Resistance to apoptosis is one of the hallmarks of cancer. Cancer cells circumvent cell death through the mitochondrial apoptosis pathway to ensure tumor growth, maintenance and resistance to current treatments. The mitochondrial apoptosis pathway is governed by the expression levels and interactions of the BCL-2 family proteins, which comprise the pro-apoptotic effector proteins BAX and BAK, the anti-apoptotic proteins e.g. BCL- 2, BCL-XL, MCL-1 and the pro-apoptotic BH3-only proteins. Cancer cells most commonly block mitochondrial apoptosis by upregulating the anti-apoptotic BCL-2 proteins that neutralize BH3-only proteins and activated BAX and BAK. Therefore, efforts have focused on the development of selective inhibitors of anti-apoptotic BCL-2 proteins to re-activate apoptosis. Inhibitors such as the FDA-approved Venetoclax rely on the function and availability of BH3-only proteins to activate BAX and BAK. However, in many tumors, BH3-only proteins can be downregulated, suppressed or deleted, making these tumors insensitive to these inhibitors and limiting their broader clinical application. We hypothesized that small-molecule direct activation of pro-apoptotic BAX via the BAX trigger site is an alternative and possibly complementary pharmacological strategy to promote apoptosis in cancer cells. This approach can promote BAX activation independently of BH3-only proteins and therefore should have the potential to overcome apoptosis blockade in resistant tumors. Our laboratory recently used unique structural and molecular insights and medicinal chemistry to develop a potent and selective compound, termed BAX Trigger Site Activator 1(BTSA1) that promotes BAX activation and induces mitochondrial dysfunction and apoptosis. Using BTSA1, we provided proof-of-concept for direct BAX activation as a therapeutic target in Acute Myeloid Leukemia and demonstrated that direct BAX activation is well tolerated in vivo. Here, we hypothesized to generate BTSA1 analogues with improved potency, oral bioavailability and pharmacokinetics. Our goal is to evaluate their activity and mechanism of action in diverse cancer models as single agents or combination treatments and investigate mechanisms of sensitivity and resistance to BAX activation and apoptosis. Moreover, we aim to identify a clinical candidate BAX activator with favorable cellular, pharmacological and safety properties. Therefore, we propose the following specific aims: 1) characterize potency, selectivity, pro-apoptotic activity, in vitro ADME/Tox and pharmacokinetic properties of BTSA1 analogues, 2) determine cellular efficacy and mechanism of action of 2nd generation BTSAs, including BTSA1.2, alone and in combination treatments using various leukemia and solid tumor cells and investigate determinants of sensitivity and resistance, 3) determine safety and therapeutic potential of select BTSAs alone or in combination therapy and investigate novel biomarkers and mechanisms of apoptosis regulation. This proposal will advance an innovative therapeutic strategy and therapeutics and inform the most suitable context for targeting BAX activation in cancer.

Public Health Relevance

Evasion of apoptosis is a hallmark of cancer development, maintenance and chemoresistance. Pro-apoptotic BAX functions as gateway to apoptosis induction and our previous work has highlighted direct BAX activation as a promising therapeutic strategy. The goal of this proposal is to investigate small-molecule BAX activators in different type of tumors and investigate novel mechanisms and biomarkers that will facilitate the application of BAX activators to cancer patients.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
2R01CA178394-07A1
Application #
9974012
Study Section
Mechanisms of Cancer Therapeutics - 2 Study Section (MCT2)
Program Officer
Kondapaka, Sudhir B
Project Start
2014-07-01
Project End
2025-05-31
Budget Start
2020-06-01
Budget End
2021-05-31
Support Year
7
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Albert Einstein College of Medicine
Department
Type
DUNS #
081266487
City
Bronx
State
NY
Country
United States
Zip Code
10461
Rocha, Agostinho G; Franco, Antonietta; Krezel, Andrzej M et al. (2018) MFN2 agonists reverse mitochondrial defects in preclinical models of Charcot-Marie-Tooth disease type 2A. Science 360:336-341
Galluzzi, Lorenzo; Vitale, Ilio; Aaronson, Stuart A et al. (2018) Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ 25:486-541
Reyna, Denis E; Gavathiotis, Evripidis (2018) Pulling the BAX trigger for tumor cell death. Oncotarget 9:8204-8205
Garner, Thomas P; Lopez, Andrea; Reyna, Denis E et al. (2017) Progress in targeting the BCL-2 family of proteins. Curr Opin Chem Biol 39:133-142
Reyna, Denis E; Garner, Thomas P; Lopez, Andrea et al. (2017) Direct Activation of BAX by BTSA1 Overcomes Apoptosis Resistance in Acute Myeloid Leukemia. Cancer Cell 32:490-505.e10
Karoulia, Zoi; Gavathiotis, Evripidis; Poulikakos, Poulikos I (2017) New perspectives for targeting RAF kinase in human cancer. Nat Rev Cancer 17:676-691
Karoulia, Zoi; Wu, Yang; Ahmed, Tamer A et al. (2016) An Integrated Model of RAF Inhibitor Action Predicts Inhibitor Activity against Oncogenic BRAF Signaling. Cancer Cell 30:485-498
Klionsky, Daniel J (see original citation for additional authors) (2016) Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy 12:1-222
Cotto-Rios, Xiomaris M; Gavathiotis, Evripidis (2016) Chemical genetics: Unraveling cell death mysteries. Nat Chem Biol 12:470-1
Uchime, Onyinyechukwu; Dai, Zhou; Biris, Nikolaos et al. (2016) Synthetic Antibodies Inhibit Bcl-2-associated X Protein (BAX) through Blockade of the N-terminal Activation Site. J Biol Chem 291:89-102

Showing the most recent 10 out of 19 publications