With a worldwide incidence of 1 in 3000, neurofibromatosis type 1 (NF1) is the most common inherited cancer predisposition syndrome. NF1 is caused by germ line mutations in the NF1 tumor suppressor gene (TSG), which encodes a GTPase activating protein (GAP) called neurofibromin that forms a molecular complex with activated Ras-GTP and negatively regulates Ras signaling by accelerating GTP hydrolysis. NF1, the most common rasopathy, has a propensity to develop neoplastic diseases that progress to aggressive cancers and frequently affect children, adolescents, and young adults. A common feature of NF1-associated neoplasms and malignant tumors is somatic loss of the normal NF1 allele. Importantly, limited epidemiologic data support the hypothesis that patients with NF1 who are cured of a primary cancer are at increased risk of developing treatment-induced secondary neoplasms (SNs). Together, the neoplastic diseases and aggressive malignancies that develop in NF1 are a substantial cause of morbidity and premature mortality. In addition to its role as an initiating mutation in NF1-associated cancers, recent genome-wide sequencing studies uncovered frequent somatic NF1 mutations in glioblastoma, acute myeloid leukemia, adenocarcinoma of the lung, and other sporadic cancers. Importantly, there are currently no mechanism-based therapies for cancers with mutations in genes encoding components of the Ras/GAP molecular switch (KRAS, NRAS, HRAS, and NF1). Thus, our focus will be developing effective higher-quality healthcare delivery options for children, adolescents and young adults with neurofibromatosis (NF) and provide insights that will benefit the entire Ras community. The overall goal of this Developmental and Hyperactive Ras Tumor (DHART) SPORE is to implement effective targeted molecular therapies for neoplasms and cancers by conducting integrated, mechanistically based translational research. The overarching objectives of this highly-qualified, collaborative group are : 1) to evaluate novel therapeutics in validated preclinical models and in the treatment of patients with NF1; 2) to identify risk factors of individuals with NF1 to acquire spontaneous and treatment-associated second malignancies; and 3) to decrease tumor associated morbidity and mortality of patients with NF1. This application draws on the expertise of an accomplished team of clinical investigators, physician/ scientists, and basic researchers with an extensive track record of productive collaborations. This program encompasses four highly integrated projects and three cores. The theme of translational therapeutics informs Projects 1 through 3, and the focus of project 4 exemplifies the role of cancer epidemiology and prevention. State-of-the-art core facilities will inform the patient-oriented cancer therapeutic and prevention aspects of this SPORE by elucidating mutations that contribute to of cancer pathogenesis and by defining biomarkers of drug response and resistance that will inform next generation treatments.

Public Health Relevance

? OVERALL This Developmental and Hyperactive Ras Tumor (DHART) SPORE application focuses on evaluating novel therapeutics in validated preclinical models and in the treatment of patients with neurofibromatosis type 1 and proposes to identify risk factors of individuals with NF1 to tumors and a strategy to decrease tumor associated morbidity and mortality.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Specialized Center--Cooperative Agreements (U54)
Project #
5U54CA196519-04
Application #
9543410
Study Section
Special Emphasis Panel (ZCA1)
Program Officer
Hubbard, Leah
Project Start
2015-09-01
Project End
2020-08-31
Budget Start
2018-09-01
Budget End
2019-08-31
Support Year
4
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Indiana University-Purdue University at Indianapolis
Department
Pediatrics
Type
Schools of Medicine
DUNS #
603007902
City
Indianapolis
State
IN
Country
United States
Zip Code
46202
Brosseau, Jean-Philippe; Liao, Chung-Ping; Wang, Yong et al. (2018) NF1 heterozygosity fosters de novo tumorigenesis but impairs malignant transformation. Nat Commun 9:5014
Liao, Chung-Ping; Booker, Reid C; Brosseau, Jean-Philippe et al. (2018) Contributions of inflammation and tumor microenvironment to neurofibroma tumorigenesis. J Clin Invest 128:2848-2861
Lu, Jia; Bera, Asim K; Gondi, Sudershan et al. (2018) KRAS Switch Mutants D33E and A59G Crystallize in the State 1 Conformation. Biochemistry 57:324-333
Dvorak, Christopher C; Satwani, Prakash; Stieglitz, Elliot et al. (2018) Disease burden and conditioning regimens in ASCT1221, a randomized phase II trial in children with juvenile myelomonocytic leukemia: A Children's Oncology Group study. Pediatr Blood Cancer 65:e27034
Yoshimi, Akihide; Balasis, Maria E; Vedder, Alexis et al. (2017) Robust patient-derived xenografts of MDS/MPN overlap syndromes capture the unique characteristics of CMML and JMML. Blood 130:397-407
Strowd 3rd, Roy E; Blakeley, Jaishri O (2017) Common Histologically Benign Tumors of the Brain. Continuum (Minneap Minn) 23:1680-1708
Maertens, Ophélia; McCurrach, Mila E; Braun, Benjamin S et al. (2017) A Collaborative Model for Accelerating the Discovery and Translation of Cancer Therapies. Cancer Res 77:5706-5711
Stieglitz, Elliot; Mazor, Tali; Olshen, Adam B et al. (2017) Genome-wide DNA methylation is predictive of outcome in juvenile myelomonocytic leukemia. Nat Commun 8:2127
Sherborne, Amy L; Lavergne, Vincent; Yu, Katharine et al. (2017) Somatic and GermlineTP53Alterations in Second Malignant Neoplasms from Pediatric Cancer Survivors. Clin Cancer Res 23:1852-1861
Davidson, Philip R; Sherborne, Amy L; Taylor, Barry et al. (2017) A pooled mutational analysis identifies ionizing radiation-associated mutational signatures conserved between mouse and human malignancies. Sci Rep 7:7645

Showing the most recent 10 out of 12 publications