Skip to main content
Log in

Einfluss der Molekularpathologie auf die onkologische Chirurgie von Tumoren des oberen Gastrointestinaltraktes

Influence of molecular pathology on oncological surgery of tumors of the upper gastrointestinal tract

  • Leitthema
  • Published:
Der Chirurg Aims and scope Submit manuscript

Zusammenfassung

Hintergrund

In jüngster Vergangenheit konnten die Tumoren des oberen Gastrointestinaltraktes (ösophageales Plattenepithelkarzinom [ESCC], ösophageales Adenokarzinom [EAC], gastrales Adenokarzinom) zunehmend molekulargenetisch charakterisiert werden. Die resultierenden Subtypen bieten hierbei gemäß ihren biologischen Eigenschaften mögliche Angriffspunkte für zielgerichtete Therapien.

Ziel der Arbeit

Es wird eine Übersicht über den aktuellen Stand der molekularen Subtypen aller drei Tumorentitäten vermittelt und hiervon abgeleitete individualisierte Therapiestrategien diskutiert.

Material und Methoden

Es wurde eine selektive Literaturrecherche insbesondere hinsichtlich der bestehenden molekularen Subtypisierung, der derzeitig bestehenden „targeted therapies“ und der Auswirkung auf die onkologischen Therapiekonzepte durchgeführt. Dabei wurden Datenbanken wie PubMed genutzt.

Ergebnisse

Anhand der molekularen Charakteristika wird das ESCC in drei Subtypen kategorisiert. Genetisch ähnelt hierbei zumindest eine Subgruppe eher den Oropharynxkarzinomen als denjenigen des oberen Gastrointestinal(GI)-Traktes. Das EAC zeichnet sich dagegen durch seine hohe Tumorlast und die große chromosomale Instabilität aus. Beim Magenkarzinom werden zudem vier Subtypen unterschieden: 1. Epstein-Barr-Virus(EBV)-positive, 2. mikrosatelliteninstabile (MSI), 3. genomisch stabile (GS) und 4. chromosomal instabile (CIN) Tumoren.

Schlussfolgerung

Bisher konnten anhand der genetischen Charakterisierung nur wenige zielgerichtete Therapieoptionen für die drei Tumorentitäten in die tägliche Routine überführt werden. Hierzu gehören die selektive ERBB2-Inhibition, die Immuntherapie mittels PD-L1-Inhibition oder kombinierte Blockade von ERBB2/VEGF.

Abstract

Background

In the recent past tumors of the upper gastrointestinal (GI) tract, e.g. esophageal squamous cell carcinoma (ESCC), esophageal adenocarcinoma (EAC) and gastric adenocarcinoma, have been increasingly characterized by molecular genetics. The resulting subtypes provide potential targets for novel targeted treatment based on the biological characteristics.

Objective

This article summarizes the current state of molecular subtyping in all three tumor entities and the individualized treatment strategies derived from these categories are discussed.

Material and methods

A selective literature search was performed focusing on molecular subtyping, current targeted treatment and the impact on the oncological treatment concepts. Databases such as PubMed were used.

Results

Based on molecular characteristics ESCC is nowadays categorized into three subtypes. Genetically, at least one subgroup resembles oropharyngeal carcinomas rather than those of the upper GI tract. In contrast, EAC is characterized by its high tumor burden and large chromosomal instability. In gastric carcinoma 4 subtypes are distinguished: 1) Epstein-Barr virus (EBV) positive, 2) microsatellite instability (MSI), 3) genomically stable (GS) and 4) chromosomally unstable (CIN) tumors.

Conclusion

To date, only a few targeted therapeutic options have been successfully transferred into daily routine derived from the genetic characterization of the three tumor entities. These include selective ERBB2 inhibition, immunotherapy using PD-L1 inhibition or combined blockade of ERBB2/VEGF.

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. Al-Batran S‑E, Hartmann JT, Hofheinz R et al (2008) Biweekly fluorouracil, leucovorin, oxaliplatin, and docetaxel (FLOT) for patients with metastatic adenocarcinoma of the stomach or esophagogastric junction: a phase II trial of the Arbeitsgemeinschaft Internistische Onkologie. Ann Oncol 19:1882–1887. https://doi.org/10.1093/annonc/mdn403

    Article  PubMed  Google Scholar 

  2. Arnold M, Soerjomataram I, Ferlay J, Forman D (2015) Global incidence of oesophageal cancer by histological subtype in 2012. Gut 64:381–387. https://doi.org/10.1136/gutjnl-2014-308124

    Article  PubMed  Google Scholar 

  3. Bandla S, Pennathur A, Luketich JD et al (2012) Comparative genomics of esophageal adenocarcinoma and squamous cell carcinoma. Ann Thorac Surg 93:1101–1106. https://doi.org/10.1016/j.athoracsur.2012.01.064

    Article  PubMed  PubMed Central  Google Scholar 

  4. Bang YJ, Van Cutsem E, Feyereislova A et al (2010) Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet 376:687–697. https://doi.org/10.1016/S0140-6736(10)61121-X

    Article  CAS  PubMed  Google Scholar 

  5. Bass AJ, Thorsson V, Shmulevich I et al (2014) Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513:202–209. https://doi.org/10.1038/nature13480

    Article  CAS  Google Scholar 

  6. Cancer Genome Atlas Research Network, Analysis Working Group: Asan University, BC Cancer Agency et al (2017) Integrated genomic characterization of oesophageal carcinoma. Nature 541:169–175. https://doi.org/10.1038/nature20805

    Article  CAS  Google Scholar 

  7. Choi IJ, Kook M‑C, Kim Y‑I et al (2018) Helicobacter pylori therapy for the prevention of metachronous gastric cancer. N Engl J Med 378:1085–1095. https://doi.org/10.1056/nejmoa1708423

    Article  CAS  PubMed  Google Scholar 

  8. Coleman HG, Xie SH, Lagergren J (2018) The epidemiology of esophageal adenocarcinoma. Gastroenterology 154:390–405. https://doi.org/10.1053/j.gastro.2017.07.046

    Article  PubMed  Google Scholar 

  9. Contino G, Vaughan TL, Whiteman D, Fitzgerald RC (2017) The evolving genomic landscape of Barrett’s esophagus and esophageal adenocarcinoma. Gastroenterology 153:657–673.e1. https://doi.org/10.1053/j.gastro.2017.07.007

    Article  PubMed  Google Scholar 

  10. Dulak AM, Stojanov P, Peng S et al (2013) Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity. Nat Genet 45:478–486. https://doi.org/10.1038/ng.2591

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Fatehi Hassanabad A, Chehade R, Breadner D, Raphael J (2020) Esophageal carcinoma: towards targeted therapies. Cell Oncol (Dordr) 43:195–209. https://doi.org/10.1007/s13402-019-00488-2

    Article  Google Scholar 

  12. Frankell AM, Jammula S, Li X et al (2019) The landscape of selection in 551 esophageal adenocarcinomas defines genomic biomarkers for the clinic. Nat Genet. https://doi.org/10.1038/s41588-018-0331-5

    Article  PubMed  PubMed Central  Google Scholar 

  13. Fuchs CS, Tomasek J, Yong CJ et al (2014) Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 383:31–39. https://doi.org/10.1016/S0140-6736(13)61719-5

    Article  CAS  PubMed  Google Scholar 

  14. Fuchs CS, Shitara K, Di Bartolomeo M et al (2019) Ramucirumab with cisplatin and fluoropyrimidine as first-line therapy in patients with metastatic gastric or junctional adenocarcinoma (RAINFALL): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol 20:420–435. https://doi.org/10.1016/S1470-2045(18)30791-5

    Article  CAS  PubMed  Google Scholar 

  15. Haffner I, Schierle K, Raimúndez E et al (2021) HER2 expression, test deviations, and their impact on survival in metastatic gastric cancer: results from the prospective multicenter VARIANZ study. J Clin Oncol 39:1468–1478. https://doi.org/10.1200/jco.20.02761

    Article  PubMed  PubMed Central  Google Scholar 

  16. van Hagen P, Hulshof MCCM, van Lanschot JJB et al (2012) Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 366:2074–2084. https://doi.org/10.1056/NEJMoa1112088

    Article  PubMed  Google Scholar 

  17. Huang ZH, Ma XW, Zhang J et al (2018) Cetuximab for esophageal cancer: an updated meta-analysis of randomized controlled trials 11 medical and health sciences 1112 oncology and carcinogenesis. BMC Cancer 18:1–13. https://doi.org/10.1186/s12885-018-5040-z

    Article  Google Scholar 

  18. von Loga K, Woolston A, Punta M et al (2020) Extreme intratumour heterogeneity and driver evolution in mismatch repair deficient gastro-oesophageal cancer. Nat Commun 11:139. https://doi.org/10.1038/s41467-019-13915-7

    Article  CAS  Google Scholar 

  19. Martincorena I, Fowler JC, Wabik A et al (2018) Somatic mutant clones colonize the human esophagus with age. Science 362:911–917. https://doi.org/10.1126/science.aau3879

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Mourikis TP, Benedetti L, Foxall E et al (2019) Patient-specific cancer genes contribute to recurrently perturbed pathways and establish therapeutic vulnerabilities in esophageal adenocarcinoma. Nat Commun 10:3101. https://doi.org/10.1038/s41467-019-10898-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ohtsu A, Shah MA, Van Cutsem E et al (2011) Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. J Clin Oncol 29:3968–3976. https://doi.org/10.1200/JCO.2011.36.2236

    Article  CAS  PubMed  Google Scholar 

  22. Pakzad R, Mohammadian-Hafshejani A, Khosravi B et al (2016) The incidence and mortality of esophageal cancer and their relationship to development in Asia. Ann Transl Med 4:1–11. https://doi.org/10.3978/j.issn.2305-5839.2016.01.11

    Article  CAS  Google Scholar 

  23. Parsonnet J, Friedman GD, Vandersteen DP et al (1991) Helicobacter pylori infection and the risk of gastric carcinoma. N Engl J Med 325:1127–1131. https://doi.org/10.1056/NEJM199110173251603

    Article  CAS  PubMed  Google Scholar 

  24. Plum PS, Gebauer F, Krämer M et al (2019) HER2/neu (ERBB2) expression and gene amplification correlates with better survival in esophageal adenocarcinoma. BMC Cancer 19:38. https://doi.org/10.1186/s12885-018-5242-4

    Article  PubMed  PubMed Central  Google Scholar 

  25. Reid BJ, Li X, Galipeau PC, Vaughan TL (2010) Barrett’s oesophagus and oesophageal adenocarcinoma: time for a new synthesis. Nat Rev Cancer 10:87–101. https://doi.org/10.1038/nrc2773

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Repana D, Nulsen J, Dressler L et al (2019) The network of cancer genes (NCG): a comprehensive catalogue of known and candidate cancer genes from cancer sequencing screens. Genome Biol 20:1. https://doi.org/10.1186/s13059-018-1612-0

    Article  PubMed  PubMed Central  Google Scholar 

  27. Rubenstein JH, Shaheen NJ (2015) Epidemiology, diagnosis, and management of esophageal adenocarcinoma. Gastroenterology 149:302–317.e1. https://doi.org/10.1053/j.gastro.2015.04.053

    Article  PubMed  Google Scholar 

  28. Rumiato E, Pasello G, Montagna M et al (2011) DNA copy number profile discriminates between esophageal adenocarcinoma and squamous cell carcinoma and represents an independent prognostic parameter in esophageal adenocarcinoma. Cancer Lett 310:84–93. https://doi.org/10.1016/j.canlet.2011.06.017

    Article  CAS  PubMed  Google Scholar 

  29. Rustgi AK, El-Serag HB (2014) Esophageal carcinoma. N Engl J Med 371:2499–2509. https://doi.org/10.1056/NEJMra1314530

    Article  CAS  PubMed  Google Scholar 

  30. Secrier M, Li X, de Silva N et al (2016) Mutational signatures in esophageal adenocarcinoma define etiologically distinct subgroups with therapeutic relevance. Nat Genet 48:1131–1141. https://doi.org/10.1038/ng.3659

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Shah MA, Kojima T, Hochhauser D et al (2019) Efficacy and safety of pembrolizumab for heavily pretreated patients with advanced, metastatic adenocarcinoma or squamous cell carcinoma of the esophagus: the phase 2 KEYNOTE-180 study. JAMA Oncol 5:546–550. https://doi.org/10.1001/jamaoncol.2018.5441

    Article  PubMed  Google Scholar 

  32. Wang K, Johnson A, Ali SM et al (2015) Comprehensive genomic profiling of advanced esophageal squamous cell carcinomas and esophageal adenocarcinomas reveals similarities and differences. Oncologist 20:1132–1139. https://doi.org/10.1634/theoncologist.2015-0156

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wilke H, Muro K, Van Cutsem E et al (2014) Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): A double-blind, randomised phase 3 trial. Lancet Oncol 15:1224–1235. https://doi.org/10.1016/S1470-2045(14)70420-6

    Article  CAS  PubMed  Google Scholar 

  34. Yoon HH, Shi Q, Sukov WR et al (2012) Association of HER2/ErbB2 expression and gene amplification with pathologic features and prognosis in esophageal adenocarcinomas. Clin Cancer Res 18:546–554. https://doi.org/10.1158/1078-0432.CCR-11-2272

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Patrick S. Plum.

Ethics declarations

Interessenkonflikt

P.S. Plum, A. Quaas, H. Alakus und C.J. Bruns 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

I. Gockel, Leipzig

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

Plum, P.S., Quaas, A., Alakus, H. et al. Einfluss der Molekularpathologie auf die onkologische Chirurgie von Tumoren des oberen Gastrointestinaltraktes. Chirurg 92, 981–985 (2021). https://doi.org/10.1007/s00104-021-01466-x

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00104-021-01466-x

Schlüsselwörter

Keywords

Navigation