Genomic characterization of a well-differentiated grade 3 pancreatic neuroendocrine tumor

Laura M Williamson, Michael Steel, Jasleen K Grewal, My Lihn Thibodeau, Eric Y Zhao, Jonathan M Loree, Kevin C Yang, Sharon M Gorski, Andrew J Mungall, Karen L Mungall, Richard A Moore, Marco A Marra, Janessa Laskin, Daniel J Renouf, David F Schaeffer, Steven J M Jones, Laura M Williamson, Michael Steel, Jasleen K Grewal, My Lihn Thibodeau, Eric Y Zhao, Jonathan M Loree, Kevin C Yang, Sharon M Gorski, Andrew J Mungall, Karen L Mungall, Richard A Moore, Marco A Marra, Janessa Laskin, Daniel J Renouf, David F Schaeffer, Steven J M Jones

Abstract

Pancreatic neuroendocrine neoplasms (PanNENs) represent a minority of pancreatic neoplasms that exhibit variability in prognosis. Ongoing mutational analyses of PanNENs have found recurrent abnormalities in chromatin remodeling genes (e.g., DAXX and ATRX), and mTOR pathway genes (e.g., TSC2, PTEN PIK3CA, and MEN1), some of which have relevance to patients with related familial syndromes. Most recently, grade 3 PanNENs have been divided into two groups based on differentiation, creating a new group of well-differentiated grade 3 neuroendocrine tumors (PanNETs) that have had a limited whole-genome level characterization to date. In a patient with a metastatic well-differentiated grade 3 PanNET, our study utilized whole-genome sequencing of liver metastases for the comparative analysis and detection of single-nucleotide variants, insertions and deletions, structural variants, and copy-number variants, with their biologic relevance confirmed by RNA sequencing. We found that this tumor most notably exhibited a TSC1-disrupting fusion, showed a novel CHD7-BEND2 fusion, and lacked any somatic variants in ATRX, DAXX, and MEN1.

Trial registration: ClinicalTrials.gov NCT02155621.

Keywords: neoplasm of the pancreas; neuroendocrine neoplasm.

© 2019 Williamson et al.; Published by Cold Spring Harbor Laboratory Press.

Figures

Figure 1.
Figure 1.
Tumor histopathology and Ki-67 immunohistochemistry. (A) Core biopsy of a liver metastasis stained for hematoxylin and eosin showing a metastatic well-differentiated neuroendocrine tumor clinicoradiologically consistent with pancreatic origin (magnification at 10× [left] and 40× [right]). (B) Paired Ki-67 immunohistochemistry showing a proliferative index of 30%, classifying the tumor as a well-differentiated grade 3 (G3) neuroendocrine tumor (magnification at 10× [left] and 40× [right]).
Figure 2.
Figure 2.
Copy-number aberration and TSC1–TMEM71 structural rearrangement in the G3 PanNET. (A) Circos plot illustrating copy-number alterations and loss of heterozygosity (LOH) observed in the tumor sample. (B) Genomic sequencing read alignment of the genomic regions affected by the rearrangement. Green reads that aligned to TSC1 and purple reads that aligned to TMEM71 indicate split reads that support the rearrangement. (C) Illustration of TSC1–TMEM71 rearrangement generated by MAVIS (Reisle et al. 2018). The TMEM71 and TSC1 breakpoints are indicated at the chromosome, gene, and transcript level (B1 and B2, respectively, upper box). Protein-coding sequence associated with the respective transcripts are indicated by the black line and the amino acids included in the fusion product are indicated. Pfam (http://pfam.xfam.org) protein domains are indicated in the track below the transcript: Hamartin (PF04388). The predicted fusion transcript and protein-coding sequence are shown in the lower box (TSC1 exons are colored green; TMEM71 exons are colored blue).
Figure 3.
Figure 3.
CHD7–BEND2 structural rearrangement and increased BEND2 expression. (A) Genomic sequencing read alignment of the genomic regions affected by the rearrangement. Gold reads that aligned to CHD7 and purple reads that aligned to BEND2 indicate reads that support the rearrangement. (B) Illustration of CHD7–BEND2 rearrangement. The CHD7 and BEND2 breakpoints are indicated at the chromosome, gene, and transcript level (B1 and B2, respectively, upper box). The protein-coding sequences associated with the respective transcripts are indicated by the black line, and the amino acids included in the fusion product are indicated. The predicted fusion transcript and protein-coding sequence are shown in the lower box (CHD7 exons are colored blue; BEND2 exons are colored green). Pfam protein domains are indicated by the tracks below the transcript: SNF2_N (F00176), helicase_C (PF00271), Chromo (PF00385), BRK (PF07533), BEN (PF10523). (C) Genomic sequencing reads (top) and RNA sequencing reads (bottom) aligned to the BEND2 gene. Genomic reads that are affected by the rearrangement are colored purple. The BEND2 fusion breakpoint is indicated by the dashed line. RNA-seq coverage downstream from the breakpoint (exons 5–14) is increased compared to upstream of the breakpoint (exons 1–4). (D) Boxplot of BEND2 RPKM (log10 transformed) across a panel of 40 TCGA data sets. BEND2 log10(RPKM) for this patient sample is indicated by the red horizontal line.
Figure 4.
Figure 4.
Low expression of SSTR genes in the G3 PanNET. SSTR1, SSTR2, SSTR3, and SSTR5 RPKM normalized to the mean of the cohort were plotted for seven metastatic PanNET samples. Relative SSTR gene expression for this patient sample is highlighted in red.

References

    1. Abhiman S, Iyer LM, Aravind L. 2008. BEN: a novel domain in chromatin factors and DNA viral proteins. Bioinformatics 24: 458–461. 10.1093/bioinformatics/btn007
    1. Alexandrov LB, Nik-Zainal S, Wedge DC, Campbell PJ, Stratton MR. 2013. Deciphering signatures of mutational processes operative in human cancer. Cell Rep 3: 246–259. 10.1016/j.celrep.2012.12.008
    1. Bajpai R, Chen DA, Rada-Iglesias A, Zhang J, Xiong Y, Helms J, Chang CP, Zhao Y, Swigut T, Wysocka J. 2010. CHD7 cooperates with PBAF to control multipotent neural crest formation. Nature 463: 958–962. 10.1038/nature08733
    1. Bergsland EK, Roy R, Stephens P, Ross JS, Bailey M, Olshen A. 2016. Genomic profiling to distinguish poorly differentiated neuroendocrine carcinomas arising in different sites. J Clin Oncol 34: 4020 10.1200/JCO.2016.34.15_suppl.4020
    1. Birol I, Jackman SD, Nielsen CB, Qian JQ, Varhol R, Stazyk G, Morin RD, Zhao Y, Hirst M, Schein JE, et al. 2009. De novo transcriptome assembly with ABySS. Bioinformatics 25: 2872–2877. 10.1093/bioinformatics/btp367
    1. Caplin ME, Pavel M, Ćwikla JB, Phan AT, Raderer M, Sedláčková E, Cadiot G, Wolin EM, Capdevila J, Wall L, et al. 2014. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med 371: 224–233. 10.1056/NEJMoa1316158
    1. Castro AF, Rebhun JF, Clark GJ, Quilliam LA. 2003. Rheb binds tuberous sclerosis complex 2 (TSC2) and promotes S6 kinase activation in a rapamycin- and farnesylation-dependent manner. J Biol Chem 278: 32493–32496. 10.1074/jbc.C300226200
    1. Coriat R, Walter T, Terris B, Couvelard A, Ruszniewski P. 2016. Gastroenteropancreatic well-differentiated grade 3 neuroendocrine tumors: review and position statement. Oncologist 21: 1191–1199. 10.1634/theoncologist.2015-0476
    1. Dai Q, Ren A, Westholm JO, Serganov AA, Patel DJ, Lai EC. 2013. The BEN domain is a novel sequence-specific DNA-binding domain conserved in neural transcriptional repressors. Genes Dev 27: 602–614. 10.1101/gad.213314.113
    1. Dai Q, Ren A, Westholm JO, Duan H, Patel DJ, Lai EC. 2015. Common and distinct DNA-binding and regulatory activities of the BEN-solo transcription factor family. Genes Dev 29: 48–62. 10.1101/gad.252122.114
    1. Dasari A, Shen C, Halperin D, Zhao B, Zhou S, Xu Y, Shih T, Yao JC. 2017. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol 3: 1335–1342. 10.1001/jamaoncol.2017.0589
    1. Ding J, Bashashati A, Roth A, Oloumi A, Tse K, Zeng T, Haffari G, Hirst M, Marra MA, Condon A, et al. 2012. Feature-based classifiers for somatic mutation detection in tumour–normal paired sequencing data. Bioinformatics 28: 167–175. 10.1093/bioinformatics/btr629
    1. Edge S, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A. 2010. AJCC Cancer Staging Manual. Springer, France.
    1. Feng W, Khan MA, Bellvis P, Zhu Z, Bernhardt O, Herold-Mende C, Liu HK. 2013. The chromatin remodeler CHD7 regulates adult neurogenesis via activation of SoxC transcription factors. Cell Stem Cell 13: 62–72. 10.1016/j.stem.2013.05.002
    1. Florio T. 2008. Molecular mechanisms of the antiproliferative activity of somatostatin receptors (SSTRs) in neuroendocrine tumors. Front Biosci 13: 822–840. 10.2741/2722
    1. Ha G, Roth A, Lai D, Bashashati A, Ding J, Goya R, Giuliany R, Rosner J, Oloumi A, Shumansky K, et al. 2012. Integrative analysis of genome-wide loss of heterozygosity and monoallelic expression at nucleotide resolution reveals disrupted pathways in triple-negative breast cancer. Genome Res 22: 1995–2007. 10.1101/gr.137570.112
    1. Huang J, Manning BD. 2008. The TSC1–TSC2 complex: a molecular switchboard controlling cell growth. Biochem J 412: 179–190. 10.1042/BJ20080281
    1. Inoki K, Li Y, Xu T, Guan KL. 2003. Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. Genes Dev 17: 1829–1834. 10.1101/gad.1110003
    1. Jensen RT, Berna MJ, Bingham DB, Norton JA. 2008. Inherited pancreatic endocrine tumor syndromes: advances in molecular pathogenesis, diagnosis, management, and controversies. Cancer 113: 1807–1843. 10.1002/cncr.23648
    1. Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A, Schulick RD, Tang LH, Wolfgang CL, Choti MA, et al. 2011. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 331: 1199–1203. 10.1126/science.1200609
    1. John M, Meyerhof W, Richter D, Waser B, Schaer JC, Scherübl H, Boese-Landgraf J, Neuhaus P, Ziske C, Mölling K, et al. 1996. Positive somatostatin receptor scintigraphy correlates with the presence of somatostatin receptor subtype 2. Gut 38: 33–39. 10.1136/gut.38.1.33
    1. Jones SJ, Laskin J, Li YY, Griffith OL, An J, Bilenky M, Butterfield YS, Cezard T, Chuah E, Corbett R, et al. 2010. Evolution of an adenocarcinoma in response to selection by targeted kinase inhibitors. Genome Biol 11: R82 10.1186/gb-2010-11-8-r82
    1. Jones MR, Lim H, Shen Y, Pleasance E, Ch'ng C, Reisle C, Leelakumari S, Zhao C, Yip S, Ho J, et al. 2017. Successful targeting of the NRG1 pathway indicates novel treatment strategy for metastatic cancer. Ann Oncol 28: 3092–3097. 10.1093/annonc/mdx523
    1. Klimstra DS, Modlin IR, Coppola D, Lloyd RV, Suster S. 2010. The pathologic classification of neuroendocrine tumors: a review of nomenclature, grading, and staging systems. Pancreas 39: 707–712. 10.1097/MPA.0b013e3181ec124e
    1. Kulke MH, Siu LL, Tepper JE, Fisher G, Jaffe D, Haller DG, Ellis LM, Benedetti JK, Bergsland EK, Hobday TJ, et al. 2011. Future directions in the treatment of neuroendocrine tumors: consensus report of the National Cancer Institute Neuroendocrine Tumor clinical trials planning meeting. J Clin Oncol 29: 934–943. 10.1200/JCO.2010.33.2056
    1. Larson AM, Hedgire SS, Deshpande V, Stemmer-Rachamimov AO, Harisinghani MG, Ferrone CR, Shah U, Thiele EA. 2012. Pancreatic neuroendocrine tumors in patients with tuberous sclerosis complex. Clin Genet 82: 558–563. 10.1111/j.1399-0004.2011.01805.x
    1. Laskin J, Jones S, Aparicio S, Chia S, Ch'ng C, Deyell R, Eirew P, Fok A, Gelmon K, Ho C, et al. 2015. Lessons learned from the application of whole-genome analysis to the treatment of patients with advanced cancers. Cold Spring Harb Mol Case Stud 1: a000570 10.1101/mcs.a000570
    1. Li H, Durbin R. 2010. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics 26: 589–595. 10.1093/bioinformatics/btp698
    1. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R, 1000 Genome Project Data Processing Subgroup. 2009. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25: 2078–2079. 10.1093/bioinformatics/btp352
    1. Lloyd RV, Osamura RY, Klöppel G, Rosai J. 2017. WHO classification of tumours of endocrine organs. IARC Press, France.
    1. Osipovich AB, Long Q, Manduchi E, Gangula R, Hipkens SB, Schneider J, Okubo T, Stoeckert CJ, Takada S, Magnuson MA. 2014. Insm1 promotes endocrine cell differentiation by modulating the expression of a network of genes that includes Neurog3 and Ripply3. Development 141: 2939–2949. 10.1242/dev.104810
    1. Papotti M, Bongiovanni M, Volante M, Allìa E, Landolfi S, Helboe L, Schindler M, Cole SL, Bussolati G. 2002. Expression of somatostatin receptor types 1–5 in 81 cases of gastrointestinal and pancreatic endocrine tumors. A correlative immunohistochemical and reverse-transcriptase polymerase chain reaction analysis. Virchows Arch 440: 461–475. 10.1007/s00428-002-0609-x
    1. Rausch T, Zichner T, Schlattl A, Stutz AM, Benes V, Korbel JO. 2012. DELLY: structural variant discovery by integrated paired-end and split-read analysis. Bioinformatics 28: i333–i339. 10.1093/bioinformatics/bts378
    1. Reisle C, Mungall KL, Choo C, Paulino D, Bleile DW, Muhammadzadeh A, Mungall AJ, Moore RA, Shlafman I, Coope R, et al. 2018. MAVIS: merging, annotation, validation, and illustration of structural variants. Bioinformatics. 10.1093/bioinformatics/bty621
    1. Rinke A, Müller HH, Schade-Brittinger C, Klose KJ, Barth P, Wied M, Mayer C, Aminossadati B, Pape UF, Bläker M, et al. 2009. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol 27: 4656–4663. 10.1200/JCO.2009.22.8510
    1. Robertson G, Schein J, Chiu R, Corbett R, Field M, Jackman SD, Mungall K, Lee S, Okada HM, Qian JQ, et al. 2010. De novo assembly and analysis of RNA-seq data. Nat Methods 7: 909–912.
    1. Santiago Lima AJ, Hoogeveen-Westerveld M, Nakashima A, Maat-Kievit A, van den Ouweland A, Halley D, Kikkawa U, Nellist M. 2014. Identification of regions critical for the integrity of the TSC1-TSC2-TBC1D7 complex. PLoS One 9: e93940 10.1371/journal.pone.0093940
    1. Sathyan KM, Shen Z, Tripathi V, Prasanth KV, Prasanth SG. 2011. A BEN-domain-containing protein associates with heterochromatin and represses transcription. J Cell Sci 124: 3149–3163. 10.1242/jcs.086603
    1. Saunders CT, Wong WS, Swamy S, Becq J, Murray LJ, Cheetham RK. 2012. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinformatics 28: 1811–1817. 10.1093/bioinformatics/bts271
    1. Scarpa A, Chang DK, Nones K, Corbo V, Patch AM, Bailey P, Lawlor RT, Johns AL, Miller DK, Mafficini A, et al. 2017. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature 543: 65–71. 10.1038/nature21063
    1. Schnetz MP, Handoko L, Akhtar-Zaidi B, Bartels CF, Pereira CF, Fisher AG, Adams DJ, Flicek P, Crawford GE, Laframboise T, et al. 2010. CHD7 targets active gene enhancer elements to modulate ES cell-specific gene expression. PLoS Genet 6: e1001023 10.1371/journal.pgen.1001023
    1. Sheffield BS, Tinker AV, Shen Y, Hwang H, Li-Chang HH, Pleasance E, Ch'ng C, Lum A, Lorette J, McConnell YJ, et al. 2015. Personalized oncogenomics: clinical experience with malignant peritoneal mesothelioma using whole genome sequencing. PLoS One 10: e0119689 10.1371/journal.pone.0119689
    1. Simpson JT, Wong K, Jackman SD, Schein JE, Jones SJ, Birol I. 2009. ABySS: a parallel assembler for short read sequence data. Genome Res 19: 1117–1123. 10.1101/gr.089532.108
    1. Sturm D, Orr BA, Toprak UH, Hovestadt V, Jones DTW, Capper D, Sill M, Buchhalter I, Northcott PA, Leis I, et al. 2016. New brain tumor entities emerge from molecular classification of CNS-PNETs. Cell 164: 1060–1072. 10.1016/j.cell.2016.01.015
    1. Tang LH, Basturk O, Sue JJ, Klimstra DS. 2016. A practical approach to the classification of WHO grade 3 (G3) well-differentiated Neuroendocrine Tumor (WD-NET) and Poorly Differentiated Neuroendocrine Carcinoma (PD-NEC) of the pancreas. Am J Surg Pathol 40: 1192–1202. 10.1097/PAS.0000000000000662
    1. Wong HL, Yang KC, Shen Y, Zhao EY, Loree JM, Kennecke HF, Kalloger SE, Karasinska JM, Lim HJ, Mungall AJ, et al. 2018. Molecular characterization of metastatic pancreatic neuroendocrine tumors (PNETs) using whole-genome and transcriptome sequencing. Cold Spring Harb Mol Case Stud 4 10.1101/mcs.a002329
    1. Yachida S, Vakiani E, White CM, Zhong Y, Saunders T, Morgan R, de Wilde RF, Maitra A, Hicks J, Demarzo AM, et al. 2012. Small cell and large cell neuroendocrine carcinomas of the pancreas are genetically similar and distinct from well-differentiated pancreatic neuroendocrine tumors. Am J Surg Pathol 36: 173–184. 10.1097/PAS.0b013e3182417d36
    1. Yao JC, Phan AT, Chang DZ, Wolff RA, Hess K, Gupta S, Jacobs C, Mares JE, Landgraf AN, Rashid A, et al. 2008. Efficacy of RAD001 (everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. J Clin Oncol 26: 4311–4318. 10.1200/JCO.2008.16.7858
    1. Yao JC, Lombard-Bohas C, Baudin E, Kvols LK, Rougier P, Ruszniewski P, Hoosen S, St. Peter J, Haas T, Lebwohl D, et al. 2010. Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. J Clin Oncol 28: 69–76. 10.1200/JCO.2009.24.2669
    1. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, Hobday TJ, Okusaka T, Capdevila J, de Vries EG, et al. 2011. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 364: 514–523. 10.1056/NEJMoa1009290
    1. Yao JC, Pavel M, Lombard-Bohas C, Van Cutsem E, Voi M, Brandt U, He W, Chen D, Capdevila J, de Vries EGE, et al. 2016. Everolimus for the treatment of advanced pancreatic neuroendocrine tumors: overall survival and circulating biomarkers from the randomized, phase III RADIANT-3 study. J Clin Oncol 34: 3906–3913. 10.1200/JCO.2016.68.0702
    1. Zhao EY, Shen Y, Pleasance E, Kasaian K, Leelakumari S, Jones M, Bose P, Ch'ng C, Reisle C, Eirew P, et al. 2017. Homologous recombination deficiency and platinum-based therapy outcomes in advanced breast cancer. Clin Cancer Res 23: 7521–7530. 10.1158/1078-0432.CCR-17-1941

Source: PubMed

3
Abonnieren