Network


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

Hotspot


Dive into the research topics where David Apelian is active.

Publication


Featured researches published by David Apelian.


Vaccine | 2014

Safety, tolerability and immunogenicity of GS-4774, a hepatitis B virus-specific therapeutic vaccine, in healthy subjects: a randomized study.

A. Gaggar; Claire Coeshott; David Apelian; Timothy C. Rodell; Brian R. Armstrong; Gong Shen; G. Mani Subramanian; John G. McHutchison

BACKGROUND GS-4774 is a recombinant, heat-killed, yeast-based immunotherapy engineered to express hepatitis B virus (HBV)-specific antigens. GS-4774 is being developed as a therapeutic vaccine for chronic HBV infection. The aim of this study was to assess the safety, tolerability and immunogenicity of GS-4774 in healthy subjects. DESIGN This was a randomized, open-label, dose-ascending study. Subjects were allocated to one of three dose groups (n=20 per group) to receive 10, 40 or 80 yeast units (YU; 1YU=10(7) yeast) of GS-4774 in two immunization regimens (five subcutaneous injections at weekly intervals with one monthly booster or three subcutaneous injections at monthly intervals). T-cell-mediated responses were determined by interferon (IFN)-γ enzyme-linked immunospot (ELISpot) assay and lymphocyte-proliferation assay (LPA). RESULTS Adverse events were reported by 39 of 60 (65%) subjects; all were mild or moderate and none was serious. Adverse events occurred most frequently in the highest dose group, 80YU, and the number of individual events was higher after weekly immunization than monthly. The most common adverse events were injection-site reactions. Most (88%) subjects responded to GS-4774 by at least one of the T-cell assays. Following immunization with GS-4774, IFN-γ-producing T-cells specific for HBV antigens were detectable in 30 (51%) subjects. The ELISpot response was observed at all doses, with the highest frequency of responders occurring at the highest dose (10YU: 45%; 40YU: 35%; 80YU: 74%). Proliferative responses to HBV recombinant antigens were observed in 90% subjects; responses were mainly independent of GS-4774 dose and immunization regimen. CONCLUSIONS GS-4774 was safe and well-tolerated in healthy subjects with injection-site reactions being the most frequently reported adverse events. With both weekly and monthly regimens, GS-4774 provided HBV-specific immune responses at all doses evaluated. Further evaluation of GS-4774 is ongoing in patients with chronic HBV infection. CLINICAL TRIAL REGISTRY Clinicaltrials.gov (NCT01779505).


Clinical Cancer Research | 2013

Combination Therapy with a Second-Generation Androgen Receptor Antagonist and a Metastasis Vaccine Improves Survival in a Spontaneous Prostate Cancer Model

Andressa Ardiani; Benedetto Farsaci; Connie J. Rogers; Andrew Asher Protter; Zhimin Guo; Thomas H. King; David Apelian; James W. Hodge

Purpose: Enzalutamide, a second-generation androgen antagonist, was approved by the U.S. Food and Drug Administration (FDA) for castration-resistant prostate cancer (CRPC) treatment. Immunotherapy has been shown to be a promising strategy for prostate cancer. This study was performed to provide data to support the combination of enzalutamide and immunotherapy for CRPC treatment. Experimental Design: Male C57BL/6 or TRAMP (transgenic adenocarcinoma of the mouse prostate) prostate cancer model mice were exposed to enzalutamide and/or a therapeutic vaccine targeting Twist, an antigen involved in epithelial-to-mesenchymal transition and metastasis. The physiologic and immunologic effects of enzalutamide were characterized. The generation of Twist-specific immunity by Twist-vaccine was assessed. Finally, the combination of enzalutamide and Twist-vaccine to improve TRAMP mice overall survival was evaluated. Results: Enzalutamide mediated immunogenic modulation in TRAMP-C2 cells. In vivo, enzalutamide mediated reduced genitourinary tissue weight, enlargement of the thymus, and increased levels of T-cell excision circles. Because no changes were seen in T-cell function, as determined by CD4+ T-cell proliferation and regulatory T cell (Treg) functional assays, enzalutamide was determined to be immune inert. Enzalutamide did not diminish the ability of Twist-vaccine to generate Twist-specific immunity. Twist was confirmed as a valid tumor antigen in TRAMP mice by immunohistochemistry. The combination of enzalutamide and Twist-vaccine resulted in significantly increased overall survival of TRAMP mice compared with other treatment groups (27.5 vs. 10.3 weeks). Notably, the effectiveness of the combination therapy increased with disease stage, i.e., the greatest survival benefit was seen in mice with advanced-stage prostate tumors. Conclusions: These data support the combination of enzalutamide and immunotherapy as a promising treatment strategy for CRPC. Clin Cancer Res; 19(22); 6205–18. ©2013 AACR.


