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:

Extrachromosomal DNA amplifications in cancer

Abstract

Extrachromosomal DNA (ecDNA) amplification is an important driver alteration in cancer. It has been observed in most cancer types and is associated with worse patient outcome. The functional impact of ecDNA has been linked to its unique properties, such as its circular structure that is associated with altered chromatinization and epigenetic regulatory landscape, as well as its ability to randomly segregate during cell division, which fuels intercellular copy number heterogeneity. Recent investigations suggest that ecDNA is structurally more complex than previously anticipated and that it localizes to specialized nuclear bodies (hubs) and can act in trans as an enhancer for genes on other ecDNAs or chromosomes. In this Review, we synthesize what is currently known about how ecDNA is generated and how its genetic and epigenetic architecture affects proto-oncogene deregulation in cancer. We discuss how recently identified ecDNA functions may impact oncogenesis but also serve as new therapeutic vulnerabilities in cancer.

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: Oncogene amplification on ecDNA is a frequent event in cancer and promotes tumour heterogeneity.
Fig. 2: ecDNA life cycle.
Fig. 3: ecDNA movement and location during the cell cycle.
Fig. 4: Chromatin organization on ecDNA.
Fig. 5: The role of ecDNA and ecDNA hubs in transcriptional regulation.
Fig. 6: Opportunities for development of ecDNA-targeting therapeutic strategies.

Similar content being viewed by others

References

  1. ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium. Pan-cancer analysis of whole genomes. Nature 578, 82–93 (2020).

    Article  Google Scholar 

  2. Group, P. T. C. et al. Genomic basis for RNA alterations in cancer. Nature 578, 129–136 (2020).

    Article  Google Scholar 

  3. Verhaak, R. G. W., Bafna, V. & Mischel, P. S. Extrachromosomal oncogene amplification in tumour pathogenesis and evolution. Nat. Rev. Cancer 19, 283–288 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. deCarvalho, A. C. et al. Discordant inheritance of chromosomal and extrachromosomal DNA elements contributes to dynamic disease evolution in glioblastoma. Nat. Genet. 50, 708–717 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Turner, K. M. et al. Extrachromosomal oncogene amplification drives tumour evolution and genetic heterogeneity. Nature 543, 122–125 (2017). Together with deCarvalho et al. (2018), this work demonstrates that ecDNA is highly frequently observed in brain tumours, providing early suggestions that the ecDNA incidence in cancer is much higher than previously thought.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Cox, D., Yuncken, C. & Spriggs, A. I. Minute chromatin bodies in malignant tumours of childhood. Lancet 1, 55–58 (1965). This work presents the initial discovery of ecDNA in the nuclei of neoplastic cells.

    Article  CAS  PubMed  Google Scholar 

  7. Chapman, O. S. et al. The landscape of extrachromosomal circular DNA in medulloblastoma. Preprint at bioRxiv https://doi.org/10.1101/2021.10.18.464907 (2021).

    Article  Google Scholar 

  8. Zhao, X. K. et al. Focal amplifications are associated with chromothripsis events and diverse prognoses in gastric cardia adenocarcinoma. Nat. Commun. 12, 6489 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Kim, H. et al. Extrachromosomal DNA is associated with oncogene amplification and poor outcome across multiple cancers. Nat. Genet. 52, 891–897 (2020). This work presents the first whole-genome sequencing-based pan-cancer analysis of ecDNA amplifications across newly diagnosed tumours and cancer types.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Koche, R. P. et al. Extrachromosomal circular DNA drives oncogenic genome remodeling in neuroblastoma. Nat. Genet. 52, 29–34 (2020). This work uses whole-genome sequencing and, for the first time, circular DNA sequencing in neuroblastoma cancer samples to demonstrate how ecDNA impacts genome organization.

