The overexpression of procaspase-3 (PC-3) in diverse tumor types presents an exploitable weakness in cancer and an opportunity for selective induction of apoptotic death in cancer cells. This proposal is a renewal of R01-CA120439, whose major goals included the investigation of the potential of the PC-3 activator PAC-1 in the treatment of glioblastoma (GBM) and metastatic osteosarcoma. These past four years have been very successful and as a result of these studies PAC-1 is now in a single-agent Phase 1 clinical trial (for late stage human cancer patients) and a combination clinical trial (for human glioblastoma patients), with other trials to being in 2018. Our work has shown that PAC-1 treatment, through activation of PC-3 to caspase-3, can induce cleavage of key caspase-3 substrates in cancer cells, leading to pronounced synergy with certain anticancer drugs. In the proposed work we will identify the most promising PAC-1/drug combinations for the treatment of Grade II/III meningioma, taking advantage of a key feature of PAC-1 (its blood-brain barrier penetrance) and utilizing our novel translational pathway (evaluation in pet dogs with meningioma). We have also found that PAC-1 markedly synergizes with drugs that hit targets in the MAPK pathway, due to the ability of PAC-1 to induce cleavage and inactivation of MEK1 and MEK2. This discovery will be explored through investigation of PAC-1 combinations in non-small cell lung cancer models, utilizing mouse models of metastases to the CNS. Finally, based on known caspase-3 substrates and preliminary data, we hypothesize that PAC-1 will have considerable mechanism-based synergy with immune checkpoint inhibitors through the facile cleavage and inactivation of the protein MLH1 by caspase- 3. Logical extension of this data would suggest that such mechanism-based synergy with PAC-1 could lead to immune checkpoint inhibitors being active in a much higher percentage of cancer patients, an exciting possibility. This is a hypothesis-driven proposal with high potential for fundamental and translational impact.

Public Health Relevance

There is a lack of effective targeted therapies for many deadly cancers, and even those that are available typically provide only a modest benefit due to inevitable resistance. We have identified the overexpression of procaspase-3 as an exploitable defect in cancer, and we have discovered a compound (called PAC-1) that activates procaspase-3 and selectively induces apoptosis in cancer cells. We now seek to use the synergistic activity of PAC-1 and our innovative translational path to identify outstanding PAC-1/drug combinations that can be advanced for the treatment of deadly malignancies.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA120439-12
Application #
9756307
Study Section
Drug Discovery and Molecular Pharmacology Study Section (DMP)
Program Officer
Kondapaka, Sudhir B
Project Start
2007-08-06
Project End
2023-07-31
Budget Start
2019-08-01
Budget End
2020-07-31
Support Year
12
Fiscal Year
2019
Total Cost
Indirect Cost
Name
University of Illinois Urbana-Champaign
Department
Chemistry
Type
Schools of Arts and Sciences
DUNS #
041544081
City
Champaign
State
IL
Country
United States
Zip Code
61820
Peh, Jessie; Boudreau, Matthew W; Smith, Hannah M et al. (2018) Overcoming Resistance to Targeted Anticancer Therapies through Small-Molecule-Mediated MEK Degradation. Cell Chem Biol 25:996-1005.e4
Joshi, Avadhut D; Botham, Rachel C; Schlein, Lisa J et al. (2017) Synergistic and targeted therapy with a procaspase-3 activator and temozolomide extends survival in glioma rodent models and is feasible for the treatment of canine malignant glioma patients. Oncotarget 8:80124-80138
Botham, Rachel C; Roth, Howard S; Book, Alison P et al. (2016) Small-Molecule Procaspase-3 Activation Sensitizes Cancer to Treatment with Diverse Chemotherapeutics. ACS Cent Sci 2:545-59
Peh, Jessie; Fan, Timothy M; Wycislo, Kathryn L et al. (2016) The Combination of Vemurafenib and Procaspase-3 Activation Is Synergistic in Mutant BRAF Melanomas. Mol Cancer Ther 15:1859-69
Roth, Howard S; Hergenrother, Paul J (2016) Derivatives of Procaspase-Activating Compound 1 (PAC-1) and their Anticancer Activities. Curr Med Chem 23:201-41
Palchaudhuri, Rahul; Lambrecht, Michael J; Botham, Rachel C et al. (2015) A Small Molecule that Induces Intrinsic Pathway Apoptosis with Unparalleled Speed. Cell Rep 13:2027-36
Roth, Howard S; Botham, Rachel C; Schmid, Steven C et al. (2015) Removal of Metabolic Liabilities Enables Development of Derivatives of Procaspase-Activating Compound 1 (PAC-1) with Improved Pharmacokinetics. J Med Chem 58:4046-65
Botham, Rachel C; Fan, Timothy M; Im, Isak et al. (2014) Dual small-molecule targeting of procaspase-3 dramatically enhances zymogen activation and anticancer activity. J Am Chem Soc 136:1312-9
Hsu, Danny C; Roth, Howard S; West, Diana C et al. (2012) Parallel synthesis and biological evaluation of 837 analogues of procaspase-activating compound 1 (PAC-1). ACS Comb Sci 14:44-50
West, Diana C; Qin, Yan; Peterson, Quinn P et al. (2012) Differential effects of procaspase-3 activating compounds in the induction of cancer cell death. Mol Pharm 9:1425-34

Showing the most recent 10 out of 16 publications