Malignant primary brain tumors represent the most frequent cause of cancer death in children and young adults and account for more deaths than cancer of the kidney or melanoma. Glioblastoma (GBM), the most common of these tumors, is uniformly lethal. Moreover, current therapy is non-specific and produces a median overall survival of only <15 months. In contrast, immunotherapy promises an exquisitely precise approach, and substantial evidence suggests that T cells can eradicate large, well-established tumors in mice and humans even when tumors reside within the brain. Notably, tumor immunotherapy has been recently validated in phase III clinical trials, leading to pivotal approvals by the FDA of at least two prototypic immune-based cancer treatments within the past two years. Bispecific T cell Engagers (BiTEs) represent an emerging class of bispecific antibody that has been shown to effectively redirect T cells against tumor cells. BiTEs promise to overcome many critical barriers that have traditionally limited translation of immunotherapy to the clinic. Separating this platform from other available immunotherapeutic approaches, our preliminary data support that BiTEs are (1) highly-specific molecules that greatly reduce the risk of toxicity, (2) have the ability to penetrate the blood-brain barrier (BBB) and accumulate in intracerebral tumors, and (3) may potentially overcome multiple mechanisms of immunosuppression present in patients with GBM. To date, BiTEs have not previously targeted tumor-specific antigens, and until our studies, have not yet been tested for their ability to mediate activity against tumors of the central nervous system (CNS). Here, we have designed a BiTE against the EGFRvIII tumor-specific antigen and perform preclinical tests to determine its efficacy against EGFRvIII- expressing GBM.
In Aim 1 of this proposal, we will evaluate the risk of toxicity and validate efficacy of the EGFRvIII-specific BiTE in a novel human CD3 transgenic mouse background. The unique model, along with a murine homologue of human EGFRvIII that we have created, will allow us to evaluate the actual construct we plan to take into clinic. Moreover, because the CD3 transgenic mice are heterozygotes and have a normal immune response, this model will allow us to assess the development of secondary autoimmunity and the potential for development of endogenous immune responses against tumor cells that do not express EGFRvIII.
In Aim 2, we will explore the possibility that, unlike other macromolecules, BiTEs have the unique ability to penetrate an intact BBB and determine whether this effect can be modulated to improve antitumor efficacy. Finally, in Aim 3, we will conduct early clinical studies to assess biodistribution, early toxicity, and potential efficacy of the EGFRvIII-specific BiTE in patients with EGFRvIII-expressing GBM. Overall, this work has the potential to improve the clinical management of patients with GBM by generating a novel therapeutic.

Public Health Relevance

Glioblastoma (GBM) is uniformly lethal;it is also the most common malignant primary brain tumor. Current therapy is incapacitating and limited by non-specific toxicity to systemic tissue or surrounding eloquent brain. We have developed a method to specifically target GBM using a tumor-specific, bispecific T cell engager that redirects patients'own T cells to recognize and destroy tumors, which has significant potential to improve public health and quality of life for patients affected by GBM and other cancers.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
5R01NS085412-02
Application #
8742019
Study Section
Clinical Neuroimmunology and Brain Tumors Study Section (CNBT)
Program Officer
Fountain, Jane W
Project Start
2013-09-30
Project End
2018-05-31
Budget Start
2014-06-01
Budget End
2015-05-31
Support Year
2
Fiscal Year
2014
Total Cost
$419,152
Indirect Cost
$152,176
Name
Duke University
Department
Surgery
Type
Schools of Medicine
DUNS #
044387793
City
Durham
State
NC
Country
United States
Zip Code
27705
Gedeon, Patrick C; Schaller, Teilo H; Chitneni, Satish K et al. (2018) A Rationally Designed Fully Human EGFRvIII:CD3-Targeted Bispecific Antibody Redirects Human T Cells to Treat Patient-derived Intracerebral Malignant Glioma. Clin Cancer Res 24:3611-3631
Swartz, Adam M; Reap, Elizabeth; Norberg, Pamela et al. (2018) A simple and enzyme-free method for processing infiltrating lymphocytes from small mouse tumors for ELISpot analysis. J Immunol Methods 459:90-93
Atik, Ahmet F; Suryadevara, Carter M; Schweller, Ryan M et al. (2018) Hyaluronic acid based low viscosity hydrogel as a novel carrier for Convection Enhanced Delivery of CAR T cells. J Clin Neurosci 56:163-168
Batich, Kristen A; Reap, Elizabeth A; Archer, Gary E et al. (2017) Long-term Survival in Glioblastoma with Cytomegalovirus pp65-Targeted Vaccination. Clin Cancer Res 23:1898-1909
Schaller, Teilo H; Sampson, John H (2017) Advances and challenges: dendritic cell vaccination strategies for glioblastoma. Expert Rev Vaccines 16:27-36
Suryadevara, Carter M; Riccione, Katherine A; Sampson, John H (2016) Immunotherapy Gone Viral: Bortezomib and oHSV Enhance Antitumor NK-Cell Activity. Clin Cancer Res 22:5164-5166
Suryadevara, Carter M; Gedeon, Patrick C; Sanchez-Perez, Luis et al. (2015) Are BiTEs the ""missing link"" in cancer therapy? Oncoimmunology 4:e1008339
Lin, Regina; Chen, Ling; Chen, Gang et al. (2014) Targeting miR-23a in CD8+ cytotoxic T lymphocytes prevents tumor-dependent immunosuppression. J Clin Invest 124:5352-67
Congdon, Kendra L; Gedeon, Patrick C; Suryadevara, Carter M et al. (2014) Epidermal growth factor receptor and variant III targeted immunotherapy. Neuro Oncol 16 Suppl 8:viii20-5
Gedeon, Patrick C; Riccione, Katherine A; Fecci, Peter E et al. (2014) Antibody-based immunotherapy for malignant glioma. Semin Oncol 41:496-510

Showing the most recent 10 out of 12 publications