    Article  CAS  PubMed  Google Scholar 

  11. Morton, A. R. et al. Functional enhancers shape extrachromosomal oncogene amplifications. Cell 179, 1330–1341.e13 (2019). This study demonstrates that co-amplification of enhancers and oncogenes determines the genomic break points that define ecDNA boundaries.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wu, S. et al. Circular ecDNA promotes accessible chromatin and high oncogene expression. Nature 575, 699–703 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yi, E. et al. Live-cell imaging shows uneven segregation of extrachromosomal DNA elements and transcriptionally active extrachromosomal DNA hubs in cancer. Cancer Discov. 12, 468–483 (2022).

    Article  CAS  PubMed  Google Scholar 

  14. Weller, M. et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): a randomised, double-blind, international phase 3 trial. Lancet Oncol. 18, 1373–1385 (2017).

    Article  CAS  PubMed  Google Scholar 

  15. Liehr, T., Claussen, U. & Starke, H. Small supernumerary marker chromosomes (sSMC) in humans. Cytogenet. Genome Res. 107, 55–67 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Moller, H. D., Parsons, L., Jorgensen, T. S., Botstein, D. & Regenberg, B. Extrachromosomal circular DNA is common in yeast. Proc. Natl Acad. Sci. USA 112, E3114–E3122 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Paulsen, T., Kumar, P., Koseoglu, M. M. & Dutta, A. Discoveries of extrachromosomal circles of DNA in normal and tumor cells. Trends Genet. 34, 270–278 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Shibata, Y. et al. Extrachromosomal microDNAs and chromosomal microdeletions in normal tissues. Science 336, 82–86 (2012). This landmark study reports on the presence of small circular DNA elements in human cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Shoura, M. J. et al. Intricate and cell type-specific populations of endogenous circular DNA (eccDNA) in Caenorhabditis elegans and Homo sapiens. G3 7, 3295–3303 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Moller, H. D. et al. Circular DNA elements of chromosomal origin are common in healthy human somatic tissue. Nat. Commun. 9, 1069 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Noer, J. B., Horsdal, O. K., Xiang, X., Luo, Y. & Regenberg, B. Extrachromosomal circular DNA in cancer: history, current knowledge, and methods. Trends Genet. 38, 766–781 (2022).

    Article  CAS  PubMed  Google Scholar 

  22. Zhu, Y. et al. Oncogenic extrachromosomal DNA functions as mobile enhancers to globally amplify chromosomal transcription. Cancer Cell 39, 694–707.e7 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kanda, T., Sullivan, K. F. & Wahl, G. M. Histone–GFP fusion protein enables sensitive analysis of chromosome dynamics in living mammalian cells. Curr. Biol. 8, 377–385 (1998).

    Article  CAS  PubMed  Google Scholar 

  24. Hung, K. L. et al. ecDNA hubs drive cooperative intermolecular oncogene expression. Nature 600, 731–736 (2021). Together with Yi et al. (2022), this work reports the discovery that ecDNA molecules form clusters in which cargo gene transcription is strongly increased.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Balaban-Malenbaum, G. & Gilbert, F. Double minute chromosomes and the homogeneously staining regions in chromosomes of a human neuroblastoma cell line. Science 198, 739–741 (1977).

    Article  CAS  PubMed  Google Scholar 

  26. Nik-Zainal, S. et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature 534, 47–54 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Li, Y. et al. Patterns of somatic structural variation in human cancer genomes. Nature 578, 112–121 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hadi, K. et al. Distinct classes of complex structural variation uncovered across thousands of cancer genome graphs. Cell 183, 197–210.e32 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zack, T. I. et al. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 45, 1134–1140 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Scully, R., Panday, A., Elango, R. & Willis, N. A. DNA double-strand break repair-pathway choice in somatic mammalian cells. Nat. Rev. Mol. Cell Biol. 20, 698–714 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. L’Abbate, A. et al. MYC-containing amplicons in acute myeloid leukemia: genomic structures, evolution, and transcriptional consequences. Leukemia 32, 2152–2166 (2018).

    Article  PubMed Central  Google Scholar 

  32. Alt, F. W., Kellems, R. E., Bertino, J. R. & Schimke, R. T. Selective multiplication of dihydrofolate reductase genes in methotrexate-resistant variants of cultured murine cells. J. Biol. Chem. 253, 1357–1370 (1978).

    Article  CAS  PubMed  Google Scholar 

  33. Haber, D. A., Beverley, S. M., Kiely, M. L. & Schimke, R. T. Properties of an altered dihydrofolate reductase encoded by amplified genes in cultured mouse fibroblasts. J. Biol. Chem. 256, 9501–9510 (1981).

    Article  CAS  PubMed  Google Scholar 

  34. Haber, D. A. & Schimke, R. T. Unstable amplification of an altered dihydrofolate reductase gene associated with double-minute chromosomes. Cell 26, 355–362 (1981).

    Article  CAS  PubMed  Google Scholar 

  35. Beverley, S. M., Coderre, J. A., Santi, D. V. & Schimke, R. T. Unstable DNA amplifications in methotrexate-resistant Leishmania consist of extrachromosomal circles which relocalize during stabilization. Cell 38, 431–439 (1984).

    Article  CAS  PubMed  Google Scholar 

  36. Schimke, R. T. Gene amplification in cultured animal cells. Cell 37, 705–713 (1984).

    Article  CAS  PubMed  Google Scholar 

  37. Von Hoff, D. D. et al. Hydroxyurea accelerates loss of extrachromosomally amplified genes from tumor cells. Cancer Res. 51, 6273–6279 (1991).

    Google Scholar 

  38. Shoshani, O. et al. Chromothripsis drives the evolution of gene amplification in cancer. Nature 591, 137–141 (2021).

    Article  CAS  PubMed  Google Scholar 

  39. Storlazzi, C. T. et al. Gene amplification as double minutes or homogeneously staining regions in solid tumors: origin and structure. Genome Res. 20, 1198–1206 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kohl, N. E. et al. Transposition and amplification of oncogene-related sequences in human neuroblastomas. Cell 35, 359–367 (1983). This work identifies oncogene sequences on ecDNA amplifications.

    Article  CAS  PubMed  Google Scholar 

  41. L’Abbate, A. et al. Genomic organization and evolution of double minutes/homogeneously staining regions with MYC amplification in human cancer. Nucleic Acids Res. 42, 9131–9145 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Nonet, G. H., Carroll, S. M., DeRose, M. L. & Wahl, G. M. Molecular dissection of an extrachromosomal amplicon reveals a circular structure consisting of an imperfect inverted duplication. Genomics 15, 543–558 (1993).

    Article  CAS  PubMed  Google Scholar 

  43. Carroll, S. M. et al. Double minute chromosomes can be produced from precursors derived from a chromosomal deletion. Mol. Cell Biol. 8, 1525–1533 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Stark, G. R., Debatisse, M., Giulotto, E. & Wahl, G. M. Recent progress in understanding mechanisms of mammalian DNA amplification. Cell 57, 901–908 (1989).

    Article  CAS  PubMed  Google Scholar 

  45. Nathanson, D. A. et al. Targeted therapy resistance mediated by dynamic regulation of extrachromosomal mutant EGFR DNA. Science 343, 72–76 (2014). This work identifies ecDNA modulation as a mechanism of resistance to targeted drug treatment.

    Article  CAS  PubMed  Google Scholar 

  46. Song, K. et al. Plasticity of extrachromosomal and intrachromosomal BRAF amplifications in overcoming targeted therapy dosage challenges. Cancer Discov. 12, 1046–1069 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Deshpande, V. et al. Exploring the landscape of focal amplifications in cancer using AmpliconArchitect. Nat. Commun. 10, 392 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Steele, C. D. Signatures of copy number alterations in human cancer. Nature 606, 984–991 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Drews, R. M. A pan-cancer compendium of chromosomal instability. Nature 606, 976–983 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Bergstrom, E. N. et al. Mapping clustered mutations in cancer reveals APOBEC3 mutagenesis of ecDNA. Nature 602, 510–517 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Rosswog, C. et al. Chromothripsis followed by circular recombination drives oncogene amplification in human cancer. Nat. Genet. 53, 1673–1685 (2021).

    Article  CAS  PubMed  Google Scholar 

  52. Levan, A. & Levan, G. Have double minutes functioning centromeres? Hereditas 88, 81–92 (1978).

    Article  CAS  PubMed  Google Scholar 

  53. Shimizu, N., Itoh, N., Utiyama, H. & Wahl, G. M. Selective entrapment of extrachromosomally amplified DNA by nuclear budding and micronucleation during S phase. J. Cell Biol. 140, 1307–1320 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Shimizu, N., Kanda, T. & Wahl, G. M. Selective capture of acentric fragments by micronuclei provides a rapid method for purifying extrachromosomally amplified DNA. Nat. Genet. 12, 65–71 (1996).

    Article  CAS  PubMed  Google Scholar 

  55. Tanaka, T. & Shimizu, N. Induced detachment of acentric chromatin from mitotic chromosomes leads to their cytoplasmic localization at G1 and the micronucleation by lamin reorganization at S phase. J. Cell Sci. 113, 697–707 (2000).

    Article  CAS  PubMed  Google Scholar 

  56. Kanda, T., Otter, M. & Wahl, G. M. Mitotic segregation of viral and cellular acentric extrachromosomal molecules by chromosome tethering. J. Cell Sci. 114, 49–58 (2001).

    Article  CAS  PubMed  Google Scholar 

  57. Pope, B. D. et al. Topologically associating domains are stable units of replication-timing regulation. Nature 515, 402–405 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Barker, P. E., Drwinga, H. L., Hittelman, W. N. & Maddox, A. M. Double minutes replicate once during S phase of the cell cycle. Exp. Cell Res. 130, 353–360 (1980).

    Article  CAS  PubMed  Google Scholar 

  59. Itoh, N. & Shimizu, N. DNA replication-dependent intranuclear relocation of double minute chromatin. J. Cell Sci. 111, 3275–3285 (1998).

    Article  CAS  PubMed  Google Scholar 

  60. Barker, P. E. & Hsu, T. C. Are double minutes chromosomes? Exp. Cell Res. 113, 456–458 (1978).

    Article  CAS  PubMed  Google Scholar 

  61. Lundberg, G. et al. Binomial mitotic segregation of MYCN-carrying double minutes in neuroblastoma illustrates the role of randomness in oncogene amplification. PLoS ONE 3, e3099 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Xue, Y. et al. An approach to suppress the evolution of resistance in BRAFV600E-mutant cancer. Nat. Med. 23, 929–937 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Lange, J. T. et al. Principles of ecDNA random inheritance drive rapid genome change and therapy resistance in human cancers. Preprint at bioRxiv https://doi.org/10.1101/2021.06.11.447968 (2021).

    Article  Google Scholar 

  64. Clarke, T. L. et al. Histone lysine methylation dynamics control EGFR DNA copy-number amplification. Cancer Discov. 10, 306–325 (2020).

    Article  CAS  PubMed  Google Scholar 

  65. Johnson, K. C. et al. Single-cell multimodal glioma analyses identify epigenetic regulators of cellular plasticity and environmental stress response. Nat. Genet. 53, 1456–1468 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Nikolaev, S. et al. Extrachromosomal driver mutations in glioblastoma and low-grade glioma. Nat. Commun. 5, 5690 (2014).

    Article  CAS  PubMed  Google Scholar 

  67. Bannister, A. J. & Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 21, 381–395 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Spielmann, M., Lupianez, D. G. & Mundlos, S. Structural variation in the 3D genome. Nat. Rev. Genet. 19, 453–467 (2018).