PLOS ONE | 2014

A Whole Recombinant Yeast-Based Therapeutic Vaccine Elicits HBV X, S and Core Specific T Cells in Mice and Activates Human T Cells Recognizing Epitopes Linked to Viral Clearance

Thomas H. King; Charles B. Kemmler; Zhimin Guo; Derrick Mann; Yingnian Lu; Claire Coeshott; Adam J. Gehring; Antonio Bertoletti; Zi Z. Ho; William Delaney; A. Gaggar; G. Mani Subramanian; John G. McHutchison; Shikha Shrivastava; Yu Jin L Lee; Shyamasundaran Kottilil; Donald Bellgrau; Timothy C. Rodell; David Apelian

Chronic hepatitis B infection (CHB) is characterized by sub-optimal T cell responses to viral antigens. A therapeutic vaccine capable of restoring these immune responses could potentially improve HBsAg seroconversion rates in the setting of direct acting antiviral therapies. A yeast-based immunotherapy (Tarmogen) platform was used to make a vaccine candidate expressing hepatitis B virus (HBV) X, surface (S), and Core antigens (X-S-Core). Murine and human immunogenicity models were used to evaluate the type and magnitude of HBV-Ag specific T cell responses elicited by the vaccine. C57BL/6J, BALB/c, and HLA-A*0201 transgenic mice immunized with yeast expressing X-S-Core showed T cell responses to X, S and Core when evaluated by lymphocyte proliferation assay, ELISpot, intracellular cytokine staining (ICS), or tumor challenge assays. Both CD4+ and CD8+ T cell responses were observed. Human T cells transduced with HBc18–27 and HBs183–91 specific T cell receptors (TCRs) produced interferon gamma (IFNγ following incubation with X-S-Core-pulsed dendritic cells (DCs). Furthermore, stimulation of peripheral blood mononuclear cells (PBMCs) isolated from CHB patients or from HBV vaccine recipients with autologous DCs pulsed with X-S-Core or a related product (S-Core) resulted in pronounced expansions of HBV Ag-specific T cells possessing a cytolytic phenotype. These data indicate that X-S-Core-expressing yeast elicit functional adaptive immune responses and supports the ongoing evaluation of this therapeutic vaccine in patients with CHB to enhance the induction of HBV-specific T cell responses.


Clinical Lung Cancer | 2014

Phase II Study of the GI-4000 KRAS Vaccine After Curative Therapy in Patients With Stage I-III Lung Adenocarcinoma Harboring a KRAS G12C, G12D, or G12V Mutation

Jamie E. Chaft; Anya Litvak; Maria E. Arcila; Payal R. Patel; Sandra P. D'Angelo; Lee M. Krug; Valerie W. Rusch; Alicia Mattson; Claire Coeshott; Bernard J. Park; David Apelian; Mark G. Kris; Christopher G. Azzoli

