Effective new cancer therapies have been developed based on agents with novel mechanisms of action, and those that target the pathophysiology of malignances. Clinically active in phase I studies in relapsed/refractory acute myeloid leukemia and myelodysplastic syndromes, Sapacitabine, the orally bioavailable form of 2'-C- cyano-2'-deoxy-1-2-D-arabino-pentofuranosylcytosine (CNDAC), acts by the unique mechanism of action of causing a single strand break in DNA. In the process, the incorporated analog is converted to a chain- terminating dideoxynucleotide, CNddC, a nucleotide that cannot be extended. Importantly, our initial results demonstrated that these nicks in DNA are relatively stable, and that they are converted to one-ended double- strand breaks when cells attempt to replicate their DNA across the nick. Further, our studies suggest that cells are mainly dependent upon homologous recombination (HR) to repair double-strand breaks that arise at collapsed replication forks. Thus, strategies that increase CNDAC incorporation into DNA, or approaches that diminish expression of HR pathway proteins would likely be synergistic with CNDAC. A corollary is that diseases with inherent deficiencies in homologous recombination repair would be selectively sensitive to the actions of CNDAC and its congeners. The overall goal of our proposal is to develop a thorough understanding of the mechanism by which CNDAC causes cell death, and to investigate strategies that will extend its toxicity to quiescent cells, overcome resistance due to highly expressed homologous recombination proteins, and provide rationale for the design combination strategies for translation to the design of clinical trials. We hypothesize that answers to questions posed in the following specific aims will clarify the mechanism of CNDAC action and indicate strategies to direct its application in the clinic. 1. What DNA damage repair mechanisms recognize and process CNDAC-induced DNA damage? By what mechanism is the CNddC lesion excised from DNA? Can mechanistic interactions be identified to provide a rationale for the design of combinations with CNDAC? By what mechanisms do histone deacetylase inhibitors (HDACi) sensitize cells to the action of CNDAC?

Public Health Relevance

This application proposes to develop a thorough understanding of the mechanism by which the small molecule CNDAC and related compounds cause cell death, and to investigate how tumors respond to avoid being killed. This information will be used to develop rationales for combinations with agents targeted at inhibiting such potential resistance mechanisms for translation to the design of clinical trials. Rationales for mechanism-based combinations with active anti-cancer drugs, and with the epigenetic regulatory pathways for DNA repair protein expression will be developed with a view to broadening the potential clinical application and impact of CNDAC (Sapacitabine).

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA028596-31
Application #
8657784
Study Section
Basic Mechanisms of Cancer Therapeutics Study Section (BMCT)
Program Officer
Arya, Suresh
Project Start
1980-08-01
Project End
2016-04-30
Budget Start
2014-05-01
Budget End
2015-04-30
Support Year
31
Fiscal Year
2014
Total Cost
Indirect Cost
Name
University of Texas MD Anderson Cancer Center
Department
Miscellaneous
Type
Hospitals
DUNS #
City
Houston
State
TX
Country
United States
Zip Code
77030
Liu, Xiaojun; Jiang, Yingjun; Nowak, Billie et al. (2018) Targeting BRCA1/2 deficient ovarian cancer with CNDAC-based drug combinations. Cancer Chemother Pharmacol 81:255-267
Al Abo, Muthana; Sasanuma, Hiroyuki; Liu, Xiaojun et al. (2017) TDP1 is Critical for the Repair of DNA Breaks Induced by Sapacitabine, a Nucleoside also Targeting ATM- and BRCA-Deficient Tumors. Mol Cancer Ther 16:2543-2551
Liu, Xiaojun; Jiang, Yingjun; Nowak, Billie et al. (2016) Mechanism-Based Drug Combinations with the DNA Strand-Breaking Nucleoside Analog CNDAC. Mol Cancer Ther 15:2302-2313
Lai, Tsung-Huei; Ewald, Brett; Zecevic, Alma et al. (2016) HDAC Inhibition Induces MicroRNA-182, which Targets RAD51 and Impairs HR Repair to Sensitize Cells to Sapacitabine in Acute Myelogenous Leukemia. Clin Cancer Res 22:3537-49
Liu, Xiao-Jun; Nowak, Billie; Wang, Ya-Qing et al. (2012) Sapacitabine, the prodrug of CNDAC, is a nucleoside analog with a unique action mechanism of inducing DNA strand breaks. Chin J Cancer 31:373-80
Liu, Xiaojun; Kantarjian, Hagop; Plunkett, William (2012) Sapacitabine for cancer. Expert Opin Investig Drugs 21:541-55
Guo, Lei; Liu, Xiaojun; Jiang, Yingjun et al. (2011) DNA-dependent protein kinase and ataxia telangiectasia mutated (ATM) promote cell survival in response to NK314, a topoisomerase II? inhibitor. Mol Pharmacol 80:321-7
Zecevic, Alma; Sampath, Deepa; Ewald, Brett et al. (2011) Killing of chronic lymphocytic leukemia by the combination of fludarabine and oxaliplatin is dependent on the activity of XPF endonuclease. Clin Cancer Res 17:4731-41
Liu, Xiaojun; Wang, Yaqing; Benaissa, Sherri et al. (2010) Homologous recombination as a resistance mechanism to replication-induced double-strand breaks caused by the antileukemia agent CNDAC. Blood 116:1737-46
Wang, Yaqing; Liu, Xiaojun; Matsuda, Akira et al. (2008) Repair of 2'-C-cyano-2'-deoxy-1-beta-D-arabino-pentofuranosylcytosine-induced DNA single-strand breaks by transcription-coupled nucleotide excision repair. Cancer Res 68:3881-9

Showing the most recent 10 out of 85 publications