Help

Michele Kutzler; Tara M. Robinson; Michael A. Chattergoon; Daniel K. Choo; Andrew Y. Choo; Philip Y. Choe; Mathura P. Ramanathan; Rose Parkinson; Sagar Kudchodkar; Yutaka Tamura; Maninder K. Sidhu; Vidia Roopchand; J. Joseph Kim; George N. Pavlakis; Barbara K. Felber; Thomas A. Waldmann; Jean D. Boyer; David B. Weiner

DNA vaccines are a promising technology for the induction of Ag-specific immune responses, and much recent attention has gone into improving their immune potency. In this study we test the feasibility of delivering a plasmid encoding IL-15 as a DNA vaccine adjuvant for the induction of improved Ag-specific CD8+ T cellular immune responses. Because native IL-15 is poorly expressed, we used PCR-based strategies to develop an optimized construct that expresses 80-fold higher than the native IL-15 construct. Using a DNA vaccination model, we determined that immunization with optimized IL-15 in combination with HIV-1gag DNA constructs resulted in a significant enhancement of Ag-specific CD8+ T cell proliferation and IFN-γ secretion, and strong induction of long-lived CD8+ T cell responses. In an influenza DNA vaccine model, coimmunization with plasmid expressing influenza A PR8/34 hemagglutinin with the optimized IL-15 plasmid generated improved long term CD8+ T cellular immunity and protected the mice against a lethal mucosal challenge with influenza virus. Because we observed that IL-15 appeared to mostly adjuvant CD8+ T cell function, we show that in the partial, but not total, absence of CD4+ T cell help, plasmid-delivered IL-15 could restore CD8 secondary immune responses to an antigenic DNA plasmid, supporting the idea that the effects of IL-15 on CD8+ T cell expansion require the presence of low levels of CD4 T cells. These data suggest a role for enhanced plasmid IL-15 as a candidate adjuvant for vaccine or immunotherapeutic studies.


The Lancet | 2015

Safety, efficacy, and immunogenicity of VGX-3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2b trial

Cornelia L. Trimble; Matthew P. Morrow; Kimberly A. Kraynyak; Xuefei Shen; Michael J. Dallas; Jian Yan; Lance Edwards; R Lamar Parker; Lynette Denny; Mary Giffear; Ami Shah Brown; Kathleen Marcozzi-Pierce; Divya Shah; Anna Slager; Albert Sylvester; Amir R. Khan; Kate E. Broderick; Robert J Juba; Timothy A Herring; Jean D. Boyer; Jessica Lee; Niranjan Y. Sardesai; David B. Weiner; Mark L. Bagarazzi

BACKGROUND Despite preventive vaccines for oncogenic human papillomaviruses (HPVs), cervical intraepithelial neoplasia (CIN) is common, and current treatments are ablative and can lead to long-term reproductive morbidity. We assessed whether VGX-3100, synthetic plasmids targeting HPV-16 and HPV-18 E6 and E7 proteins, delivered by electroporation, would cause histopathological regression in women with CIN2/3. METHODS Efficacy, safety, and immunogenicity of VGX-3100 were assessed in CIN2/3 associated with HPV-16 and HPV-18, in a randomised, double-blind, placebo-controlled phase 2b study. Patients from 36 academic and private gynaecology practices in seven countries were randomised (3:1) to receive 6 mg VGX-3100 or placebo (1 mL), given intramuscularly at 0, 4, and 12 weeks. Randomisation was stratified by age (<25 vs ≥25 years) and CIN2 versus CIN3 by computer-generated allocation sequence (block size 4). Funder and site personnel, participants, and pathologists were masked to treatment. The primary efficacy endpoint was regression to CIN1 or normal pathology 36 weeks after the first dose. Per-protocol and modified intention-to-treat analyses were based on patients receiving three doses without protocol violations, and on patients receiving at least one dose, respectively. The safety population included all patients who received at least one dose. The trial is registered at ClinicalTrials.gov (number NCT01304524) and EudraCT (number 2012-001334-33). FINDINGS Between Oct 19, 2011, and July 30, 2013, 167 patients received either VGX-3100 (n=125) or placebo (n=42). In the per-protocol analysis 53 (49·5%) of 107 VGX-3100 recipients and 11 (30·6%) of 36 placebo recipients had histopathological regression (percentage point difference 19·0 [95% CI 1·4-36·6]; p=0·034). In the modified intention-to-treat analysis 55 (48·2%) of 114 VGX-3100 recipients and 12 (30·0%) of 40 placebo recipients had histopathological regression (percentage point difference 18·2 [95% CI 1·3-34·4]; p=0·034). Injection-site reactions occurred in most patients, but only erythema was significantly more common in the VGX-3100 group (98/125, 78·4%) than in the placebo group (24/42, 57·1%; percentage point difference 21·3 [95% CI 5·3-37·8]; p=0·007). INTERPRETATION VGX-3100 is the first therapeutic vaccine to show efficacy against CIN2/3 associated with HPV-16 and HPV-18. VGX-3100 could present a non-surgical therapeutic option for CIN2/3, changing the treatment outlook for this common disease. FUNDING Inovio Pharmaceuticals.


