International efforts to characterize cancer genomes now provide us with an initial view of the mutations and copy number alterations that occur in human cancers. These efforts have expanded our knowledge of known oncogenic pathways and have identified new classes of oncogenes and tumor suppressor genes. However, it is now also clear that most epithelial cancers harbor hundreds of genetic alterations as a consequence of genomic instability, which complicates efforts to identify mutations critical for tumor maintenance and drives tumor heterogeneity. Moreover, it remains unclear which of these alterations confers on the tumor cell the ability to evade the immune system. The emerging clinical success of checkpoint blockade is tempered by the observation that most patients do not respond to immunotherapy. New immunotherapy targets are needed to improve tumor responses and guide rational combination immunotherapy to overcome resistance. Identifying genes that are essential for tumor survival and immune evasion will accelerate the development of new molecularly targeted therapeutics. Over the past several years, we have developed and deployed high throughput genetic and bioinformatics approaches to identify and credential cancer targets. Specifically, we have performed genome scale loss of function and gain of function screens in large sets of human cancer cell lines and patient-derived models and have identified new oncogenes and synthetic lethal interactions. The scale of these experiments has allowed us to overcome the inherent heterogeneity of cancers and to classify cancer dependencies and their context, which is essential for the initiation of drug discovery efforts. In this application, we propose to use these studies as a foundation for a new Dana-Farber Cancer Institute CTD2 Center. This Center will focus on the identification of and credentialing cancer targets, developing the means to rationally define combination therapies, and the identification of genes that modulate the response to immunotherapeutics, all through the use of novel in vitro and in vivo HT genetic and bioinformatic approaches. We will continue to make the outputs of these studies readily available to the scientific community and to participate in CTD2 Network activities. We anticipate that this Center will provide the cancer research community with information that will facilitate the prioritization of targets based on both genomic and functional evidence, inform the most appropriate genetic context for downstream mechanistic studies and facilitate the translation of this information into therapeutics and diagnostics.

Public Health Relevance

The overarching goals of this Center are to identify and credential new cancer targets through the use of high throughput genetic technologies and to disseminate both the information and approaches to the cancer research community. The cancer and cancer immune response genes identified using these approaches represent prioritized candidates for investigator-initiated research programs and targets of particular promise for future therapeutic efforts. These studies will leverage prior investments in cancer genome characterization and provide outputs that will facilitate basic and translational studies and accelerate the development of new therapeutic approaches.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project--Cooperative Agreements (U01)
Project #
5U01CA176058-07
Application #
9745538
Study Section
Special Emphasis Panel (ZCA1)
Program Officer
Gerhard, Daniela
Project Start
2013-05-01
Project End
2022-07-31
Budget Start
2019-08-01
Budget End
2020-07-31
Support Year
7
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Dana-Farber Cancer Institute
Department
Type
DUNS #
076580745
City
Boston
State
MA
Country
United States
Zip Code
02215
Chen, Liying; Alexe, Gabriela; Dharia, Neekesh V et al. (2018) CRISPR-Cas9 screen reveals a MYCN-amplified neuroblastoma dependency on EZH2. J Clin Invest 128:446-462
Pan, Joshua; Meyers, Robin M; Michel, Brittany C et al. (2018) Interrogation of Mammalian Protein Complex Structure, Function, and Membership Using Genome-Scale Fitness Screens. Cell Syst 6:555-568.e7
Lo, Justin H; Hao, Liangliang; Muzumdar, Mandar D et al. (2018) iRGD-guided Tumor-penetrating Nanocomplexes for Therapeutic siRNA Delivery to Pancreatic Cancer. Mol Cancer Ther 17:2377-2388
Gannon, Hugh S; Zou, Tao; Kiessling, Michael K et al. (2018) Identification of ADAR1 adenosine deaminase dependency in a subset of cancer cells. Nat Commun 9:5450
Viswanathan, Srinivas R; Nogueira, Marina F; Buss, Colin G et al. (2018) Genome-scale analysis identifies paralog lethality as a vulnerability of chromosome 1p loss in cancer. Nat Genet 50:937-943
Li, Ji; Choi, Peter S; Chaffer, Christine L et al. (2018) An alternative splicing switch in FLNB promotes the mesenchymal cell state in human breast cancer. Elife 7:
Aguirre, Andrew J; Hahn, William C (2018) Synthetic Lethal Vulnerabilities in KRAS-Mutant Cancers. Cold Spring Harb Perspect Med 8:
Zhang, Xiaoyang; Choi, Peter S; Francis, Joshua M et al. (2018) Somatic Superenhancer Duplications and Hotspot Mutations Lead to Oncogenic Activation of the KLF5 Transcription Factor. Cancer Discov 8:108-125
Takeda, David Y; Spisák, Sándor; Seo, Ji-Heui et al. (2018) A Somatically Acquired Enhancer of the Androgen Receptor Is a Noncoding Driver in Advanced Prostate Cancer. Cell 174:422-432.e13
Giacomelli, Andrew O; Yang, Xiaoping; Lintner, Robert E et al. (2018) Mutational processes shape the landscape of TP53 mutations in human cancer. Nat Genet 50:1381-1387

Showing the most recent 10 out of 57 publications