    Article  CAS  PubMed  Google Scholar 

  69. Chen, W. et al. Sequencing of methylase-accessible regions in integral circular extrachromosomal DNA reveals differences in chromatin structure. Epigenetics Chromatin 14, 40 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Helmsauer, K. et al. Enhancer hijacking determines extrachromosomal circular MYCN amplicon architecture in neuroblastoma. Nat. Commun. 11, 5823 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Blumrich, A. et al. The FRA2C common fragile site maps to the borders of MYCN amplicons in neuroblastoma and is associated with gross chromosomal rearrangements in different cancers. Hum. Mol. Genet. 20, 1488–1501 (2011).

    Article  CAS  PubMed  Google Scholar 

  72. Purshouse, K. et al. Oncogene expression from extrachromosomal DNA is driven by copy number amplification and does not require spatial clustering. Preprint at bioRxiv https://doi.org/10.1101/2022.01.29.478046 (2022).

    Article  Google Scholar 

  73. Meldi, L. & Brickner, J. H. Compartmentalization of the nucleus. Trends Cell Biol. 21, 701–708 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Chuang, C. H. et al. Long-range directional movement of an interphase chromosome site. Curr. Biol. 16, 825–831 (2006).

    Article  CAS  PubMed  Google Scholar 

  75. Osborne, C. S. et al. Active genes dynamically colocalize to shared sites of ongoing transcription. Nat. Genet. 36, 1065–1071 (2004).

    Article  CAS  PubMed  Google Scholar 

  76. Delmore, J. E. et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146, 904–917 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Roskoski, R. Jr. Properties of FDA-approved small molecule protein kinase inhibitors: a 2021 update. Pharmacol. Res. 165, 105463 (2021).

    Article  CAS  PubMed  Google Scholar 

  78. Umate, P., Tuteja, N. & Tuteja, R. Genome-wide comprehensive analysis of human helicases. Commun. Integr. Biol. 4, 118–137 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Szczesny, R. J. et al. Yeast and human mitochondrial helicases. Biochim. Biophys. Acta 1829, 842–853 (2013).

    Article  CAS  PubMed  Google Scholar 

  80. Yu, L. et al. Gemcitabine eliminates double minute chromosomes from human ovarian cancer cells. PLoS ONE 8, e71988 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Von Hoff, D. D. et al. Elimination of extrachromosomally amplified MYC genes from human tumor cells reduces their tumorigenicity. Proc. Natl Acad. Sci. USA 89, 8165–8169 (1992).

    Article  Google Scholar 

  82. Rajendran, L., Knolker, H. J. & Simons, K. Subcellular targeting strategies for drug design and delivery. Nat. Rev. Drug Discov. 9, 29–42 (2010).

    Article  CAS  PubMed  Google Scholar 

  83. Petros, R. A. & DeSimone, J. M. Strategies in the design of nanoparticles for therapeutic applications. Nat. Rev. Drug Discov. 9, 615–627 (2010).

    Article  CAS  PubMed  Google Scholar 

  84. Pouton, C. W., Wagstaff, K. M., Roth, D. M., Moseley, G. W. & Jans, D. A. Targeted delivery to the nucleus. Adv. Drug Deliv. Rev. 59, 698–717 (2007).

    Article  CAS  PubMed  Google Scholar 

  85. Luebeck, J. et al. AmpliconReconstructor integrates NGS and optical mapping to resolve the complex structures of focal amplifications. Nat. Commun. 11, 4374 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Prada-Luengo, I., Krogh, A., Maretty, L. & Regenberg, B. Sensitive detection of circular DNAs at single-nucleotide resolution using guided realignment of partially aligned reads. BMC Bioinforma. 20, 663 (2019).

