?Project4 Melanomaisthedeadliestformofskincancer.Whilesignificantprogresshasbeenmadetreatingmelanoma, drug resistant represents one of the greatest challenges to achieve optimal responses and improve patient outcomes. Our long-term goal is to understand mechanisms underlying dysregulated signaling and drug resistance in melanoma to form the pre-clinical basis for improved treatment options. In this project, we are focusing on a clinical unmet need, the treatment of cutaneous melanomas that are wild-type (WT) for BRAF (both WT BRAF/WT NRAS and mutant NRAS). MEK-ERK1/2 signaling is activated in WT BRAF melanoma but the response to MEK inhibitors is poor. Furthermore, immune checkpoint agents elicit responses in only 30-40% of cases and patients who are non-responsive have no effective treatment options. The goals of this project are to identify drugtolerance mechanisms in subsetsof WTBRAF melanomas to provide the basis for new strategies to improve targeted inhibitor treatments and provide salvage options for melanomas that are non-responsive to immune checkpoint agents. Our preliminary data indicate that MEK inhibition triggers a receptor tyrosine kinase-mediated adaptive response in WT/WT melanoma. Based on these data, we hypothesize that the efficacy of MEK inhibitors will be improved with agents targeting either adaptive ErbB3 responses or epigenetic BET/BRD ?reader? proteins, as these epigenetic readers regulate multiple receptor kinasetyrosinekinaseandoncogenicpathwaysassociatedwithdrugresistance.
We aim toidentifyandtarget mechanisms underlying enhanced activation of the growth factor receptor, ErbB3, in MEK inhibitor-treated melanoma. Additionally, we will test the efficacy of BET inhibitor-based combinations to mitigate stress/therapytolerancemechanismsanddevelopoptimalcombinatorialapproachestooffsetdrugresistance. We will primarily focus on WT/WT melanomabut, where possible, extend our observation to mutant NRAS melanoma. To achieve these goals, we will leverage our unique genetically and clinically annotated models including in vitro 3D/T cell autologous organoids, immune checkpoint inhibitor-resistant patient-derived xenografts and syngeneic mouse models. Through synergistic interactions withother projects and cores in the P01, we will identify mechanisms underlying tumor cell intrinsic and stromal-mediated adaptive responses to targeted and immune therapy and inform future combinatorial targeted/epigenetic inhibitor strategies that can be translated into effective treatments. This project meets the NCI mission by conducting research into the treatmentofthedeadliestformofskincancer.

Public Health Relevance

?Project4 In Pennsylvania (the state served by our Cancer Centers), the 5 year incidence of melanoma is above the national average and is the fastest rising across cancer types. This proposal aims to address the issue of increased melanoma by providing the preclinical basis for targeted inhibitor and epigenetic inhibitor combinationswithinsubsetsofgenetically-definedwild-typeBRAFmelanomapatients.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
2P01CA114046-11A1
Application #
9791687
Study Section
Special Emphasis Panel (ZCA1)
Project Start
2008-04-01
Project End
2024-08-31
Budget Start
2019-09-01
Budget End
2020-08-31
Support Year
11
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Wistar Institute
Department
Type
DUNS #
075524595
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Liu, Shujing; Zhang, Gao; Guo, Jianping et al. (2018) Loss of Phd2 cooperates with BRAFV600E to drive melanomagenesis. Nat Commun 9:5426
Pathria, Gaurav; Scott, David A; Feng, Yongmei et al. (2018) Targeting the Warburg effect via LDHA inhibition engages ATF4 signaling for cancer cell survival. EMBO J 37:
Reyes-Uribe, Patricia; Adrianzen-Ruesta, Maria Paz; Deng, Zhong et al. (2018) Exploiting TERT dependency as a therapeutic strategy for NRAS-mutant melanoma. Oncogene 37:4058-4072
Rebecca, Vito W; Nicastri, Michael C; Fennelly, Colin et al. (2018) PPT1 promotes tumor growth and is the molecular target of chloroquine derivatives in cancer. Cancer Discov :
Kaur, Amanpreet; Ecker, Brett L; Douglass, Stephen M et al. (2018) Remodeling of the Collagen Matrix in Aging Skin Promotes Melanoma Metastasis and Affects Immune Cell Motility. Cancer Discov :
Chen, Gang; Huang, Alexander C; Zhang, Wei et al. (2018) Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 560:382-386
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 :

Showing the most recent 10 out of 144 publications