De-regulation of the C-MYC oncogene is among the most frequent molecular abnormalities in human cancer. c-Myc is a bHLH-ZIP transcription factor whose dimerization with another bHLH-ZIP protein, Max, is necessary for transformation and all other biological activities. That c-Myc is indispensable for cell proliferation, that most normal cells are non-cycling and thus non-c-Myc-expressing, and that transient inhibition of c-Myc may be sufficient to achieve permanent tumor regression, makes therapeutic targeting of the oncoprotein highly appealing. We have previously identified 7 low molecular weight compounds that abrogate/prevent bHLH-ZIP-mediated c-Myc-Max heterodimerization (""""""""Myc compounds""""""""). Non structure- guided (i.e. """"""""unbiased"""""""") modifications of one of these has resulted in the generation of """"""""2nd generation"""""""" analogs with up to 8-fold enhanced potencies, thus demonstrating that significant improvements in compound efficacy can be obtained even in the absence of a model of the compound in association with its target site. In order to identify even better analogs in a more rational and meaningful way, we have demonstrated that 3 of our original Myc compounds bind to distinct sites on the intrinsically disordered c- Myc bHLH-ZIP monomer, alter its secondary structure, and prevent its association with Max. The remaining 4 compounds are redundant and bind to one of these sites. Using NMR spectroscopy, we have generated structural models of the first three compounds in association with synthetic peptides comprising their cognate binding sites. Thus, in Specific Aim 1, we will utilize computational methods and high throughput in silico screens to identify new and more potent """"""""3rd generation"""""""" analogs whose structures are rationally predicated on those of the original 3 founding members.
In Specific Aim 2, we will compare the abilities of these new compounds to inhibit c-Myc-Max association and c-Myc-mediated neoplastic growth.
In Specific Aim 3, we will conduct in vivo pharmacologic testing of the most promising analogs prioritized by their """"""""ADMET"""""""" profiles. Finally, in Specific Aim 4, selected analogs identified in Specific Aims 2 and 3 will be chemically linked in order to generate """"""""4th generation"""""""" compounds capable of simultaneous binding to two distinct sites on the c-Myc bHLH-ZIP domain. This proposal is highly interdisciplinary and interactive in that it utilizes the expertise of a pediatric oncologist/molecular biologist (Prochownik), an organic chemist (Metallo), computational biologists (Behar and Mustata), and pharmacologists (Lazo, Eiseman and Egorin). Together, this diverse, complementary, and synergistic group of investigators, from two institutions, proposes studies that promise new insights regarding the rational design of Myc compounds and the mechanisms by which they disable this critical oncoprotein.

Public Health Relevance

This highly focused application is a multidisciplinary translational effort by seven investigators from two institutions to develop rationally designed small molecules that target the c-Myc oncoprotein. We have already determined the 3D NMR structures of three compounds that bind directly to different sites on the c-Myc monomer and inhibit its activity. Using these as a starting point, we propose to design and characterize a series of more potent analogs, some of which will be chemically linked to provide synergistic binding and biological activities.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA140624-02
Application #
7877789
Study Section
Drug Discovery and Molecular Pharmacology Study Section (DMP)
Program Officer
Lees, Robert G
Project Start
2009-07-01
Project End
2014-05-31
Budget Start
2010-07-23
Budget End
2011-05-31
Support Year
2
Fiscal Year
2010
Total Cost
$558,533
Indirect Cost
Name
University of Pittsburgh
Department
Pediatrics
Type
Schools of Medicine
DUNS #
004514360
City
Pittsburgh
State
PA
Country
United States
Zip Code
15213
Li, Youjun; Zhu, Yahui; Prochownik, Edward V (2016) MicroRNA-based screens for synthetic lethal interactions with c-Myc. RNA Dis 3:
Wang, Huabo; Sharma, Lokendra; Lu, Jie et al. (2015) Structurally diverse c-Myc inhibitors share a common mechanism of action involving ATP depletion. Oncotarget 6:15857-70
Jung, Kwan-Young; Wang, Huabo; Teriete, Peter et al. (2015) Perturbation of the c-Myc-Max protein-protein interaction via synthetic ?-helix mimetics. J Med Chem 58:3002-24
Fletcher, Steven; Prochownik, Edward V (2015) Small-molecule inhibitors of the Myc oncoprotein. Biochim Biophys Acta 1849:525-43
Wang, Huabo; Ramakrishnan, Anand; Fletcher, Steven et al. (2015) A quantitative, surface plasmon resonance-based approach to evaluating DNA binding by the c-Myc oncoprotein and its disruption by small molecule inhibitors. J Biol Methods 2:
Wang, Huabo; Teriete, Peter; Hu, Angela et al. (2015) Direct inhibition of c-Myc-Max heterodimers by celastrol and celastrol-inspired triterpenoids. Oncotarget 6:32380-95
Chauhan, Jay; Wang, Huabo; Yap, Jeremy L et al. (2014) Discovery of methyl 4'-methyl-5-(7-nitrobenzo[c][1,2,5]oxadiazol-4-yl)-[1,1'-biphenyl]-3-carboxylate, an improved small-molecule inhibitor of c-Myc-max dimerization. ChemMedChem 9:2274-2285
Edmunds, Lia R; Sharma, Lokendra; Kang, Audry et al. (2014) c-Myc programs fatty acid metabolism and dictates acetyl-CoA abundance and fate. J Biol Chem 289:25382-92
Wang, Huabo; Chauhan, Jay; Hu, Angela et al. (2013) Disruption of Myc-Max heterodimerization with improved cell-penetrating analogs of the small molecule 10074-G5. Oncotarget 4:936-47
Elster, Jennifer D; McGuire, Terence F; Lu, Jie et al. (2013) Rapid in vitro derivation of endothelium directly from human cancer cells. PLoS One 8:e77675

Showing the most recent 10 out of 20 publications