J Pathol Transl Med.  2021 May;55(3):192-201. 10.4132/jptm.2021.01.23.

Identification of PI3K-AKT signaling as the dominant altered pathway in intestinal type ampullary cancers through whole-exome sequencing

Affiliations
  • 1Departments of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
  • 2Departments of Surgical Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
  • 3Departments of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
  • 4Department of Pathology & Lab Medicine, All India Institute of Medical Sciences, Raebareli, India

Abstract

Background
The genetic landscape of intestinal (INT) and pancreatobiliary (PB) type ampullary cancer (AC) has been evolving with distinct as well as overlapping molecular profiles.
Methods
We performed whole-exome sequencing in 37 cases of AC to identify the targetable molecular profiles of INT and PB tumors. Paired tumor-normal sequencing was performed on the HiSeq 2500 Illumina platform.
Results
There were 22 INT, 13 PB, and two cases of mixed differentiation of AC that exhibited a total of 1,263 somatic variants in 112 genes (2–257 variants/case) with 183 somatic deleterious variants. INT showed variations in 78 genes (1–31/case), while PB showed variations in 51 genes (1–29/case). Targetable mutations involving one or more major pathways were found in 86.5% of all ACs. Mutations in APC, CTNNB1, SMAD4, KMT2, EPHA, ERBB, and Notch genes were more frequent in INT tumors, while chromatin remodeling complex mutations were frequent in PB tumors. In the major signaling pathways, the phosphoinositide 3-kinase (PI3)/AKT and RAS/mitogen-activated protein kinase (MAPK) pathways were significantly mutated in 70% of cases (82% INT, 46% PB, p = .023), with PI3/AKT mutation being more frequent in INT and RAS/MAPK in PB tumors. Tumor mutation burden was low in both differentiation types, with 1.6/Mb in INT and 0.8/Mb in PB types (p =.217).
Conclusions
The exome data suggest that INT types are genetically more unstable than PB and involve mutations in tumor suppressors, oncogenes, transcription factors, and chromatin remodeling genes. The spectra of the genetic profiles of INT and PB types suggested primary targeting of PI3/AKT in INT and RAS/RAF and PI3/AKT pathways in PB carcinomas.

Keyword

Ampullary; Exome; Intestinal; Pancreatobiliary

Figure

  • Fig. 1 Microphotograph of intestinal differentiation. Low-power (A) and high-power (B) views showing tall columnar cells with elongated to oblong basal nuclei and nuclear stratification.

  • Fig. 2 Microphotograph of pancreatobiliary differentiation. Low-power (A) and high-power (B) views showing cuboidal to low columnar cells with rounded centrally placed nuclei with no nuclear stratification. Desmoplastic stroma can be observed between the tumor glands.

  • Fig. 3 Immunohistochemical stains (A, CDX2; B, cytokeratin 20; C, MUC2) expressed in the intestinal type of ampullary cancer.

  • Fig. 4 Immunohistochemical stains (A, MUC1; B, cytokeratin [CK] 17; C, CK7) expressed in the pancreatobiliary type of ampullary cancer.

  • Fig. 5 Schema representing the workflow for whole-exome sequencing and data analysis. UTR, untranslated region.

  • Fig. 6 (A) Deleterious somatic variants in intestinal (INT) ampullary cancers. (B) Deleterious somatic variants in pancreatobiliary (PB) ampullary cancers.

  • Fig. 7 (A) Genes forming part of the cellular component in intestinal and pancreatobiliary types of ampullary carcinoma. (B) Genes responsible for different classes of proteins in intestinal and pancreatobiliary types of ampullary carcinoma. (C) Genes involved in different biological functions and processes in intestinal and pancreatobiliary types of ampullary carcinoma.

  • Fig. 8 Pathways involving the significantly most frequently mutated genes in intestinal and pancreatobiliary subtypes of ampullary carcinoma (p < .002).


Cited by  1 articles

Histologic subtyping of ampullary carcinoma for targeted therapy
Seung-Mo Hong
J Pathol Transl Med. 2021;55(3):235-235.    doi: 10.4132/jptm.2021.04.28.


Reference

References

1. Adsay V, Ohike N, Tajiri T, et al. Ampullary region carcinomas: definition and site specific classification with delineation of four clinicopathologically and prognostically distinct subsets in an analysis of 249 cases. Am J Surg Pathol. 2012; 36:1592–608.
2. Benhamiche AM, Jouve JL, Manfredi S, Prost P, Isambert N, Faivre J. Cancer of the ampulla of Vater: results of a 20-year population-based study. Eur J Gastroenterol Hepatol. 2000; 12:75–9.
3. Demeure MJ, Craig DW, Sinari S, et al. Cancer of the ampulla of Vater: analysis of the whole genome sequence exposes a potential therapeutic vulnerability. Genome Med. 2012; 4:56.
Article
4. Kim WS, Choi DW, Choi SH, Heo JS, You DD, Lee HG. Clinical significance of pathologic subtype in curatively resected ampulla of vater cancer. J Surg Oncol. 2012; 105:266–72.
Article
5. Colussi O, Voron T, Pozet A, et al. Prognostic score for recurrence after Whipple’s pancreaticoduodenectomy for ampullary carcinomas; results of an AGEO retrospective multicenter cohort. Eur J Surg Oncol. 2015; 41:520–6.
Article
6. Neoptolemos JP, Moore MJ, Cox TF, et al. Effect of adjuvant chemotherapy with fluorouracil plus folinic acid or gemcitabine vs observation on survival in patients with resected periampullary adenocarcinoma: the ESPAC-3 periampullary cancer randomized trial. JAMA. 2012; 308:147–56.
7. Shoji H, Morizane C, Hiraoka N, et al. Twenty-six cases of advanced ampullary adenocarcinoma treated with systemic chemotherapy. Jpn J Clin Oncol. 2014; 44:324–30.
Article
8. Kimura W, Futakawa N, Yamagata S, et al. Different clinicopathologic findings in two histologic types of carcinoma of papilla of Vater. Jpn J Cancer Res. 1994; 85:161–6.
Article
9. Albores-Saveedra J, Henson DE, Klimstra DS. Tumors of the gallbladder, extrahepatic bile ducts, and ampulla of Vater. Atlas of tumor pathology. 3rd series. 27. Washington, DC: Armed Forces Institute of Pathology;2000. p. 259–316.
10. Kumari N, Prabha K, Singh RK, Baitha DK, Krishnani N. Intestinal and pancreatobiliary differentiation in periampullary carcinoma: the role of immunohistochemistry. Hum Pathol. 2013; 44:2213–9.
Article
11. Yachida S, Wood LD, Suzuki M, et al. Genomic sequencing identifies ELF3 as a driver of ampullary carcinoma. Cancer Cell. 2016; 29:229–40.
Article
12. Gingras MC, Covington KR, Chang DK, et al. Ampullary cancers harbor ELF3 tumor suppressor gene mutations and exhibit frequent WNT dysregulation. Cell Rep. 2016; 14:907–19.
Article
13. Lundgren S, Hau SO, Elebro J, et al. Mutational landscape in resected periampullary adenocarcinoma: relationship with morphology and clinical outcome. JCO Precis Oncol. 2019; 3:PO1800323.
Article
14. Perkins G, Svrcek M, Bouchet-Doumenq C, et al. Can we classify ampullary tumours better? Clinical, pathological and molecular features: results of an AGEO study. Br J Cancer. 2019; 120:697–702.
Article
15. Overman MJ, Soifer HS, Schueneman AJ, et al. Performance and prognostic utility of the 92-gene assay in the molecular subclassification of ampullary adenocarcinoma. BMC Cancer. 2016; 16:668.
Article
16. The Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012; 487:330–7.
17. Cancer Genome Atlas Research Network. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell. 2017; 32:185–203.
Full Text Links
  • JPTM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr