Cancer is caused by the activation of oncogenes and the inactivation of tumor suppressor genes. The targeted inactivation and repair of these gene products may be a specific and effective therapy for cancer. Previously, we have shown that the inactivation of the MYC oncogene is sufficient to induce sustained regression of hematopoietic tumors [2]. Subsequently, we reported that even brief inactivation of MYC is sufficient to induce sustained tumor regression of osteosarcoma [3, 4]. Our results support the general hypothesis that tumors exhibit the phenomena of oncogene addiction [5, 6]. To explain our findings, we reasoned that MYC inactivation induces a permanent change in the ability of cells to induce a cancer-associated gene expressionprogram. Upon MYC inactivation, there are specific and sustained changes in gene expression [Wu et al, PLoS Genetics [1];and see Appendix, Shachaf et al, Cancer Research, 2008]. These changes in gene expression are frequently accompanied by permanent changes in the ability of MYC binding to promoter loci, as shown by ChIP. We performed a preliminary ChIP-on-chip analysis for MYC and interrogated changes in binding of other transcription factors in a genome-wide scale. Moreover, we also found that changes in gene expression are associated with specific alterations in chromatin modifications. Importantly, MYC appears to regulate gene expression not just through interactions with the canonical DNA binding sequence (E-Box), but additionally, its binding specificity may be regulated by DNA methylation. We provide new results that illustrate that the TGF-n signaling pathway may play an important role in the mechanism by which MYC inactivation induces changes in gene expression and cellular senescence. Moreover, we provide evidence that MYC regulates DNA methylation. Hence, we hypothesize that MYC inactivation restores auto-regulatory programs, including the induction of CDKIs, through effects on DNA methylation, resulting in the induction of a cellular senescence program. Hence, both cell extrinsic receptor based mechanisms, including TGF-U signaling, as well as cell intrinsic mechanisms, including regulation of DNA methylation, may be critical to oncogene addiction. We now propose experiments and request salary and grant support for a team of 1 principal investigator, 2 post-doctoral fellows and 1 graduate student and a research associates to delineate the role of TGF-13 signaling and DNA methylation on the mechanism of tumor regression upon MYC inactivation. Our results are consistent with the notion that MYC may directly regulate the global chromatin structure;and suggest the surprising idea that MYC-induced tumor cells remain unaware or """"""""amnesic"""""""" - as we have recently described [10] - of their cancerous state, yet remain poised to undergo senescence. The results of our proposed experiments will have important implications for the mechanisms by which the MYC oncogene maintains tumorigenesis and the development of new therapies for the treatment of cancer.

Public Health Relevance

Previously, we have shown that the inactivation of the MYC oncogene is sufficient to induce the sustained regression of cancer. We have just found that when MYC is inactivated in cancers this results in the induction of a program of terminal differentiation called cellular senescence. We have proposed studies to investigate how this switch is turned on, why it results in the regression of tumors and if this switch is also relevant to human cancers.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA105102-06
Application #
7895092
Study Section
Tumor Cell Biology Study Section (TCB)
Program Officer
Spalholz, Barbara A
Project Start
2004-02-01
Project End
2011-06-30
Budget Start
2010-07-01
Budget End
2011-06-30
Support Year
6
Fiscal Year
2010
Total Cost
$328,804
Indirect Cost
Name
Stanford University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
009214214
City
Stanford
State
CA
Country
United States
Zip Code
94305
Shroff, Emelyn H; Eberlin, Livia S; Dang, Vanessa M et al. (2015) MYC oncogene overexpression drives renal cell carcinoma in a mouse model through glutamine metabolism. Proc Natl Acad Sci U S A 112:6539-44
Casey, Stephanie C; Vaccari, Monica; Al-Mulla, Fahd et al. (2015) The effect of environmental chemicals on the tumor microenvironment. Carcinogenesis 36 Suppl 1:S160-83
Casey, Stephanie C; Amedei, Amedeo; Aquilano, Katia et al. (2015) Cancer prevention and therapy through the modulation of the tumor microenvironment. Semin Cancer Biol 35 Suppl:S199-S223
Gabay, Meital; Li, Yulin; Felsher, Dean W (2014) MYC activation is a hallmark of cancer initiation and maintenance. Cold Spring Harb Perspect Med 4:
Li, Y; Casey, S C; Felsher, D W (2014) Inactivation of MYC reverses tumorigenesis. J Intern Med 276:52-60
Ansari, Celina; Tikhomirov, Grigory A; Hong, Su Hyun et al. (2014) Development of novel tumor-targeted theranostic nanoparticles activated by membrane-type matrix metalloproteinases for combined cancer magnetic resonance imaging and therapy. Small 10:566-75, 417
Casey, Stephanie C; Li, Yulin; Felsher, Dean W (2014) An essential role for the immune system in the mechanism of tumor regression following targeted oncogene inactivation. Immunol Res 58:282-91
Das, Bikul; Kashino, Suely S; Pulu, Ista et al. (2013) CD271(+) bone marrow mesenchymal stem cells may provide a niche for dormant Mycobacterium tuberculosis. Sci Transl Med 5:170ra13
Casey, Stephanie C; Bellovin, David I; Felsher, Dean W (2013) Noncanonical roles of the immune system in eliciting oncogene addiction. Curr Opin Immunol 25:246-58
Bisikirska, B C; Adam, S J; Alvarez, M J et al. (2013) STK38 is a critical upstream regulator of MYC's oncogenic activity in human B-cell lymphoma. Oncogene 32:5283-91

Showing the most recent 10 out of 33 publications