MTAP deletion confers enhanced dependency on the PRMT5 arginine methyltransferase in cancer cells

Tumors put in a vulnerable position Cancer cells often display alterations in metabolism that help fuel their growth. Such metabolic “rewiring” may also work against the cancer cells, however, by creating new vulnerabilities that can be exploited therapeutically. A variety of human tumors show changes in methionine metabolism caused by loss of the gene coding for 5-methylthioadenosine phosphorylase (MTAP). Mavrakis et al. and Kryukov et al. found that the loss of MTAP renders cancer cell lines sensitive to growth inhibition by compounds that suppress the activity of a specific arginine methyltransferase called PRMT5. Conceivably, drugs that inhibit PRMT5 activity could be developed into a tailored therapy for MTAP-deficient tumors. Science, this issue pp. 1208 and 1214 Tumors cope with a genomic change by rewiring their metabolism, but this makes them more susceptible to certain drugs. The discovery of cancer dependencies has the potential to inform therapeutic strategies and to identify putative drug targets. Integrating data from comprehensive genomic profiling of cancer cell lines and from functional characterization of cancer cell dependencies, we discovered that loss of the enzyme methylthioadenosine phosphorylase (MTAP) confers a selective dependence on protein arginine methyltransferase 5 (PRMT5) and its binding partner WDR77. MTAP is frequently lost due to its proximity to the commonly deleted tumor suppressor gene, CDKN2A. We observed increased intracellular concentrations of methylthioadenosine (MTA, the metabolite cleaved by MTAP) in cells harboring MTAP deletions. Furthermore, MTA specifically inhibited PRMT5 enzymatic activity. Administration of either MTA or a small-molecule PRMT5 inhibitor showed a modest preferential impairment of cell viability for MTAP-null cancer cell lines compared with isogenic MTAP-expressing counterparts. Together, our findings reveal PRMT5 as a potential vulnerability across multiple cancer lineages augmented by a common “passenger” genomic alteration.

[1]  Konstantinos J. Mavrakis,et al.  Abstract LB-017: Disordered methionine metabolism in MTAP/CDKN2A-deleted cancers leads to marked dependence on PRMT5 , 2016 .

[2]  Konstantinos J. Mavrakis,et al.  Disordered methionine metabolism in MTAP/CDKN2A-deleted cancers leads to dependence on PRMT5 , 2016, Science.

[3]  Pierre-Jacques Hamard,et al.  Arginine methyltransferase PRMT5 is essential for sustaining normal adult hematopoiesis. , 2015, The Journal of clinical investigation.

[4]  Robert A Copeland,et al.  A selective inhibitor of PRMT5 with in vivo and in vitro potency in MCL models. , 2015, Nature chemical biology.

[5]  C. Koh,et al.  MYC regulates the core pre-mRNA splicing machinery as an essential step in lymphomagenesis , 2015, Nature.

[6]  D. Choi,et al.  PRMT5 is essential for the eIF4E-mediated 5'-cap dependent translation. , 2014, Biochemical and biophysical research communications.

[7]  Ellen T. Gelfand,et al.  Parallel genome-scale loss of function screens in 216 cancer cell lines for the identification of context-specific genetic dependencies , 2014, Scientific Data.

[8]  J. Bertino,et al.  6-thioguanine: a drug with unrealized potential for cancer therapy. , 2014, The oncologist.

[9]  K. Howitz,et al.  Assay development for histone methyltransferases. , 2013, Assay and drug development technologies.

[10]  M. Bedford,et al.  Protein arginine methyltransferases and cancer , 2012, Nature Reviews Cancer.

[11]  Benjamin E. Gross,et al.  The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. , 2012, Cancer discovery.

[12]  Adam A. Margolin,et al.  The Cancer Cell Line Encyclopedia enables predictive modeling of anticancer drug sensitivity , 2012, Nature.

[13]  Randall W. King,et al.  A Bioinformatics Method Identifies Prominent Off-targeted Transcripts in RNAi Screens , 2012, Nature Methods.

[14]  Yu-Jie Hu,et al.  Versatility of PRMT5-induced methylation in growth control and development. , 2011, Trends in biochemical sciences.

[15]  J. Mesirov,et al.  Systematic investigation of genetic vulnerabilities across cancer cell lines reveals lineage-specific dependencies in ovarian cancer , 2011, Proceedings of the National Academy of Sciences.

[16]  J. Bertino,et al.  Targeting tumors that lack methylthioadenosine phosphorylase (MTAP) activity , 2011, Cancer biology & therapy.

[17]  A. Sickmann,et al.  RioK1, a New Interactor of Protein Arginine Methyltransferase 5 (PRMT5), Competes with pICln for Binding and Modulates PRMT5 Complex Composition and Substrate Specificity* , 2010, The Journal of Biological Chemistry.

[18]  Xinbin Chen,et al.  PRMT5 is required for cell-cycle progression and p53 tumor suppressor function , 2009, Nucleic acids research.

[19]  Robert L Moritz,et al.  PRMT5-mediated methylation of histone H4R3 recruits DNMT3A, coupling histone and DNA methylation in gene silencing , 2009, Nature Structural &Molecular Biology.

[20]  H. Burris,et al.  A phase II multicenter study of L-alanosine, a potent inhibitor of adenine biosynthesis, in patients with MTAP-deficient cancer , 2009, Investigational New Drugs.

[21]  L. Lim,et al.  Widespread siRNA "off-target" transcript silencing mediated by seed region sequence complementarity. , 2006, RNA.

[22]  J. Eshleman,et al.  Homozygous deletions of methylthioadenosine phosphorylase in human biliary tract cancers , 2005, Molecular Cancer Therapeutics.

[23]  E. Montgomery,et al.  Concordant Loss of MTAP and p16/CDKN2A Expression in Gastroesophageal Carcinogenesis: Evidence of Homozygous Deletion in Esophageal Noninvasive Precursor Lesions and Therapeutic Implications , 2005, The American journal of surgical pathology.

[24]  Mark T Bedford,et al.  Arginine methylation an emerging regulator of protein function. , 2005, Molecular cell.

[25]  J. Cameron,et al.  Homozygous deletion of the MTAP gene in invasive adenocarcinoma of the pancreas and in periampullary cancer: A potential new target for therapy , 2005, Cancer biology & therapy.

[26]  M. Ladanyi,et al.  Homozygous deletion of CDKN2A and codeletion of the methylthioadenosine phosphorylase gene in the majority of pleural mesotheliomas. , 2003, Clinical cancer research : an official journal of the American Association for Cancer Research.

[27]  P. Diegelman,et al.  Methylthioadenosine phosphorylase, a gene frequently codeleted with p16(cdkN2a/ARF), acts as a tumor suppressor in a breast cancer cell line. , 2002, Cancer research.

[28]  M. Mann,et al.  A Novel WD Repeat Protein Component of the Methylosome Binds Sm Proteins* , 2002, The Journal of Biological Chemistry.

[29]  Juri Rappsilber,et al.  The Methylosome, a 20S Complex Containing JBP1 and pICln, Produces Dimethylarginine-Modified Sm Proteins , 2001, Molecular and Cellular Biology.

[30]  Steven Clarke,et al.  PRMT5 (Janus Kinase-binding Protein 1) Catalyzes the Formation of Symmetric Dimethylarginine Residues in Proteins* , 2001, The Journal of Biological Chemistry.

[31]  S. Pestka,et al.  The Human Homologue of the Yeast Proteins Skb1 and Hsl7p Interacts with Jak Kinases and Contains Protein Methyltransferase Activity* , 1999, The Journal of Biological Chemistry.

[32]  P. Tran,et al.  Genomic cloning of methylthioadenosine phosphorylase: a purine metabolic enzyme deficient in multiple different cancers. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[33]  T. Savarese,et al.  Gene deletion chemoselectivity: codeletion of the genes for p16(INK4), methylthioadenosine phosphorylase, and the alpha- and beta-interferons in human pancreatic cell carcinoma lines and its implications for chemotherapy. , 1996, Cancer research.

[34]  J. Seidenfeld,et al.  Trends in the biochemical pharmacology of 5'-deoxy-5'-methylthioadenosine. , 1982, Biochemical pharmacology.