Skip to main content

Advertisement

Log in

Histo-molecular characterization of pancreatic cancer with microsatellite instability: intra-tumor heterogeneity, B2M inactivation, and the importance of metastatic sites

  • Brief Report
  • Published:
Virchows Archiv Aims and scope Submit manuscript

Abstract

Pancreatic ductal adenocarcinoma (PDAC) with microsatellite instability (MSI)/defective mismatch repair (dMMR) is the only subtype of pancreatic cancer with potential response to immunotherapy. Here, we report the histo-molecular characterization of MSI/dMMR PDAC with immunohistochemistry, MSI-based PCR, and next-generation sequencing. Five paradigmatic cases have been identified. The main results include the first report in pancreatic cancer of MSI/dMMR intra-tumor heterogeneity, the presence of microsatellite-stable metastases from MSI/dMMR primary and recurrent B2M gene inactivation, which may confer resistance to immunotherapy. In addition to the classic PDAC drivers, ARID1A was the most common mutated gene in the cohort. Intra-tumor heterogeneity, B2M inactivation, and metastatic sites should be carefully considered in MSI/dMMR PDAC, which should also be investigated in routine diagnostic practice with specific molecular analysis. The chromatin remodeler ARID1A represents another potential driver gene in this context.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Data availability

All data/information are available in the manuscript and in the supplementary material.

References

  1. Le DT, Uram JN, Wang H et al (2015) PD-1 Blockade in tumors with mismatch-repair deficiency. N Engl J Med 372:2509–2520. https://doi.org/10.1056/NEJMoa1500596

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Le DT, Durham JN, Smith KN et al (2017) Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357:409–413. https://doi.org/10.1126/science.aan6733

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Luchini C, Bibeau F, Ligtenberg MJL et al (2019) ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: a systematic review-based approach. Ann Oncol 30:1232–1243. https://doi.org/10.1093/annonc/mdz116

    Article  CAS  PubMed  Google Scholar 

  4. Luchini C, Brosens LAA, Wood LD et al (2021) Comprehensive characterisation of pancreatic ductal adenocarcinoma with microsatellite instability: histology, molecular pathology and clinical implications. Gut 70:148–156. https://doi.org/10.1136/gutjnl-2020-320726

    Article  CAS  PubMed  Google Scholar 

  5. Grant RC, Denroche R, Jang GH et al (2020) Clinical and genomic characterisation of mismatch repair deficient pancreatic adenocarcinoma. Gut. https://doi.org/10.1136/gutjnl-2020-320730

    Article  PubMed  Google Scholar 

  6. Luchini C, Grant RC, Scarpa A, Gallinger S (2021) Microsatellite instability/mismatch repair deficiency in pancreatic cancers: the same or different? Gut. https://doi.org/10.1136/gutjnl-2020-323805

    Article  PubMed  Google Scholar 

  7. Marabelle A, Le DT, Ascierto PA et al (2019) Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair–deficient cancer: results from the phase II KEYNOTE-158 Study. JCO 38:1–10. https://doi.org/10.1200/JCO.19.02105

    Article  Google Scholar 

  8. Mafficini A, Lawlor RT, Ghimenton C et al (2021) Solid pseudopapillary neoplasm of the pancreas and abdominal desmoid tumor in a patient carrying two different BRCA2 germline mutations: new horizons from tumor molecular profiling. Genes 12:481. https://doi.org/10.3390/genes12040481

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Papke DJ, Nowak JA, Yurgelun MB et al (2018) Validation of a targeted next-generation sequencing approach to detect mismatch repair deficiency in colorectal adenocarcinoma. Mod Pathol 31:1882–1890. https://doi.org/10.1038/s41379-018-0091-x

    Article  CAS  PubMed  Google Scholar 

  10. Ahdesmäki MJ, Chapman BA, Cingolani P et al (2017) Prioritisation of structural variant calls in cancer genomes. PeerJ 5:e3166. https://doi.org/10.7717/peerj.3166

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Richards S, Aziz N, Bale S et al (2015) Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med 17:405–423. https://doi.org/10.1038/gim.2015.3012

    Article  PubMed  PubMed Central  Google Scholar 

  12. Lupinacci RM, Goloudina A, Buhard O et al (2018) Prevalence of microsatellite instability in intraductal papillary mucinous neoplasms of the pancreas. Gastroenterology 154:1061–1065. https://doi.org/10.1053/j.gastro.2017.11.009

    Article  CAS  PubMed  Google Scholar 

  13. Kim ST, Cristescu R, Bass AJ et al (2018) Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat Med 24:1449–1458. https://doi.org/10.1038/s41591-018-0101-z

    Article  CAS  PubMed  Google Scholar 

  14. Loupakis F, Maddalena G, Depetris I et al (2019) Treatment with checkpoint inhibitors in a metastatic colorectal cancer patient with molecular and immunohistochemical heterogeneity in MSI/dMMR status. J Immunother Cancer 7:297. https://doi.org/10.1186/s40425-019-0788-5

    Article  PubMed  PubMed Central  Google Scholar 

  15. Sade-Feldman M, Jiao YJ, Chen JH et al (2017) Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat Commun 8:1136. https://doi.org/10.1038/s41467-017-01062-w

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Middha S, Yaeger R, Shia J et al (2019) Majority of B2M-mutant and -deficient colorectal carcinomas achieve clinical benefit from immune checkpoint inhibitor therapy and are microsatellite instability-high. JCO Precis Oncol 3:PO.18.00321. https://doi.org/10.1200/PO.18.00321

    Article  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study was supported by Associazione Italiana Ricerca sul Cancro (AIRC 5x1000 n. 12182); Fondazione Cariverona: Oncology Biobank Project “Antonio Schiavi” (prot. 203885/2017); Fondazione Italiana Malattie Pancreas (FIMP-Ministero Salute J38D19000690001); and Italian Ministry of Health (RF CO-2019-12369662: CUP: B39C21000370001).

Author information

Authors and Affiliations

Authors

Contributions

CL: study conception and design; CL, DC, PN, VNA, AS: histological examination; CL, AM, GP, PM, RTL, AS: immunohistochemical analysis; CL, AM, LP, CS, RTL, AS: molecular analysis; CL, GMal, GMar, AP, RS, MM, AS: clinic-pathologic data; all authors: data elaboration and interpretation; CL, AM, RTL, AS: paper writing; all authors: final editing and approval of the present version.

Corresponding authors

Correspondence to Claudio Luchini or Aldo Scarpa.

Ethics declarations

Ethics approval

This study was approved by the Verona Ethics Committee, date of approval: 04–08-2020, project 2610-CESC, code: MN-2019.

Conflict of interest

The authors declare no competing interests.

Additional information

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Supplementary Figure 1.

The different components of the primary tumor of case #1, including low grade IPMN (A), high grade IPMN (B), ductal adenocarcinoma (C), and colloid carcinoma (D) (Hematoxylin-eosin, 20x magnification). (PNG 687 kb)

High resolution image (TIF 3320 kb)

Supplementary Figure 2.

A representative histological / immunohistochemical image of the primary tumor of case #1. The histology clearly shows an IPMN with an associated adenocarcinoma. All tumor components show immunohistochemical loss of MSH2 and MSH6, and retained expression of MLH1 and PMS2 (10x magnification; A: hematoxylin-eosin; B: MSH2 loss; C: MLH1 retained expression; D: PMS2 retained expression; E: MSH6 loss). (PNG 664 kb)

High resolution image (TIF 2977 kb)

Supplementary Figure 3.

A representative image of case #2. The histology shows a medullary carcinoma. At immunohistochemistry, tumor cells exhibited MSH2, and MSH6 loss, and retained expression of MLH1 and PMS2 (10x magnification; A: hematoxylin-eosin; B: MSH2 loss; C: MLH1 retained expression; D: PMS2 retained expression; E: MSH6 loss). (PNG 656 kb)

High resolution image (TIF 2748 kb)

Supplementary Figure 4.

Graphical representation of two heterozygous frameshift mutations, each affecting a different copy of the B2M gene, in a lymph node metastasis of medullary carcinoma of the pancreas (case no. 2). The mutational data supports biallelic inactivation of the gene. Of note, the primary tumor also harbored B2M inactivation, but this was due to homozygous mutation. (PNG 106 kb)

High resolution image (TIF 132 kb)

Supplementary Figure 5.

A representative histological / immunohistochemical image of the primary tumor of case #3. The histology shows a biphasic neoplasm, displaying medullary (A), and ductal (B) features (hematoxylin-eosin, A: 10x magnification, B: 20x magnification). To illustrate the immunohistochemical profile, an area with both components (medullary on the left, ductal on the right) was selected: MSH6 loss of expression is limited to the medullary component (C, 10x magnification, with retained expression in intra-tumor lymphocytes); conversely MSH2 (D, 10x magnification), MLH1 (E, 10x magnification), and PMS2 (F,10x magnification) show retained expression in both components. (PNG 1647 kb)

High resolution image (TIF 2474 kb)

Supplementary Figure 6.

Graphical representation of two heterozygous mutations, each affecting a different copy of the B2M gene (exon 1) in nodal metastases of a mixed ductal/medullary carcinoma of the pancreas (case no. 3). A) One mutation causes a frameshift and thus a truncated protein. The second mutation causes an amino acid change at codon 8 from Ala to Asp, which was further investigated. B) Analysis of the conservation of amino acids of B2M exon 1 among primates. Codon 8 is one of the most conserved residues, residing in a stretch of hydrophobic amino acids (purple color). The sequence from residues 1 to 20 is a signal peptide that targets B2M gene product to the endoplasmic reticulum, where it joins the other components of the major histocompatibility complex I. C) Analysis of the change in hydrophobicity (hydropathy) of the leader peptide due to the mutation of Ala at codon 8 to Asp. Dots represent hydropathy scores of single residues, lines illustrate local hydropathy computed on a window of three amino acids, accounting for interactions between side chains. The hydrophobicity loss of signal peptides has been shown to inhibit translocation of proteins. D) Prediction of the recognition of B2M exon1 as a signal peptide upon mutation of Ala 8 to Asp, performed with the SignalP 5.0 software. The prediction score drops from 0.99 to 0.80, showing a damaging effect of the amino acid substitution. (PNG 158 kb)

High resolution image (TIF 181 kb)

Supplementary Figure 7.

A representative histological/immunohistochemical image of case #5. Histologically, this is a conventional pancreatic ductal adenocarcinoma with a very small, metastatic lymph node (A, Hematoxylin-eosin, 4x magnification). At immunohistochemistry, MSH2 (B, 10x magnification), and MSH6 (C, 10x magnification) show retained expression, while MLH1 (D, 10x magnification), and PMS2 (E, 10x magnification) show loss of expression. (PNG 526 kb)

High resolution image (TIF 2326 kb)

Supplementary Table 1

(PDF 42 kb)

Supplementary Table 2

(DOCX 13 kb)

Supplementary Table 3

(DOCX 35 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Luchini, C., Mafficini, A., Chatterjee, D. et al. Histo-molecular characterization of pancreatic cancer with microsatellite instability: intra-tumor heterogeneity, B2M inactivation, and the importance of metastatic sites. Virchows Arch 480, 1261–1268 (2022). https://doi.org/10.1007/s00428-021-03205-3

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00428-021-03205-3

Keywords

Navigation