Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Evolutionary dynamics in Barrett oesophagus: implications for surveillance, risk stratification and therapy

Abstract

Cancer development is a dynamic evolutionary process characterized by marked intratumoural heterogeneity at the genetic, epigenetic and phenotypic levels. Barrett oesophagus, the pre-malignant condition to oesophageal adenocarcinoma (EAC), is an exemplary system to longitudinally study the evolution of malignancy. Evidence has emerged of Barrett oesophagus lesions pre-programmed for progression to EAC many years before clinical detection, indicating a considerable window for therapeutic intervention. In this Review, we explore the mechanisms underlying clonal expansion and contraction that establish the Barrett oesophagus clonal mosaicism over time and space and discuss intrinsic genotypic and extrinsic environmental drivers that direct the evolutionary trajectory of Barrett oesophagus towards a malignant phenotype. We propose that understanding and exploiting the evolutionary dynamics of Barrett oesophagus will identify novel therapeutic targets, improve prognostic tools and offer the opportunity for personalized surveillance programmes geared to prevent progression to EAC.

Key points

  • Longitudinal surveillance of Barrett oesophagus offers an exemplary opportunity to study lesion evolution over time and space and during the progression to oesophageal adenocarcinoma.

  • Distinct evolutionary patterns exist between non-progressing and progressing Barrett oesophagus with evidence for protracted evolutionary dynamics occurring over many years.

  • Chronic inflammation due to reflux accelerates evolution through mutagenesis and provides a selective pressure for mutant clones that are viable in the harsh environment and can rapidly repopulate the ulcerated mucosa.

  • The measurement of evolutionary dynamics in Barrett oesophagus lesions is a potentially powerful prognostic tool and identifies a wide window for therapeutic intervention and prevention of cancer in patients with Barrett oesophagus.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Evolutionary models in Barrett oesophagus.
Fig. 2: The role of inflammation in Barrett oesophagus.

Similar content being viewed by others

References

  1. Cook, M. B., Chow, W. H. & Devesa, S. S. Oesophageal cancer incidence in the United States by race, sex, and histologic type, 1977–2005. Br. J. Cancer 101, 855–859 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Sung, H. et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209–249 (2021).

    Article  PubMed  Google Scholar 

  3. Arnold, M. et al. Progress in cancer survival, mortality, and incidence in seven high-income countries 1995–2014 (ICBP SURVMARK-2): a population-based study. Lancet Oncol. 20, 1493–1505 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Morgan, E. et al. International trends in oesophageal cancer survival by histological subtype between 1995 and 2014. Gut 70, 234–242 (2021).

    CAS  PubMed  Google Scholar 

  5. Arnold, M., Ferlay, J., van Berge Henegouwen, M. I. & Soerjomataram, I. Global burden of oesophageal and gastric cancer by histology and subsite in 2018. Gut 69, 1564–1571 (2020).

    Article  PubMed  Google Scholar 

  6. Arnold, M., Laversanne, M., Brown, L. M., Devesa, S. S. & Bray, F. Predicting the future burden of esophageal cancer by histological subtype: international trends in incidence up to 2030. Am. J. Gastroenterol. 112, 1247–1255 (2017). Predictions fitting and extrapolating age–period–cohort models expect the number of new EAC cases to increase rapidly between 2005 and 2030, thereby becoming the predominant type of oesophageal cancer in a growing number of high-income countries.

    Article  PubMed  Google Scholar 

  7. Smyth, E. C. et al. Oesophageal cancer. Nat. Rev. Dis. Prim. 3, 17048 (2017).

    Article  PubMed  Google Scholar 

  8. Naini, B. V., Souza, R. F. & Odze, R. D. Barrett’s esophagus: a comprehensive and contemporary review for pathologists. Am. J. Surg. Pathol. 40, e45–e66 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Spechler, S. J., Fitzgerald, R. C., Prasad, G. A. & Wang, K. K. History, molecular mechanisms, and endoscopic treatment of Barrett’s esophagus. Gastroenterology 138, 854–869 (2010).

    Article  PubMed  Google Scholar 

  10. Ronkainen, J. et al. Prevalence of Barrett’s esophagus in the general population: an endoscopic study. Gastroenterology 129, 1825–1831 (2005).

    Article  PubMed  Google Scholar 

  11. Zagari, R. M. et al. Gastro-oesophageal reflux symptoms, oesophagitis and Barrett’s oesophagus in the general population: the Loiano-Monghidoro study. Gut 57, 1354–1359 (2008).

    Article  CAS  PubMed  Google Scholar 

  12. Runge, T. M., Abrams, J. A. & Shaheen, N. J. Epidemiology of Barrett’s esophagus and esophageal adenocarcinoma. Gastroenterol. Clin. North. Am. 44, 203–231 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Fitzgerald, R. C. et al. Cytosponge-trefoil factor 3 versus usual care to identify Barrett’s oesophagus in a primary care setting: a multicentre, pragmatic, randomised controlled trial. Lancet 396, 333–344 (2020). A multicentre randomized controlled trial showed superior performance of Barrett oesophagus detection in the primary care setting using a novel non-endoscopic Cytosponge-TFF3 device compared with standard care in the primary care setting of patients with gastro-oesophageal reflux.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Shaheen, N. J. & Richter, J. E. Barrett’s oesophagus. Lancet 373, 850–861 (2009).

    Article  CAS  PubMed  Google Scholar 

  15. Hamade, N. et al. Significant decline in the prevalence of Barrett’s esophagus among patients with gastroesophageal reflux disease. Dis. Esophagus 34, doaa131 (2021).

    Article  PubMed  Google Scholar 

  16. O’Donovan, M. & Fitzgerald, R. C. Screening for Barrett’s esophagus: are new high-volume methods feasible? Dig. Dis. Sci. 63, 2105–2114 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Harrison, R. et al. Detection of intestinal metaplasia in Barrett’s esophagus: an observational comparator study suggests the need for a minimum of eight biopsies. Am. J. Gastroenterol. 102, 1154–1161 (2007).

    Article  PubMed  Google Scholar 

  18. Weusten, B. et al. Endoscopic management of Barrett’s esophagus: European Society of Gastrointestinal Endoscopy (ESGE) position statement. Endoscopy 49, 191–198 (2017).

    Article  PubMed  Google Scholar 

  19. Shaheen, N. J., Falk, G. W., Iyer, P. G. & Gerson, L. B., American College of Gastroenterology. ACG Clinical Guideline: diagnosis and management of Barrett’s esophagus. Am. J. Gastroenterol. 111, 30–50 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. di Pietro, M., Alzoubaidi, D. & Fitzgerald, R. C. Barrett’s esophagus and cancer risk: how research advances can impact clinical practice. Gut Liver 8, 356–370 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Levine, D. S., Blount, P. L., Rudolph, R. E. & Reid, B. J. Safety of a systematic endoscopic biopsy protocol in patients with Barrett’s esophagus. Am. J. Gastroenterol. 95, 1152–1157 (2000).

    Article  CAS  PubMed  Google Scholar 

  22. Sharma, P. et al. The development and validation of an endoscopic grading system for Barrett’s esophagus: the Prague C & M criteria. Gastroenterology 131, 1392–1399 (2006).

    Article  PubMed  Google Scholar 

  23. Pohl, H. et al. Length of Barrett’s oesophagus and cancer risk: implications from a large sample of patients with early oesophageal adenocarcinoma. Gut 65, 196–201 (2016).

    Article  PubMed  Google Scholar 

  24. Antony, A. et al. Adherence to quality indicators in endoscopic surveillance of Barrett’s esophagus and correlation to dysplasia detection rates. Clin. Res. Hepatol. Gastroenterol. 42, 591–596 (2018).

    Article  PubMed  Google Scholar 

  25. Verbeek, R. E. et al. Surveillance of Barrett’s esophagus and mortality from esophageal adenocarcinoma: a population-based cohort study. Am. J. Gastroenterol. 109, 1215–1222 (2014).

    Article  PubMed  Google Scholar 

  26. Holmberg, D., Ness-Jensen, E., Mattsson, F. & Lagergren, J. Adherence to clinical guidelines for Barrett’s esophagus. Scand. J. Gastroenterol. 54, 945–952 (2019).

    Article  PubMed  Google Scholar 

  27. Peters, F. P. et al. Histologic evaluation of resection specimens obtained at 293 endoscopic resections in Barrett’s esophagus. Gastrointest. Endosc. 67, 604–609 (2008).

    Article  PubMed  Google Scholar 

  28. Schlemper, R. J. et al. The Vienna classification of gastrointestinal epithelial neoplasia. Gut 47, 251–255 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Fitzgerald, R. C. et al. British Society of Gastroenterology guidelines on the diagnosis and management of Barrett’s oesophagus. Gut 63, 7–42 (2014).

    Article  PubMed  Google Scholar 

  30. Vennalaganti, P. et al. Discordance among pathologists in the United States and Europe in diagnosis of low-grade dysplasia for patients with Barrett’s esophagus. Gastroenterology 152, 564–570.e4 (2017).

    Article  PubMed  Google Scholar 

  31. Salomao, M. A., Lam-Himlin, D. & Pai, R. K. Substantial interobserver agreement in the diagnosis of dysplasia in Barrett esophagus upon review of a patient’s entire set of biopsies. Am. J. Surg. Pathol. 42, 376–381 (2018).

    Article  PubMed  Google Scholar 

  32. Montgomery, E. et al. Reproducibility of the diagnosis of dysplasia in Barrett esophagus: a reaffirmation. Hum. Pathol. 32, 368–378 (2001).

    Article  CAS  PubMed  Google Scholar 

  33. Spechler, S. J., Sharma, P., Souza, R. F., Inadomi, J. M. & Shaheen, N. J. American Gastroenterological Association medical position statement on the management of Barrett’s esophagus. Gastroenterology 140, 1084–1091 (2011).

    Article  PubMed  Google Scholar 

  34. Hvid-Jensen, F., Pedersen, L., Drewes, A. M., Sørensen, H. T. & Funch-Jensen, P. Incidence of adenocarcinoma among patients with Barrett’s esophagus. N. Engl. J. Med. 365, 1375–1383 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Hvid-Jensen, F. & Drewes, A. M. Should aspirin and PPIs be recommended for patients with Barrett’s oesophagus? Lancet 392, 362–364 (2018).

    Article  PubMed  Google Scholar 

  36. Zeki, S. & Fitzgerald, R. C. Targeting care in Barrett’s oesophagus. Clin. Med. 14 (Suppl. 6), s78–s83 (2014).

    Article  Google Scholar 

  37. Wenker, T. N., Tan, M. C., Liu, Y., El-Serag, H. B. & Thrift, A. P. Prior diagnosis of Barrett’s esophagus is infrequent, but associated with improved esophageal adenocarcinoma survival. Dig. Dis. Sci. 63, 3112–3119 (2018).

    Article  PubMed  Google Scholar 

  38. Visrodia, K. et al. Magnitude of missed esophageal adenocarcinoma after Barrett’s esophagus diagnosis: a systematic review and meta-analysis. Gastroenterology 150, 599–607.e7 (2016).

