Skip to main content
Log in

Molekulare Testung und Liquid Biopsies bei kolorektalen Karzinomen

Molecular testing and liquid biopsies in colorectal cancer

  • Schwerpunkt
  • Published:
Die Gastroenterologie Aims and scope

Zusammenfassung

Hintergrund

Die Einführung neuer DNA-Sequenzierungstechnologien führte zur Entdeckung einer Vielzahl von prognostisch und prädiktiv relevanten Biomarkern in Tumorgewebe und Blut von Patienten mit kolorektalen Karzinom.

Ziel der Arbeit

Darstellung einer rationalen gewebebasierten molekularpathologischen Diagnostik und Diskussion des klinischen Anwendungsbereichs von zirkulierender Tumor-DNA (ctDNA) aus Flüssigbiopsien im kolorektalen Karzinom.

Material und Methoden

Auswertung der existierenden Literatur und Kongressveröffentlichungen, Diskussion von post-hoc-Analysen klinischer Studien und Expertenempfehlungen.

Ergebnisse

In den Stadien II und III gemäß Union for International Cancer Control (UICC) trägt die gewebebasierte Evaluation der Mikrosatelliteninstabilität (MSI-H) im Rahmen einer Beratung bezüglich einer adjuvanten Chemotherapie (Kolonkarzinom, UICC II) oder ggf. individueller neoadjuvanter Therapiekonzepte einschließlich der Anwendung von Immuntherapie (Kolon‑, Rektumkarzinom, UICC II/III) zur individuellen Therapieoptimierung bei. Aus Flüssigbiopsien ist der Nachweis von ctDNA mit einer minimalen Resterkrankung assoziiert, die die Wahrscheinlichkeit des krankheitsfreien Überlebens beeinflusst. Im metastasierten Stadium (UICC IV) sollte eine gewebebasierte Bestimmung von RAS- und BRAF-V600E-Mutationen, MSI‑H und perspektivisch auch einer HER2/neu-Überexpression erfolgen. Eine breitere molekulare Diagnostik zur Optimierung der Erstlinientherapie (molekulare Hyperselektion) zeigt einen nur geringen zusätzlichen Vorteil. Die ctDNA kann für eine longitudinale Verlaufsbeobachtung der klonalen Tumorevolution oder als Alternative zur invasiven Diagnostik eingesetzt werden.

Diskussion

Eine molekularpathologische Diagnostik aus Gewebe und Blut ergänzt sich komplementär und sollte entsprechend der zugrundeliegenden Fragestellung gezielt und sinnhaft eingesetzt werden.

Abstract

Background

The introduction of new DNA sequencing technologies has led to the discovery of many prognostically and predictively relevant biomarkers in tumor tissue and blood from patients with colorectal cancer.

Objectives

Presentation of meaningful tissue-based molecular pathological diagnostics and discussion of the clinical application of circulating tumor DNA (ctDNA) from liquid biopsies in colorectal cancer.

Materials and methods

Evaluation of existing literature and congress publications, discussion of post hoc analyses of clinical studies and expert recommendations.

Results

In Union for International Cancer Control (UICC) stages II/III, the tissue-based evaluation of microsatellite instability (MSI-H) contributes to individual therapy optimization through advice on adjuvant chemotherapy (colon cancer, UICC II) or, if necessary, individual neo-adjuvant therapy concepts via the use of immunotherapy (colon, rectal cancer, UICC II/III). From liquid biopsies, ctDNA was associated with minimal residual disease, which influences disease-free survival. In the metastatic stage (UICC IV), tissue-based determination of RAS and BRAF V600E mutations, MSI‑H and in the near future also HER2/neu overexpression should be performed. Broader molecular diagnostics to optimize first-line therapy (molecular hyperselection) shows little additional benefit. ctDNA can be used for longitudinal monitoring of clonal tumor evolution or as an alternative to invasive diagnostics in patients.

Conclusions

Molecular pathological diagnostics from tissue and blood complement each other and should be used in a targeted and meaningful way according to the underlying question.

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

Literatur

  1. Andre T, Elez E, Cutsem EV et al (2024) Nivolumab (NIVO) plus ipilimumab (IPI) vs chemotherapy (chemo) as first-line (1L) treatment for microsatellite instability-high/mismatch repair-deficient (MSI-H/dMMR) metastatic colorectal cancer (mCRC): First results of the CheckMate 8HW study. JCO 42:LBA768–LBA768

    Article  Google Scholar 

  2. Bettegowda C, Sausen M, Leary RJ et al (2014) Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 6:224ra224

    Article  Google Scholar 

  3. Bond MJG, Bolhuis K, Loosveld OJL et al (2023) First-line systemic treatment strategies in patients with initially unresectable colorectal cancer liver metastases (CAIRO5): an open-label, multicentre, randomised, controlled, phase 3 study from the Dutch Colorectal Cancer Group. Lancet Oncol 24:757–771

    Article  CAS  PubMed  Google Scholar 

  4. Cercek A, Lumish M, Sinopoli J et al (2022) PD‑1 blockade in mismatch repair-deficient, locally advanced rectal cancer. N Engl J Med 386:2363–2376

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Cervantes A, Adam R, Roselló S et al (2023) Metastatic colorectal cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up〈sup〉☆〈/sup〉. Ann Oncol 34:10–32

    Article  CAS  PubMed  Google Scholar 

  6. Chalabi M, Fanchi LF, Dijkstra KK et al (2020) Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat Med 26:566–576

    Article  CAS  PubMed  Google Scholar 

  7. Chan HT, Nagayama S, Otaki M et al (2022) Tumor-informed or tumor-agnostic circulating tumor DNA as a biomarker for risk of recurrence in resected colorectal cancer patients. Front Oncol 12:1055968

    Article  CAS  PubMed  Google Scholar 

  8. Cremolini C, Morano F, Moretto R et al (2017) Negative hyper-selection of metastatic colorectal cancer patients for anti-EGFR monoclonal antibodies: the PRESSING case-control study. Ann Oncol 28:3009–3014

    Article  CAS  PubMed  Google Scholar 

  9. De Roock W, Claes B, Bernasconi D et al (2010) Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol 11:753–762

    Article  CAS  PubMed  Google Scholar 

  10. Diaz LA Jr., Shiu KK, Kim TW et al (2022) Pembrolizumab versus chemotherapy for microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer (KEYNOTE-177): final analysis of a randomised, open-label, phase 3 study. Lancet Oncol 23:659–670

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Guidoboni M, Gafa R, Viel A et al (2001) Microsatellite instability and high content of activated cytotoxic lymphocytes identify colon cancer patients with a favorable prognosis. Am J Pathol 159:297–304

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Heinrich K, Miller-Phillips L, Ziemann F et al (2022) Lessons learned: the first consecutive 1000 patients of the CCCMunich(LMU) Molecular Tumor Board. J Cancer Res Clin Oncol

  13. Kopetz S, Grothey A, Yaeger R et al (2019) Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer. N Engl J Med 381:1632–1643

    Article  CAS  PubMed  Google Scholar 

  14. 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 

  15. Lonardi S, Pietrantonio F, Tarazona Llavero N et al (2023) LBA28 The PEGASUS trial: post-surgical liquid biopsy-guided treatment of stage III and high-risk stage II colon cancer patients. Ann Oncol 34:S1268–S1269

    Article  Google Scholar 

  16. Lonardi S, Rasola C, Lobefaro R et al (2023) Initial panitumumab plus fluorouracil, leucovorin, and oxaliplatin or plus fluorouracil and leucovorin in elderly patients with RAS and BRAF wild-type metastatic colorectal cancer: the PANDA trial by the GONO foundation. J Clin Oncol 41:5263–5273

    Article  CAS  PubMed  Google Scholar 

  17. Morton D, Seymour M, Magill L et al (2023) Preoperative chemotherapy for operable colon cancer: mature results of an international randomized controlled trial. J Clin Oncol 41:1541–1552

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Osumi H, Vecchione L, Keilholz U et al (2021) NeoRAS wild-type in metastatic colorectal cancer: myth or truth?-case series and review of the literature. Eur J Cancer 153:86–95

    Article  CAS  PubMed  Google Scholar 

  19. Pietrantonio F, Bergamo F, Rossini D et al (2023) Negative hyperselection of elderly patients with RAS and BRAF wild-type metastatic colorectal cancer receiving initial panitumumab plus FOLFOX or 5‑FU/LV. Eur J Cancer 195:113396

    Article  CAS  PubMed  Google Scholar 

  20. Rossini D, Antoniotti C, Lonardi S et al (2022) Upfront modified fluorouracil, leucovorin, oxaliplatin, and irinotecan plus panitumumab versus fluorouracil, leucovorin, and oxaliplatin plus panitumumab for patients with RAS/BRAF wild-type metastatic colorectal cancer: the phase III TRIPLETE study by GONO. J Clin Oncol 40:2878–2888

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sartore-Bianchi A, Pietrantonio F, Lonardi S et al (2022) Circulating tumor DNA to guide rechallenge with panitumumab in metastatic colorectal cancer: the phase 2 CHRONOS trial. Nat Med 28:1612–1618

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Shitara K, Muro K, Watanabe J et al (2024) Baseline ctDNA gene alterations as a biomarker of survival after panitumumab and chemotherapy in metastatic colorectal cancer. Nat Med

  23. Shitara K, Muro K, Watanabe J et al (2023) Negative hyperselection of patients with RAS wild-type metastatic colorectal cancer for panitumumab: a biomarker study of the phase III PARADIGM trial. JCO 41:11–11

    Article  Google Scholar 

  24. Siena S, Di Bartolomeo M, Raghav K et al (2021) Trastuzumab deruxtecan (DS-8201) in patients with HER2-expressing metastatic colorectal cancer (DESTINY-CRC01): a multicentre, open-label, phase 2 trial. Lancet Oncol 22:779–789

    Article  CAS  PubMed  Google Scholar 

  25. Siravegna G, Marsoni S, Siena S et al (2017) Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol 14:531–548

    Article  CAS  PubMed  Google Scholar 

  26. Stintzing S, Heinemann V, Weikersthal LFV et al (2023) Phase III FIRE-4 study (AIO KRK-0114): Influence of baseline liquid biopsy results in first-line treatment efficacy of FOLFIRI/cetuximab in patients with tissue RAS-WT mCRC. JCO 41:3507–3507

    Article  Google Scholar 

  27. Taieb J, Kourie HR, Emile JF et al (2018) Association of prognostic value of primary tumor location in stage III colon cancer with RAS and BRAF mutational status. JAMA Oncol 4:e173695

    Article  PubMed  Google Scholar 

  28. Tie J, Cohen JD, Lahouel K et al (2022) Circulating tumor DNA analysis guiding adjuvant therapy in stage II colon cancer. N Engl J Med 386:2261–2272

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Tie J, Wang Y, Tomasetti C et al (2016) Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci Transl Med 8:346r

    Article  Google Scholar 

  30. Van Cutsem E, Cervantes A, Adam R et al (2016) ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol 27:1386–1422

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Arndt Stahler.

Ethics declarations

Interessenkonflikt

A. Stahler: Beratertätigkeit: Bristol-Myers Squibb/Pfizer, Novocure, Takeda; Reisekosten: Amgen, Roche, Lilly, Pfizer; Honorare: Roche, Servier, Taiho Pharmaceutical, Takeda, Merck KGaA, Amgen, Daiichi Sankyo. S. Stintzing: Beratertätigkeit: Merck KGaA, Roche, Amgen, Pierre Fabre, MSD, AstraZeneca, SERVIER, GlaxoSmithKline, TERUMO, Nordic Bioscience, Seagen; Reisekosten: Merck KGaA, Roche, Sanofi, Bayer, Sirtex Medical, Amgen, Lilly, Takeda, Pierre Fabre, AstraZeneca; Honorare: Merck KGaA, Roche, Amgen, SERVIER, MSD, Pfizer, Pierre Fabre, Bristol-Myers Squibb, Nordic Bioscience, AstraZeneca; Forschungsförderung: Pierre Fabre, Roche Molecular Diagnostics, Merck Serono, Amgen.

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

Additional information

Redaktion

Ralf Jakobs, Ludwigshafen

Thomas Seufferlein, Ulm

Hinweis des Verlags

Der Verlag bleibt in Hinblick auf geografische Zuordnungen und Gebietsbezeichnungen in veröffentlichten Karten und Institutsadressen neutral.

figure qr

QR-Code scannen & Beitrag online lesen

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Stahler, A., Stintzing, S. Molekulare Testung und Liquid Biopsies bei kolorektalen Karzinomen. Gastroenterologie (2024). https://doi.org/10.1007/s11377-024-00797-5

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s11377-024-00797-5

Schlüsselwörter

Keywords

Navigation