Off-Target Effects of c-MET Inhibitors on Thyroid Cancer Cells

Aberrantly activated c-MET signaling occurs in several cancers, promoting the development of c-MET inhibitors. In this study, we found that eight of eight thyroid cancer cell lines (including six anaplastic thyroid cell lines) have prominent expression of c-MET protein. Fifty percent of the thyroid cancer cell lines (four of eight) were growth inhibited by two small molecule c-MET inhibitors (tivantinib and crizotinib) associated with apoptosis and G2–M cell-cycle arrest. However, crizotinib did not inhibit 50% proliferation of thyroid cancer cells (SW1736 and TL3) at a concentration at which the drug completely inhibited ligand-stimulated c-MET phosphorylation. However, tivantinib was less potent than crizotinib at inhibiting c-MET phosphorylation, but was more potent than crizotinib at decreasing cell growth. Suppressing c-MET protein expression and phosphorylation using siRNA targeting c-MET did not induce cell-cycle arrest and apoptosis. Taken together, tivantinib and crizotinib have off-target(s) activity, contributing to their antitumor activity. In vivo study showed that crizotinib markedly inhibited the growth of thyroid cancer cells (SW1736) in immunodeficient mice. In summary, c-MET inhibitors (tivantinib and crizotinib) suppress the growth of aggressive thyroid cancer cells, and this potential therapeutic benefit results from their non–MET-targeting effects. Mol Cancer Ther; 13(1); 134–43. ©2013 AACR.

[1]  Ryohei Katayama,et al.  Cytotoxic activity of tivantinib (ARQ 197) is not due solely to c-MET inhibition. , 2013, Cancer research.

[2]  A. Bardelli,et al.  Tivantinib (ARQ197) Displays Cytotoxic Activity That Is Independent of Its Ability to Bind MET , 2013, Clinical Cancer Research.

[3]  W. Birchmeier,et al.  Targeting MET in cancer: rationale and progress , 2012, Nature Reviews Cancer.

[4]  K. Al-Kuraya,et al.  c-Met Inhibitor Synergizes with Tumor Necrosis Factor-Related Apoptosis-Induced Ligand to Induce Papillary Thyroid Carcinoma Cell Death , 2012, Molecular medicine.

[5]  N. Senzer,et al.  A Phase I Dose-Escalation Study of Tivantinib (ARQ 197) in Adult Patients with Metastatic Solid Tumors , 2011, Clinical Cancer Research.

[6]  D. Collins,et al.  Phase I trial of a selective c-MET inhibitor ARQ 197 incorporating proof of mechanism pharmacodynamic studies. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[7]  L. Trusolino,et al.  MET signalling: principles and functions in development, organ regeneration and cancer , 2010, Nature Reviews Molecular Cell Biology.

[8]  J. Copland,et al.  Anaplastic thyroid carcinoma: pathogenesis and emerging therapies. , 2010, Clinical oncology (Royal College of Radiologists (Great Britain)).

[9]  S. Jeay,et al.  ARQ 197, a Novel and Selective Inhibitor of the Human c-Met Receptor Tyrosine Kinase with Antitumor Activity , 2010, Molecular Cancer Therapeutics.

[10]  D. Bottaro,et al.  Targeting the HGF/Met signalling pathway in cancer. , 2010, European journal of cancer.

[11]  M. Ladanyi,et al.  Mutational profile of advanced primary and metastatic radioactive iodine-refractory thyroid cancers reveals distinct pathogenetic roles for BRAF, PIK3CA, and AKT1. , 2009, Cancer research.

[12]  G. V. Vande Woude,et al.  Novel Therapeutic Inhibitors of the c-Met Signaling Pathway in Cancer , 2009, Clinical Cancer Research.

[13]  Christopher Korch,et al.  Deoxyribonucleic acid profiling analysis of 40 human thyroid cancer cell lines reveals cross-contamination resulting in cell line redundancy and misidentification. , 2008, The Journal of clinical endocrinology and metabolism.

[14]  Gary L Clayman,et al.  Small molecule c‐MET inhibitor PHA665752: Effect on cell growth and motility in papillary thyroid carcinoma , 2008, Head & neck.

[15]  Shinji Yamazaki,et al.  An orally available small-molecule inhibitor of c-Met, PF-2341066, exhibits cytoreductive antitumor efficacy through antiproliferative and antiangiogenic mechanisms. , 2007, Cancer research.

[16]  Mieke Schutte,et al.  Phosphatidylinositol-3-OH Kinase or RAS Pathway Mutations in Human Breast Cancer Cell Lines , 2007, Molecular Cancer Research.

[17]  G. Braunstein,et al.  Antitumor Activity of Suberoylanilide Hydroxamic Acid against Thyroid Cancer Cell Lines In vitro and In vivo , 2006, Clinical Cancer Research.

[18]  A. Costantino,et al.  Activation of the hepatocyte growth factor (HGF)-Met system in papillary thyroid cancer: biological effects of HGF in thyroid cancer cells depend on Met expression levels. , 2004, Endocrinology.

[19]  A. Pasquini,et al.  Increased expression of Met protein is associated with up‐regulation of hypoxia inducible factor‐1 (HIF‐1) in tumour cells in papillary carcinoma of the thyroid , 2004, The Journal of pathology.

[20]  F. Meric,et al.  Expression profile of tyrosine kinases in breast cancer. , 2002, Clinical cancer research : an official journal of the American Association for Cancer Research.

[21]  J. Samet,et al.  Food and Drug Administration , 2007, BMJ : British Medical Journal.

[22]  C Eng,et al.  Gene expression in papillary thyroid carcinoma reveals highly consistent profiles , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[23]  T. Ståhl,et al.  Non-autocrine, constitutive activation of Met in human anaplastic thyroid carcinoma cells in culture , 1999, British Journal of Cancer.

[24]  P. Comoglio,et al.  Expression of Met protein and urokinase‐type plasminogen activator receptor (uPA‐R) in papillary carcinoma of the thyroid , 1998, The Journal of pathology.

[25]  F. Trimarchi,et al.  Expression of the hepatocyte growth factor and c-met in normal thyroid, non-neoplastic, and neoplastic nodules. , 1998, Thyroid : official journal of the American Thyroid Association.

[26]  P. Comoglio,et al.  EXPRESSION OF Met PROTEIN IN THYROID TUMOURS , 1996, The Journal of pathology.

[27]  M. Pierotti,et al.  Overexpression of the C-MET/HGF receptor in human thyroid carcinomas derived from the follicular epithelium , 1995, Journal of endocrinological investigation.

[28]  W. Bodmer,et al.  p53 mutations in colorectal cancer. , 1990, Proceedings of the National Academy of Sciences of the United States of America.