The long-term goal of this P01 is to promote research to better understand the biology of melanomas and to translate this research to better therapies with the ultimate goal of achieving cures. Our overarching hypothesis is that intrinsic resistance to signaling inhibitors can be overcome if we take into account the genetic signatures of melanoma cells and the biological properties that are driven by intrinsic and extrinsic signals. For this competitive renewal, we will shift our focus away from the traditional targeting of kinase active sites that control canonical growth factor signaling. To achieve our goals we have developed three overarching aims, which require close coordination of projects and cores:
In Aim 1 we will define cell fates for therapy of melanoma. While cell death due to apoptosis or necrosis is widely regarded to be the ultimate goal of any cancer therapy, in melanoma we now take into account two additional cell states that emerging data indicate are critical: autophagy (Projects 2 and 3) and senescence (Project 1). Our goal is to block cells from entering dormant survival states such as quiescence or pseudo-senescence and force them to die as a result of apoptosis or necrosis.
In Aim 2, we will investigate the mechanism of action of signaling inhibitors. Project 4 will generate a series of novel targeted inhibitors to prevent dimer formatio of BRAF or CRAF and investigate a novel STAT3 inhibitor that resulted from recent screens. Under this aim we will combine the expertise of chemists, structural biologists and cell biologists to develop a new generation of compounds that promise a major impact on melanoma therapy in the future.
In Aim 3, we will develop combination strategies for melanoma therapy. Since melanomas evolve through genetic alterations of multiple driver genes and are highly responsive to signals from the tumor microenvironment, the field has learned that single agents cannot cure by killing all malignant cells. In each project we will explore synergy among signaling inhibitor therapies using unique models for selection. The Cores for this P01 are essential for our progress in the coming funding cycle. To account for the increased needs for compound synthesis and modification, we have added a Medicinal Chemistry Core (D) in this renewal application.

Public Health Relevance

This P01 focuses on understanding the biology of melanoma and on developing new therapeutic strategies against this disease. Our goal is to eliminate all tumor cells and avoid the survival of any small subpopulation of resistant cells that could repopulate the patient and give rise to more aggressive and resistant tumors. This program consists of four projects and five cores. One of the four projects extends from our previous proposal but now additionally focuses on the design of novel inhibitors that target mutant and wild type BRAF, as well as the related CRAF protein which allows cells to escape BRAF signal inhibition. The other three projects move beyond RAF inhibition to target new processes and pathways based on our decades-long experience in signaling in melanoma. Autophagy (self-digestion due to stress), proper protein assembly (heat shock proteins), and senescence (aging and quiescence) are novel areas to focus on and target in melanoma.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
5P01CA114046-07
Application #
8741934
Study Section
Special Emphasis Panel (ZCA1-RPRB-2 (M1))
Program Officer
Arya, Suresh
Project Start
2005-04-01
Project End
2018-08-31
Budget Start
2014-09-01
Budget End
2015-08-31
Support Year
7
Fiscal Year
2014
Total Cost
$2,404,014
Indirect Cost
$688,336
Name
Wistar Institute
Department
Type
DUNS #
075524595
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Ojha, Rani; Leli, Nektaria M; Onorati, Angelique et al. (2018) ER translocation of the MAPK pathway drives therapy resistance in BRAF mutant melanoma. Cancer Discov :
Kugel 3rd, Curtis H; Douglass, Stephen M; Webster, Marie R et al. (2018) Age Correlates with Response to Anti-PD1, Reflecting Age-Related Differences in Intratumoral Effector and Regulatory T-Cell Populations. Clin Cancer Res 24:5347-5356
Nicastri, Michael C; Rebecca, Vito W; Amaravadi, Ravi K et al. (2018) Dimeric quinacrines as chemical tools to identify PPT1, a new regulator of autophagy in cancer cells. Mol Cell Oncol 5:e1395504
Nti, Akosua A; Serrano, Leona W; Sandhu, Harpal S et al. (2018) FREQUENT SUBCLINICAL MACULAR CHANGES IN COMBINED BRAF/MEK INHIBITION WITH HIGH-DOSE HYDROXYCHLOROQUINE AS TREATMENT FOR ADVANCED METASTATIC BRAF MUTANT MELANOMA: Preliminary Results From a Phase I/II Clinical Treatment Trial. Retina :
Perego, M; Maurer, M; Wang, J X et al. (2018) A slow-cycling subpopulation of melanoma cells with highly invasive properties. Oncogene 37:302-312
Echevarría-Vargas, Ileabett M; Reyes-Uribe, Patricia I; Guterres, Adam N et al. (2018) Co-targeting BET and MEK as salvage therapy for MAPK and checkpoint inhibitor-resistant melanoma. EMBO Mol Med 10:
Hammerlindl, Heinz; Ravindran Menon, Dinoop; Hammerlindl, Sabrina et al. (2018) Acetylsalicylic Acid Governs the Effect of Sorafenib in RAS-Mutant Cancers. Clin Cancer Res 24:1090-1102
Ecker, Brett L; Kaur, Amanpreet; Douglass, Stephen M et al. (2018) Age-Related Changes in HAPLN1 Increase Lymphatic Permeability and Affect Routes of Melanoma Metastasis. Cancer Discov :
Cañadas, Israel; Thummalapalli, Rohit; Kim, Jong Wook et al. (2018) Tumor innate immunity primed by specific interferon-stimulated endogenous retroviruses. Nat Med 24:1143-1150
Grasso, Michael; Estrada, Michelle A; Berrios, Kiara N et al. (2018) N-(7-Cyano-6-(4-fluoro-3-(2-(3-(trifluoromethyl)phenyl)acetamido)phenoxy)benzo[d]thiazol-2-yl)cyclopropanecarboxamide (TAK632) Promotes Inhibition of BRAF through the Induction of Inhibited Dimers. J Med Chem 61:5034-5046

Showing the most recent 10 out of 144 publications