Recurrent loss of heterozygosity correlates with clinical outcome in pancreatic neuroendocrine cancer

Pancreatic neuroendocrine tumors (pNETs) are uncommon cancers arising from pancreatic islet cells. Here we report the analysis of gene mutation, copy number, and RNA expression of 57 sporadic well-differentiated pNETs. pNET genomes are dominated by aneuploidy, leading to concordant changes in RNA expression at the level of whole chromosomes and chromosome segments. We observed two distinct patterns of somatic pNET aneuploidy that are associated with tumor pathology and patient prognosis. Approximately 26% of the patients in this series had pNETs with genomes characterized by recurrent loss of heterozygosity (LoH) of 10 specific chromosomes, accompanied by bi-allelic MEN1 inactivation and generally poor clinical outcome. Another ~40% of patients had pNETs that lacked this recurrent LoH pattern but had chromosome 11 LoH, bi-allelic MEN1 inactivation, and universally good clinical outcome. The somatic aneuploidy allowed pathogenic germline variants (e.g., ATM) to be expressed unopposed, with RNA expression patterns showing inactivation of downstream tumor suppressor pathways. No prognostic associations were found with tumor morphology, single gene mutation, or expression of RNAs reflecting the activity of immune, differentiation, proliferative or tumor suppressor pathways. In pNETs, single gene mutations appear to be less important than aneuploidy, with MEN1 the only statistically significant recurrently mutated driver gene. In addition, only one pNET in the series had clearly actionable single nucleotide variants (SNVs) (in PTEN and FLCN) confirmed by corroborating RNA expression changes. The two clinically relevant patterns of LoH described here define a novel oncogenic mechanism and a plausible route to genomic precision oncology for this tumor type.Cancer: Frequent chromosome loss in rare pancreatic tumorsThe loss of entire chromosomes seems to be a fundamental driver of tumors arising from the hormone-producing cells of the pancreas. A team led by Cristin Print and Ben Lawrence from the University of Auckland, New Zealand, performed genomic and pathological analysis of 57 pancreatic neuroendocrine tumors, a rare form of cancer caused by the abnormal growth of hormone-producing islet cells within the pancreas. The researchers observed two distinct patterns of chromosome loss, with 26% of the samples missing one copy of 10 specific chromosomes and another 40% lacking a copy of chromosome 11. In both groups, the abnormal chromosome count prompts abnormal gene activity patterns, with recessive mutations unleashed and expressed unopposed. Single gene mutations seem to play only a minor role, suggesting that single gene-targeted drugs will provide little benefit in this disease setting, with more nuanced approaches required.

[1]  C. Larsson,et al.  Multiple endocrine neoplasia type 1 gene maps to chromosome 11 and is lost in insulinoma , 1988, Nature.

[2]  James Allan,et al.  DNA repair methyltransferase (Mgmt) knockout mice are sensitive to the lethal effects of chemotherapeutic alkylating agents. , 1999, Mutagenesis.

[3]  P. Beck‐Peccoz,et al.  Circulating ghrelin levels in patients with pancreatic and gastrointestinal neuroendocrine tumors: identification of one pancreatic ghrelinoma. , 2003, The Journal of clinical endocrinology and metabolism.

[4]  T. Speed,et al.  Summaries of Affymetrix GeneChip probe level data. , 2003, Nucleic acids research.

[5]  Benjamin M. Bolstad,et al.  affy - analysis of Affymetrix GeneChip data at the probe level , 2004, Bioinform..

[6]  S. Gerson MGMT: its role in cancer aetiology and cancer therapeutics , 2004, Nature Reviews Cancer.

[7]  Gordon K Smyth,et al.  Statistical Applications in Genetics and Molecular Biology Linear Models and Empirical Bayes Methods for Assessing Differential Expression in Microarray Experiments , 2011 .

[8]  Jeffrey T. Chang,et al.  GATHER: a systems approach to interpreting genomic signatures , 2006, Bioinform..

[9]  A. Rashid,et al.  Allelic alterations in pancreatic endocrine tumors identified by genome-wide single nucleotide polymorphism analysis. , 2007, Endocrine-related cancer.

[10]  Hadley Wickham,et al.  ggplot2 - Elegant Graphics for Data Analysis (2nd Edition) , 2017 .

[11]  Gonçalo R. Abecasis,et al.  The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..

[12]  Jeffrey W. Clark,et al.  O-Methylguanine DNA Methyltransferase Deficiency and Response toTemozolomide-BasedTherapy in Patients with Neuroendocrine Tumors , 2008 .

[13]  F. Bosman,et al.  WHO Classification of Tumours of the Digestive System , 2010 .

[14]  S. Beghelli,et al.  MEN1 in pancreatic endocrine tumors: analysis of gene and protein status in 169 sporadic neoplasms reveals alterations in the vast majority of cases. , 2010, Endocrine-related cancer.

[15]  C. Croce,et al.  Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[16]  Michael A. Choti,et al.  DAXX/ATRX, MEN1, and mTOR Pathway Genes Are Frequently Altered in Pancreatic Neuroendocrine Tumors , 2011, Science.

[17]  Marcel Martin Cutadapt removes adapter sequences from high-throughput sequencing reads , 2011 .

[18]  Y. Bang,et al.  Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. , 2011, The New England journal of medicine.

[19]  Jun Ma,et al.  Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. , 2011, The Lancet. Oncology.

[20]  D. Coppola,et al.  First‐line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas , 2011, Cancer.

[21]  Gonçalo R. Abecasis,et al.  The variant call format and VCFtools , 2011, Bioinform..

[22]  Ben Lawrence,et al.  The epidemiology of gastroenteropancreatic neuroendocrine tumors. , 2011, Endocrinology and metabolism clinics of North America.

[23]  E. D. de Vries,et al.  Everolimus for advanced pancreatic neuroendocrine tumors. , 2011, The New England journal of medicine.

[24]  S. Salzberg,et al.  TopHat-Fusion: an algorithm for discovery of novel fusion transcripts , 2011, Genome Biology.

[25]  Colin N. Dewey,et al.  RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome , 2011, BMC Bioinformatics.

[26]  R. McLendon,et al.  Altered Telomeres in Tumors with ATRX and DAXX Mutations , 2011, Science.

[27]  Thomas Zichner,et al.  DELLY: structural variant discovery by integrated paired-end and split-read analysis , 2012, Bioinform..

[28]  Christopher A. Miller,et al.  VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. , 2012, Genome research.

[29]  J. Chabot,et al.  Capecitabine and temozolomide (CAPTEM) for metastatic, well-differentiated neuroendocrine cancers: The Pancreas Center at Columbia University experience , 2013, Cancer Chemotherapy and Pharmacology.

[30]  Wendy S. W. Wong,et al.  Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs , 2012, Bioinform..

[31]  Steven L Salzberg,et al.  Fast gapped-read alignment with Bowtie 2 , 2012, Nature Methods.

[32]  Hiromitsu Araki,et al.  GeneSetDB: A comprehensive meta-database, statistical and visualisation framework for gene set analysis , 2012, FEBS open bio.

[33]  S. Elledge,et al.  Cumulative Haploinsufficiency and Triplosensitivity Drive Aneuploidy Patterns and Shape the Cancer Genome , 2013, Cell.

[34]  G. Getz,et al.  Inferring tumour purity and stromal and immune cell admixture from expression data , 2013, Nature Communications.

[35]  David T. W. Jones,et al.  Signatures of mutational processes in human cancer , 2013, Nature.

[36]  B. Ebert,et al.  Deletion 5q MDS: molecular and therapeutic implications. , 2013, Best practice & research. Clinical haematology.

[37]  Jiaqian Wang,et al.  Whole exome sequencing of insulinoma reveals recurrent T372R mutations in YY1 , 2013, Nature Communications.

[38]  Heng Li Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM , 2013, 1303.3997.

[39]  S. Kreth,et al.  Personalized treatment strategies in glioblastoma: MGMT promoter methylation status , 2013, OncoTargets and therapy.

[40]  Jianmin Wu,et al.  Somatic Point Mutation Calling in Low Cellularity Tumors , 2013, PloS one.

[41]  Helga Thorvaldsdóttir,et al.  Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration , 2012, Briefings Bioinform..

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

[43]  Anushya Muruganujan,et al.  PANTHER in 2013: modeling the evolution of gene function, and other gene attributes, in the context of phylogenetic trees , 2012, Nucleic Acids Res..

[44]  S. Halgamuge,et al.  Inferring copy number and genotype in tumour exome data , 2014, BMC Genomics.

[45]  E. Speel,et al.  Loss of DAXX and ATRX are associated with chromosome instability and reduced survival of patients with pancreatic neuroendocrine tumors. , 2014, Gastroenterology.

[46]  Thomas Lengauer,et al.  Comprehensive Analysis of DNA Methylation Data with RnBeads , 2014, Nature Methods.

[47]  Sohrab P. Shah,et al.  TITAN: inference of copy number architectures in clonal cell populations from tumor whole-genome sequence data , 2014, Genome research.

[48]  Andreas Krämer,et al.  Causal analysis approaches in Ingenuity Pathway Analysis , 2013, Bioinform..

[49]  R Core Team,et al.  R: A language and environment for statistical computing. , 2014 .

[50]  C. Romualdi,et al.  Prognostic Relevance of Aberrant DNA Methylation in G1 and G2 Pancreatic Neuroendocrine Tumors , 2014, Neuroendocrinology.

[51]  Andrew E. Teschendorff,et al.  ChAMP: 450k Chip Analysis Methylation Pipeline , 2014, Bioinform..

[52]  Rafael A. Irizarry,et al.  Minfi: a flexible and comprehensive Bioconductor package for the analysis of Infinium DNA methylation microarrays , 2014, Bioinform..

[53]  A. Amon,et al.  Short- and long-term effects of chromosome mis-segregation and aneuploidy , 2015, Nature Reviews Molecular Cell Biology.

[54]  D. Hanahan,et al.  A Cross-Species Analysis in Pancreatic Neuroendocrine Tumors Reveals Molecular Subtypes with Distinctive Clinical, Metastatic, Developmental, and Metabolic Characteristics. , 2015, Cancer discovery.

[55]  Sabine Tejpar,et al.  Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[56]  Ash A. Alizadeh,et al.  Robust enumeration of cell subsets from tissue expression profiles , 2015, Nature Methods.

[57]  V. Paradis,et al.  Gly388Arg FGFR4 Polymorphism Is Not Predictive of Everolimus Efficacy in Well-Differentiated Digestive Neuroendocrine Tumors , 2015, Neuroendocrinology.

[58]  Julian Gehring,et al.  SomaticSignatures: inferring mutational signatures from single-nucleotide variants , 2014, bioRxiv.

[59]  Rezvan Ehsani,et al.  EpiFactors: a comprehensive database of human epigenetic factors and complexes , 2015, Database J. Biol. Databases Curation.

[60]  A. Meeker,et al.  Analysis of potential response predictors to capecitabine/temozolomide in metastatic pancreatic neuroendocrine tumors. , 2016, Endocrine-related cancer.

[61]  E. Mardis,et al.  pVAC-Seq: A genome-guided in silico approach to identifying tumor neoantigens , 2016, Genome Medicine.

[62]  James Y. Zou Analysis of protein-coding genetic variation in 60,706 humans , 2015, Nature.

[63]  Xiaoyu Chen,et al.  Manta: rapid detection of structural variants and indels for germline and cancer sequencing applications , 2016, Bioinform..

[64]  Z. Storchová,et al.  Effects of aneuploidy on gene expression: implications for cancer , 2016, The FEBS journal.

[65]  Obi L. Griffith,et al.  GenVisR: Genomic Visualizations in R , 2016, bioRxiv.

[66]  Andrew J. Hill,et al.  Analysis of protein-coding genetic variation in 60,706 humans , 2015, bioRxiv.

[67]  V. Paradis,et al.  MGMT expression predicts response to temozolomide in pancreatic neuroendocrine tumors. , 2016, Endocrine-related cancer.

[68]  Ignace Vergote,et al.  Niraparib Maintenance Therapy in Platinum-Sensitive, Recurrent Ovarian Cancer. , 2016, The New England journal of medicine.

[69]  Shivashankar H. Nagaraj,et al.  Whole-genome landscape of pancreatic neuroendocrine tumours , 2017, Nature.

[70]  T. Speed,et al.  GRIDSS: sensitive and specific genomic rearrangement detection using positional de Bruijn graph assembly. , 2017, Genome research.

[71]  J. Berlin,et al.  Phase 3 Trial of 177Lu‐Dotatate for Midgut Neuroendocrine Tumors , 2017, The New England journal of medicine.

[72]  M. Mahajan,et al.  Insights into beta cell regeneration for diabetes via integration of molecular landscapes in human insulinomas , 2017, Nature Communications.

[73]  M. Fiorentino,et al.  Correlation between MGMT promoter methylation and response to temozolomide-based therapy in neuroendocrine neoplasms: an observational retrospective multicenter study , 2018, Endocrine.

[74]  E. Raymond,et al.  Clinical and Biomarker Evaluations of Sunitinib in Patients with Grade 3 Digestive Neuroendocrine Neoplasms , 2018, Neuroendocrinology.

[75]  Ying Cheng,et al.  Osimertinib in Untreated EGFR‐Mutated Advanced Non–Small‐Cell Lung Cancer , 2018, The New England journal of medicine.