Click For The Home Page Of The Neuro-Oncology Branch
Neuro-Oncology Branch
Brain X-ray
NOB - Dedicated To The Treatment Of Adult & Pediatric Brain Tumors
Contact Us National Cancer Institute National Institute of Neurological Disorders and Stroke
About The Clinic
About NOB
Laboratory Research
Clinical Trials
Patient Referrals
Career & Fellowship Opportunities
Education & Resources

Laboratory Research

Howard A. Fine, M.D., Lab and Branch Chief

The overall goal of the Neuro-Oncology Laboratory is to develop novel therapeutic strategies for the treatment of gliomas through an understanding, exploitation, and eventual clinical translation of the principles underlying the molecular and genetic pathogenesis of these tumors. Our approach is to leverage the unique resources of the intramural NIH program, including its tremendous scientific and clinical freedom to explore high risk yet high pay off projects, to build an NIH-wide pre-clinical and clinical brain tumor experimental therapeutics center. This Center works collaboratively and synergistically with both the NIH extramural community as well as with the private sector to ensure the most efficient and rapid development of novel approaches to the treatment of these devastating tumors. Below is a brief summary of several of our current translational initiatives.


  1. Glioma Molecular Diagnostic Initiative:
    Although it is well recognized that human gliomas are a heterogeneous group of tumors, there are to date no pathologic classification schemas that reproducibly allow the identification of biologically similar tumors or predict for tumor-specific therapies. The lack of such a classification schema significantly limits the ability of scientists to unravel the molecular pathogenesis of different glioma subtypes and precludes clinicians from offering therapeutic options that are specific for a patient's particular tumor. We have therefore initiated a large national effort in collaboration with the Cancer Genome Anatomy Project (CGAP) and NCI's Cancer Therapy Evaluation Program (CTEP) to develop a comprehensive and novel molecular classification schema for human gliomas based on gene expression profiles using cDNA microarray technology, comparative genomic hybridization (CGH), SNP analyses, and high throughput sequencing. Based on cDNA and SAGE libraries from more then 50 human gliomas, we have constructed a 48,000 element cDNA/oligonucleotide microarray chip enriched for genes that appear to be important in glioma biology. In conjunction with the NCI's CTEP-sponsored national Brain Tumor Consortia, we will receive hundreds of tumor specimens and correlative, prospective corollary clinical data. The data generated from this prospective study will be assimilated into the molecular/clinical database we are currently generating from the nearly 500 glioma specimens and historical clinical data we have received from our extramural collaborators. It is our expectation that within 5 years we will define a new molecular classification schema for human gliomas that has both prognostic and predictive therapeutic utility. Additionally, this project will generate a public international molecular and clinical database offering an unprecedented opportunity for gene discovery, elucidation of signal transduction pathways, and molecular target identification and validation that will be available to investigators through out the world. Finally, these data will be the first data to populate an exciting new initiative known as the Cancer Molecular Analysis Program (CMAP), a web-based bio-informatics platform that will allow researchers and clinicians the ability identify and evaluate molecular targets in cancer through integration of basic and clinical cancer research programs.

    Back to Top


  2. Bone Marrow-Derived Neural Stem Cells (NSC):
    NSC hold great promise for the treatment of a number of serious neurological illnesses including brain tumors. Nevertheless, the ability to obtain sufficient numbers of NSC from adult tissue represents a significant technical challenge. Although embryonic stem (ES) cells represent a potential source of cells with NSC-like properties, therapeutic use of ES cells are limited by questions regarding tumorigenic potential, immunological tolerance, and ethical concerns. We have now demonstrated that a subpopulation of adult human bone marrow-derived cells that can be propagated in large numbers in vitro, have morphology, gene expression profiles, and migratory properties similar to human fetal brain-derived NSC. These bone marrow-derived NSC (BMNSC) can be induced to differentiate into all of the primary lineages that make up the central nervous system including astrocytes, oligodendrocytes, and different neuronal subtypes. Additionally, our microarray gene expression studies have allowed us to identify a set of 73 genes that appear to play a pivotal role in the differentiation of both bone marrow and brain-derived NSC toward the differentiated neural lineages. Finally, we have demonstrated the ability of BMNSC to migrate both to sites of tumor cell infiltration within the brain and to sites of neural tissue damage. Our data demonstrate that through their migratory capabilities, these BMNSC transduced with therapeutic genes can mediate brain tissue repair and brain tumor destruction. We have a number of ongoing studies investigating issues related to the therapeutic use of these cells for anti-tumor purposes, for neural damage repair, and for better understanding the molecular biology and genetics of these cells. In particular, we are investigating how the set of genes we have identified as being pivotal in NSC differentiation are affected by specific mutations found within human gliomas. On the clinical side, we will soon be initiating the first clinical trial of these cells in which bone marrow will be harvested from patients will malignant brain tumors who are scheduled for surgery. BMNSC will be generated from their marrows, genetically marked and re-administered to the patient prior to surgery. Following surgery, the efficiency of migration and transgene expression of the genetically marked BMNSC will be evaluated by examining the resected tumor specimen. This study will hopefully establish the groundwork for a series of clinical studies designed to evaluate the ability of these cells to mediate brain tumor destruction and/or repair of damaged neural tissue.

    Back to Top


  3. Glioma-Selective Gene Therapy:
    Although therapeutic gene transfer offers a potentially promising new therapeutic strategy for the treatment of malignant gliomas, its clinical application has been limited by issues related to the non-tumor selectivity and distribution inefficiencies of current genetic delivery vector systems. We have addressed the problem of tumor selectivity in two different ways. First, we have developed a technology that allows us to exploit the observation that nearly 100% of malignant gliomas have deregulated retinoblastoma protein (rb) function secondary to genetic or epigenetic mutations in the P16/cyclinD/CDK4/RB pathway. Thus, a prediction, that until now, had not been demonstrated experimentally in vivo, is that E2F responsive promoters should be more active in tumor cells relative to normal cells due to an excess of 'free' E2F and loss of pRB/E2F repressor complexes. We have demonstrated that adenoviral and adenoviral associated viral (AAV)-based vectors, containing transgenes driven by the E2F-1 promoter, can mediate tumor selective gene expression in vivo, allowing for eradication of established gliomas with significantly less normal tissue toxicity than seen with standard viral vectors. In a second strategy to achieve glioma-selective vector transduction we, in collaboration with Robert Kotin of NHLBI, have recently demonstrated that a newly cloned and never before utilized subtype of AAV (AAV-5), allows for highly selective glioma cell transduction in vitro and in vivo secondary to the expression of the AAV-5 co-receptor (a specific 2,3-linked sialic acid glycopolysaccharide), on glioma cells but not only normal cells within the central nervous system. Finally, in collaboration with Ed Oldfield and the Surgical Neurology Branch of the NIH, we have addressed the problem of inefficient vector delivery by demonstrating that a new delivery technology developed here at the NIH, known as enhanced convection delivery, can distribute particles as large as adenoviral virions uniformly, at high concentrations, and safely throughout a region of the brain as large as the total cerebral hemisphere. All these studies have led us to the verge of initiating a series of clinical studies that will be designed to test the safety, vector distribution, glioma selective transgene expression, and eventual efficacy of E2F-responsive AAV-5 based vectors administered by direct intracerebral enhanced convection in patients with malignant gliomas.

    Back to Top


  4. Endothelial Progenitor Cells:
    We have identified a methodology for isolating and expanding in vitro a sub-population of human hematopoietic cells that are in fact endothelial progenitor cells (EPCs) or angioblasts. We have demonstrated that we can genetically engineer these cells ex-vivo to express marker genes or the thymidine kinase (TK) gene using retrovirus-mediated gene transfer. Genetically labeled EPCs were transplanted into wild type and sub-lethally irradiated mice and found to migrate and incorporate into the angiogenic vasculature of growing tumors while maintaining transgene expression. Ganciclovir (GCV) treatment resulted in tumor vascular collapse with resultant tumor necrosis in animals previously administered TK-expressing EPCs. These results demonstrate the feasibility of utilizing genetically modified EPCs as angiogenesis-selective gene-targeting vectors and demonstrate the potential of this approach to mediate non-toxic and systemic anti-tumor responses. Additionally, our EPC marker experiments have demonstrated that as much as 15-30% of tumor vasculature may in fact be bone marrow-derived. If validated, these experiments suggests a paradigm shift in that tumor-associated neovasculature may not be derived just through angiogenic mechanisms as previously believed, but also through the embryonic process of vasculogenesis. We are exploring a number of in vitro and in vivo experiments to better understand this process and to elucidate the cellular and molecular biology of the EPCs, including our recent demonstration that these cells appear to contribute to the repair of the endothelial component of the blood-brain barrier. Finally, we are preparing a pilot clinical study where EPCs will be harvested from patients, propagated and genetically marked ex vivo. These autologous cells will then be re-administered back into the systemic circulation from the donor patients with recurrent malignant gliomas, scheduled for repeat tumor resection. We will evaluate the resected tumor for incorporation of the genetically marked EPCs into tumor vascular and for evidence of transgene expression. This initial marker study will hopefully provide the biological basis that will pave the way for similar studies using EPCs carrying therapeutic genes.

    Back to Top


  5. Molecular Therapeutics:
    The Neuro-Oncology Laboratories are dedicated to the development of novel molecularly targeted anti-tumor agents. Toward this end there are currently two major programs:

    1. Selectively Targeted Ubiquination. We refined a technology that allows us to select a cellular protein (i.e. cell cycle regulator, oncogene, anti-apoptotic gene) and selectively target it for ubiquination and proteosome-mediated destruction. We have an active program that utilizes viral vectors to transduce a molecule that directs specific components of the cell cycle into proteosome degradation initiating rapid glioma cell apoptosis in vitro and tumor destruction in vivo without causing any toxicity to normal cells. We are currently working to bring this technology to the clinic while developing similar strategies, targeting other novel proteins, in the laboratory.

    2. Phase Display: We have an active program to identify novel molecular targets of 'signatures" on both glioma cell membranes as well as on tumor-associated endothelium utilizing random phage technology. We have already identified a number of exciting candidates targets and are currently confirming their tumor and/or tumor endothelial specificity. We will be linking these novel peptides to positron emitting nucleotides for PET scan imaging as a novel diagnostic modality and covalently binding these peptides to both cytotoxic agents and high energy radionucleotides as a tumor selective targeting technology.

    Back to Top


  6. Animal Brain Tumor Experimental Therapeutics and Diagnostic Core:
    One of the newest initiatives of the Neuro-Oncology Branch is an animal brain tumor experimental therapeutics and diagnostic core. The impetus for this initiative grows out of realization that most academic centers and pharmaceutical/biotechnology companies do not routinely evaluate new agents or diagnostic modalities in brain tumor cell lines, or in vivo models. The reasons for this are many, but include the technical difficulties in establishing reliable orthotopic models, and the relatively poor brain tumor models that were historically available. This has resulted in inadequate pre-clinical data for properly screening and selecting appropriate agents to bring to clinical trials for brain tumor patients, and for inadequate pharmacokinetic/toxicity data for designing the optimal initial trials. For example, as locally infiltrative, but non-metastasizing tumors, alternate methods of drug delivery may be preferable to standard intravenous administration, particular given issues related to the blood-brain barrier. One such delivery technology known as enhanced convection diffusion was developed here at the NIH and represents a way of homogeneously delivering an agent throughout the brain by direct infusion into the brain parenchyma. There are few if any pharmaceutical companies, however, that have gone to the effort to set up the methodologies that could test whether convection delivery is a better (more efficient, less toxic) method for delivering a novel anti-tumor agents to a brain tumor. Furthermore, the lack of thoughtful pre-clinical testing and modeling has made the development of surrogate markers of the biologic activity of any given drug virtual impossible; something particularly pertinent for the clinical development of new classes of drugs with cytostatic (rather then cytotoxic) properties (i.e. differentiating and anti-angiogenic agents).

    It is, therefore, our belief that this brain tumor therapeutics core will be invaluable for the purpose of designing more rationale methods of drug selection, for generating new clinically useful surrogate markers of drug activity, and for establishing better clinical study designs. This core is available for testing new therapeautic agents through collabarations with investigators both within the intramural and extramural NCI program, investigators at other academic medical institutions, and within the private sector through special arrangements.

    The experimental brain tumor therapeutics core consists of five major components:

    1. New Cytotoxic, Cytostatic, and Radiation Sensitizing Agent Evaluation: This consists of the testing of these new agents in vitro against a number of different adult and pediatric glioma, primitive neuro-ectodermal, neuronal tumor cell lines. The agents would be further testing in orthotopic brain tumor models using stereotactic technologies and evaluating a number of newer delivery technologies (besides standard oral or intravenous administration) including intracarotid administration, delivery with or without selective or gross blood brain barrier disruption, convection delivery, etc. The most promising of these agents will then have extensive pharmacology performed on them in animals pretreated with anti-epileptic agents know to induce the cytochrome P450 system in order to evaluate whether the brain tumor population is likely to experience altered pharmacokinetics of the agents compared to patients with systemic tumors.

    2. Central Nervous System Pharmacology (Blood-Brain Barrier): In conjunction with exploring alternate delivery modalities, the core has the capability of performing extensive pharmacology with an emphasis on assessing the ability of an agent to cross both an intact and a compromised blood-brain barrier. Along with cerebral spinal fluid pharmacology, the core has the ability to study brain/spinal cord drug penetration using techniques such as microdialysis and autoradiography. Finally, in collaboration with Frank Balis in the Pediatric Oncology Branch (NCI), the core has the capability of performing central nervous system pharmacology, including microdialysis, in non-human primates for agents that demand such an evaluation.

    3. Neurotoxicity: Evaluation of neurotoxicity of anticancer agents alone or in combination (with other agents and/or radiation) is rarely if ever undertaken as part of the pre-clinical development of any new agent. Neurotoxicity, particularly as it relates to the developing central nervous system (i.e. pediatric patients), however, is of vital importance for the ultimate clinical utility of any new anticancer agent. Thus, in conjunction with experts at NINDS and NIMH, a series of behavior, radiographic, and pathologic screens for therapy-induced neurotoxicity will be developed and used as part of a standard screen for agents that appear most promising for clinical development. Information gained through these screens will hopefully be presented to intramural and extramural investigators who might be interested in exploring the mechanism of injury in more depth. Having a routine animal screen in place, will be a great incentive for the future development of neuroprotective agents through both academia and the pharmaceutical industry.

    4. Evaluation of Novel Endpoints: As mentioned above, many of the new classes of anti-tumor therapeutics will have cytostatic rather then cytotoxic properties. Evaluating which of these agents will have biologic activity in humans in small, early clinical trials is a challenge since the standard "response" criteria are based on the determination of cytotoxic responses (i.e. the tumor shrinks by 50%). The only truly valid clinical parameter available for evaluating the activity of a truly cytostatic agent is patient survival or tumor progression-free survival. These, however, are not useful parameters for screening drug activity in small, early phase clinical trials. Thus, if surrogate markers of biologic activity could be identified, one could utilize these as early endpoints for screening out agents with little or no activity in vivo. Toward that end, the experimental brain tumor therapeutics core will actively develop surrogate markers of drug anti-tumor activity that can be utilized and validated in clinical trials. There are three major areas that well be explored:

      1. Imaging: In collaboration with investigators in NCI and NINDS, we will take advantage of the enormous small animal imaging facilities that are currently being constructed. Examples of the kind of endpoints we are interested in developing include techniques to quantitate microvasculature, vascular permeability, cerebral edema, and tumor metabolism. Currently we have the capabilities of doing high resolution MRI, MR spectroscopy, and PET scanning on small animals.

      2. Gene Expression Profiling: A major effort of the core is to generate gene expression profiles using microarray technology, from given glioma cell lines treated with a specific class of agents (i.e. farnesyl transferase inhibitors). If characteristic patterns could be identified that correspond with anti-tumor activity, then clinical trials can/will be devised to administer one of these agents to patients with brain tumors immediately prior to biopsy/surgery in order to attempt and identify a similar genetic profile clinically. This effort is complimented by the recently completed construction of the brain tumor microarray chip built in the laboratories of the Neuro-Oncology Branch on conjunction with the Cancer Genome Anatomy project (CGAP). Identification of characteristic expression profiles may also lead to gene discovery and the identification of new molecular targets for future therapeutic intervention.

      3. Proteinomics: Another major effort is to exploit the technology of cell-wide protein assessment through the developing initiatives within the Center for Cancer Research (CCR) at the NCI. In collaboration with investigators like Lance Liotta in the Pathology Branch of the NCI, we will begin to look at a wide array of proteins (i.e. abundance, post-translational modification) that are thought to be potentially relevant to the mechanisms of action of the drug being evaluated and/or proteins important to the biology of the tumor cell in an effort to identify potentially clinically useful surrogate markers of drug activity.

      4. Serum Markers: Serum markers of anti-tumor activity would be of enormous utility given their ready accessibility. We and others have already begun to evaluate the serum levels of angiogenic peptides such as VEGF and bFGF as potential markers of tumor "response", however, pre-clinical modeling is imperative as is the continued identification of additional markers for other classes of compounds.

    5. Brain Tumor Repository of Mouse Models and Cell Lines: Recently, a number of mouse brain tumor models (medulloblastoma, oligodendroglioma, NF/schwanoma) have been generated as have human brain tumor cell lines. These reagents have been generally constructed in individual laboratories, and there currently is no central place to acquire such reagents. Furthermore, these tumors and cell lines have not been well characterized. Thus, it is our intention to collect these reagents from our collaborators throughout the word and characterize these tumors and cell lines, morphologically, cytogenetically, immunohistochemically, and genetically through expression profiling. We will utilize the most appropriate of these models for our pre-clinical evaluations. The animal brain tumor therapeutic core offers an outstanding opportunity for the extramural NCI-sponsored brain tumor consortia, the intramural NIH-wide Brain Tumor Program, and the pharmaceutical industry to more thoroughly evaluate and model novel anti-tumor agents in a pre-clinical setting prior to moving into clinical trials. This should result in superior clinical trial design with greater certainty of obtaining the maximal amount of useful data from every treated patient. Furthermore, the core represents an outstanding resource to entice potential partnerships between NCI and pharmaceutical/biotechnology companies for the expressed purpose of using the core to develop new anti-tumor and/or neural protective agents.

    Back to Top


  7. Pediatric Laboratory Research:

    Childhood brain tumors are diverse in their biology, histology, and propensity for dissemination. A major research objective in the Neuro-Oncology Branch is the development of noninvasive methods of evaluating childhood brain tumors using new imaging methods. These include Proton Nuclear Magnetic Resonance Spectroscopic Imaging (1H-MRSI), 3-dimensional imaging, and newer MR sequences. 1H-MRSI is a noninvasive method of monitoring biochemical markers in vivo within normal brain tissue and tumor. The biochemical markers measured include N-acetyl aspartate (NAA), a normal neural marker, creatine (Cr), a marker of cell energy, choline (CHO), a constituent of the cell membrane and therefore an indicator of cell number and turnover, and lactate (LAC), a marker of anaerobic carbohydrate metabolism. Data from our pilot 1H-MRSI study indicate that the CHO:NAA ratio may be predictive of outcome in children with recurrent primary tumors. We are continuing these spectroscopy studies in an effort to define specific spectroscopic patterns for tumor growth, tumor edema, tumor necrosis and tumor response to chemotherapy in children with brain tumors. Comparative studies using PET (Positron Emission Tomography) scanning are planned. 1H-MRSI is also being used to study neurotoxic effects of cancer treatment in pediatric patients. Patients with or at risk for neurotoxicity are currently being evaluated by MRI, 1H-MRSI and neuropsychologic testing. Correlation of spectroscopic findings with the results of neuropsychological testing will then be performed to determine if neurotoxicity can be objectively defined.

    Another important research objective is the development of novel experimental therapeutic agents for children with CNS tumors. We are particularly interested and committed to development of innovative approaches, newer agents that can overcome drug resistance, and agents that can block or overcome the blood:brain barrier, thereby enhancing drug delivery to the tumor. We have a strong collaboration with the NCI Pediatric Oncology Branch and the Pharmacology and Experimental Therapeutics Section in development of new pharmacologic agents. Clinical pharmacokinetic and pharmacodynamic studies are performed in the majority of children enrolled on investigational studies.

    Back to Top


 

 
Home Page | For Patients | For Professionals | About NOB | Visitor Info | Disclaimer
 
National Cancer Institute National Institutes of Health Dept. Of Health And Human Services
National Cancer Institute National Institutes of Health Department of Health and Human Services