INTRODUCTION Patients with early-stage lung cancer have a high risk of recurrence despite multimodality therapy. KRAS-mutant lung adenocarcinomas are the largest genetically defined subgroup, representing 25% of patients. GI-4000 is a heat-killed recombinant Saccharomyces cerevisiae yeast-derived vaccine expressing mutant KRAS proteins. The present phase II study assessed the feasibility, immunogenicity, and safety of the GI-4000 vaccine in patients with early-stage, KRAS-mutant lung cancer. MATERIALS AND METHODS Patients with stage I-III KRAS-mutant lung cancer who completed curative therapy were enrolled. The patients received the genotype matched GI-4000 vaccine for ≤ 3 years or until intolerance, disease recurrence, or death. The KRAS antigen T-cell response was assessed using the interferon-gamma enzyme-linked immunospot assay in peripheral blood mononuclear cells. The study was powered to detect an immune response in ≥ 25% of patients. RESULTS A total of 24 patients were enrolled over 28 months. No vaccine-related serious adverse events occurred. One patient withdrew consent because of pain at the injection site. The study met its primary endpoint, with 50% of patients developing an immune response to mutant KRAS. The median number of vaccinations received was 15 (range, 1-19). Ten patients experienced disease recurrence, and 6 died. Compared with the genotypically matched historical controls, the recurrence rates were equivalent but overall survival showed a favorable trend. CONCLUSION GI-4000 was well tolerated and immunogenic when used as consolidation therapy in patients with stage I-III KRAS-mutant lung cancer. The patterns of recurrence and death observed in the present study can be used to design a randomized study of GI-4000 with overall survival as the primary endpoint.


Vaccine | 2011

Maturation of human dendritic cells with Saccharomyces cerevisiae (yeast) reduces the number and function of regulatory T cells and enhances the ratio of antigen-specific effectors to regulatory T cells.

Vittore Cereda; Matteo Vergati; Ngar-Yee Huen; Maria Giovanna di Bari; Caroline Jochems; Chiara Intrivici; James L. Gulley; David Apelian; Jeffrey Schlom; Kwong Y. Tsang

We compared the effects of yeast-treated human dendritic cells (DCs) with CD40L-matured human DCs for the induction of effector cells and the number and functionality of CD4(+)CD25(+)CD127(-)FoxP3(+) regulatory T cells (Tregs). DCs were treated with yeast or CD40L and cocultured with isolated autologous CD4(+) T cells. CD4(+)CD25(+)CD127(-) T cells isolated from the coculture of CD4(+) T cells plus yeast-treated DCs (yeast coculture) had a lower expression of FoxP3 and decreased suppressive function compared to CD4(+)CD25(+)CD127(-) T cells isolated from the coculture of CD4(+) T cells plus CD40L-treated DCs (CD40L coculture). Also, compared to the CD40L coculture, the yeast coculture showed increases in the ratio of CD4(+)CD25(+) activated T cells to Tregs and in the production of Th1-related cytokines (IL-2, TNF-α, IFN-γ) and IL-6. In addition, yeast-treated DCs used as antigen-presenting cells (APCs) incubated with the tumor antigen CEA enhanced the proliferation of CEA-specific CD4(+) T cells compared to the use of CD40L-matured DCs used as APCs. This is the first study to report on the role of yeast-treated/matured human DCs in reducing Treg frequency and functionality and in enhancing effector to Treg ratios. These results provide an additional rationale for the use of yeast as a vector in cancer vaccines.


Cancer Research | 2011

Abstract 758: Cancer vaccine immunotherapy employing Saccharomyces cerevisiae (yeast) as a vector can modulate the balance between CD4+ T cells and regulatory T cells (Tregs) and enhance the specific antitumor immune response

Matteo Vergati; Vittore Cereda; Chiara Intrivici; Ngar-Yee Huen; Maria Giovanna di Bari; Caroline Jochems; James L. Gulley; David Apelian; Jeffrey Schlom; Kwong Y. Tsang

Regulatory T cells (Tregs) have been broadly related to modulation of the antitumor immune response. We have previously shown that whole, heat-killed, recombinant Saccharomyces cerevisiae yeast genetically modified to express carcinoembryonic antigen (yeast-CEA) can efficiently activate human dendritic cells (DCs) and stimulate CEA-specific CD8+ T cells. In the present study, we further investigated how the balance between CD4+ T cells and Tregs induced by yeast-treated DCs might affect the generation of a CEA-specific immune response. Autologous CD4+ T cells were co-cultured with human DCs matured by yeast vs. CD40L. Activated CD4+CD25+ effector T cells (Teffs) and CD4+CD25+CD127- Tregs isolated from the two co-cultures were analyzed. A higher Teff/Treg ratio, greater levels of Th1-related cytokines, lower expression of FoxP3 in Tregs, decreased FoxP3 demethylation levels in the Treg-specific demethylated region, and decreased Treg suppressive function were found in the cells isolated from the yeast co-culture compared to those from the CD40L co-culture. DCs isolated from the yeast co-culture showed a significant decrease in surface expression of programmed death ligand 2 (PD-L2) vs. the CD40L co-culture (55.7% vs. 31.2%). No differences were observed in programmed death ligand 1 (PD-L1) levels. Teffs isolated from the yeast co-culture had higher levels of PD-L1 (22.9% vs. 13.7%), but significantly lower levels of programmed death 1 (PD-1, 2.4% vs. 11.6%), compared to the co-culture using CD40L-matured DCs. Tregs isolated from the yeast co-culture showed a significant increase in surface expression of PD-L1 (25.5% vs. 3.6%) and no difference in levels of PD-1 (0.7% vs. 1.4%) compared to the CD40L co-culture. These pro-inflammatory changes may partially explain both the increased CEA-specific CD4+ T-cell proliferation using yeast- vs. CD40L-treated DCs pulsed with CEA protein, and the higher percentage of CEA-specific CD4+ T cells generated when yeast-matured DCs vs. CD40L-treated DCs were used as antigen-presenting cells in the presence of CEA protein. This is the first study to report on the role of yeast-treated human DCs in modulating the balance between Teffs and Tregs and thus enhancing the antitumor immune response. Altogether, these preliminary findings provide a rationale for further evaluation of yeast constructs in cancer vaccine immunotherapy. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 758. doi:10.1158/1538-7445.AM2011-758


Vaccine | 2007

Whole recombinant yeast-based immunotherapy induces potent T cell responses targeting HCV NS3 and Core proteins

Aurelia Haller; Georg M. Lauer; Thomas H. King; Charles B. Kemmler; Valerie Fiolkoski; Yingnian Lu; Don Bellgrau; Timothy C. Rodell; David Apelian; Alex Franzusoff; Richard C. Duke


Oncotarget | 2013

Immunological targeting of tumor cells undergoing an epithelial-mesenchymal transition via a recombinant brachyury-yeast vaccine

Duane H. Hamilton; Mary T. Litzinger; Alessandra Jales; Bruce Huang; Romaine I. Fernando; James W. Hodge; Andressa Ardiani; David Apelian; Jeffrey Schlom; Claudia Palena


Cancer Immunology, Immunotherapy | 2014

Phase I trial of a recombinant yeast-CEA vaccine (GI-6207) in adults with metastatic CEA-expressing carcinoma

Marijo Bilusic; Christopher R. Heery; Philip M. Arlen; Myrna Rauckhorst; David Apelian; Kwong Y. Tsang; Jo A. Tucker; Caroline Jochems; Jeffrey Schlom; James L. Gulley; Ravi A. Madan


Archive | 2006

COMPOSITIONS AND METHODS FOR ELICITING AN IMMUNE RESPONSE TO ESCAPE MUTANTS OF TARGETED THERAPIES

David Apelian; Alex Franzusoff; Timothy C. Rodell

Collaboration


Dive into the David Apelian's collaboration.

Top Co-Authors

Avatar

Alex Franzusoff

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Thomas H. King

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Jeffrey Schlom

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Zhimin Guo

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Richard C. Duke

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Yingnian Lu

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

James L. Gulley

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kwong Y. Tsang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

James W. Hodge

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge