Most human tumors are either inherently insensitive to chemotherapy or acquire resistance upon relapse. Apoptosis is a major cytotoxic mechanism of chemotherapy, and defective apoptosis regulation in cancer cells contributes to therapeutic resistance. Apoptosis-based anticancer agents, such as BH3 and SMAC mimetics, have shown promising results in recent preclinical studies. However, the mechanisms of chemotherapy- induced apoptosis remain poorly understood. The agents in current clinical development have weak efficacy and limited applications due to their target limitations. Our long-term objective is to understand how apoptosis is initiated in human cancer cells, and develop more effective anticancer agents accordingly in a rational way. The ongoing studies have elucidated an essential role of PUMA, a BH3-only proapoptotic Bcl-2 family member, in chemotherapy-induced and p53-dependent apoptosis. Induction of PUMA by anticancer agents is often defective in cancer cells due to p53 deficiencies. Deletion of PUMA led to chemoresistance, and elevated PUMA expression elicited profound apoptotic and antitumor effects. These activities of PUMA are dependent upon its unique BH3 domain, and ability to induce mitochondrial dysfunction through the mitochondrial apoptogenic protein SMAC. Furthermore, PUMA is also activated by targeted anticancer drugs through p53- independent mechanisms. These observations suggest that the potent anticancer activities of PUMA, mediated by a unique BH3 domain structure and SMAC-regulated mitochondrial dysfunction, are broadly involved in anticancer therapy and can be manipulated to develop novel anticancer agents. This competitive renewal seeks to: 1) delineate the key determinants of the potent proapoptotic and antitumor activities of PUMA;2) determine the roles of PUMA and SMAC in modulating the response to targeted therapy;and 3) identify novel small-molecule PUMA inducers to kill p53-deficient cancer cells. These studies will provide new insight into the anticancer effects of chemotherapeutic agents. In the long run, the results of these studies may lead to improved therapeutic strategies and more effective anticancer agents.

Public Health Relevance

Deregulation in apoptosis is a hallmark of cancer and renders cancer cells resistant to anticancer therapy. This project seeks to understand the anticancer mechanisms of PUMA, an essential mediator of therapeutic response, and to identify novel agents that activate PUMA to kill cancer cells. The results will be useful for developing improved therapeutic strategies and more effective anticancer therapy.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA106348-08
Application #
8257564
Study Section
Drug Discovery and Molecular Pharmacology Study Section (DMP)
Program Officer
Salnikow, Konstantin
Project Start
2004-04-01
Project End
2015-04-30
Budget Start
2012-05-01
Budget End
2013-04-30
Support Year
8
Fiscal Year
2012
Total Cost
$262,120
Indirect Cost
$87,339
Name
University of Pittsburgh
Department
Pharmacology
Type
Schools of Medicine
DUNS #
004514360
City
Pittsburgh
State
PA
Country
United States
Zip Code
15213
Knickelbein, Kyle; Tong, Jingshan; Chen, Dongshi et al. (2018) Restoring PUMA induction overcomes KRAS-mediated resistance to anti-EGFR antibodies in colorectal cancer. Oncogene 37:4599-4610
Tong, Jingshan; Wang, Peng; Tan, Shuai et al. (2017) Mcl-1 Degradation Is Required for Targeted Therapeutics to Eradicate Colon Cancer Cells. Cancer Res 77:2512-2521
Tong, J; Tan, S; Zou, F et al. (2017) FBW7 mutations mediate resistance of colorectal cancer to targeted therapies by blocking Mcl-1 degradation. Oncogene 36:787-796
He, Kan; Chen, Dongshi; Ruan, Hang et al. (2016) BRAFV600E-dependent Mcl-1 stabilization leads to everolimus resistance in colon cancer cells. Oncotarget 7:47699-47710
Wei, Liang; Leibowitz, Brian J; Wang, Xinwei et al. (2016) Inhibition of CDK4/6 protects against radiation-induced intestinal injury in mice. J Clin Invest 126:4076-4087
Chen, Dongshi; Yu, Jian; Zhang, Lin (2016) Necroptosis: an alternative cell death program defending against cancer. Biochim Biophys Acta 1865:228-36
He, K; Zheng, X; Li, M et al. (2016) mTOR inhibitors induce apoptosis in colon cancer cells via CHOP-dependent DR5 induction on 4E-BP1 dephosphorylation. Oncogene 35:148-57
Chen, Xiaojun; Song, Xiaomeng; Yue, Wen et al. (2015) Fibulin-5 inhibits Wnt/?-catenin signaling in lung cancer. Oncotarget 6:15022-34
Brown, M F; Leibowitz, B J; Chen, D et al. (2015) Loss of caspase-3 sensitizes colon cancer cells to genotoxic stress via RIP1-dependent necrosis. Cell Death Dis 6:e1729
Wang, Xinwei; Wei, Liang; Cramer, Julie M et al. (2015) Pharmacologically blocking p53-dependent apoptosis protects intestinal stem cells and mice from radiation. Sci Rep 5:8566

Showing the most recent 10 out of 60 publications