Hepatocellular carcinoma (HCC) accounts for nearly 29,000 deaths annually in the United States alone. However, the molecular mechanisms that drive HCC development remain elusive and current HCC therapies provide negligible clinical benefit. Factors that epigenetically silence an HCC tumor suppressor gene have the potential to promote tumorigenesis and thus may provide novel drug targets for HCC therapies. T o discover such factors, we performed an innovative genome-wide human RNA interference (RNAi) screen to identify factors that mediate epigenetic silencing of the HCC tumor suppressor gene Hedgehog-Interacting Protein (HHIP). HHIP is a negative regulator of Sonic hedgehog (SHH) signaling and loss of HHIP due to epigenetic silencing aberrantly activates SHH signaling, which has been proposed to promote tumor growth in multiple cancers including HCC. One of the factors identified in our screen is CDC-like kinase 1 (CLK1), a dual specificity protein kinase that phosphorylates serine/arginine-rich proteins involved in pre-mRNA splicing. We found that CLK1 transforms cultured immortalized hepatocytes and promotes HCC tumor growth in mouse subcutaneous xenografts, and these effects are dependent upon CLK1 protein kinase activity. Notably, epigenetic silencing of HHIP and CLK1 overexpression occur frequently in HCC patient samples, supporting the clinical relevance of our results. Based on these collective findings, we hypothesize that CLK1 is a driver of HCC and functions by epigenetically silencing the tumor suppressor HHIP. The results of the experiments proposed in this application will establish the role of CLK1 as a driver of HCC, determine the mechanism by which CLK1 promotes tumor growth, and evaluate CLK1 as a drug target for HCC therapy.
In Aim 1, we will establish the role of CLK1 in initiation and progression of hepatic tumorigenesis using a series of complementary mouse models that recapitulate characteristic features of HCC.
In Aim 2, we will test our hypothesis that CLK1 promotes hepatic tumor growth through epigenetic silencing of HHIP, resulting in aberrant activation of SHH signaling. We will also investigate other mechanisms by which CLK1 may promote tumor growth.
In Aim 3, we will evaluate CLK1 as a novel drug target for the development of HCC therapeutics. In preliminary experiments, we have found that inhibition of CLK1 enhances natural killer (NK) cell-mediated eradication of HCC cells. Therefore, we predict that reactivation of HHIP by pharmacological inhibition of CLK1 will: (1) directly inhibit tumor growth by blocking oncogenic SHH signaling, and (2) augment NK cell-mediated eradication of tumor cells. To test these predictions we will determine whether the highly specific CLK1 small molecule inhibitor KH-CB19 can effectively inhibit growth of hepatic tumors and enhance the ability of NK cells to eradicate tumors using a complementary series of immunocompromised and immunocompetent mouse models of HCC. Collectively, the results of the experiments proposed in this application will elucidate a novel druggable pathway that promotes HCC development and evaluate a new approach for treating HCC.

Public Health Relevance

Nearly 29,000 Americans die from hepatocellular (liver) carcinoma (HCC) every year and current treatments for HCC provide no long-term benefit, highlighting the urgent need for new and effective therapies. We have discovered that a protein called CLK1 is required for the growth of HCC tumors, and found that inhibiting CLK1 not only blocks HCC tumor growth but also enhances the ability of the immune system to destroy the tumor. The results of the experiments proposed in this application will establish CLK1 as a novel drug target for the development of new and effective HCC therapies.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
1R01CA218008-01A1
Application #
9524316
Study Section
Cancer Genetics Study Section (CG)
Program Officer
Johnson, Ronald L
Project Start
2018-04-01
Project End
2023-03-31
Budget Start
2018-04-01
Budget End
2019-03-31
Support Year
1
Fiscal Year
2018
Total Cost
Indirect Cost
Name
University of Massachusetts Medical School Worcester
Department
Anatomy/Cell Biology
Type
Schools of Medicine
DUNS #
603847393
City
Worcester
State
MA
Country
United States
Zip Code
Bugide, Suresh; Janostiak, Radoslav; Wajapeyee, Narendra (2018) Epigenetic Mechanisms Dictating Eradication of Cancer by Natural Killer Cells. Trends Cancer 4:553-566
Nagarajan, Arvindhan; Malvi, Parmanand; Wajapeyee, Narendra (2018) Heparan Sulfate and Heparan Sulfate Proteoglycans in Cancer Initiation and Progression. Front Endocrinol (Lausanne) 9:483