
Clinical Trials of Dendritic Cell Therapies for Cancer: Biotech's Bumpy Road to the Market
Monday, October 28, 2013
Dendritic cells (DCs) are the most potent antigen-presenting cells known; owing to their ability to stimulate antigen-specific cytolytic and memory T-cell responses, their use as cancer vaccines is rapidly increasing. While clinical trials provide evidence that dendritic cells vaccines are safe and elicit immunological responses in most patients, few complete tumor remissions have been reported and further technological advances are required. An effective dendritic cell vaccine must possess and maintain several characteristics: it must migrate to lymph nodes, have a mature, Th1-polarizing phenotype expressed stably after infusion and present antigen for sufficient time to produce a T-cell response capable of eliminating a tumor. While dendritic cells are readily matured ex vivo, their phenotype and fate after infusion are rarely evaluable; therefore, strategies to ensure that dendritic cells access lymphoid tissues and retain an immunostimulatory phenotype are required. Recently, the FDA approved the first dendritic cell therapy, Provenge, developed by Dendreon. These efforts have helped to establish proof of principle that properly activated DCs, loaded with the proper form and dose of antigen and properly activated, as well as properly migrating to lymph nodes, can initiate and expand tumor-specific CD4+ and CD8+ T cell responses to induce meaningful therapeutic responses. So far, despite induction of robust tumor-specific T cell responses by DC cancer vaccines in many patients and occasional spectacular complete tumor regressions, particularly in patients with melanoma, the promise of this new therapeutic approach has not been fully realized. This symposium will highlight recent clinical trial results and provide a biotech perspective on the remaining bottlenecks and roadblocks that must be solved in order for DC therapy to be successfully commercialized.
*Reception to follow.
Registration and Webinar Pricing
Member | $30 |
Student/Postdoc Member | $15 |
Nonmember (Academia) | $65 |
Nonmember (Corporate) | $85 |
Nonmember (Non-profit) | $65 |
Nonmember (Student / Postdoc / Resident / Fellow) | $45 |
Mission Partner support for the Frontiers of Science program provided by 
Agenda
* Presentation titles and times are subject to change.
Monday, October 28, 2013 | |
8:00 AM | Registration and Continental Breakfast |
8:30 AM | Welcome and Introduction |
9:00 AM | Cellular Immunotherapy for T Cell Lymphoma: Advantages of Maturationally Synchronized, Physiologically Induced Dendritic Cells |
9:40 AM | Early Dendritic Cell Trials at Stanford: setting the stage for Sipuleucil-T |
10:20 AM | Coffee break |
10:50 AM | Dendritic Cell Vaccines Targeting Cancer Stem Cells |
11:30 AM | Biomarkers to Indicate Potentially Provenge-Responsive Prostate Cancer Patients |
12:10 PM | Interleukin-12 Secreting Dendritic Cells in Cancer Immune Therapy: A Randomised Clinical Trial for Brain Cancer |
12:50 PM | Networking lunch |
1:30 PM | From Nobel Prize to Bedside: Innovative Approaches to Personalized Dendritic Cell Therapies |
2:10 PM | Using the Tumor Immunopeptidome to Guide the Design of Personalized DC-Based Cancer Vaccines |
2:50 PM | Dendritic Cell Based Immunotherapy of Melanoma: the Brussels Experience |
3:10 PM | Coffee break |
3:40 PM | Collaborative Clinical Studies on Immunotherapies to Treat Ovarian Cancer |
4:20 PM | CVac™ Phase 2 Clinical Study for Treatment of Ovarian Cancer Patients in First & Second Remission (CAN-003 protocol): Manufacturing & Clinical Outcomes |
5:00 PM | Closing Remarks Networking reception |
6:00 PM | Close |
Speakers
Organizers
John E. Hambor, PhD
Boehringer Ingelheim Pharmaceuticals
Dr. John Hambor is currently a Distinguished Research Fellow at Boehringer Ingelheim where he coordinates a strategic postdoctoral research program focused on developing new drug concepts in collaboration with academic investigators. Previously, Dr. Hambor was a consultant with the Cell Therapy Group, specializing in stem cell-based drug discovery. Prior to serving as CEO of CellDesign, a developer of next generation stem cell technologies, he contributed 17 years of research at Pfizer where he identified and validated new drug targets in the areas of inflammation and immunology and developed stem cell-based assays for drug efficacy and safety studies. Dr. Hambor received both a BA and MS degree in Microbiology from Miami University of Ohio, and earned a PhD in Pathology from Case Western Reserve University. As a postdoctoral fellow at Yale University in the Department of Immunobiology, he researched the molecular basis of CD8 expression during T cell development. He has been an Adjunct Assistant Professor at Connecticut College since 2000 as where he teaches Immunology. He also serves as a member of the board of directors for the VA Connecticut Research and Education Foundation.
Jennifer S. Henry, PhD
The New York Academy of Sciences
Speakers
Richard L. Edelson, MD
Yale School of Medicine
Three interlocking themes form the supporting legs of the academic, research and clinical career of Richard Edelson, MD., and are germane to this symposium. He has focused on the development and implementation of cellular immunotherapy for a lymphoma, has elucidated the clinically relevant fundamental biology of malignant T cells and has been responsible for strengthening synergies between basic scientists and translational clinicians. He was the originator of extracorporeal photochemo-therapy (ECPs), the first FDA-approved cellular immunotherapy for cancer. That therapy has to date been administered more than a million times in over 500 university medical centers, as a primary treatment for T cell lymphoma and as a therapy for graft-versus-host disease and organ transplant rejection. He led the team that determined that the scientific basis of ECP’s efficacy is its efficient generation, without addition of maturational cytokines, of monocyte conversion to cross-presenting antigen presenting cells. That discovery has stimulated his group’s current efforts to restructure and refine that therapy for potential use in other types of cancer. Dr. Edelson is a graduate of the Yale University School of Medicine, trained in Internal Medicine at the University of Chicago and in Dermatology at Harvard and did a dual fellowship in the Immunology Laboratory of the National Institute of Allergy and Infectious Diseases and the Medical Oncology Branch of the National Cancer Institute of the NIH. After a decade on the faculty of Columbia University’s College of Physicians and Surgeons, where he served as Deputy Director of that institution’s General Clinical Research Center and as Leader of the Immunology Program of its Cancer Center, he relocated to Yale University, as Professor and Chairman of its Department of Dermatology, a position he has held continuously from 1986 to present. From 2003 to 2009, he was also the Director of the Yale University Comprehensive Cancer Center. He has organized two past New York Academy of Science Symposia: “Antigen and Clone-Specific Immuno-regulation” and “Clinically Relevant Basic Biology of Cutaneous T Cell Lymphoma”.
Edgar G. Engleman, MD
Stanford University School of Medicine
Dr. Engleman is Professor of Pathology and Medicine at Stanford University School of Medicine, where he directs the Stanford Blood Center. The Center, with its 300 person staff, supplies blood products to Stanford and nearby community hospitals, and performs histocompatibility and genetic testing in support of the organ transplant programs at Stanford. Dr. Engleman has supervised more than 200 research trainees, authored more than 275 scientific articles and has been an editor of multiple scientific journals. Although his early research was directed at identifying and characterizing subsets of human T cells, for the past 25 years his group has been studying dendritic cell biology. After developing methods for isolating and arming human dendritic cells, he conceived of the idea of using these cells to vaccinate patients against their own tumors. Twenty years ago, Dr. Engleman and his collaborators at Stanford began performing dendritic cell trials in cancer patients. Based on encouraging results, he co-founded Dendreon Corporation, a biotech company dedicated to developing dendritic cell based cancer vaccines. He was Chairman of the company’s scientific advisory board for more than 10 years, and a number of his former trainees joined the company and led its research and development effort. The company’s first vaccine, Sipuleucel-T or Provenge, was approved by the FDA in 2010 for the treatment of patients with advanced prostate cancer. In addition to Dendreon, Dr. Engleman has been a founder of several other biotech companies. In 1996 he co-founded Vivo Ventures, an investment firm that invests in biomedical companies in the U.S. and China.
Thomas Felzmann, MD
Activartis Biotech GmbH, Austria
Dr. Felzmann graduated in 1987 at the Medical University Wien where he worked as a postdoctoral fellow at the MUW’s Institute of Immunology. His clinical training in oncology began in 1990. In 1992 he returned to full-time research as a postdoc at the NIH, Bethesda, MD, where he stayed for three years. Returning to Austria in 1995, he began training in paediatric oncology and was charged with the establishment of the Laboratory of Tumour Immunology at the St. Anna Children’s Cancer Research Institute. During the next 5 years his team succeeded in developing a proprietary Dendritic Cell based Cancer Vaccine technology for which patents have been granted in the meantime. Starting in 2000 he conducted a series of phase he trials and in 2010 initiated a randomised efficacy study. Since 2003, the clinical development is conducted by Activartis, Vienna, AT, that is financed by a group of private investors.
Lana E. Kandalaft, PharmD, MTR, PhD
University of Pennsylvania
Dr. Lana Kandalaft has joined the Ovarian Cancer Research Center at the University of Pennsylvania in 2008 as Director of Clinical Development and Operations and as an Assistant Professor of Obstetrics and Gynecology with a special focus on whole tumor lysate vaccine development. Dr. Kandalaft has a Pharm.D and a Ph.D in Cell biology and drug delivery from The United Kingdom. She also holds a masters in translational research (MTR) from the University of Pennsylvania. She completed her postdoctoral fellowship training at The National Cancer Institute focusing on preclinical animal models and cancer therapeutics. She then continued working at The NCI as a senior research fellow before joining Penn. She recently took a new position as The Director of the Developmental Therapeutics Center at the Department of Oncology and The Ludwig Branch in Lausanne, Switzerland. She continues to be an Adjunct Assistant Professor at Penn working on whole tumor lysate vaccine approaches.
Matthew Lehman
Prima Biomed, Australia
Mr Lehman served as Prima BioMed’s Chief Operating Officer from February 2010 until his appointment as Chief Executive Officer in September 2012. Mr Lehman has experience in clinical research, development programs and obtaining drug approval. He has specific expertise in clinical development strategies, operations and in-outsourcing. From 2000 until 2010, Mr Lehman was Chief Operating Officer for SPRI Clinical Trials in the US and Europe where he managed teams in all areas of clinical operations. Mr Lehman is based in San Francisco, USA and plays a key role in leading our research and development plans, and clinical trials for its CVac ovarian cancer therapy vaccine. Mr Lehman has a Master of Science from Columbia University in New York, and a Bachelor of Arts from the University of Louisville, Kentucky. He is also a member of the European Business Association and Association for Clinical Research Professionals.
Gregory Lizee, PhD
MD Anderson Cancer Center
Dr. Lizée was born and raised in Vancouver, Canada. He earned his Ph.D. at the University of British Columbia in 2000, working on characterizing the role of the MHC class I (MHC-I) cytoplasmic tail in murine antiviral immune responses. That work defined a highly conserved tyrosine-based endocytic motif as being essential for dendritic cell (DC) antigen-cross presentation and cytotoxic T lymphocyte (CTL) priming. Following his Ph.D. studies, Dr. Lizée relocated to Washington, D.C. for a postdoctoral fellowship at the Surgery Branch of the National Cancer Institute, led by Dr. Steven Rosenberg and best known for their pioneering immunotherapy trials for the treatment of human cancer. During his tenure there, Dr. Lizée defined mutated peptides derived from the melanoma-associated BRAF oncogene as constituting targets for CD4+ T-cell responses, and also helped to develop novel lentiviral vector-based systems for gene transfer into antigen-presenting cells. In 2005, Dr. Lizée established his own research effort at M.D. Anderson Cancer Center in Houston, Texas. There, he has continued his mechanistic studies on human DC antigen presentation, with the ultimate goal of improving DC-based vaccines for the treatment of cancer. In particular, his group has shown that there are two opposing motifs within the cytoplasmic domain of MHC-I molecules, and that alteration of these motifs can dramatically alter CTL priming efficiency by DCs, findings that could have important implications for human cancer vaccines. More recently, Dr. Lizée has been focused on identifying MHC-I ligands directly from the surface of patient tumor cell lines and biopsies using a combined approach encompassing MS-based proteomics, genomics, and bioinformatics. The ultimate goal of this project is to develop personalized cancer vaccines that are not restricted by tumor type, patient HLA haplotype, or limited by a lack of known target antigens.
Charles A. Nicolette, PhD
Argos Therapeutics Inc.
Charles Nicolette received his PhD in biochemistry and cellular and developmental biology from the State University of New York at Stony Brook, completing his doctoral dissertation and post-doctoral fellowship at Cold Spring Harbor Laboratory. Dr. Nicolette was the Director of Antigen Discovery for cancer vaccine development at Genzyme Corporation where he directed pre-clinical, translational and clinical stage programs for 6 years. He joined Argos Therapeutics in August 2003 and has overseen the development of all of Argos' pipeline products from preclinical through Phase 3 clinical development. He is the inventor on dozens of patent applications primarily relating to vaccine development for malignant and infectious diseases.
Sarah J. Schlesinger, MD
The Rockefeller University
Sarah J. Schlesinger is a research associate professor in the laboratory of cellular immunology and physiology at The Rockefeller University and a research scientist at The Aaron Diamond AIDS Research Center, a world-renowned biomedical research institute. Dr. Schlesinger has been actively engaged in HIV/AIDS and HIV vaccine research for 10 years, and has published over 50 papers on the subject. Dr. Schlesinger led the Dendritic Cell program at the Division of Retrovirology at the Walter Reed Army Institute of Research (1990-2002). She is now an active member of the research team at Aaron Diamond that is devoting considerable efforts to develop a vaccine to halt the spread of the AIDS epidemic.
Adam Steinman
Kris Thielemans, MD, PhD
Vrije Universiteit Brussel, Belgium
Kris Thielemans has been one of the first to use dendritic cells for the immunotherapy of cancer In Belgium. For more than 10 years, his lab has worked on and perfected several methods to modify dendritic cells to make them more immunogenic against tumor antigens. Those methods of modification include mRNA electroporation and lentiviral transduction. Both the transfection conditions of mouse and human dendritic cells and the stability of the mRNA for highest possible expression have been optimized. The lab has an EU-approved GMP facility in Brussels to produce clinical-grade mRNA for use either as an API or an IMP in various clinical trials (cancer and infectious diseases).
James B. Trager, PhD
Dendreon Corporation
James Trager is Vice President of Research at Dendreon Corporation. Dr. Trager earned his doctorate at the University of California at Berkeley, studying the role of auto-phosphorylation on signaling by the v-src tyrosine kinase in the lab of G. Steven Martin. At Geron, Dr. Trager was part of a team that cloned hTERT, the protein component of human telomerase; identified key interactions between the protein and RNA components of the enzyme; and brought the first telomerase inhibitor to the clinic. Dr. Trager has maintained his focus on novel cancer therapeutics at Dendreon, where has taken roles of increasing scope over the course of the last 9 years, with responsibilities for new product development, biomarker identification, and clinical immunology. He’s been instrumental in bringing Provenge, the first FDA approved cellular immunotherapy for cancer, through clinical study and to the market. Current challenges include in-depth characterization of Provenge mechanism of action, identification of biomarkers predictive of patient response to Provenge, and the application of lessons learned to development of next generation cellular immunotherapies for treatment of cancer. Dr. Trager is a graduate of St. John’s College in Santa Fe, New Mexico, and a former Peace Corps volunteer in the Central African Republic.
John S. Yu, MD
Cedars-Sinai Medical Center and Immunocellular Therapeutics, Ltd.
John S. Yu, MD is the Medical Director of the Brain Tumor Center at Cedars-Sinai Medical Center. He is Professor and Vice Chair in the Department of Neurosurgery.. He is also conducting extensive research in immune and stem cell therapy for brain tumors. Dr. Yu earned his bachelor's degree from Stanford University and spent a year at the Sorbonne in Paris studying French literature. He completed his fellowship in immunology at the Institut Pasteur in Paris. He earned his medical degree from Harvard Medical School and master's degree from the Harvard University Department of Genetics. He completed his neurosurgical residency at Massachusetts General Hospital in Boston. In addition, he was a neuroscience fellow at the National Institute of Mental Health in the Neuroimmunology Unit at Massachusetts General Hospital, and he was a Culpeper Scholar at the Molecular Neurogenetics Unit at that hospital.
In 2006, he founded Immunocellular Therapeutics, a clinical stage biotechnology public company focused on dendritic cell therapy for cancer.
Sponsors
Academy Friends
Promotional Partners
American Society of Hematology
The New York Academy of Medicine
Mission Partner support for the Frontiers of Science program provided by 
Abstracts
Cellular Immunotherapy for T Cell Lymphoma: Advantages of Maturationally Synchronized, Physiologically Induced DC
Richard L. Edelson, MD, Yale School of Medicine
Early Dendritic Cell Trials at Stanford: Setting the Stage for Sipuleucel-T
Edgar G. Engleman, MD, Stanford University School of Medicine
Danger Signals in Cancer Immune Therapy with Dendritic Cells: Experience and First Trends from a Randomized Clinical Trial
Thomas Felzmann, MD, Activartis Biotech GmbH, Austria
We demonstrated in pre-clinical experiments that limiting LPS/IFN-g-mediated DC maturation to 6 hours enables priming of T-cells in vitro or in vivo, which is superior compared to a maturation of 24-48 hours. This strategy was employed in a randomized clinical efficacy trial for the treatment of glioblastoma multiforme (GBM), the most frequent and most aggressive form of brain cancer, using an individualized DC cancer immune therapy (CIT) concept (GBM-Vax, EudraCT 2009-015979-27, NCT01213407). We randomized 78 patients aged 18-70 years. All patients received first line treatment for GBM, surgery, radiotherapy, and chemotherapy with Temozolomide; in the treatment group, AV0113 was applied as add-on. Primary and secondary objectives were PFS and OS. The study still collects follow up information. The current trend based on approximately two thirds of the patients suggests a strong reduction of deaths in the first year after diagnosis (AV0113 treatment: 18% deaths; control: 45% deaths).
Reactions at the AV0113 DC-CIT injection site were mild and included redness and swelling. Some patients developed fever above 40°C that could be therapeutically controlled. The majority of severe adverse events concerned the nervous system but was not different in the two groups (AV0113 treatment: 39%; control: 41%).
Randomization for the GBM-Vax study was completed in May 2013; hence we expect a complete data set for the one-year survival in May 2014. If the current trend is confirmed, we expect a first demonstration of efficacy for IL-12-secreting DCs in the treatment of brain cancer.
From Nobel Prize to Bedside: Innovative Approaches to Personalized Dendritic Cell Therapies
Charles A. Nicolette, PhD, Argos Therapeutics Inc.
Using the Tumor Immunopeptidome to Guide the Design of Personalized DC-based Cancer Vaccines
Gregory Lizee, PhD, MD Anderson Cancer Center
The specific objective of this research project is to develop an epitope identification method that can reliably and routinely identify MHC-I ligands presented by cancer cells from individual patients. Since recent evidence has shown that the most effective and immunogenic tumor antigens are those containing tumor-specific mutations, we have been specifically searching for those antigens using a combination of next generation sequencing, mass spectrometry-based proteomics, and HLA bioinformatics. We are focusing our initial efforts on melanomas and tumors from lung and colon cancer patients, which accumulate the highest number of somatic mutations. We have now already used our comprehensive antigen identification approach to successfully identify hundreds of non-mutated tumor-associated antigens from individual patient tumor cell lines and surgical biopsies; in addition, we have also identified a number of mutated epitopes, including some that elicit strong autologous T-cell reactivity. Having knowledge of the antigenic peptide targets on patient tumors will allow for the development of more specific and effective immunotherapies, and will greatly broaden the patient base that can be treated with such therapies: most importantly, it will enable us to leave behind the historical limitations such as only being able to target a few known, shared tumor antigens restricted to HLA-A*0201 positive patients. This personalized tumor antigen identification method holds the promise of increasing the efficacy of autologous DC-based cancer vaccines substantially by adding much-needed specificity to current immunotherapeutic approaches.
