Gliomas are genotypically heterogeneous, both in biomarker expression (or lack thereof) and in their ability to support replication of genetically modified herpes simplex virus (HSV) vectors. In addition to heterogeneity in Gene Expression Profiles (GEP), a subpopulation of glioma cells has been identified that has properties resembling those of neural stem cells and these cells are believed to be responsible for glioma recurrence and to exclusively maintain the neoplastic clone. This subpopulation of cells, referred to as Brain Tumor-lnitiating Cells or Glioma Progenitor Cells (GPC), contribute significantly to chemoresistance and radioresistance of malignant gliomas. These GPC may also display differential susceptibility to oncolysis by genetically engineered HSV alone or combined with radiation. Anti-tumor activity of genetically engineered HSV vectors is dependent on the ability of the host tumor cell to support HSV infection and replication. A clear understanding of the mechanisms of enhanced tumor killing mediated by both y{1}34.5-deleted HSV and receptor-targeted wild-type HSV vectors is critical to achieve improved clinical efficacy. We have already shown that IL-12 expression in a Ay[1}34.5 HSV background results in enhanced anti-tumor activity and this enhanced effect is immune-mediated. The question remains whether this benefit would also be seen with a targeted virus in which the virus'ability to overcome the PKR response remains intact or is restored. Project 3 has three fundamental aims.
Aim 1 will ascertain the relative oncolytic potential of each of the novel mutant HSV engineered by Projects 1 and 2 to use different entry mechanisms or to overcome innate host responses. Their antitumor activity on glioma cells, alone or combined with irradiation, will be compared with that of our current Ay{1}34.5 HSV vectors. These studies will (i) determine which modified viruses are best able to infect cells and overcome natural host defenses, and, (ii) lead to the engineering of viruses that combine these optimal features.
Aim 2 will focus on defining the comparative susceptibility of a subpopulation of glioma progenitor cells to oncolytic Ay{1}34.5 HSV alone and with irradiation. HSV specifically targeted to CD133 will be tested for efficacy on GPC while retaining safety for normal neuroglial cells. GEP of GPC and non-GPC tumor cells will be compared to reveal potential causes for resistance to HSV.
Aim 3 will complete preclinical testing of genetically engineered HSV that incorporate all genetic modifications.

Public Health Relevance

Malignant gliomas are radiation- and chemotherapy-resistant brain tumors that are universally fatal. We seek to optimize application of oncolytic mutant Herpes Simplex Viruses to treat malignant gliomas safely and effectively. These studies will establish the most effective HSV-based strategy to kill both glioma tumor cells comprising the bulk of the tumor and malignant glioma progenitor cells that give rise to glioma tumor cells. Strategies for improved virus oncolytic activity that utilize native and unique entry molecules on tumor cells and transfer of genetic information are expected to amplify the oncolytic effect safely.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
5P01CA071933-14
Application #
8378337
Study Section
Special Emphasis Panel (ZCA1-GRB-S)
Project Start
Project End
Budget Start
2012-07-01
Budget End
2013-06-30
Support Year
14
Fiscal Year
2012
Total Cost
$170,931
Indirect Cost
$37,954
Name
University of Alabama Birmingham
Department
Type
DUNS #
063690705
City
Birmingham
State
AL
Country
United States
Zip Code
35294
Friedman, Gregory K; Bernstock, Joshua D; Chen, Dongquan et al. (2018) Enhanced Sensitivity of Patient-Derived Pediatric High-Grade Brain Tumor Xenografts to Oncolytic HSV-1 Virotherapy Correlates with Nectin-1 Expression. Sci Rep 8:13930
Waters, Alicia M; Johnston, James M; Reddy, Alyssa T et al. (2017) Rationale and Design of a Phase 1 Clinical Trial to Evaluate HSV G207 Alone or with a Single Radiation Dose in Children with Progressive or Recurrent Malignant Supratentorial Brain Tumors. Hum Gene Ther Clin Dev 28:7-16
Ring, Eric K; Markert, James M; Gillespie, G Yancey et al. (2017) Checkpoint Proteins in Pediatric Brain and Extracranial Solid Tumors: Opportunities for Immunotherapy. Clin Cancer Res 23:342-350
Foreman, Paul M; Friedman, Gregory K; Cassady, Kevin A et al. (2017) Oncolytic Virotherapy for the Treatment of Malignant Glioma. Neurotherapeutics 14:333-344
Ring, Eric K; Li, Rong; Moore, Blake P et al. (2017) Newly Characterized Murine Undifferentiated Sarcoma Models Sensitive to Virotherapy with Oncolytic HSV-1 M002. Mol Ther Oncolytics 7:27-36
Patel, Daxa M; Foreman, Paul M; Nabors, L Burt et al. (2016) Design of a Phase I Clinical Trial to Evaluate M032, a Genetically Engineered HSV-1 Expressing IL-12, in Patients with Recurrent/Progressive Glioblastoma Multiforme, Anaplastic Astrocytoma, or Gliosarcoma. Hum Gene Ther Clin Dev 27:69-78
Friedman, Gregory K; Moore, Blake P; Nan, Li et al. (2016) Pediatric medulloblastoma xenografts including molecular subgroup 3 and CD133+ and CD15+ cells are sensitive to killing by oncolytic herpes simplex viruses. Neuro Oncol 18:227-35
McFarland, Braden C; Marks, Margaret P; Rowse, Amber L et al. (2016) Loss of SOCS3 in myeloid cells prolongs survival in a syngeneic model of glioma. Oncotarget 7:20621-35
Jackson, Joshua D; Markert, James M; Li, Li et al. (2016) STAT1 and NF-?B Inhibitors Diminish Basal Interferon-Stimulated Gene Expression and Improve the Productive Infection of Oncolytic HSV in MPNST Cells. Mol Cancer Res 14:482-92
Friedman, Gregory K; Beierle, Elizabeth A; Gillespie, George Yancey et al. (2015) Pediatric cancer gone viral. Part II: potential clinical application of oncolytic herpes simplex virus-1 in children. Mol Ther Oncolytics 2:

Showing the most recent 10 out of 172 publications