    Article  CAS  Google Scholar 

  87. Zheng, S. et al. A survey of intragenic breakpoints in glioblastoma identifies a distinct subset associated with poor survival. Genes Dev. 27, 1462–1472 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Hung, K. L. et al. Targeted profiling of human extrachromosomal DNA by CRISPR-CATCH. Preprint at bioRxiv https://doi.org/10.1101/2021.11.28.470285 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Kumar, P. et al. ATAC-seq identifies thousands of extrachromosomal circular DNA in cancer and cell lines. Sci. Adv. 6, eaba2489 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Rajkumar, U. et al. ecSeg: semantic segmentation of metaphase images containing extrachromosomal DNA. iScience 21, 428–435 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank K. Seburn for providing constructive feedback while writing the manuscript (The Jackson Laboratory (JAX), Research Program Development). They thank C.-l. Wei (JAX) and R. Koche (Memorial Sloan–Kettering Cancer Center) for helpful discussions. This work was supported by grants from the US National Institutes of Health (NIH) (R01 CA237208, R21 NS114873, R21 CA256575, R33 CA236681, OT2 CA278649 and Cancer Center Support Grant P30 CA034196 (R.G.W.V.)), Cancer Research UK (reference 270422-0523 to R.G.W.V.) and a grant from the Brain Tumour Charity (R.G.W.V.). E.Y. is supported by a basic research fellowship from the American Brain Tumour Association (BRF1800014). A.G.H. is supported by the Deutsche Forschungsgemeinschaft (DFG; German Research Foundation) (398299703, the eDynamic Cancer Grand Challenge) and the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (grant agreement No. 949172). R.C.G. is supported by a fellowship from the ‘la Caixa’ Foundation (ID 100010434). The fellowship code is LCF/BQ/EU20/11810051.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to all aspects of the article.

Corresponding authors

Correspondence to Anton G. Henssen or Roel G. W. Verhaak.

Ethics declarations

Competing interests

R.G.W.V. is a co-founder and/or adviser of Boundless Bio and Stellanova Therapeutics. There is no commercial interest or intellectual property associated with this work. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Genetics thanks Lukas Chavez, Ofer Shoshani and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note

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

Glossary

Proto-oncogenes

Genes involved in normal cell growth that, when abnormally activated, lead or contribute to cancer development.

Focal amplification

A DNA region that only spans a sub-chromosomal arm proportion of the chromosome and is amplified at a high level; that is, more than eight copies.

Intratumoural heterogeneity

The differences among cancer cells within the same tumour.

Cargo gene

Any gene (or genes) harboured on the sequence of an extrachromosomal DNA (ecDNA) element.

Homogeneously staining regions

(HSRs). Chromosomal regions with DNA amplification presenting a uniformed staining pattern with Giemsa nucleic acid stain.

Enhancer hijacking

A process in which a somatic structural genomic rearrangement brings an enhancer into physical proximity of a gene it does not normally interact with, and activates it ectopically.

Hemizygous

The type of zygosity in which only one allele contains a gene or mutation.

Phasing

The process of inferring haplotype information of a sequence from genomic data.

Chromothripsis

A massive chromosomal rearrangement resulting from a chromosome shattering event, characterized by more than 20 DNA fragments stitched together in an abnormal order.

Breakage–fusion–bridge cycles

(BFBs). A mechanism of chromosomal instability caused by a cycle of telomere breaks and dicentric chromosome formation.

Mitosis

A cellular process in which replicated genetic information in a single cell is divided into two identical nuclei.

Micronuclei

The small nuclear structures that reside in the cytoplasm and contain damaged DNA fragments which were not incorporated into the main nucleus after mitosis.

ecDNA concatenation

(Extrachromosomal DNA concatenation). A structure in which two or more closed circular DNAs are interlinked.

Chromatin accessibility

The extent to which proteins are able to interact with chromatinized DNA, which is regulated through nucleosome occupancy and other factors occluding access to DNA.

Transcription factories

Molecular complexes consisting of extrachromosomal DNAs (ecDNAs) and transcription machinery components, with high transcriptional activity of ecDNA sequences.

Synthetic aneuploidy effect

Increased transcriptional activity of regions of the genome where extrachromosomal DNAs (ecDNAs) make a physical connection, similar to the effect of aneuploidy.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yi, E., Chamorro González, R., Henssen, A.G. et al. Extrachromosomal DNA amplifications in cancer. Nat Rev Genet 23, 760–771 (2022). https://doi.org/10.1038/s41576-022-00521-5

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41576-022-00521-5

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer