Skip to main content
Log in

MSI-Testung

Was ist neu? Was ist zu beachten?

MSI testing

What is new? What should be considered? German version

  • Übersichten
  • Published:
Der Pathologe Aims and scope Submit manuscript

Zusammenfassung

Aufgrund neuer Studiendaten mit Immuncheckpoint-Inhibitoren (ICI) gewinnt der Nachweis einer hochgradigen Mikrosatelliteninstabilität (MSI-H) bzw. der zugrunde liegenden Mismatch-Repair-Protein-Defizienz (dMMR) nun auch zur Prädiktion von Therapieansprechen zunehmend an Bedeutung. Aktuell wurde von der Europäischen Medizinischen Agentur (EMA) erstmals ein PD-1-ICI (Pembrolizumab) für die Erstlinientherapie des fortgeschrittenen (Stadium IV) dMMR/MSI‑H kolorektalen Karzinoms (KRK) zugelassen. Weitere Indikationen, wie z. B. für das metastastasierte dMMR/MSI-H-Endometriumkarzinom (EK) in der Zweitlinientherapie (Dostarlimab) sind bereits gefolgt, andere werden noch im laufenden Jahr 2021 erwartet. Dies erfordert eine Neubewertung der Frage nach der optimalen Testung in der Routinediagnostik. Auf Basis einer Abwägung von Stärken und Schwächen der in der Breite zur Verfügung stehenden Methoden (Immunhistochemie und PCR) wird ein Testalgorithmus vorgeschlagen, der qualitätsgesichert eine zuverlässige und kosteneffektive dMMR/MSI-H-Testung erlaubt. Für das KRK und EK ist damit analog internationaler Empfehlungen (NICE, NCCN) eine Testung bereits bei der Primärdiagnose möglich. Somit wird der Kliniker von vornherein in die Lage versetzt, neben den prädiktiven auch die prognostischen und prädispositionsbedingten Implikationen eines solchen Tests bei der Beratung von Patienten und Erarbeitung von Therapieempfehlungen zu berücksichtigen. Als Grundlage der Qualitätssicherung wird die Teilnahme an Ringversuchen und eine fortlaufende Ergebnisdokumentation (z. B. QuIP-Monitor) empfohlen.

Abstract

Based on new trial data regarding immune checkpoint inhibitors (ICIs), the detection of high-grade microsatellite instability (MSI-H) or underlying deficient mismatch repair protein (dMMR) is now becoming increasingly important for predicting treatment response. For the first time, a PD‑1 ICI (pembrolizumab) has been approved by the European Medicines Agency (EMA) for first-line treatment of advanced (stage IV) dMMR/MSI‑H colorectal cancer (CRC). Further indications, such as dMMR/MSI‑H endometrial carcinoma (EC), have already succeeded (Dostarlimab, 2nd line treatment) and others are expected to follow before the end of 2021. The question of optimal testing in routine diagnostics should therefore be re-evaluated. Based on a consideration of the strengths and weaknesses of the widely available methods (immunohistochemistry and PCR), a test algorithm is proposed that allows quality assured, reliable, and cost-effective dMMR/MSI‑H testing. For CRC and EC, testing is therefore already possible at the primary diagnosis stage, in line with international recommendations (NICE, NCCN). The clinician is therefore enabled from the outset to consider not only the predictive but also the prognostic and predispositional implications of such a test when counseling patients and formulating treatment recommendations. As a basis for quality assurance, participation in interlaboratory comparisons and continuous documentation of results (e.g., QuIP Monitor) are strongly recommended.

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.

Abb. 1
Abb. 2
Abb. 3

Literatur

  1. André T, Shiu KK, Kim TW et al (2020) Pembrolizumab in microsatellite-instability-high advanced colorectal cancer. N Engl J Med 383:2207–2218

    Article  PubMed  Google Scholar 

  2. AWMF (2018) S3-Leitlinie Diagnostik, Therapie und Nachsorge der Patientinnen mit Endometriumkarzinom (AWMF-Registernummer: 032/034-OL)

    Google Scholar 

  3. AWMF (2019) S3-Leitlinie Kolorektales Karzinom (AWMF-Registernummer: 021/007OL)

    Google Scholar 

  4. Bass BP, Engel KB, Greytak SR, Moore HM (2014) A review of preanalytical factors affecting molecular, protein, and morphological analysis of formalin-fixed, paraffin-embedded (FFPE) tissue: how well do you know your FFPE specimen? Arch Pathol Lab Med 138(11):1520–1530

    Article  PubMed  Google Scholar 

  5. Bläker H, Haupt S, Morak M et al (2020) Age-dependent performance of BRAF mutation testing in Lynch syndrome diagnostics. Int J Cancer 147:2801–2810

    Article  PubMed  CAS  Google Scholar 

  6. Britton H, Huang L, Lum A et al (2019) Molecular classification defines outcomes and opportunities in young women with endometrial carcinoma. Gynecol Oncol 153:487–495

    Article  PubMed  Google Scholar 

  7. Buhard O, Suraweera N, Lectard A et al (2004) Quasimonomorphic mononucleotide repeats for high-level microsatellite instability analysis. Dis Markers 20:251–257

    Article  PubMed  PubMed Central  Google Scholar 

  8. Cancer Genome Atlas Network (2012) Comprehensive molecular characterization of human colon and rectal cancer. Nature 487:330–337

    Article  CAS  Google Scholar 

  9. Cancer Genome Atlas Research Network (2014) Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513:202–209

    Article  CAS  Google Scholar 

  10. Cancer Genome Atlas Research Network, Kandoth C, Schultz N, Cherniack AD et al (2013) Integrated genomic characterization of endometrial carcinoma. Nature 497:67–73

    Article  CAS  Google Scholar 

  11. Cancer Genome Atlas Research Network, Weinstein JN, Collisson EA, Mills GB et al (2013) The cancer genome atlas pan-cancer analysis project. Nat Genet 45:1113–1120

    Article  PubMed Central  CAS  Google Scholar 

  12. Carethers JM (2017) Microsatellite instability pathway and EMAST in colorectal cancer. Curr Colorectal Cancer Rep 13:73–80

    Article  PubMed  PubMed Central  Google Scholar 

  13. Casey L, Singh N (2021) POLE, MMR, and MSI testing in endometrial cancer: proceedings of the ISGyP companion society session at the USCAP 2020 annual meeting. Int J Gynecol Pathol 40(1):5–16

    Article  PubMed  Google Scholar 

  14. Chen W, Frankel WL (2019) A practical guide to biomarkers for the evaluation of colorectal cancer. Mod Pathol 32:S1–S15

    Article  CAS  Google Scholar 

  15. Cho KR, Cooper K, Croce S et al (2019) International society of gynecological pathologists (ISGyP) endometrial cancer project: guidelines from the special techniques and ancillary studies group. Int J Gynecol Pathol 38(1):S114–S122

    Article  PubMed  Google Scholar 

  16. Darvin P, Toor SM, Sasidharan Nair V, Elkord E (2018) Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med 50:1–11

    Article  PubMed  CAS  Google Scholar 

  17. Devaraj B et al (2010) Relationship of EMAST and microsatellite instability among patients with rectal cancer. J Gastrointest Surg. https://doi.org/10.1007/s11605-010-1340-6

    Article  PubMed  PubMed Central  Google Scholar 

  18. Dietmaier W, Büttner R, Rüschoff J (2019) Mikrosatelliteninstabilität: Aktueller Überblick über Methoden und Anwendungen. Pathologe 40(3):313–327

    Article  CAS  PubMed  Google Scholar 

  19. Dominguez-Valentin M et al (2020) Cancer risks by gene, age, and gender in 6350 carriers of pathogenic mismatch repair variants. Genet Med 22(1):15–25

    Article  CAS  PubMed  Google Scholar 

  20. Engel C, Forberg J, Holinski-Feder E et al (2006) Novel strategy for optimal sequential application of clinical criteria, immunohistochemistry and microsatellite analysis in the diagnosis of hereditary nonpolyposis colorectal cancer. Int J Cancer 118:115–122

    Article  CAS  PubMed  Google Scholar 

  21. Green AK, Feinberg J, Makker V (2020) A review of immune checkpoint blockade therapy in endometrial cancer. Am Soc Clin Oncol Educ Book 40:1–7

    PubMed  Google Scholar 

  22. Hayashi H, Nakagawa K (2020) Combination therapy with PD‑1 or PD-L1 inhibitors for cancer. Int J Clin Oncol 25(5):818–830

    Article  CAS  PubMed  Google Scholar 

  23. Horn LC, Emons G, Aretz S et al (2019) S3-Leitlinie Diagnostik und Therapie des Endometriumkarzinoms: Anforderungen an die Pathologie. Pathologe 40:21–35

    Article  PubMed  Google Scholar 

  24. https://www.clincaltrials.gov. Zugegriffen: 03.05.2021

  25. Iivanainen S, Koivunen JP (2020) Possibilities of improving the clinical value of immune checkpoint inhibitor therapies in cancer care by optimizing patient selection. Int J Mol Sci 21:556

    Article  CAS  PubMed Central  Google Scholar 

  26. Jöhrens K, Dietmaier W, Utpatel K et al (2021) Qualitätssicherung in der MMRD und MSI Diagnostik. Pathologe, im Druck

    Google Scholar 

  27. Joost P et al (2014) Heterogenous mismatch-repair status in colorectal cancer. Diagn Pathol 9:126

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Kim J, Park WY, Kim NKD et al (2017) Good laboratory standards for clinical next-generation sequencing cancer panel tests. J Pathol Transl Med 51:191–204

    Article  PubMed  PubMed Central  Google Scholar 

  29. Kloth M, Ruesseler V, Engel C et al (2016) Activating ERBB2/HER2 mutations indicate susceptibility to pan-HER inhibitors in Lynch and Lynch-like colorectal cancer. Gut 65:1296–1305

    Article  CAS  PubMed  Google Scholar 

  30. Latham A, Srinivasan P, Kemel Y et al (2019) Microsatellite instability is associated with the presence of Lynch syndrome pan-cancer. J Clin Oncol 37:286–295

    Article  CAS  PubMed  Google Scholar 

  31. 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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Le DT, Kim TW, Van Cutsem E et al (2020) Phase II open-label study of pembrolizumab in treatment-refractory, microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: keynote-164. J Clin Oncol 38:11–19

    Article  CAS  PubMed  Google Scholar 

  33. 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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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

    Article  CAS  PubMed  Google Scholar 

  35. Marabelle A, Le DT, Ascierto PA et al (2020) Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II keynote-158 study. J Clin Oncol 38:1–10

    Article  CAS  PubMed  Google Scholar 

  36. Mills AM, Liou S, Ford JM et al (2014) Lynch syndrome screening should be considered for all patients with newly diagnosed endometrial cancer. Am J Surg Pathol 38:1501–1509

    Article  PubMed  PubMed Central  Google Scholar 

  37. Mojtahed A, Schrijver I, Ford JM et al (2011) A two-antibody mismatch repair protein immunohistochemistry screening approach for colorectal carcinomas, skin sebaceous tumors, and gynecologic tract carcinomas. Mod Pathol 24:1004–1014

    Article  PubMed  Google Scholar 

  38. National Institute for Health and Care Excellence (NICE) (2021) NICE guidance on molecular testing strategies for Lynch syndrome in people with colorectal cancer. https://www.bsg.org.uk/clinical-resource/nice-guidance-on-molecular-testing-strategies-for-lynch-syndrome-in-people-with-colorectal-cancer/#:~:text=NICE%20recommends%20that%20microsatellite%20instability,the%20presence%20of%20Lynch%20Syndrome.Andhttps://www.nice.org.uk/guidance/dg42/chapter/3-Evidence. Zugegriffen: 21. Jan. 2021

  39. NCCN (2021) https://www.nccn.org/professionals/physician_gls/pdf/colon.pdf. Zugegriffen: 22. Jan. 2021

  40. Niu BT, Hammond RFL, Leen SLS, Gilks CB, Singh N (2019) Two versus four immunostains for Lynch syndrome screening in endometrial carcinoma. Histopathology 75:442–445

    Article  PubMed  Google Scholar 

  41. Oaknin A, Gilbert L, Tinker AV et al (2020) LBA36—safety and antitumor activity of dostarlimab in patients (pts) with advanced or recurrent DNA mismatch repair deficient (dMMR) or proficient (MMRp) endometrial cancer (EC): results from garnet. Ann Oncol 31(4):S1142–S1215. https://doi.org/10.1016/annonc/annonc325

    Article  Google Scholar 

  42. Overman MJ, Lonardi S, Wong KYM et al (2018) Durable clinical benefit with nivolumab plus Ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J Clin Oncol 36:773–779

    Article  CAS  PubMed  Google Scholar 

  43. Pearlman R, Markow M, Knight D et al (2018) Two-stain immunohistochemical screening for Lynch syndrome in colorectal cancer may fail to detect mismatch repair deficiency. Mod Pathol 31:1891–1900

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Peltomäki P (2016) Update on Lynch syndrome genomics. Fam Cancer 15:385–393

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Ryan NAJ, McMahon R, Tobi S et al (2020) The proportion of endometrial tumours associated with lynch syndrome (petals): a prospective cross-sectional study. PLoS Med 17:e1003263

    Article  PubMed  PubMed Central  Google Scholar 

  46. Shia J, Tang LH, Vakiani E et al (2009) Immunohistochemistry as first-line screening for detecting colorectal cancer patients at risk for hereditary nonpolyposis colorectal cancer syndrome: a 2-antibody panel may be as predictive as a 4-antibody panel. Am J Surg Pathol 33:1639–1645

    Article  PubMed  Google Scholar 

  47. Siemanowski J, Schömig-Markiefka B, Buhl T et al (2021) Managing difficulties of microsatellite instability testing in endometrial cancer-limitations and advantages of four different PCR based approaches. Cancers 13(6):1268. https://doi.org/10.3390/cancers13061268

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sohn BH et al (2017) Clinical significance of four molecular subtypes of gastric cancer identified by the cancer genome atlas project. Clin Cancer Res 23:4441–4449

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Soslow RA, Tornos C, Park KJ et al (2019) Endometrial carcinoma diagnosis: use of FIGO grading and genomic subcategories in clinical practice: recommendations of the international society of gynecological pathologists. Int J Gynecol Pathol 38(1):S64–S74

    Article  PubMed  Google Scholar 

  50. Stelloo E, Jansen AML, Osse EM et al (2017) Practical guidance for mismatch repair-deficiency testing in endometrial cancer. Ann Oncol 28:96–102

    Article  CAS  PubMed  Google Scholar 

  51. Talhouk A, McConechy MK, Leung S et al (2017) Confirmation of ProMisE: a simple, genomics-based clinical classifier for endometrial cancer. Cancer 123:802–813

    Article  CAS  PubMed  Google Scholar 

  52. Xicola RM, Llor X, Pons E et al (2007) Performance of different microsatellite marker panels for detection of mismatch repair-deficient colorectal tumors. J Natl Cancer Inst 99:244–252

    Article  CAS  PubMed  Google Scholar 

  53. Yang G, Zheng R‑Y, Jin Z‑S (2019) Correlations between microsatellite instability and the biological behaviour of tumors. J Cancer Res Clin Oncol 145:2891–2899

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Josef Rüschoff.

Ethics declarations

Interessenkonflikt

S. Merkelbach-Bruse: Honorar für Vorträge und Advisory Boards von AstraZeneca, Roche, Novartis, GSK, MSD, Targos, Molecular Health und Merck. R. Büttner: Honorar für Vorträge und Advisory Boards von AbbVie, AstraZeneca, Bayer, BMS, Boehringer-Ingelheim, Illumina, Lilly, MSD, Novartis, Qiagen, Pfizer und Roche. Mitgründer und Scientific Advisor von Targos Mol. Path. Gmbh. A. Hartmann: Honorar für Vorträge und Advisory Boards von Abbvie, AstraZeneca, Biontech, BMS, Boehringer Ingelheim, Cepheid, Diaceutics, Illumina, Ipsen, Janssen, MSD, Nanostring, Novartis, Qiagen, QUIP GmbH, Roche und 3DHistotech. L.-C. Horn: Honorar für Vorträge und Advisory Boards von AstraZeneca, Roche, Dako, Novartis, Exact Sciences und GSK. D. Mayr: Honorar für Vorträge und Advisory Boards von AstraZeneca, MSD, Roche, Novartis und BMS. R. Knüchel: Honorar für Vorträge und Advisory Board von MSD. J. Rüschoff: Honorar für Vorträge und Advisory Boards von MSD/Merck, GSK, BMS, AstraZeneca, Roche, Exact Sciences und QuIP GmbH. Mitgründer und medizin. Leiter von Targos Mol. Path. Gmbh. H.-U. Schildhaus: Honorar für Vorträge und Advisory Boards von MSD, BMS, Pfizer, Novartis Oncology, Roche Pharma und Molecular Health. Angestellter von Targos Mol. Path. Gmbh. W. Weichert: Honorar für Vorträge und Advisory Boards von Roche, MSD, BMS, AstraZeneca, Pfizer, Merck, Lilly, Boehringer, Novartis, Takeda, Bayer, Amgen, Astellas, Illumina, Siemens, Agilent, GSK, ADC und Molecular Health. M. Tiemann: Honorar für Vorträge und Advisory Boards von BMS und MSD. G. Baretton, M. Dietel, K. Jöhrens, H. Bläker, W. Dietmaier, T. Kirchner, P. Schirmacher und K. Tiemann geben an, dass kein Interessenkonflikt besteht.

Für diesen Beitrag wurden von den Autoren keine Studien an Menschen oder Tieren durchgeführt. Für die aufgeführten Studien gelten die jeweils dort angegebenen ethischen Richtlinien.

Additional information

Redaktion

W. Roth, Mainz

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rüschoff, J., Baretton, G., Bläker, H. et al. MSI-Testung. Pathologe 42, 414–423 (2021). https://doi.org/10.1007/s00292-021-00944-7

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00292-021-00944-7

Schlüsselwörter

Keywords

Navigation