National Cancer Institute U.S. National Institutes of Health | www.cancer.gov
SPORES Logo - Translational Research, Clinical, Pre-Clinical, Basic, Pre-Clinical SPOREs - Specialized Programs of Research Excellence
Home Organization of the OSB Organ-Specific SPORE Programs Information for the Public Patient Advocate Research Team Information for Applicants IntraSPORE Communications  
THE JOHNS HOPKINS UNIVERSITY

Overall Abstract

Principal Investigator(s): Richard F. Ambinder, M.D., Ph.D.

This application of the Specialized Program of Research Excellence (SPORE) in Lymphoma at the Johns Hopkins Medical Institutions supports a highly interactive, multidisciplinary, and inter-institutional program. Four research projects, four cores, a career development and developmental research program are proposed to further our translational research in Lymphoma. Research Project 1: EBV and Hodgkins Disease, Dr. Richard Ambinder (basic) and Dr. Cindy Schwartz (clinical); Project 2: Epidemiology of Lymphoma in HIV-infected Populations, Dr. Otoniel Martinez-Maza (basic) and Dr. Richard Ambinder (clinical); Project 3: Treatment of Lymphoma by Death Receptor-induced Apoptosis, Dr. Atul Bedi (basic) and Drs. Ian Flinn and Richard Wahl (clinical); Project 4: Immunotherapy for B Cell Lymphoma, Dr. Hyam Levitsky (basic) and Drs. Ian Flinn and Ivan Borrello (clinical). The Career Development Program (Dr. Diane Hayward and Dr. Georgia Vogelsang) aid the emergence of new investigators and the Research Developmental Program (Dr. Chi Dang) provides rapid funding of innovative directions.


Project 1
Epstein-Barr Virus and Hodgkin's Disease

Principal Investigator(s): Richard F. Ambinder, M.D., Ph.D.; Cindy Schwartz, M.D. Co-Investigator(s): Meghan Higman, M.D.; Yiping Yang, M.D.; Hyam Levitsky, M.D.; Peter Mauch, M.D.; David C. Fisher, M.D.

Our goal is to investigate whether in Hodgkin's lymphoma (HL) molecular markers can inform decisions regarding the duration or intensity of therapy and to determine whether vaccination and/or adoptive immunotherapy strategies may ultimately allow standard therapies to be shortened or de-escalated in some patients. HL is particularly well suited for these investigations in several regards. With present therapies some patients fail while others are cured yet continue to receive treatment for weeks or months afterwards. New prognostic markers and techniques to monitor tumor response may help identify those who are likely to be cured and should ultimately receive less therapy and those who are not yet cured and may need high dose or experimental therapies. Similarly, new therapeutic modalities to augment cytotoxic chemotherapy or radiation therapy are attractive insofar as they may result in cures in patients who are presently not cured or might partially substitute for these therapies in patients who are being cured and so allow a reduction in toxicity. Epstein-Barr virus (EBV) is present in the malignant cells of HL in many patients. Measurement of viral DNA may be useful in tumor monitoring. EBV DNA is detectable in plasma from patients with EBV-associated tumors including HL and has been reported to be useful in therapeutic monitoring of post-transplant lymphoma and nasopharyngeal carcinoma. In the first aim, we seek to characterize the origins of viral DNA detected in plasma from patients with HL, determine the association with the presence of viral nucleic acid and protein in the tumor, and assess the possible utility of viral DNA detection in therapeutic monitoring. These studies will involve prospective collections from the Children's Oncology Group (COG) and from patients treated at the Johns Hopkins Hospital, the Dana Farber Cancer Institute, and the Massachusetts General Hospital. The 2nd and 3rd aim involve immunotherapeutic strategies. The EBV proteins LMP2 and EBNA1 are both expressed in EBV(+) HL tumor cells and are established as targets for CD8(+) cytotoxic T cells and CD4(+) helper T cells, respectively. Both therapeutic approaches will be investigated in association with high dose therapy and autologous peripheral stem cell rescue insofar as this has emerged as the standard salvage therapy for HL patients and reduces tumor bulk and tumor-associated cytokines, chemokines and other molecules that may compromise the efficacy of the immune response. In the second aim, a therapeutic vaccine strategy in the treatment of EBV(+) HL will be investigated. A phase I/pilot trial in patients with EBV(+) HL will involve vaccination with an irradiated LMP2/EBNA1/GM-CSF expressing cell line. The goal of the trial will be to establish efficacy in achieving immunologic response endpoints and to assess safety. In the third aim, an adoptive cellular immunotherapy with autologous T cells specific for EBV proteins expressed in HL in the context of high dose therapy with peripheral stem cell transplantation. Aims 2 and 3 will be conducted at the Johns Hopkins Hospital, the Brigham and Women's Hospital and the Massachusetts General Hospital. These studies should illuminate the use of a new approaches to tumor monitoring and immunotherapy. Although focused on EBV(+) HL, the approaches to monitoring tumor-specific DNA in serum and the use of antigen-specific targeted immune interventions should be generalizable as specific molecular markers for Hodgkin's and other lymphomas are identified.


Project 2
Epidemiology of Lymphoma in HIV-infected Populations

Principal Investigator(s): Otoniel Martinez-Maza, Ph.D. (Basic); Richard F. Ambinder, M.D., Ph.D. (Clinical)
Co-Investigator(s): Lisa Jacobson, Ph.D.; Michael Dean, Ph.D.; Liz Breen, Ph.D.

Non-Hodgkin's B cell lymphoma (AIDS-NHL) is seen in greatly-elevated frequency in HIV-infected people. In this proposal, studies are presented to elucidate the molecular epidemiology of AIDS-NHL. In preliminary studies, elevated levels of several immune system molecules that are associated with B cell activation (IL6, IL10, sCD23, sCD27, sCD44, and IgE), were seen prior to the clinical detection of AIDS-NHL. Notably, there were clear differences in the patterns of expression of such B cell-stimulatory molecules seen in different subtypes of AIDS-NHL, suggesting that there are differences in the character of the immune dysfunction that precedes the development of different subsets of these cancers. In addition to this, genetic polymorphisms in the genes encoding IL10, SDF1, and CCR5 have been seen to be associated with the development of AIDS-NHL. These findings are of great significance, since few molecular risk factors have been identified for AIDS-NHL. The proposed studies will: 1) determine if enhanced B cell stimulation (elevated serum levels of B cell-stimulatory cytokines or of immune system molecules associated with B cell activation), and/or increased in vivo EBV levels, precede the development of AIDS-NHL, 2) determine if HAART affects the pre-lymphoma expression of B cell stimulatory molecules and/or in vivo EBV levels, 3) determine if polymorphisms in the genes encoding B cell-stimulatory cytokines or cytokine receptors are associated with an elevated risk for the development of AIDS-NHL, and 4) to define a pattern of molecular markers that identify those HIV+ people who are at increased risk for the development of AIDS-NHL, and then carry out a pilot clinical study to assess treatment of such high-risk individuals with prophylactic anti-lymphoma therapy (rituximab). Determination of characteristics preceding the development of AIDS-NHL will involve a case-control study, nested within two well-characterized cohort studies (the MACS and the ALIVE study), in which serial specimens have been collected and frozen. The same nested design will be utilized to examine the association between polymorphisms in genes encoding B cell-stimulatory cytokines and cytokine receptors and the development of AIDS-NHL, using specimens from the MACS and ALIVE study, as well as from the AIDS Malignancies Consortium (AMC). These studies will add valuable new information, which could form the foundation for future studies on the pathogenesis of AIDS-NHL, and is expected to lead to new risk-assessment schemes to allow earlier and more effective clinical intervention. The proposed studies will be carried out as a collaboration between workers at UCLA (Martínez-Maza and Breen), JHU (Jacobson and Ambinder), and NCI-Frederick (Dean).


Project 3B
Id Pulsed Dendritic Cell Vaccine

Principal Investigator(s): John Timmerman, M.D.; Sven de Vos, M.D./ Ph.D.

Human B cell lymphomas are highly susceptible to immunotherapeutic interventions, making them an attractive setting in which to develop and test new immune-based cancer treatments and determine their mechanisms of action. The tumor-specific immunoglobulin expressed by B cell lymphomas (idiotype, Id) can serve as a target for active immunotherapy. Traditional Id vaccines consist of Id protein chemically conjugated to the highly immunogenic carrier protein keyhole limpet hemocyanin (Id-KLH) and injected subcutaneously along with an immunologic adjuvant. Timmerman et al have recently demonstrated that immunization with Id-loaded dendritic cells (DCs) can induce durable tumor regressions in patients with lymphoma (see attached reprint). In contrast, such tumor regressions have been observed much less frequently using Id-KLH protein vaccines, so further studies of DC-based Id vaccines are warranted. We hypothesize that a "prime-and-boost" vaccination schedule in which immunization with mature DCs loaded with recombinant Id protein is followed by booster Id-KLH protein plus GM-CSF injections will lead to tumor regression responses in at least 30% of patients with measurable follicular lymphoma. This clinical intervention will provide a platform in which to study anti-tumor immune effector mechanisms in responder and non-responder subjects. We hypothesize that multiple immune effectors, including T cells and antibodies, can contribute to tumor regression in patients following this DC-based Id vaccine, and that particular patterns of effector responses will correlate with regression. Identifying and characterizing the immune effector mechanisms successfully recruited to mediate tumor regressions should lead to improved lymphoma immunotherapies. Our study includes the following aims:

Specific Aim 1: Determine the tumor regression rate following an Id-pulsed DC "prime" and Id-KLH protein "boost" vaccine in patients with measurable, relapsed follicular lymphoma.
Specific Aim 2: Determine if loading human DCs with tumor-antigen-antibody immune complexes increases their immunogenicity in vitro and in vivo.
Specific Aim 3: Characterize the humoral anti-Id immune responses elicited by Id/DC prime-boost vaccination. Serum anti-Id antibodies will be tested for their ability to bind selectively to tumor cells and mediate antibody-dependent cellular cytotoxicity, complement-mediated cytotoxicity, signal transduction, and apoptosis.
Specific Aim 4: Characterize the T-cell anti-tumor immune responses elicited by Id/DC prime-boost vaccination, including the process of "determinant spreading" to non-Id lymphoma antigens. We theorize that the dominant mechanism(s) of tumor regression will become apparent using our chosen set of immune effector analyses. Immune and clinical responses will be correlated with the gene expression profile of pre-treatment tumor cells.


Project 4
Immunotherapy for B Cell Lymphoma

Principal Investigator(s): Hyam Levitsky, M.D. (Basic); Ivan Borrello, M.D. (Co-Clinical); Ian Flinn, M.D., Ph.D. (Co-Clinical)

Both active and passive immunotherapies have shown increasing promise as treatments for human B cell lymphomas. A fundamental feature of the biology of these diseases that may influence the response to immunotherapy is the fact that lymphoma cells are the transformed counterpart of key components of the immune system itself. For example, as a tumor derived from antigen presenting cells (APCs), B cell lymphomas are well equipped to process antigen and engage in cognate interactions with lymphoma antigen-specific T cells. Consequently, therapeutic strategies seeking to manipulate anti-tumor immunity will effect both the normal host response to lymphoma antigens as well as the tumor directly. In this proposal, we seek to understand and exploit this aspect of lymphoma biology in our efforts to develop effective immunotherpies for B cell lymphoma. These projects will utilize well-characterized murine models for the purpose of comparing therapeutic efficacy of several vaccine and antibody based therapies, and identifying their underlying mechanism of action. Those strategies showing the greatest promise in preclinical models will be directly evaluated in early phase clinical trials. Specifically, we will:

  1. Seek to enhance the efficacy of GM-CSF tumor cell-based vaccination through the use of a universal GM-CSF producing bystander cell engineered to co-express tumor necrosis factor (TNF)/TNF receptor family members. Vaccination with irradiated autologous lymphoma cells mixed with such a bystander cell line will be compared to our current vaccine formulation (tumor + GM-CSF bystander alone). The influence of TNF/TNFr family member signaling on host APCs recruited to the vaccine site versus on the vaccinating lymphoma cells will be compared.
  2. Evaluate systemic treatment with monoclonal antibodies (mAb) targeting CD40, OX40, and CTLA-4 as single agents and in combination with tumor cell-based vaccines. The influence of each of these on the function of host immunity (e.g. APC activation, and enhanced T cell priming respectively) will be compard to direct effects on lymphoma cells in vivo.
  3. Explore antibody therapy and tumor vaccines for the treatment of lymphoma during immune reconstitution following autologous (or in the mouse, syngeneic), hematopoietic stem cell transplantation (HSCT). In addtion to these in vivo therapies, we will seek to augment the anti-lymphoma immune response through the ex vivo manipulation of autologous lymphocytes that accompany the graft during transplantation.

Project 5
Tumor Stem Cells in Hodgkin's Disease

Principal Investigator(s): William Matsui, M.D.; Richard Jones, M.D.

Classical Hodgkin lymphoma (HL) is characterized by pathognomonic Hodgkin-Reed Sternberg (H/RS) cells that contain rearranged and somatically hypermutated immunoglobulin gene sequences indicative of a germinal center B cell origin. However, these cells appear to be quiescent, suggesting that they may not represent the replicating component of the disease. This is reminiscent of our previous findings that neoplastic plasma cells in multiple myleoma (MM) are terminally differentiated and arise from the differentiation of a small population of myeloma stem cells that resemble memory B cells. Analysis of HL cell lines revealed that the majority of cells were CD30+CD15+ like typical H/RS cells, but small populations of CD30dimCD15dim cells that expressed B cell surface antigens were also found. Evaluation of clonogenic growth in vitro demonstrated that colony forming potential was restricted to CD30dimCD15dimCD22+ cells. Preliminary in vitro studies of clinical HL specimens also suggest that CD19+ B cells can form colonies consisting of H/RS cells. Therefore, HL may be similar to MM and myeloid leukemias in which small populations of clonogenic cancer stem cells are likely responsible for the initiation, maintenance and relapse of clinical disease. Furthermore, our previous studies demonstrated that the developmental signaling pathway Hedgehog (Hh) is aberrantly activated in MM and regulates stem cell self-renewal. Similarly, HL cell lines were found to express high levels of Hh pathway components. The study and charaterization of HL stem cells may lead to the development of novel therapies that inhibit their self-renewal and ability to produce new tumor cells resulting in long-term remissions. Accordingly, we propose to: 1. Study the biology of cancer stem cells in classical Hodgkin lymphoma; 2. Study mechanisms governing the self-renewal of Hodgkin lymphoma stem cells; and 3. Develop novel therapeutic strategies based on the biology of Hodgkin lymphoma stem cells.


Core 1
Administration and Communication Core

Principal Investigator(s): Richard F. Ambinder, M.D., Ph.D.

This core will be responsible for managing the SPORE and disseminating information within the SPORE and for external interactions. The Core is designed for low-cost, yet efficient administration and communication in order to focus funds on research activities. The administrative component of the Core follows an organizational diagram for management activities. Monitoring of research will occur via 1) Research Project Teams, 2) Committee of Research Project Principal Investigators, 3) Core Investigators Committee, 4) Developmental Projects Program, 5) Career Development Committee, and 6) the central SPORE Steering Committee. An institutional Advisory Board and External Advisory Board will provide yearly formal evaluations and reports to the Steering Committee.

The communication component of the Core is directed at intra-SPORE, inter-SPORE, and National Cancer Institute research activities. The Core provides travel funds for Johns Hopkins Lymphoma SPORE participation in the NCI SPORE annual meetings, to include the SPORE Director/PI, members of the Committee of Research Project Principal Investigators and other SPORE investigators. In addition, communication of validated results to the medical community in order to impact on the incidence and mortality of colorectal and pancreatic cancer will occur via publication of SPORE findings in medical journals, occasional inter-institutional think tanks and organized public conferences through the Office of Continuing Medical Education of the Johns Hopkins Medical Institutions, news releases to medical writers and press conferences for announcement of vital advances through the Office of Public Affairs of The Johns Hopkins Medical Institutions, and when appropriate, alerts coordinated through the NCI program staff.


Core 2
Human Specimen Resource and "Laboratory Core"

Principal Investigator(s): Michael Borowitz, M.D., Ph.D.; Hyam Levitsky, M.D.
Co-Investigator(s): Richard Schulick, M.D.; Janice Davis-Sproul, Ph.D; Edward Weir, M.D.; Risa Mann, M.D.

The Laboratory core is an interdepartmental collaborative effort designed to support basic and translational research on the etiology, early detection, prevention, and treatment of lymphoma. The Core provides a human specimen resource of neoplastic and normal tissue from patients with lymphoma with quality assurance measures, inventory and tracking, and correlative clinical data. This data will be stored in a secure relational database with web interface and both tape back -up of data and back-up server. In addition, the laboratory core will support the certification and preparation of clinical lots of genetically altered cell lines to be used as vaccines in lymphoma-related clinical trials. Storage of frozen specimens, laboratory processing, and data management will take place in the newly constructed Weinberg building of the Johns Hopkins cancer Center in immediate proximity to the operating rooms and surgical pathology laboratory in close collaboration with the breast and gastrointestinal SPOREs tissue banking efforts. The Core is directed by an experienced hematopathologist with prior tissue banking experience. The Co-PI is an experienced translational immunologist who will oversee production of the vaccines and immunologic assays performed on patient materials supported through this core.


Core 3
Clinical Core

Principal Investigator(s): Ian Flinn, M.D., Ph.D.
Co-Investigator(s): Richard Jones, M.D.

The Clinical Trials Core is a multidisciplinary effort designed to support the translational research of this SPORE. This resource will provide assistance in the design, conduct, and analysis of the clinical trials conducted within the spore. Specifically, assistance with translating laboratory findings into questions answerable within a clinical trial, clinical trial development, regulatory assistance with IND and IRB approvals, and clinical trial conduct will be provided. The core will provide research nursing, data management, adverse event reporting, and trial monitoring for the clinical trials. The core will assure the safety and confidentiality of the data within a pre-existing relational database with web interface. This core will support the laboratory core by managing the human investigation issues including obtaining informed consent from patients for the human tissue repository. The Core is directed by an experienced lymphoma clinical investigator with prior experience in database design. The Core Co-investigator is an experienced laboratory and clinical investigator experienced at moving laboratory findings into the clinic. Staff include Society of Clinical Research Associates certified data managers and experienced research nurses.


Core 4
Biostatistics Core

Principal Investigator(s): Steven Piantadosi, M.D., Ph.D.
Co-Investigator(s): Elizabeth Garrett, Ph.D.; Lisa Jacobson, Ph.D.

The Biostatistics Core of the proposed Johns Hopkins SPORE in lymphoma is designed to:

  1. Provide biostatistical consultation and support to all projects in the program, by assisting in the design, collection, visualization, analysis, quantitative modeling, interpretation and publication of the data arising in the course of program activities.
  2. Provide consultation for the design and analysis of clinical trials and other cohort studies to all the projects in the program by working with the Clinical Trials Core.
  3. Provide the infrastructural support, programming, and computer maintenance necessary to biostatistical activities within the program.

The Core will have an integral role in the scientific development, execution, and analysis of all projects in the SPORE, including the clinical trials. Core investigators have extensive and complementary experiences in quantitative methods for biomedical applications, including both clinical and basic science studies. They are committed to taking a direct interest in the substantive issues being investigated; to participating in regular project and program meetings, and to providing rigorous and innovative input on all quantitative matters arising in the projects. By contributing to multiple projects, they will also be in a position to promote interdisciplinary interactions among projects.


Developmental Research Program
Principal Investigator(s): Chi Dang, M.D., Ph.D.; Stephen Desiderio, M.D., Ph.D.

No abstract available.

The Developmental Research Program is a major focus for the SPORE because this mechanism provides a continuous flow of new ideas and projects to stimulate lymphoma research in the context of the SPORE. It provides an opportunity to encourage and facilitate the development of new research directions, methodologies, and collaborations. It also allows the SPORE broad access to investigators across The Johns Hopkins University and outside collaborators by providing support to pilot projects with the potential to develop into full-fledged translational projects. The scientific review process and programmatic review will assure that only the projects with both the highest scientific merit and translational potential worthy of future consideration as full projects within the SPORE are funded.


Career Development Program
Principal Investigator(s): S. Diane Hayward, Ph.D.; Georgia Vogelsang, M.D.

No abstract available.

The goal of this program is to facilitate the career development of investigators with a focus on translational research from the talent pool throughout the Johns Hopkins University. New investigators or established investigators in relevant areas who refocus their research on lymphoma prevention, diagnosis or treatment are encouraged to apply.


National Cancer Institute Department of Health and Human Services National Institutes of Health FirstGov.gov