    Article  PubMed  Google Scholar 

  39. Nowell, P. C. The clonal evolution of tumor cell populations. Science 194, 23–28 (1976).

    Article  CAS  PubMed  Google Scholar 

  40. Greaves, M. & Maley, C. C. Clonal evolution in cancer. Nature 481, 306–313 (2012). This Review explains fundamental principles of clonal evolution in cancer such as genetic diversification, genetic drivers, the clonal architecture, clonal dynamics and the influence of the cancer ecosystem.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Nik-Zainal, S. et al. The life history of 21 breast cancers. Cell 149, 994–1007 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sottoriva, A. et al. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc. Natl Acad. Sci. USA 110, 4009–4014 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. de Bruin, E. C. et al. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science 346, 251–256 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Gundem, G. et al. The evolutionary history of lethal metastatic prostate cancer. Nature 520, 353–357 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Landau, D. A. et al. Evolution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell 152, 714–726 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Merlo, L. M. F., Pepper, J. W., Reid, B. J. & Maley, C. C. Cancer as an evolutionary and ecological process. Nat. Rev. Cancer 6, 924–935 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Souza, R. F. Reflux esophagitis and its role in the pathogenesis of Barrett’s metaplasia. J. Gastroenterol. 52, 767–776 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. McQuaid, K. R., Laine, L., Fennerty, M. B., Souza, R. & Spechler, S. J. Systematic review: the role of bile acids in the pathogenesis of gastro-oesophageal reflux disease and related neoplasia. Aliment. Pharmacol. Ther. 34, 146–165 (2011).

    Article  CAS  PubMed  Google Scholar 

  50. Que, J., Garman, K. S., Souza, R. F. & Spechler, S. J. Pathogenesis and cells of origin of Barrett’s esophagus. Gastroenterology 157, 349–364.e1 (2019).

    Article  PubMed  Google Scholar 

  51. Jiang, M. et al. Transitional basal cells at the squamous-columnar junction generate Barrett’s oesophagus. Nature 550, 529–533 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Leedham, S. J. et al. Individual crypt genetic heterogeneity and the origin of metaplastic glandular epithelium in human Barrett’s oesophagus. Gut 57, 1041–1048 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. von Furstenberg, R. J. et al. Porcine esophageal submucosal gland culture model shows capacity for proliferation and differentiation. Cell Mol. Gastroenterol. Hepatol. 4, 385–404 (2017).

    Article  Google Scholar 

  54. Wang, X. et al. Residual embryonic cells as precursors of a Barrett’s-like metaplasia. Cell 145, 1023–1035 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Nowicki-Osuch, K. et al. Molecular phenotyping reveals the identity of Barrett’s esophagus and its malignant transition. Science 373, 760–767 (2021).

    Article  CAS  PubMed  Google Scholar 

  56. Peters, Y. et al. Barrett oesophagus. Nat. Rev. Dis. Prim. 5, 35 (2019).

    Article  PubMed  Google Scholar 

  57. Galipeau, P. C., Prevo, L. J., Sanchez, C. A., Longton, G. M. & Reid, B. J. Clonal expansion and loss of heterozygosity at chromosomes 9p and 17p in premalignant esophageal (Barrett’s) tissue. J. Natl Cancer Inst. 91, 2087–2095 (1999).

    Article  CAS  PubMed  Google Scholar 

  58. Wong, D. J. et al. p16(INK4a) lesions are common, early abnormalities that undergo clonal expansion in Barrett’s metaplastic epithelium. Cancer Res. 61, 8284–8289 (2001).

    CAS  PubMed  Google Scholar 

  59. Martinez, P. et al. Dynamic clonal equilibrium and predetermined cancer risk in Barrett’s oesophagus. Nat. Commun. 7, 12158 (2016). A longitudinal single-cell fluorescent in situ hybridization analysis of non-dysplastic Barrett oesophagus biopsy samples, including 300 non-progressing and 20 progressing Barrett oesophagus lesions, showing that clones exist in a dynamic equilibrium with relatively stable levels of genetic diversity over time, whereby clonal expansions are rare events.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Li, X. et al. Assessment of esophageal adenocarcinoma risk using somatic chromosome alterations in longitudinal samples in Barrett’s esophagus. Cancer Prev. Res. 8, 845–856 (2015). A multi-region SNP array analysis of CNAs in 248 patients with Barrett oesophagus over space and time showing that non-progressing Barrett oesophagus typically harbours a low burden of large-scale CNAs that remain low over time, whereas progressing Barrett oesophagus develops high levels of CNAs.

    Article  CAS  Google Scholar 

  61. Maley, C. C. et al. Genetic clonal diversity predicts progression to esophageal adenocarcinoma. Nat. Genet. 38, 468–473 (2006). A fluorescent in situ hybridization study including 268 patients with non-progressing Barrett oesophagus and 37 patients with progressing Barrett oesophagus identifies genetic clonal diversity as a predictor of progression to EAC in patients with Barrett oesophagus.

    Article  CAS  PubMed  Google Scholar 

  62. Ross-Innes, C. S. et al. Whole-genome sequencing provides new insights into the clonal architecture of Barrett’s esophagus and esophageal adenocarcinoma. Nat. Genet. 47, 1038–1046 (2015). Whole-genome sequencing on 23 paired Barrett oesophagus and EAC samples and one in-depth Barrett oesophagus case sampled over time and space reveals the presence of mutations and copy number changes in non-dysplastic Barrett prior to the phenotypic evolution of the clonally related HGD descendent, questioning the adequacy of dysplasia as a biomarker for cancer risk.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Slaughter, D. P., Southwick, H. W. & Smejkal, W. Field cancerization in oral stratified squamous epithelium; clinical implications of multicentric origin. Cancer 6, 963–968 (1953).

    Article  CAS  PubMed  Google Scholar 

  64. Braakhuis, B. J. M., Tabor, M. P., Kummer, J. A., Leemans, C. R. & Brakenhoff, R. H. A genetic explanation of Slaughter’s concept of field cancerization: evidence and clinical implications. Cancer Res. 63, 1727–1730 (2003).

    CAS  PubMed  Google Scholar 

  65. Galandiuk, S. et al. Field cancerization in the intestinal epithelium of patients with Crohn’s ileocolitis. Gastroenterology 142, 855–864.e8 (2012).

    Article  PubMed  Google Scholar 

  66. Eads, C. A. et al. Fields of aberrant CpG island hypermethylation in Barrett’s esophagus and associated adenocarcinoma. Cancer Res. 60, 5021–5026 (2000).

    CAS  PubMed  Google Scholar 

  67. Gu, J. et al. Genome-wide catalogue of chromosomal aberrations in Barrett’s esophagus and esophageal adenocarcinoma: a high-density single nucleotide polymorphism array analysis. Cancer Prev. Res. 3, 1176–1186 (2010).

    Article  CAS  Google Scholar 

  68. Reid, B. J. et al. Barrett’s esophagus: ordering the events that lead to cancer. Eur. J. Cancer Prev. 5 (Suppl. 2), 57–65 (1996).

    Article  PubMed  Google Scholar 

  69. Li, X. et al. Temporal and spatial evolution of somatic chromosomal alterations: a case-cohort study of Barrett’s esophagus. Cancer Prev. Res. 7, 114–127 (2014). A large longitudinal study that examines multi-region CNA profiles of 79 patients with Barrett oesophagus that progressed to EAC and 169 patients with Barrett oesophagus that did not progress and showed that progressing Barrett oesophagus has more CNAs than non-progressing Barrett oesophagus and that progressors display a sudden sharp increase in genetic alterations 2–4 years before cancer detection.

    Article  Google Scholar 

  70. Hardie, L. J. et al. p16 expression in Barrett’s esophagus and esophageal adenocarcinoma: association with genetic and epigenetic alterations. Cancer Lett. 217, 221–230 (2005).

    Article  CAS  PubMed  Google Scholar 

  71. Bian, Y.-S., Osterheld, M.-C., Fontolliet, C., Bosman, F. T. & Benhattar, J. p16 inactivation by methylation of the CDKN2A promoter occurs early during neoplastic progression in Barrett’s esophagus. Gastroenterology 122, 1113–1121 (2002).

    Article  CAS  PubMed  Google Scholar 

  72. Stachler, M. D. et al. Paired exome analysis of Barrett’s esophagus and adenocarcinoma. Nat. Genet. 47, 1047–1055 (2015). A paired whole-exome study of Barrett oesophagus and EAC suggesting that many EACs emerge through TP53 mutation with subsequent genome doubling, followed by the acquisition of oncogenic amplifications.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Weaver, J. M. J. et al. Ordering of mutations in preinvasive disease stages of esophageal carcinogenesis. Nat. Genet. 46, 837–843 (2014). A whole-genome sequencing study including EAC, high-grade dysplasia and non-dysplastic Barrett oesophagus shows that only mutations in TP53 and SMAD4 occur in a stage-specific manner, whereas the majority of recurrently mutated genes in EAC are also mutated in non-dysplastic Barrett oesophagus and might therefore occur early in Barrett development.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Dulak, A. M. et al. Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity. Nat. Genet. 45, 478–486 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Galipeau, P. C. et al. 17p (p53) allelic losses, 4N (G2/tetraploid) populations, and progression to aneuploidy in Barrett’s esophagus. Proc. Natl Acad. Sci. USA 93, 7081–7084 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Reid, B. J. et al. Predictors of progression in Barrett’s esophagus II: baseline 17p (p53) loss of heterozygosity identifies a patient subset at increased risk for neoplastic progression. Am. J. Gastroenterol. 96, 2839–2848 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Maley, C. C. et al. The combination of genetic instability and clonal expansion predicts progression to esophageal adenocarcinoma. Cancer Res. 64, 7629–7633 (2004).

    Article  CAS  PubMed  Google Scholar 

  78. Killcoyne, S. et al. Genomic copy number predicts esophageal cancer years before transformation. Nat. Med. 26, 1726–1732 (2020). Using shallow whole-genome sequencing of 777 Barrett oesophagus biopsy samples from a retrospective cohort of 88 patients undergoing Barrett oesophagus surveillance, a risk stratification model based on CNA profile complexity was able to identify 50% of patients at high risk ≥8 years before clinical progression and showed improving detection rates to 78% and 85% at 2 years and 1 year prior, respectively.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Quante, M., Graham, T. A. & Jansen, M. Insights into the pathophysiology of esophageal adenocarcinoma. Gastroenterology 154, 406–420 (2018).

    Article  CAS  PubMed  Google Scholar 

  80. McDonald, S. A. C. et al. Mechanisms of field cancerization in the human stomach: the expansion and spread of mutated gastric stem cells. Gastroenterology 134, 500–510 (2008).

    Article  CAS  PubMed  Google Scholar 

  81. Theodorou, D. et al. Intraluminal pH and goblet cell density in Barrett’s esophagus. J. Gastrointest. Surg. 16, 469–474 (2012).