BMC Immunology | 2005

Standardization of cytokine flow cytometry assays

Holden T. Maecker; Aline Rinfret; Patricia D'Souza; Janice Darden; Eva Roig; Claire Landry; Peter Hayes; Josephine Birungi; Omu Anzala; Miguel Garcia; Alexandre Harari; Ian Frank; Ruth Baydo; Megan Baker; Jennifer Holbrook; Janet Ottinger; Laurie Lamoreaux; C. Lorrie Epling; Elizabeth Sinclair; Maria A. Suni; Kara Punt; Sandra A. Calarota; Sophia El-Bahi; Gailet Alter; Hazel Maila; Ellen Kuta; Josephine H. Cox; Clive M. Gray; Marcus Altfeld; Nolwenn Nougarede

BackgroundCytokine flow cytometry (CFC) or intracellular cytokine staining (ICS) can quantitate antigen-specific T cell responses in settings such as experimental vaccination. Standardization of ICS among laboratories performing vaccine studies would provide a common platform by which to compare the immunogenicity of different vaccine candidates across multiple international organizations conducting clinical trials. As such, a study was carried out among several laboratories involved in HIV clinical trials, to define the inter-lab precision of ICS using various sample types, and using a common protocol for each experiment (see additional files online).ResultsThree sample types (activated, fixed, and frozen whole blood; fresh whole blood; and cryopreserved PBMC) were shipped to various sites, where ICS assays using cytomegalovirus (CMV) pp65 peptide mix or control antigens were performed in parallel in 96-well plates. For one experiment, antigens and antibody cocktails were lyophilised into 96-well plates to simplify and standardize the assay setup. Results (CD4+cytokine+ cells and CD8+cytokine+ cells) were determined by each site. Raw data were also sent to a central site for batch analysis with a dynamic gating template.Mean inter-laboratory coefficient of variation (C.V.) ranged from 17–44% depending upon the sample type and analysis method. Cryopreserved peripheral blood mononuclear cells (PBMC) yielded lower inter-lab C.V.s than whole blood. Centralized analysis (using a dynamic gating template) reduced the inter-lab C.V. by 5–20%, depending upon the experiment. The inter-lab C.V. was lowest (18–24%) for samples with a mean of >0.5% IFNγ + T cells, and highest (57–82%) for samples with a mean of <0.1% IFNγ + cells.ConclusionICS assays can be performed by multiple laboratories using a common protocol with good inter-laboratory precision, which improves as the frequency of responding cells increases. Cryopreserved PBMC may yield slightly more consistent results than shipped whole blood. Analysis, particularly gating, is a significant source of variability, and can be reduced by centralized analysis and/or use of a standardized dynamic gating template. Use of pre-aliquoted lyophilized reagents for stimulation and staining can provide further standardization to these assays.


Cancer Immunology, Immunotherapy | 2008

Results and harmonization guidelines from two large-scale international Elispot proficiency panels conducted by the Cancer Vaccine Consortium (CVC/SVI)

Sylvia Janetzki; Katherine S. Panageas; Leah Ben-Porat; Jean D. Boyer; Cedrik M. Britten; Timothy M. Clay; Michael Kalos; Holden T. Maecker; Pedro Romero; Jianda Yuan; W. Martin Kast; Axel Hoos

The Cancer Vaccine Consortium of the Sabin Vaccine Institute (CVC/SVI) is conducting an ongoing large-scale immune monitoring harmonization program through its members and affiliated associations. This effort was brought to life as an external validation program by conducting an international Elispot proficiency panel with 36 laboratories in 2005, and was followed by a second panel with 29 participating laboratories in 2006 allowing for application of learnings from the first panel. Critical protocol choices, as well as standardization and validation practices among laboratories were assessed through detailed surveys. Although panel participants had to follow general guidelines in order to allow comparison of results, each laboratory was able to use its own protocols, materials and reagents. The second panel recorded an overall significantly improved performance, as measured by the ability to detect all predefined responses correctly. Protocol choices and laboratory practices, which can have a dramatic effect on the overall assay outcome, were identified and lead to the following recommendations: (A) Establish a laboratory SOP for Elispot testing procedures including (A1) a counting method for apoptotic cells for determining adequate cell dilution for plating, and (A2) overnight rest of cells prior to plating and incubation, (B) Use only pre-tested serum optimized for low background: high signal ratio, (C) Establish a laboratory SOP for plate reading including (C1) human auditing during the reading process and (C2) adequate adjustments for technical artifacts, and (D) Only allow trained personnel, which is certified per laboratory SOPs to conduct assays. Recommendations described under (A) were found to make a statistically significant difference in assay performance, while the remaining recommendations are based on practical experiences confirmed by the panel results, which could not be statistically tested. These results provide initial harmonization guidelines to optimize Elispot assay performance to the immunotherapy community. Further optimization is in process with ongoing panels.


Science Translational Medicine | 2012

Immunotherapy Against HPV16/18 Generates Potent TH1 and Cytotoxic Cellular Immune Responses

Mark L. Bagarazzi; Jian Yan; Matthew P. Morrow; Xuefei Shen; Parker Rl; Jinhee Lee; Giffear M; Panyupa Pankhong; Amir S. Khan; Kate E. Broderick; Feng Lin; Jean D. Boyer; Ruxandra Draghia-Akli; White Cj; Jung-Ok Kim; David B. Weiner; Niranjan Y. Sardesai

CD8+ T cells with cytolytic activity are induced after therapeutic human papillomavirus vaccination in humans. Shocking HPV into Submission Human papillomavirus (HPV) infection is frequently asymptomatic but can lead to the development of cervical cancer in infected women. Current vaccines against HPV are quite effective at preventing infection; however, there is no vaccine to help those already infected. Now, Bagarazzi et al. report that a therapeutic vaccine for HPV can induce an immune response in a phase 1 study. VGX-3100 is a candidate vaccine for the high-risk HPV serotypes 16 and 18. Here, 18 women previously treated for cervical neoplasia—a precursor to cervical cancer—were given the DNA vaccine VGX-3100 by electroporation—where a small localized electric pulse accompanies the injection—in a range of doses. Previous attempts at DNA vaccination have proved less than successful in clinical trials; however, preclinical studies suggest that electroporation may greatly enhance the efficacy of the vaccine. The authors show that the electroporation-delivered VGX-3100 induces a robust HPV-specific immune response in previously infected individuals and that the vaccine is safe and immunogenic. Although efficacy remains to be tested in a larger trial, the enhanced immune response elicited by VGX-3100 may attack HPV-infected cells, potentially inducing cancer regression in individuals already infected with HPV. Despite the development of highly effective prophylactic vaccines against human papillomavirus (HPV) serotypes 16 and 18, prevention of cervical dysplasia and cancer in women infected with high-risk HPV serotypes remains an unmet medical need. We report encouraging phase 1 safety, tolerability, and immunogenicity results for a therapeutic HPV16/18 candidate vaccine, VGX-3100, delivered by in vivo electroporation (EP). Eighteen women previously treated for cervical intraepithelial neoplasia grade 2 or 3 (CIN2/3) received a three-dose (intramuscular) regimen of highly engineered plasmid DNA encoding HPV16 and HPV18 E6/E7 antigens followed by EP in a dose escalation study (0.3, 1, and 3 mg per plasmid). Immunization was well tolerated with reports of mild injection site reactions and no study-related serious or grade 3 and 4 adverse events. No dose-limiting toxicity was noted, and pain was assessed by visual analog scale, with average scores decreasing from 6.2/10 to 1.4 within 10 min. Average peak interferon-γ enzyme-linked immunospot magnitudes were highest in the 3 mg cohort in comparison to the 0.3 and 1 mg cohorts, suggesting a trend toward a dose effect. Flow cytometric analysis revealed the induction of HPV-specific CD8+ T cells that efficiently loaded granzyme B and perforin and exhibited full cytolytic functionality in all cohorts. These data indicate that VGX-3100 is capable of driving robust immune responses to antigens from high-risk HPV serotypes and could contribute to elimination of HPV-infected cells and subsequent regression of the dysplastic process.

Collaboration


Dive into the Jean D. Boyer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark L. Bagarazzi

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Kenneth E. Ugen

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jian Yan

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Mark G. Lewis

Southern Research Institute

View shared research outputs
Top Co-Authors

Avatar

George N. Pavlakis

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Anlan Dai

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge