Prognostic Impact of Novel Molecular Subtypes of Small Intestinal Neuroendocrine Tumor

Purpose: Small intestinal neuroendocrine tumors (SINET) are the commonest malignancy of the small intestine; however, underlying pathogenic mechanisms remain poorly characterized. Whole-genome and -exome sequencing has demonstrated that SINETs are mutationally quiet, with the most frequent known mutation in the cyclin-dependent kinase inhibitor 1B gene (CDKN1B) occurring in only ∼8% of tumors, suggesting that alternative mechanisms may drive tumorigenesis. The aim of this study is to perform genome-wide molecular profiling of SINETs in order to identify pathogenic drivers based on molecular profiling. This study represents the largest unbiased integrated genomic, epigenomic, and transcriptomic analysis undertaken in this tumor type. Experimental Design: Here, we present data from integrated molecular analysis of SINETs (n = 97), including whole-exome or targeted CDKN1B sequencing (n = 29), HumanMethylation450 BeadChip (Illumina) array profiling (n = 69), methylated DNA immunoprecipitation sequencing (n = 16), copy-number variance analysis (n = 47), and Whole-Genome DASL (Illumina) expression array profiling (n = 43). Results: Based on molecular profiling, SINETs can be classified into three groups, which demonstrate significantly different progression-free survival after resection of primary tumor (not reached at 10 years vs. 56 months vs. 21 months, P = 0.04). Epimutations were found at a recurrence rate of up to 85%, and 21 epigenetically dysregulated genes were identified, including CDX1 (86%), CELSR3 (84%), FBP1 (84%), and GIPR (74%). Conclusions: This is the first comprehensive integrated molecular analysis of SINETs. We have demonstrated that these tumors are highly epigenetically dysregulated. Furthermore, we have identified novel molecular subtypes with significant impact on progression-free survival. Clin Cancer Res; 22(1); 250–8. ©2015 AACR.

[1]  P. Hellman,et al.  Somatic Mutations and Genetic Heterogeneity at the CDKN1B Locus in Small Intestinal Neuroendocrine Tumors , 2015, Annals of Surgical Oncology.

[2]  Gangning Liang,et al.  Gene body methylation can alter gene expression and is a therapeutic target in cancer. , 2014, Cancer cell.

[3]  Johan Staaf,et al.  Genome-wide DNA Methylation Analysis of Lung Carcinoma Reveals One Neuroendocrine and Four Adenocarcinoma Epitypes Associated with Patient Outcome , 2014, Clinical Cancer Research.

[4]  J. Reubi,et al.  The Glucose-Dependent Insulinotropic Polypeptide Receptor: A Novel Target for Neuroendocrine Tumor Imaging—First Preclinical Studies , 2014, The Journal of Nuclear Medicine.

[5]  C. Larsson,et al.  Global hypomethylation and promoter methylation in small intestinal neuroendocrine tumors , 2014, Epigenetics.

[6]  Martin Vingron,et al.  Frequent mutations in chromatin-remodeling genes in pulmonary carcinoids , 2014, Nature Communications.

[7]  R. Versteeg Cancer: Tumours outside the mutation box , 2014, Nature.

[8]  G. Åkerström,et al.  Global DNA methylation patterns through an array-based approach in small intestinal neuroendocrine tumors. , 2014, Endocrine-related cancer.

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

[10]  A. Teschendorff,et al.  Using high-density DNA methylation arrays to profile copy number alterations , 2014, Genome Biology.

[11]  Robert Lowe,et al.  Marmal-aid – a database for Infinium HumanMethylation450 , 2013, BMC Bioinformatics.

[12]  Trevor J Pugh,et al.  Somatic mutation of CDKN1B in small intestine neuroendocrine tumors , 2013, Nature Genetics.

[13]  C. Larsson,et al.  Copy number alterations in small intestinal neuroendocrine tumors determined by array comparative genomic hybridization , 2013, BMC Cancer.

[14]  Jing Wang,et al.  WEB-based GEne SeT AnaLysis Toolkit (WebGestalt): update 2013 , 2013, Nucleic Acids Res..

[15]  Sean R. Davis,et al.  NCBI GEO: archive for functional genomics data sets—update , 2012, Nucleic Acids Res..

[16]  S. Duffy,et al.  Quantitative DNA methylation and recurrence of breast cancer: a study of 30 candidate genes. , 2012, Cancer biomarkers : section A of Disease markers.

[17]  A. Hartmann,et al.  Gastrointestinal , Hepatobiliary , and Pancreatic Pathology Methylation-Dependent Activation of CDX 1 through NF-B A Link from Inflammation to Intestinal Metaplasia in the Human Stomach , 2012 .

[18]  Reiner Schulz,et al.  Resources for methylome analysis suitable for gene knockout studies of potential epigenome modifiers , 2012, GigaScience.

[19]  Ning Li,et al.  Promoter Hypermethylation Mediated Downregulation of FBP1 in Human Hepatocellular Carcinoma and Colon Cancer , 2011, PloS one.

[20]  Hiroyuki Yamamoto,et al.  Co-expression of laminin β3 and γ2 chains and epigenetic inactivation of laminin α3 chain in gastric cancer. , 2011, International journal of oncology.

[21]  G. Getz,et al.  GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers , 2011, Genome Biology.

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

[23]  Andrew E. Teschendorff,et al.  Genome-wide DNA methylation analysis of archival formalin-fixed paraffin-embedded tissue using the Illumina Infinium HumanMethylation27 BeadChip. , 2010, Methods.

[24]  S. Beck,et al.  AutoMeDIP-seq: A high-throughput, whole genome, DNA methylation assay , 2010, Methods.

[25]  S. T. Mees,et al.  SERPINB5 and AKAP12 -- Expression and promoter methylation of metastasis suppressor genes in pancreatic ductal adenocarcinoma , 2010, BMC Cancer.

[26]  井伊 正則 Coexpression of laminin beta 3 and gamma 2 chains and epigenetic inactivation of laminin alpha 3 chain in gastric cancer , 2010 .

[27]  Peter A. Jones,et al.  Epigenetics in cancer. , 2010, Carcinogenesis.

[28]  Ahmed Essaghir,et al.  Novel markers for enterochromaffin cells and gastrointestinal neuroendocrine carcinomas , 2009, Modern Pathology.

[29]  Derek Y. Chiang,et al.  High‐resolution analysis of genetic alterations in small bowel carcinoid tumors reveals areas of recurrent amplification and loss , 2008, Genes, chromosomes & cancer.

[30]  Manal M. Hassan,et al.  One hundred years after "carcinoid": epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[31]  A. Scarpa,et al.  TNM staging of midgut and hindgut (neuro) endocrine tumors: a consensus proposal including a grading system , 2007, Virchows Archiv.

[32]  S. Ogino,et al.  18q loss of heterozygosity in microsatellite stable colorectal cancer is correlated with CpG island methylator phenotype-negative (CIMP-0) and inversely with CIMP-low and CIMP-high , 2007, BMC Cancer.

[33]  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 .

[34]  A. Gazdar,et al.  Aberrant promoter methylation of laminin-5-encoding genes in prostate cancers and its relationship to clinicopathological features. , 2003, Clinical cancer research : an official journal of the American Association for Cancer Research.

[35]  B Nebiyou Bekele,et al.  Aberrant promoter methylation and silencing of laminin-5-encoding genes in breast carcinoma. , 2003, Clinical cancer research : an official journal of the American Association for Cancer Research.