    Article  PubMed  Google Scholar 

  82. Quante, M. et al. Bile acid and inflammation activate gastric cardia stem cells in a mouse model of Barrett-like metaplasia. Cancer Cell 21, 36–51 (2012). In a transgenic mouse model of Barrett oesophagus, oesophageal overexpression of IL-1β led to oesophagitis, Barrett-like metaplasia and EAC, which could be accelerated by exposure to bile acids and nitrosamines.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. O’Riordan, J. M. et al. Factors influencing the development of Barrett’s epithelium in the esophageal remnant postesophagectomy. Am. J. Gastroenterol. 99, 205–211 (2004).

    Article  PubMed  Google Scholar 

  84. Lavery, D. L. et al. Evolution of oesophageal adenocarcinoma from metaplastic columnar epithelium without goblet cells in Barrett’s oesophagus. Gut 65, 907–913 (2016).

    Article  CAS  PubMed  Google Scholar 

  85. Khan, S. et al. Crypt dysplasia in Barrett’s oesophagus shows clonal identity between crypt and surface cells. J. Pathol. 231, 98–104 (2013).

    Article  CAS  PubMed  Google Scholar 

  86. Vogelstein, B. et al. Genetic alterations during colorectal-tumor development. N. Engl. J. Med. 319, 525–532 (1988).

    Article  CAS  PubMed  Google Scholar 

  87. Graham, T. A. & Sottoriva, A. Measuring cancer evolution from the genome. J. Pathol. 241, 183–191 (2017).

    Article  PubMed  Google Scholar 

  88. Eldredge, N. & Gould, S. J. On punctuated equilibria. Science 276, 338–341 (1997).

    Article  CAS  PubMed  Google Scholar 

  89. Newell, F. et al. Complex structural rearrangements are present in high-grade dysplastic Barrett’s oesophagus samples. BMC Med. Genomics 12, 31 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  90. Goldschmidt, R. The Material Basis of Evolution (Yale Univ. Press, 1940).

  91. Carter, S. L. et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 30, 413–421 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Nones, K. et al. Genomic catastrophes frequently arise in esophageal adenocarcinoma and drive tumorigenesis. Nat. Commun. 5, 5224 (2014).

    Article  CAS  PubMed  Google Scholar 

  93. Stephens, P. J. et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144, 27–40 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Notta, F. et al. A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns. Nature 538, 378–382 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Malhotra, A. et al. Breakpoint profiling of 64 cancer genomes reveals numerous complex rearrangements spawned by homology-independent mechanisms. Genome Res. 23, 762–776 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Rausch, T. et al. Genome sequencing of pediatric medulloblastoma links catastrophic DNA rearrangements with TP53 mutations. Cell 148, 59–71 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Przybytkowski, E. et al. Chromosome-breakage genomic instability and chromothripsis in breast cancer. BMC Genomics 15, 579 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  98. Finley, J. C. et al. Chromosomal instability in Barrett’s esophagus is related to telomere shortening. Cancer Epidemiol. Biomark. Prev. 15, 1451–1457 (2006).

    Article  CAS  Google Scholar 

  99. Cook, M. B. et al. Cigarette smoking and adenocarcinomas of the esophagus and esophagogastric junction: a pooled analysis from the international BEACON consortium. J. Natl Cancer Inst. 102, 1344–1353 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Coleman, H. G. et al. Tobacco smoking increases the risk of high-grade dysplasia and cancer among patients with Barrett’s esophagus. Gastroenterology 142, 233–240 (2012).

    Article  PubMed  Google Scholar 

  101. Olliver, J. R. et al. Risk factors, DNA damage, and disease progression in Barrett’s esophagus. Cancer Epidemiol. Biomark. Prev. 14, 620–625 (2005).

    Article  CAS  Google Scholar 

  102. Kaz, A. M. et al. Global DNA methylation patterns in Barrett’s esophagus, dysplastic Barrett’s, and esophageal adenocarcinoma are associated with BMI, gender, and tobacco use. Clin. Epigenetics 8, 111 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  103. Kadakia, S. C., De La Baume, H. R. & Shaffer, R. T. Effects of transdermal nicotine on lower esophageal sphincter and esophageal motility. Dig. Dis. Sci. 41, 2130–2134 (1996).

    Article  CAS  PubMed  Google Scholar 

  104. Nieto, T. et al. A systematic review of epigenetic biomarkers in progression from non-dysplastic Barrett’s oesophagus to oesophageal adenocarcinoma. BMJ Open 8, e020427 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  105. Hanahan, D. & Weinberg, R. A. The hallmarks of cancer. Cell 100, 57–70 (2000).

    Article  CAS  PubMed  Google Scholar 

  106. Taniguchi, K. & Karin, M. NF-κB, inflammation, immunity and cancer: coming of age. Nat. Rev. Immunol. 18, 309–324 (2018).

    Article  CAS  PubMed  Google Scholar 

  107. Thrift, A. P., Garcia, J. M. & El-Serag, H. B. A multibiomarker risk score helps predict risk for Barrett’s esophagus. Clin. Gastroenterol. Hepatol. 12, 1267–1271 (2014).

    Article  PubMed  Google Scholar 

  108. Lagergren, J., Bergström, R., Lindgren, A. & Nyrén, O. Symptomatic gastroesophageal reflux as a risk factor for esophageal adenocarcinoma. N. Engl. J. Med. 340, 825–831 (1999).

    Article  CAS  PubMed  Google Scholar 

  109. Schmidt, M. et al. Epidemiologic risk factors in a comparison of a Barrett Esophagus Registry (BarrettNET) and a case-control population in Germany. Cancer Prev. Res. 13, 377–384 (2020).

    Article  Google Scholar 

  110. Parsonnet, J. Molecular mechanisms for inflammation-promoted pathogenesis of cancer — The Sixteenth International Symposium of the Sapporo Cancer Seminar. Cancer Res. 57, 3620–3624 (1997).

    CAS  PubMed  Google Scholar 

  111. Feagins, L. A. et al. Mechanisms of oxidant production in esophageal squamous cell and Barrett’s cell lines. Am. J. Physiol. Gastrointest. Liver Physiol. 294, G411–G417 (2008).

    Article  CAS  PubMed  Google Scholar 

  112. Secrier, M. et al. Mutational signatures in esophageal adenocarcinoma define etiologically distinct subgroups with therapeutic relevance. Nat. Genet. 48, 1131–1141 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Fitzgerald, R. C. et al. Diversity in the oesophageal phenotypic response to gastro-oesophageal reflux: immunological determinants. Gut 50, 451–459 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Fitzgerald, R. C. et al. Inflammatory gradient in Barrett’s oesophagus: implications for disease complications. Gut 51, 316–322 (2002). This article shows that specific cytokine responses with pro-inflammatory IL-8 and IL-1β expression might contribute to the local inflammatory microenvironment in Barrett oesophagus.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Kavanagh, M. E. et al. Impact of the inflammatory microenvironment on T-cell phenotype in the progression from reflux oesophagitis to Barrett oesophagus and oesophageal adenocarcinoma. Cancer Lett. 370, 117–124 (2016).

    Article  CAS  PubMed  Google Scholar 

  116. Wolfsen, H. C., Hemminger, L. L. & DeVault, K. R. Recurrent Barrett’s esophagus and adenocarcinoma after esophagectomy. BMC Gastroenterol. 4, 18 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  117. Oberg, S., Johansson, J., Wenner, J. & Walther, B. Metaplastic columnar mucosa in the cervical esophagus after esophagectomy. Ann. Surg. 235, 338–345 (2002).

    Article  PubMed  PubMed Central  Google Scholar 

  118. Schwabe, R. F. & Jobin, C. The microbiome and cancer. Nat. Rev. Cancer 13, 800–812 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Tsilimigras, M. C. B., Fodor, A. & Jobin, C. Carcinogenesis and therapeutics: the microbiota perspective. Nat. Microbiol. 2, 17008 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Yang, L. et al. Inflammation and intestinal metaplasia of the distal esophagus are associated with alterations in the microbiome. Gastroenterology 137, 588–597 (2009).

    Article  PubMed  Google Scholar 

  121. Elliott, D. R. F., Walker, A. W., O’Donovan, M., Parkhill, J. & Fitzgerald, R. C. A non-endoscopic device to sample the oesophageal microbiota: a case-control study. Lancet Gastroenterol. Hepatol. 2, 32–42 (2017).

    Article  PubMed  Google Scholar 

  122. Yamamura, K. et al. Human microbiome fusobacterium nucleatum in esophageal cancer tissue is associated with prognosis. Clin. Cancer Res. 22, 5574–5581 (2016).

    Article  CAS  PubMed  Google Scholar 

  123. Kostic, A. D. et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 14, 207–215 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. McCoy, A. N. et al. Fusobacterium is associated with colorectal adenomas. PLoS ONE 8, e53653 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Castellarin, M. et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 22, 299–306 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Jankowski, J. A. Z. et al. Esomeprazole and aspirin in Barrett’s oesophagus (AspECT): a randomised factorial trial. Lancet 392, 400–408 (2018). A randomized controlled trial comparing permutations of low-dose and high-dose PPIs with or without aspirin as a chemopreventive strategy in patients with Barrett oesophagus shows a statistically significant benefit of high-dose over low-dose PPIs, which could be enhanced with use of aspirin with high-dose PPIs.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Jovani, M. et al. Aspirin use is associated with lower risk of Barrett’s esophagus in women. Clin. Transl. Gastroenterol. 8, e131 (2017).

    PubMed Central  Google Scholar 

  128. Galipeau, P. C. et al. NSAIDs modulate CDKN2A, TP53, and DNA content risk for progression to esophageal adenocarcinoma. PLoS Med. 4, e67 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  129. Rothwell, P. M. et al. Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomised trials. Lancet 377, 31–41 (2011).

    Article  CAS  PubMed  Google Scholar 

  130. Baron, J. A. et al. A randomized trial of aspirin to prevent colorectal adenomas. N. Engl. J. Med. 348, 891–899 (2003).

    Article  CAS  PubMed  Google Scholar 

  131. Flossmann, E. & Rothwell, P. M. Effect of aspirin on long-term risk of colorectal cancer: consistent evidence from randomised and observational studies. Lancet 369, 1603–1613 (2007).

    Article  CAS  PubMed  Google Scholar 

  132. Algra, A. M. & Rothwell, P. M. Effects of regular aspirin on long-term cancer incidence and metastasis: a systematic comparison of evidence from observational studies versus randomised trials. Lancet Oncol. 13, 518–527 (2012).

    Article  CAS  PubMed  Google Scholar 

  133. Katona, B. W. & Weiss, J. M. Chemoprevention of colorectal cancer. Gastroenterology 158, 368–388 (2020).

    Article  CAS  PubMed  Google Scholar 

  134. Picardo, S. L., Maher, S. G., O’Sullivan, J. N. & Reynolds, J. V. Barrett’s to oesophageal cancer sequence: a model of inflammatory-driven upper gastrointestinal cancer. Dig. Surg. 29, 251–260 (2012).

    Article  PubMed  Google Scholar 

  135. Galipeau, P. C. et al. NSAID use and somatic exomic mutations in Barrett’s esophagus. Genome Med. 10, 17 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  136. Kostadinov, R. L. et al. NSAIDs modulate clonal evolution in Barrett’s esophagus. PLoS Genet. 9, e1003553 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Bhat, S. et al. Risk of malignant progression in Barrett’s esophagus patients: results from a large population-based study. J. Natl Cancer Inst. 103, 1049–1057 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  138. van der Wel, M. J. et al. Improved diagnostic stratification of digitised Barrett’s oesophagus biopsies by p53 immunohistochemical staining. Histopathology 72, 1015–1023 (2018).

    Article  PubMed  Google Scholar 

  139. Kaye, P. V. et al. Dysplasia in Barrett’s oesophagus: p53 immunostaining is more reproducible than haematoxylin and eosin diagnosis and improves overall reliability, while grading is poorly reproducible. Histopathology 69, 431–440 (2016). In this study, p53 IHC is reported as having increased reliability and inter-observer reliability in the diagnosis of dysplasia but showing poor results in discrimination between LGD and HGD.

    Article  PubMed  Google Scholar 

  140. Spechler, S. J. et al. American Gastroenterological Association technical review on the management of Barrett’s esophagus. Gastroenterology 140, e18–e52 (2011).

    Article  PubMed  Google Scholar 

  141. Varghese, S., Lao-Sirieix, P. & Fitzgerald, R. C. Identification and clinical implementation of biomarkers for Barrett’s esophagus. Gastroenterology 142, 435–441.e2 (2012).

    Article  PubMed  Google Scholar 

  142. Stachler, M. D. et al. Detection of mutations in Barrett’s esophagus before progression to high-grade dysplasia or adenocarcinoma. Gastroenterology 155, 156–167 (2018).

    Article  CAS  PubMed  Google Scholar 

  143. Bird-Lieberman, E. L. et al. Population-based study reveals new risk-stratification biomarker panel for Barrett’s esophagus. Gastroenterology 143, 927–935.e3 (2012).

    Article  CAS  PubMed  Google Scholar 

  144. Kastelein, F. et al. Aberrant p53 protein expression is associated with an increased risk of neoplastic progression in patients with Barrett’s oesophagus. Gut 62, 1676–1683 (2013). This paper shows that aberrant p53 IHC staining is associated with an increased risk of neoplastic progression in patients with Barrett oesophagus and increased positive predictive value for neoplastic progression from 15% with histological diagnosis of LGD alone to 33% with LGD and concurrent aberrant p53 expression.

    Article  CAS  PubMed  Google Scholar 

  145. Hadjinicolaou, A. V. et al. Aneuploidy in targeted endoscopic biopsies outperforms other tissue biomarkers in the prediction of histologic progression of Barrett’s oesophagus: a multi-centre prospective cohort study. EBioMedicine 56, 102765 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  146. Sharma, P., Shaheen, N. J., Katzka, D. & Bergman, J. AGA Clinical Practice Update on Endoscopic Treatment of Barrett’s Esophagus with Dysplasia and/or Early Cancer: Expert Review. Gastroenterology 158, 760–769 (2020).

    Article  PubMed  Google Scholar 

  147. van Olphen, S. et al. SOX2 as a novel marker to predict neoplastic progression in Barrett’s esophagus. Am. J. Gastroenterol. 110, 1420–1428 (2015).

    Article  PubMed  Google Scholar 

  148. Hong, M. K. et al. Expansion of the Ki-67 proliferative compartment correlates with degree of dysplasia in Barrett’s esophagus. Cancer 75, 423–429 (1995).

    Article  CAS  PubMed  Google Scholar 

  149. Sikkema, M. et al. Aneuploidy and overexpression of Ki67 and p53 as markers for neoplastic progression in Barrett’s esophagus: a case-control study. Am. J. Gastroenterol. 104, 2673–2680 (2009).

    Article  CAS  PubMed  Google Scholar 

  150. Yousaf, H. et al. Surface Ki-67 expression improves reproducibility of dysplasia diagnosis in Barrett’s esophagus. Am. J. Clin. Pathol. 153, 695–704 (2020).

    Article  CAS  PubMed  Google Scholar 

  151. Altaf, K., Xiong, J. J., la Iglesia, D., Hickey, L. & Kaul, A. Meta-analysis of biomarkers predicting risk of malignant progression in Barrett’s oesophagus. Br. J. Surg. 104, 493–502 (2017).

    Article  CAS  PubMed  Google Scholar 

  152. Findlay, J. M., Middleton, M. R. & Tomlinson, I. Genetic biomarkers of Barrett’s esophagus susceptibility and progression to dysplasia and cancer: a systematic review and meta-analysis. Dig. Dis. Sci. 61, 25–38 (2016).

    Article  CAS  PubMed  Google Scholar 

  153. Choi, W. T. et al. Diagnosis and risk stratification of Barrett’s dysplasia by flow cytometric DNA analysis of paraffin-embedded tissue. Gut 67, 1229–1238 (2017).

    Article  PubMed  Google Scholar 

  154. Timmer, M. R. et al. Derivation of genetic biomarkers for cancer risk stratification in Barrett’s oesophagus: a prospective cohort study. Gut 65, 1602–1610 (2016).

    Article  CAS  PubMed  Google Scholar 

  155. Hoefnagel, S. J. M. et al. A genomic biomarker-based model for cancer risk stratification of non-dysplastic Barrett’s esophagus patients after extended follow up; results from Dutch surveillance cohorts. PLoS ONE 15, e0231419 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Rabinovitch, P. S., Longton, G., Blount, P. L., Levine, D. S. & Reid, B. J. Predictors of progression in Barrett’s esophagus III: baseline flow cytometric variables. Am. J. Gastroenterol. 96, 3071–3083 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Reid, B. J., Levine, D. S., Longton, G., Blount, P. L. & Rabinovitch, P. S. Predictors of progression to cancer in Barrett’s esophagus: baseline histology and flow cytometry identify low- and high-risk patient subsets. Am. J. Gastroenterol. 95, 1669–1676 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Curtius, K., Wright, N. A. & Graham, T. A. An evolutionary perspective on field cancerization. Nat. Rev. Cancer 18, 19–32 (2018).

    Article  CAS  PubMed  Google Scholar 

  159. Tschanz, E. R. Do 40% of patients resected for Barrett esophagus with high-grade dysplasia have unsuspected adenocarcinoma? Arch. Pathol. Lab. Med. 129, 177–180 (2005).

    Article  PubMed  Google Scholar 

  160. Corley, D. A. et al. Impact of endoscopic surveillance on mortality from Barrett’s esophagus-associated esophageal adenocarcinomas. Gastroenterology 145, 312–319.e1 (2013).

    Article  PubMed  Google Scholar 

  161. Sikkema, M., de Jonge, P. J., Steyerberg, E. W. & Kuipers, E. J. Risk of esophageal adenocarcinoma and mortality in patients with Barrett’s esophagus: a systematic review and meta-analysis. Clin. Gastroenterol. Hepatol. 8, 235–244 (2010). quiz e232.

    Article  PubMed  Google Scholar 

  162. Martinez, P. et al. Evolution of Barrett’s esophagus through space and time at single-crypt and whole-biopsy levels. Nat. Commun. 9, 794 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  163. Ross-Innes, C. S. et al. Risk stratification of Barrett’s oesophagus using a non-endoscopic sampling method coupled with a biomarker panel: a cohort study. Lancet Gastroenterol. Hepatol. 2, 23–31 (2017).

    Article  PubMed  Google Scholar 

  164. Moinova, H. R. et al. Identifying DNA methylation biomarkers for non-endoscopic detection of Barrett’s esophagus. Sci. Transl. Med. 10, eaao5848 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  165. Jin, Z. et al. A multicenter, double-blinded validation study of methylation biomarkers for progression prediction in Barrett’s esophagus. Cancer Res. 69, 4112–4115 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Alvi, M. A. et al. DNA methylation as an adjunct to histopathology to detect prevalent, inconspicuous dysplasia and early-stage neoplasia in Barrett’s esophagus. Clin. Cancer Res. 19, 878–888 (2013).

    Article  CAS  PubMed  Google Scholar 

  167. Kaz, A. M. et al. DNA methylation profiling in Barrett’s esophagus and esophageal adenocarcinoma reveals unique methylation signatures and molecular subclasses. Epigenetics 6, 1403–1412 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Iyer, P. G. et al. Highly discriminant methylated DNA markers for the non-endoscopic detection of Barrett’s esophagus. Am. J. Gastroenterol. 113, 1156–1166 (2018).

    Article  CAS  PubMed  Google Scholar 

  169. Wang, Z. et al. Methylation biomarker panel performance in esophacap cytology samples for diagnosing Barrett’s esophagus: a prospective validation study. Clin. Cancer Res. 25, 2127–2135 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Maley, C. C. et al. Selectively advantageous mutations and hitchhikers in neoplasms: p16 lesions are selected in Barrett’s esophagus. Cancer Res. 64, 3414–3427 (2004).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge funding from: German Cancer Aid Society (Deutsche Krebshilfe) (M.S.), Cancer Research UK (R.J.H.), Cancer Research UK (A19771) (A.B. and T.A.G.), the US NIH via the Cancer Systems Biology Consortium U54 scheme (CA217376) (T.A.G.), German Research Foundation (DFG 3772/1) (M.Q.), Cancer Research UK PFA award (A21446) (S.A.C.M.) and a Cancer Research UK Grand Challenge award (STORMing Cancer, A29071) (S.A.C.M.).

Author information

Authors and Affiliations

Authors

Contributions

M.S. and R.J.H. wrote the first draft of the review. A.B., S.A.C.M., M.Q. and T.A.G. edited the review. All authors approved the final draft of the manuscript.

Corresponding author

Correspondence to Trevor A. Graham.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Gastroenterology & Hepatology thanks Rebecca Fitzgerald and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Glossary

Dysplasia

Neoplastic epithelium that remains confined within the basement membrane of the epithelium.

Fitness

The average contribution of a genotype to the next generation. The fitness of a genotype is manifested through its phenotype, which is also affected by the environment. Fitness generally promotes both survival and reproduction.

Clonal expansion

Spatial expansion of a clone through increased proliferation due to an advantageous phenotype.

Genetic diversity

The multitude of genetic variation on which selection can act.

Intratumoural heterogeneity

Diversity within individual tumours at the genetic, epigenetic and transcriptomic level.

Clone

A group of cells that share a common genotype as they descend from a common ancestor.

Loss of heterozygosity

(LOH). Allelic imbalance by which a heterozygous somatic cell becomes homozygous because one of the two alleles gets lost.

Fixation

When a genetic alteration reaches 100% frequency in a cell population.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schmidt, M., Hackett, R.J., Baker, AM. et al. Evolutionary dynamics in Barrett oesophagus: implications for surveillance, risk stratification and therapy. Nat Rev Gastroenterol Hepatol 19, 95–111 (2022). https://doi.org/10.1038/s41575-021-00531-4

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41575-021-00531-4

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing