Low-grade astrocytomas (LGA) of childhood present a significant clinical challenge. While 5-year progression-free survival rate for chemotherapy plus radiotherapy is ~ 68%, significant morbidity is associated with the presence of residual tumor and the current therapy that includes neuroendocrine- cognitive deficits, visual deficits, vasculopathy and secondary tumors. Moreover, the metastatic potential and malignant transformation to a high-grade astrocytoma further contributes to the poor prognosis. Recent data show that BRAF is mutated in ~23% of LGA's, and 60% of xanthoastrocytomas. We have established two of the only childhood LGA models as direct patient- xenografts in mice. These tumors retain expression profiles and genomic alterations characteristic of the original patient's tumor, thus present unique models to develop alternative, less debilitating, curative therapy. BT-40 xenografts are heterozygous for mutated BRAF (V600E) whereas BT-35 xenografts have wild type BRAF. BT-40 tumors are exquisitely sensitive to the MEK inhibitor AZD6244, whereas BT-35 tumors are unresponsive, typical of other childhood tumor xenografts with wild type BRAF. Resistance to AZD6244 is characterized by an increase in the MEK-dependent gene signature, enhanced IL-6 transcription and secretion, and activation of STAT3 signaling. Resistance to AZD6244 is unstable, as tumors passaged in untreated mice revert to drug sensitivity, and correlates with decreased IL-6 and STAT3 activation. Further, two BRAF (V600E) mutant astrocytic cell lines intrinsically resistant to AZD6244 induce STAT3 activation as MEK is inhibited. The role of STAT3 activation in resistance will be studied in Aim 1. For BT-40 tumors sensitivity to AZD6244 is characterized by complete inhibition of TORC1 signaling, suggesting that mutant BRAF controls the PI3K/TORC1 signaling axis. The role of Akt and STAT3 signaling in MEK regulation of TORC1, and the simultaneous inhibition of MEK and STAT3 to prevent development of resistance, or reverse resistance to AZD6244, will be studied in Aim 2. We plan to exploit the control of mutant BRAF over TORC1 signaling therapeutically. In BT-40 xenografts, MEK inhibition leads to rapid and complete loss of the DNA repair protein FANCD2, downstream of TORC1, thus potentially selectively sensitizing tumor cells to cisplatin, etoposide and ionizing radiation. Further, we will test the concept that combination of cisplatin o etoposide with AZD6244 will prevent the emergence of AZD6244 resistant cells, as mutant BRAF is required to maintain Akt signaling associated with survival in cells exposed to cisplatin or etoposide. We will establish additional models of mutant BRAF LGA's, and determine whether results from BT-40 xenografts are generally applicable. These data will form the basis for rapid translation of novel non-genotoxic and also more conventional therapies that are both effective and less debilitating compared to current therapeutic approaches, for children with recurrent or surgically non-resectable LGA.

Public Health Relevance

Using unique models of childhood low-grade astrocytomas in mice, we have discovered that in BRAF-mutant tumors this oncogene has a gain of function that can be exploited to selectively kill astrocytoma cells. The studies proposed will identify how tumors become resistant to inhibitors of BRAF signaling, and exploit mutant BRAF to prevent development of resistance and synergize with drugs and ionizing radiation therapy. We anticipate that the studies proposed will lead to both increased cures for children with non-resectable astrocytomas, and reduce the toxicity of radiation and cytotoxic drug therapies, thus reducing the debilitating sequellae of current therapies.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA169368-04
Application #
9065511
Study Section
Drug Discovery and Molecular Pharmacology Study Section (DMP)
Program Officer
Kondapaka, Sudhir B
Project Start
2015-03-31
Project End
2018-04-30
Budget Start
2016-05-01
Budget End
2017-04-30
Support Year
4
Fiscal Year
2016
Total Cost
Indirect Cost
Name
University of Texas Health Science Center
Department
Other Basic Sciences
Type
Schools of Medicine
DUNS #
800772162
City
San Antonio
State
TX
Country
United States
Zip Code
78229
Bassett, Emily A; Palanichamy, Kamalakannan; Pearson, Mitchell et al. (2018) Calpastatin phosphorylation regulates radiation-induced calpain activity in glioblastoma. Oncotarget 9:14597-14607
Beyer, Sasha; Fleming, Jessica; Meng, Wei et al. (2017) The Role of miRNAs in Angiogenesis, Invasion and Metabolism and Their Therapeutic Implications in Gliomas. Cancers (Basel) 9:
Palanichamy, Kamalakannan; Kanji, Suman; Gordon, Nicolaus et al. (2017) NNMT Silencing Activates Tumor Suppressor PP2A, Inactivates Oncogenic STKs, and Inhibits Tumor Forming Ability. Clin Cancer Res 23:2325-2334
Zhou, Xinhui; Liu, Weijin; Hu, Xing et al. (2017) Regulation of CHK1 by mTOR contributes to the evasion of DNA damage barrier of cancer cells. Sci Rep 7:1535
Bell, Erica Hlavin; Pugh, Stephanie L; McElroy, Joseph P et al. (2017) Molecular-Based Recursive Partitioning Analysis Model for Glioblastoma in the Temozolomide Era: A Correlative Analysis Based on NRG Oncology RTOG 0525. JAMA Oncol 3:784-792
He, Zhengfu; Hu, Xing; Liu, Weijin et al. (2017) P53 suppresses ribonucleotide reductase via inhibiting mTORC1. Oncotarget 8:41422-41431
Palanichamy, Kamalakannan; Thirumoorthy, Krishnan; Kanji, Suman et al. (2016) Methionine and Kynurenine Activate Oncogenic Kinases in Glioblastoma, and Methionine Deprivation Compromises Proliferation. Clin Cancer Res 22:3513-23
Buckner, Jan C; Shaw, Edward G; Pugh, Stephanie L et al. (2016) Radiation plus Procarbazine, CCNU, and Vincristine in Low-Grade Glioma. N Engl J Med 374:1344-55
Phelps, Doris; Bondra, Kathryn; Seum, Star et al. (2015) Inhibition of MDM2 by RG7388 confers hypersensitivity to X-radiation in xenograft models of childhood sarcoma. Pediatr Blood Cancer 62:1345-52
Studebaker, Adam; Bondra, Kathryn; Seum, Star et al. (2015) Inhibition of MEK confers hypersensitivity to X-radiation in the context of BRAF mutation in a model of childhood astrocytoma. Pediatr Blood Cancer 62:1768-74

Showing the most recent 10 out of 12 publications