Integrative genomic analysis of mouse and human hepatocellular carcinoma

Significance Hepatocellular carcinoma (HCC) research has been hampered by the absence of consensus mouse models with clearly defined molecular features faithfully recapitulating human HCC. Here we tackle this gap by implementing a cross-species comparative analysis between a large cohort of patients and four diverse mouse models focused on clinically and therapeutically relevant aspects of genomic and transcriptomic profiles and propose two of these models as valid for the study of different stages of human HCC. Cancer genomics has enabled the exhaustive molecular characterization of tumors and exposed hepatocellular carcinoma (HCC) as among the most complex cancers. This complexity is paralleled by dozens of mouse models that generate histologically similar tumors but have not been systematically validated at the molecular level. Accurate models of the molecular pathogenesis of HCC are essential for biomedical progress; therefore we compared genomic and transcriptomic profiles of four separate mouse models [MUP transgenic, TAK1-knockout, carcinogen-driven diethylnitrosamine (DEN), and Stelic Animal Model (STAM)] with those of 987 HCC patients with distinct etiologies. These four models differed substantially in their mutational load, mutational signatures, affected genes and pathways, and transcriptomes. STAM tumors were most molecularly similar to human HCC, with frequent mutations in Ctnnb1, similar pathway alterations, and high transcriptomic similarity to high-grade, proliferative human tumors with poor prognosis. In contrast, TAK1 tumors better reflected the mutational signature of human HCC and were transcriptionally similar to low-grade human tumors. DEN tumors were least similar to human disease and almost universally carried the Braf V637E mutation, which is rarely found in human HCC. Immune analysis revealed that strain-specific MHC-I genotype can influence the molecular makeup of murine tumors. Thus, different mouse models of HCC recapitulate distinct aspects of HCC biology, and their use should be adapted to specific questions based on the molecular features provided here.

[1]  Tim F. Rayner,et al.  Mutational landscape of a chemically-induced mouse model of liver cancer , 2018, bioRxiv.

[2]  T. Ideker,et al.  MHC-I Genotype Restricts the Oncogenic Mutational Landscape , 2017, Cell.

[3]  Rohit Loomba,et al.  Inflammation-induced IgA+ cells dismantle anti-liver cancer immunity , 2017, Nature.

[4]  Jean-François Deleuze,et al.  Mutational signatures reveal the dynamic interplay of risk factors and cellular processes during liver tumorigenesis , 2017, Nature Communications.

[5]  See-Tong Pang,et al.  Aristolochic acids and their derivatives are widely implicated in liver cancers in Taiwan and throughout Asia , 2017, Science Translational Medicine.

[6]  M. Kudo,et al.  Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial , 2017, The Lancet.

[7]  Steven J. M. Jones,et al.  Comprehensive and Integrative Genomic Characterization of Hepatocellular Carcinoma , 2017, Cell.

[8]  P. Oliveira,et al.  Animal models as a tool in hepatocellular carcinoma research: A Review , 2017, Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine.

[9]  Anita Makri Back to the future , 2017, Science.

[10]  S. Elledge,et al.  Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy , 2017, Science.

[11]  Masatoshi Kudo,et al.  Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial , 2017, The Lancet.

[12]  Mingming Jia,et al.  COSMIC: somatic cancer genetics at high-resolution , 2016, Nucleic Acids Res..

[13]  Hans Clevers,et al.  Tissue-specific mutation accumulation in human adult stem cells during life , 2016, Nature.

[14]  J. Martens,et al.  A novel independence test for somatic alterations in cancer shows that biology drives mutual exclusivity but chance explains most co-occurrence , 2016, bioRxiv.

[15]  Gábor E. Tusnády,et al.  A comprehensive survey of the mutagenic impact of common cancer cytotoxics , 2016, Genome Biology.

[16]  M. Stratton,et al.  Mutational signatures associated with tobacco smoking in human cancer , 2016, Science.

[17]  Keith A. Boroevich,et al.  Whole-genome mutational landscape and characterization of noncoding and structural mutations in liver cancer , 2016, Nature Genetics.

[18]  M. Nielsen,et al.  NetMHCpan-3.0; improved prediction of binding to MHC class I molecules integrating information from multiple receptor and peptide length datasets , 2016, Genome Medicine.

[19]  G. Gores,et al.  Hepatocellular carcinoma , 2016, Nature Reviews Disease Primers.

[20]  Lloyd H. Michael,et al.  The Guide for the Care and Use of Laboratory Animals. , 2016, ILAR journal.

[21]  M. Stratton,et al.  Clock-like mutational processes in human somatic cells , 2015, Nature Genetics.

[22]  Hayato Nakagawa Recent advances in mouse models of obesity- and nonalcoholic steatohepatitis-associated hepatocarcinogenesis. , 2015, World journal of hepatology.

[23]  Jessica Zucman-Rossi,et al.  Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets , 2015, Nature Genetics.

[24]  C. Mathers,et al.  Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012 , 2015, International journal of cancer.

[25]  N. Hacohen,et al.  Molecular and Genetic Properties of Tumors Associated with Local Immune Cytolytic Activity , 2015, Cell.

[26]  W. Sung,et al.  Genomic portrait of resectable hepatocellular carcinomas: Implications of RB1 and FGF19 aberrations for patient stratification , 2014, Hepatology.

[27]  Hiromi Nakamura,et al.  Trans-ancestry mutational landscape of hepatocellular carcinoma genomes , 2014, Nature Genetics.

[28]  M. Karin,et al.  ER stress cooperates with hypernutrition to trigger TNF-dependent spontaneous HCC development. , 2014, Cancer cell.

[29]  Deepak Mittal,et al.  New insights into cancer immunoediting and its three component phases--elimination, equilibrium and escape. , 2014, Current opinion in immunology.

[30]  D. Calvisi,et al.  Hydrodynamic transfection for generation of novel mouse models for liver cancer research. , 2014, The American journal of pathology.

[31]  Kristen Jepsen,et al.  Identification of Liver Cancer Progenitors Whose Malignant Progression Depends on Autocrine IL-6 Signaling , 2013, Cell.

[32]  Steven A. Roberts,et al.  Mutational heterogeneity in cancer and the search for new cancer genes , 2014 .

[33]  A. Moorman,et al.  Hepatitis C in the United States. , 2013, The New England journal of medicine.

[34]  E. Wagner,et al.  Mouse models for liver cancer , 2013, Molecular oncology.

[35]  K. Kinzler,et al.  Cancer Genome Landscapes , 2013, Science.

[36]  Kenji Suzuki,et al.  A murine model for non-alcoholic steatohepatitis showing evidence of association between diabetes and hepatocellular carcinoma , 2013, Medical Molecular Morphology.

[37]  M. Stratton,et al.  Deciphering Signatures of Mutational Processes Operative in Human Cancer , 2013, Cell reports.

[38]  S. Imbeaud,et al.  Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma , 2012, Nature Genetics.

[39]  Benjamin J. Raphael,et al.  De novo discovery of mutated driver pathways in cancer , 2011 .

[40]  C. Sander,et al.  Mutual exclusivity analysis identifies oncogenic network modules. , 2012, Genome research.

[41]  Syed Haider,et al.  International Cancer Genome Consortium Data Portal—a one-stop shop for cancer genomics data , 2011, Database J. Biol. Databases Curation.

[42]  O. Andrisani,et al.  Gene signatures in hepatocellular carcinoma (HCC). , 2011, Seminars in cancer biology.

[43]  H. El‐Serag,et al.  Surveillance for hepatocellular carcinoma: in whom and how? , 2011, Therapeutic advances in gastroenterology.

[44]  R. Hruban,et al.  Prioritization of driver mutations in pancreatic cancer using cancer-specific high-throughput annotation of somatic mutations (CHASM) , 2010, Cancer biology & therapy.

[45]  Rocio Lopez,et al.  The incidence and risk factors of hepatocellular carcinoma in patients with nonalcoholic steatohepatitis , 2010, Hepatology.

[46]  S. Akira,et al.  Disruption of TAK1 in hepatocytes causes hepatic injury, inflammation, fibrosis, and carcinogenesis , 2009, Proceedings of the National Academy of Sciences.

[47]  Derek Y. Chiang,et al.  Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma. , 2009, Cancer research.

[48]  F. Heindryckx,et al.  Experimental mouse models for hepatocellular carcinoma research , 2009, International journal of experimental pathology.

[49]  J. Llovet,et al.  Preclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and VEGF and PDGF receptor tyrosine kinase signaling , 2008, Molecular Cancer Therapeutics.

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

[51]  H. El‐Serag,et al.  Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. , 2007, Gastroenterology.

[52]  M. Stratton,et al.  Statistical Analysis of Pathogenicity of Somatic Mutations in Cancer , 2006, Genetics.

[53]  F. X. Bosch,et al.  Epidemiology of hepatocellular carcinoma. , 2005, Clinics in liver disease.

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

[55]  Division on Earth Guide for the Care and Use of Laboratory Animals , 1996 .