mTOR Inhibitors Synergize on Regression, Reversal of Gene Expression, and Autophagy in Hepatocellular Carcinoma

Combination therapy causes gene reprogramming, autophagy, and tumor regression in a mouse model approximating human HCC. Bridging the Generation Gap Kids of every generation disdain their elders—who clearly don’t understand them and are stuck in the past. Newer is better, after all. But sometimes, a blend of the old and new may be exactly what’s needed to solve a particular problem. Thomas et al. set out to see whether the new—the phosphatidylinositol 3-kinase/mammalian target of rapamycin (mTOR) adenosine triphosphate–site competitive inhibitor BEZ235—was better than the old—the U.S. Food and Drug Administration–approved mTOR-allosteric inhibitor RAD001. What they instead found was that these two drugs worked together to treat hepatocellular carcinoma (HCC). mTOR signaling is up-regulated in about 50% of HCCs. When the authors tested two mTOR-targeting drugs, BEZ235 and RAD001, on cultured HCC cells, they unexpectedly found that the drugs acted synergistically. In a mouse model that mimics human HCC, the two drugs induced a marked regression in tumor burden through a mechanism that involved down-regulation of genes involved in autophagy—where the cell degrades its own components. In patients with HCC, dysregulation of autophagy genes correlated with poor prognosis. The authors are now taking this observation into clinical trials to determine whether it holds true in people. By working together, old and new mTOR inhibitors may provide a new therapeutic option for HCC. Hepatocellular carcinoma (HCC) affects more than half a million people worldwide and is the third most common cause of cancer deaths. Because mammalian target of rapamycin (mTOR) signaling is up-regulated in 50% of HCCs, we compared the effects of the U.S. Food and Drug Administration–approved mTOR-allosteric inhibitor, RAD001, with a new-generation phosphatidylinositol 3-kinase/mTOR adenosine triphosphate–site competitive inhibitor, BEZ235. Unexpectedly, the two drugs acted synergistically in inhibiting the proliferation of cultured HCC cells. The synergistic effect closely paralleled eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) dephosphorylation, which is implicated in the suppression of tumor cell proliferation. In a mouse model approximating human HCC, the drugs in combination, but not singly, induced a marked regression in tumor burden. However, in the tumor, BEZ235 alone was as effective as the combination in inhibiting 4E-BP1 phosphorylation, which suggests that additional target(s) may also be involved. Microarray analyses revealed a large number of genes that reverted to normal liver tissue expression in mice treated with both drugs, but not either drug alone. These analyses also revealed the down-regulation of autophagy genes in tumors compared to normal liver. Moreover, in HCC patients, altered expression of autophagy genes was associated with poor prognosis. Consistent with these findings, the drug combination had a profound effect on UNC51-like kinase 1 (ULK1) dephosphorylation and autophagy in culture, independent of 4E-BP1, and in parallel induced tumor mitophagy, a tumor suppressor process in liver. These observations have led to an investigator-initiated phase 1B-2 dose escalation trial with RAD001 combined with BEZ235 in patients with HCC and other advanced solid tumors.

[1]  M. Broggini,et al.  Combination of PI3K/mTOR inhibitors: antitumor activity and molecular correlates. , 2011, Cancer research.

[2]  F. Khuri,et al.  The Combination of RAD001 and NVP-BEZ235 Exerts Synergistic Anticancer Activity against Non-Small Cell Lung Cancer In Vitro and In Vivo , 2011, PloS one.

[3]  Daniel J. Klionsky,et al.  Relieving Autophagy and 4EBP1 from Rapamycin Resistance , 2011, Molecular and Cellular Biology.

[4]  J. Llovet,et al.  Targeted therapies for hepatocellular carcinoma. , 2011, Gastroenterology.

[5]  Keiji Tanaka,et al.  Persistent activation of Nrf2 through p62 in hepatocellular carcinoma cells , 2011, The Journal of cell biology.

[6]  Y. Eishi,et al.  Autophagy-deficient mice develop multiple liver tumors. , 2011, Genes & development.

[7]  B. Viollet,et al.  AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1 , 2011, Nature Cell Biology.

[8]  Insil Kim,et al.  Mitochondrial degradation by autophagy (mitophagy) in GFP-LC3 transgenic hepatocytes during nutrient deprivation. , 2011, American journal of physiology. Cell physiology.

[9]  D. Sabatini,et al.  mTOR: from growth signal integration to cancer, diabetes and ageing , 2010, Nature Reviews Molecular Cell Biology.

[10]  C. James,et al.  Akt and Autophagy Cooperate to Promote Survival of Drug-Resistant Glioma , 2010, Science Signaling.

[11]  S. Thorgeirsson,et al.  An Integrated Genomic and Epigenomic Approach Predicts Therapeutic Response to Zebularine in Human Liver Cancer , 2010, Science Translational Medicine.

[12]  L. Schwartz,et al.  Hepatocellular carcinoma: consensus recommendations of the National Cancer Institute Clinical Trials Planning Meeting. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[13]  W. Blonski,et al.  Non-viral causes of hepatocellular carcinoma. , 2010, World journal of gastroenterology.

[14]  N. Sonenberg,et al.  mTORC1-Mediated Cell Proliferation, But Not Cell Growth, Controlled by the 4E-BPs , 2010, Science.

[15]  N. Sonenberg,et al.  S6K1 plays a critical role in early adipocyte differentiation. , 2010, Developmental cell.

[16]  B. Viollet,et al.  Metformin, independent of AMPK, inhibits mTORC1 in a rag GTPase-dependent manner. , 2010, Cell metabolism.

[17]  Chang Hwa Jung,et al.  mTOR regulation of autophagy , 2010, FEBS letters.

[18]  M. Karin,et al.  Hepatocyte IKKbeta/NF-kappaB inhibits tumor promotion and progression by preventing oxidative stress-driven STAT3 activation. , 2010, Cancer cell.

[19]  Jun Hee Lee,et al.  Dietary and Genetic Obesity Promote Liver Inflammation and Tumorigenesis by Enhancing IL-6 and TNF Expression , 2010, Cell.

[20]  T. Chou Drug combination studies and their synergy quantification using the Chou-Talalay method. , 2010, Cancer research.

[21]  T. Noda,et al.  A subdomain of the endoplasmic reticulum forms a cradle for autophagosome formation , 2009, Nature Cell Biology.

[22]  William C Hahn,et al.  MYC activity mitigates response to rapamycin in prostate cancer through eukaryotic initiation factor 4E-binding protein 1-mediated inhibition of autophagy. , 2009, Cancer research.

[23]  P. Dennis,et al.  A novel, human Atg13 binding protein, Atg101, interacts with ULK1 and is essential for macroautophagy , 2009, Autophagy.

[24]  C. Chresta,et al.  Ku-0063794 is a specific inhibitor of the mammalian target of rapamycin (mTOR) , 2009, The Biochemical journal.

[25]  Svati H Shah,et al.  A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. , 2009, Cell metabolism.

[26]  D. Sabatini,et al.  An ATP-competitive Mammalian Target of Rapamycin Inhibitor Reveals Rapamycin-resistant Functions of mTORC1* , 2009, Journal of Biological Chemistry.

[27]  D. Guertin,et al.  mTOR complex 2 is required for the development of prostate cancer induced by Pten loss in mice. , 2009, Cancer cell.

[28]  Robbie Loewith,et al.  Active-Site Inhibitors of mTOR Target Rapamycin-Resistant Outputs of mTORC1 and mTORC2 , 2009, PLoS biology.

[29]  S. Thorgeirsson,et al.  E2F1 inhibits c-Myc-driven apoptosis via PIK3CA/Akt/mTOR and COX-2 in a mouse model of human liver cancer. , 2008, Gastroenterology.

[30]  Gareth Griffiths,et al.  Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum , 2008, The Journal of cell biology.

[31]  Dieter Häussinger,et al.  Sorafenib in advanced hepatocellular carcinoma. , 2008, The New England journal of medicine.

[32]  J. Zavadil,et al.  eIF4GI links nutrient sensing by mTOR to cell proliferation and inhibition of autophagy , 2008, The Journal of cell biology.

[33]  S. Horvath,et al.  Antitumor Activity of Rapamycin in a Phase I Trial for Patients with Recurrent PTEN-Deficient Glioblastoma , 2008, PLoS medicine.

[34]  Robin Mathew,et al.  Role of autophagy in cancer , 2007, Nature Reviews Cancer.

[35]  H. El‐Serag Epidemiology of hepatocellular carcinoma in USA , 2007, Hepatology research : the official journal of the Japan Society of Hepatology.

[36]  A. Hackshaw,et al.  A phase III randomised, double blind, placebo controlled trial of gemcitabine/carboplatin with or without thalidomide in advanced non-small cell lung cancer (NSCLC) , 2007 .

[37]  David M Sabatini,et al.  Defining the role of mTOR in cancer. , 2007, Cancer cell.

[38]  S. Carr,et al.  PRAS40 is an insulin-regulated inhibitor of the mTORC1 protein kinase. , 2007, Molecular cell.

[39]  Timothy J. Griffin,et al.  Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40 , 2007, Nature Cell Biology.

[40]  G. Thomas,et al.  Nutrient overload, insulin resistance, and ribosomal protein S6 kinase 1, S6K1. , 2006, Cell metabolism.

[41]  D. Sabatini,et al.  Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB. , 2006, Molecular cell.

[42]  F. Natt,et al.  Amino acids mediate mTOR/raptor signaling through activation of class 3 phosphatidylinositol 3OH-kinase. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[43]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[44]  H. Lane,et al.  The mTOR Inhibitor RAD001 Sensitizes Tumor Cells to DNA-Damaged Induced Apoptosis through Inhibition of p21 Translation , 2005, Cell.

[45]  H. Hung Treatment modalities for hepatocellular carcinoma. , 2005, Current cancer drug targets.

[46]  S. Thorgeirsson,et al.  Application of comparative functional genomics to identify best-fit mouse models to study human cancer , 2004, Nature Genetics.

[47]  S. Thorgeirsson,et al.  Classification and prediction of survival in hepatocellular carcinoma by gene expression profiling , 2004, Hepatology.

[48]  Terence P. Speed,et al.  A comparison of normalization methods for high density oligonucleotide array data based on variance and bias , 2003, Bioinform..

[49]  R. Abraham,et al.  The Rapamycin-Binding Domain Governs Substrate Selectivity by the Mammalian Target of Rapamycin , 2002, Molecular and Cellular Biology.

[50]  G. Thomas,et al.  Lethality of Drosophila lacking TSC tumor suppressor function rescued by reducing dS6K signaling. , 2002, Genes & development.

[51]  H. El‐Serag Epidemiology of hepatocellular carcinoma. , 2001, Clinics in liver disease.

[52]  A. Meijer,et al.  Phosphorylation of Ribosomal Protein S6 Is Inhibitory for Autophagy in Isolated Rat Hepatocytes (*) , 1995, The Journal of Biological Chemistry.

[53]  Peter Dalgaard,et al.  R Development Core Team (2010): R: A language and environment for statistical computing , 2010 .

[54]  Tara L. Kieffer,et al.  Hepatocellular Carcinoma: Epidemiology and Molecular Carcinogenesis , 2009 .

[55]  Yoon-Koo Kang,et al.  Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. , 2009, The Lancet. Oncology.

[56]  H. Lane,et al.  The mTOR Inhibitor RAD 001 Sensitizes Tumor Cells to DNA-Damaged Induced Apoptosis through Inhibition of p 21 Translation , 2005 .

[57]  K. Rao,et al.  Carcinogenicity of diethylnitrosamine in newborn, infant, and adult mice , 2004, Journal of Cancer Research and Clinical Oncology.

[58]  N. Fausto,et al.  Mouse Liver Tumorigenesis: Models, Mechanisms, and Relevance to Human Disease , 1999, Seminars in liver disease.

[59]  Y. Benjamini,et al.  Controlling the false discovery rate: a practical and powerful approach to multiple testing , 1995 .