Dendritic Cell Based Immunotherapy of Melanoma: The Brussels' Experience
Kris Thielemans, Vrije Universiteit Brussel
At our institution, clinical trials in pretreated advanced melanoma patients are being performed. These patients are treated with TriMixDC-MEL, a mixture of TriMix-DC co-electroporated with mRNA encoding a fusion of DC.LAMP and 1 of 4 melanoma associated antigens (gp100, tyrosinase, MAGE-C2 or MAGE-A3).
Ina pilot clinical trial, 24.106 TriMixDC-MEL cells were administrated solely by the intradermal (ID) route. Subsequently, a phase IB was conducted to investigate the safety of administrating TriMixDC-MEL by the intravenous (IV) and ID-route. The ratio of ID/IV administered DC was: Cohort-1: 20.106/4.106 DC [2pts], -2: 12.106/12.106 DC[3pts], -3: 4.106/20.106 [6pts], and -4: 0/24.106 DC [4pts]; DC were administered 4x q2w, and a 5th administration on w16. Local skin reactions (gr1-2) were observed in all pts receiving DC ID, flu-like symptoms (< gr2) were observed in 12/21 pts treated ID and in 8/15 pts treated ID/IV. Post IV infusion chills (gr2) were observed in 3/15 pts. Inflammatory cytokine release was documented during these chills. ID administration of TriMixDC-MEL was found to be feasible, safe, effectively stimulating CD8+ T-cell responses, but did not result in objective tumor responses. In contrast, the combined ID/IV administration of TriMixDC-MEL resulted in 2 PR and 2 CR (by RECIST) out of 15 pts (BORR of 27%; ongoing after 24+, 28+, 33+, and 34+ mths). A confirmed stable disease was documented in four additional patients (for a disease control rate of 53%). From this study we concluded that ID/IV-administration of TriMixDC-MEL as a single-agent cellular immunotherapy is associated with distinct but manageable side-effects and has seemingly superior clinical activity as compared to DC administered solely ID in patients with pretreated advanced melanoma.
Ipilimumab (ipi), an anti-CTLA-4 mAb, enhances T-cell function and has established activity in advanced melanoma pts. We aimed to investigate the safety and activity of TriMixDC-MEL combined with ipi. TriMixDC-MEL was administered IV (20.106) and ID (4.106) 1h after ipi infusion (10 mg/kg), q3w for a total of 4 administrations. Maintenance therapy with ipi was allowed q12w for pts free from progression at week 24. The primary endpoint was disease control rate (by irRC). 37 pts initiated treatment. Local skin injection reactions (gr 1-2) were observed in all pts, flu-like symptoms (gr 1-2) in 20 (54%) pts, post-infusion chills (gr 1-2) in 15 (40%) pts. Immune-related adverse events were observed in 29 (78%) pts [11 (29%) pts had grade 3 or 4 AEs]. Most common were dermatitis (24 pts); hypophysitis/hypopituitarism (6 pts), diarrhea (6 pts), and hepatitis (5 pts). irAEs necessitated systemic corticosteroids in 17 (45%) pts. The best objective tumor response (35 evaluable pts): 5 CR, 5 PR, 9 SD and 16 PD (disease control rate: 54%). Objective responses are currently ongoing in 6/10 pts (11+ -22+ months). This phase II study of TriMixDC-MEL ID/IV in combination with ipi demonstrates anti-melanoma activity in over 50% of the patients with therapy resistant advanced melanoma. Further clinical development of TriMixDC-MEL in combination with immune checkpoint modulators is warranted.
Furthermore, TriMixDC-MEL is currently under evaluation in a randomized phase II trial in the adjuvant setting following resection of macrometastases.
Collaborative Clinical Studies on Immunotherapies to Treat Ovarian Cancer
Lana E. Kandalaft, PharmD, MTR, PhD
Eleven patients have completed vaccination and T cell transfer to date, while twenty-three additional patients completed vaccination only. Vaccination was well tolerated and elicited tumor-specific T cell responses against various ovarian tumor antigens. Preliminary results demonstrate that patients’ DCs loaded with HOCl-oxidized lysate elicited strong tumor-specific IFN-γ secretions and produced high levels of Th1-priming cytokines and chemokines, including IL-12.
Preliminary results indicate a clinical benefit rate 66% in the first study and an incremental clinical benefit increase of 40% in the vaccine only cohort, 60% in the vaccine and bevacizumab cohort and finally 80% cohort 2 and 80% in the vaccine, bevcacizumab and cyclophosphamide cohort at the end of study. Progression-free survival (PFS) comparing the three cohorts with a matched control group of patients who have undergone secondary debulking and have had cyclophosphamide and bevacizumab but no immunotherapy demonstrates at 500 days from enrollment, PFS of <10% in the control group, 45% in cohort 1, 50% in cohort 2, and 80% in cohort 3 (p=0.0334).
Following lymphodepletion, adoptive transfer of vaccine-primed T-cells was well tolerated and resulted in durable reduction of T-regulatory cells and restoration of vaccine-induced antitumor immunity in patients who experienced clinical benefit. One patient exhibited a complete response at end of study and stable disease was observed in 7 out of the 11 patients who completed vaccination and T cell transfer.
Our results suggest the use of combinatorial cellular immunotherapy comprising DC vaccination with whole tumor antigen and adoptive lymphocyte transfer using tumor antigen-specific T cells for the treatment of patients with recurrent ovarian cancer is promising yet warrants further investigation.
Biomarkers to Indicate Potentially Provenge-Responsive Prostate Cancer Patients
James B. Trager, PhD, Dendreon Corporation
Extensive immunological analyses have been performed in trials evaluating sipuleucel-T in men with mCRPC. APC activation, antigen-specific immune responses, and cytokine accumulation during product manufacture were studied. Consistent with an immunological prime-boost model, APC activation, along with antigen-specific IFN-γ ELISpot, proliferative and humoral responses, were greater at the second and third infusions than at the first. Antigen-specific cellular responses increased after sipuleucel-T treatment. Following culture with PA2024 we also observed increased CD4+ and CD8+ T cell activation. The observed T cell activation phenotypes were consistent with the cytokines (IL-2, IL-5, IL-13, and IL-10) produced during manufacture; these cytokines, associated with T cell activation, were elevated at the second and third product manufacture.
This talk will provide an overview of sipuleucel-T production, with a focus on product characterization and immune response. A variety of biomarkers, both baseline and pharmacodynamic, are correlative of clinical response to sipuleucel-T. We will discuss the biological interpretations of these markers and in particular their implications in understanding the mechanism of action sipuleucel-T.
Dendritc Cell Vaccines Targeting Cancer Stem Cells
John S. Yu, MD, Cedars-Sinai Medical Center and Immunocellular Therapeutics, Ltd.
CVac™ Phase 2 Clinical Study for Treatment of Ovarian Cancer Patients in First & Second Remission (CAN-003 protocol): Manufacturing & Clinical Outcomes
Matthew Lehman, Prima Biomed, Australia
Operationally, the trial demonstrated the ability to transfer the manufacturing technology, utilize multicenter autologous cell collections, and it established the comparability of product manufacturing between two sites.
Clinically, the CAN-003 trial indicated that CVac is well tolerated and non-toxic. Although there is significant biological variability, CVac induces a broad spectrum mucin 1-specific CD4+ and CD8+ T cell response in this patient population. There is no observed effect on median progression-free survival; however, differing trends were observed between the first and second remission patient groups. Overall survival data is considered too immature for analysis as of September 2013.
Travel & Lodging
Our Location
The New York Academy of Sciences
7 World Trade Center
250 Greenwich Street, 40th floor
New York, NY 10007-2157
212.298.8600
Hotels Near 7 World Trade Center
Recommended partner hotel
Club Quarters, World Trade Center
140 Washington Street
New York, NY 10006
Phone: 212.577.1133
The New York Academy of Sciences is a member of the Club Quarters network, which offers significant savings on hotel reservations to member organizations. Located opposite Memorial Plaza on the south side of the World Trade Center, Club Quarters, World Trade Center is just a short walk to the Academy.
Use Club Quarters Reservation Password NYAS to reserve your discounted accommodations online.
Other nearby hotels
212.945.0100 | |
212.693.2001 | |
212.385.4900 | |
212.269.6400 | |
212.742.0003 | |
212.232.7700 | |
212.747.1500 | |
212.344.0800 |