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.

  • Article
  • Published:

Cancer aneuploidies are shaped primarily by effects on tumour fitness

Abstract

Aneuploidies—whole-chromosome or whole-arm imbalances—are the most prevalent alteration in cancer genomes1,2. However, it is still debated whether their prevalence is due to selection or ease of generation as passenger events1,2. Here we developed a method, BISCUT, that identifies loci subject to fitness advantages or disadvantages by interrogating length distributions of telomere- or centromere-bounded copy-number events. These loci were significantly enriched for known cancer driver genes, including genes not detected through analysis of focal copy-number events, and were often lineage specific. BISCUT identified the helicase-encoding gene WRN as a haploinsufficient tumour-suppressor gene on chromosome 8p, which is supported by several lines of evidence. We also formally quantified the role of selection and mechanical biases in driving aneuploidy, finding that rates of arm-level copy-number alterations are most highly correlated with their effects on cellular fitness1,2. These results provide insight into the driving forces behind aneuploidy and its contribution to tumorigenesis.

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: Prevalence and characteristics of different types of SCNA.
Fig. 2: BISCUT identifies known and new cancer driver genes through analysis of SCNA length distributions.
Fig. 3: Validation of genes identified by BISCUT for negative and positive selection.
Fig. 4: Pan-cancer mechanical coefficients and RF.

Similar content being viewed by others

Data availability

All SNP array data used for analysis are publicly available from TCGA’s Genomic Data Commons Data Portal at https://portal.gdc.cancer.gov/. All DNA and RNA sequencing data generated in this study are available at the NIH Sequence Read Archive (accession code PRJNA967303).

Code availability

The codes used to merge copy-number segments, call partial-SCNAs, detect loci under selection and determine RF values and mechanical coefficients are freely available for download at https://github.com/beroukhim-lab/BISCUT-py3 and https://doi.org/10.5281/zenodo.7896522.

References

  1. Weaver, B. A. & Cleveland, D. W. Does aneuploidy cause cancer? Curr. Opin. Cell Biol. 18, 658–667 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Taylor, A. M. et al. Genomic and functional approaches to understanding cancer aneuploidy. Cancer Cell 33, 676–689 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Boveri, T. Concerning the origin of malignant tumours by Theodor Boveri. Translated and annotated by Henry Harris. J. Cell Sci. 121, 1–84 (2008).

    Article  PubMed  Google Scholar 

  4. Holland, A. J. & Cleveland, D. W. Boveri revisited: chromosomal instability, aneuploidy and tumorigenesis. Nat. Rev. Mol. Cell Biol. 10, 478–487 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sheltzer, J. M. et al. Single-chromosome gains commonly function as tumor suppressors. Cancer Cell 31, 240–255 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Pavelka, N. et al. Aneuploidy confers quantitative proteome changes and phenotypic variation in budding yeast. Nature 468, 321–325 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ly, P. et al. Characterization of aneuploid populations with trisomy 7 and 20 derived from diploid human colonic epithelial cells. Neoplasia 13, 348–357 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Rutledge, S. D. et al. Selective advantage of trisomic human cells cultured in non-standard conditions. Sci. Rep. 6, 22828 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Sunshine, A. B. et al. The fitness consequences of aneuploidy are driven by condition-dependent gene effects. PLoS Biol. 13, e1002155 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Ravichandran, M. C., Fink, S., Clarke, M. N., Hofer, F. C. & Campbell, C. S. Genetic interactions between specific chromosome copy number alterations dictate complex aneuploidy patterns. Genes Dev. 32, 1485–1498 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hughes, T. R. et al. Widespread aneuploidy revealed by DNA microarray expression profiling. Nat. Genet. 25, 333–337 (2000).

    Article  CAS  PubMed  Google Scholar 

  12. Liu, G. et al. Gene essentiality is a quantitative property linked to cellular evolvability. Cell 163, 1388–1399 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Salehi, S. et al. Clonal fitness inferred from time-series modelling of single-cell cancer genomes. Nature 595, 585–590 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kimmel, G. J. et al. Intra-tumor heterogeneity, turnover rate and karyotype space shape susceptibility to missegregation-induced extinction. PLoS Comput. Biol. 19, e1010815 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lee, A. J. X. et al. Chromosomal instability confers intrinsic multidrug resistance. Cancer Res. 71, 1858–1870 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Cai, Y. et al. Loss of chromosome 8p governs tumor progression and drug response by altering lipid metabolism. Cancer Cell 29, 751–766 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Uno, N. et al. CRISPR/Cas9-induced transgene insertion and telomere-associated truncation of a single human chromosome for chromosome engineering in CHO and A9 cells. Sci. Rep. 7, 12739 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  18. Mermel, C. H. et al. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol. 12, R41 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Brennan, C. W. et al. The somatic genomic landscape of glioblastoma. Cell 155, 462–477 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cimini, D. Merotelic kinetochore orientation, aneuploidy, and cancer. Biochim. Biophys. Acta 1786, 32–40 (2008).

    CAS  PubMed  Google Scholar 

  21. Blackford, A. N. & Stucki, M. How cells respond to DNA breaks in mitosis. Trends Biochem. Sci. 45, 321–331 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Martincorena, I. et al. Universal patterns of selection in cancer and somatic tissues. Cell 171, 1029–1041 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hoadley, K. A. et al. Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell 173, 291–304 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Beroukhim, R. et al. The landscape of somatic copy-number alteration across human cancers. Nature 463, 899–905 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  Google Scholar 

  26. Xue, W. et al. A cluster of cooperating tumor-suppressor gene candidates in chromosomal deletions. Proc. Natl Acad. Sci. USA 109, 8212–8217 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chan, E. M. et al. WRN helicase is a synthetic lethal target in microsatellite unstable cancers. Nature 568, 551–556 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bailey, M. H. et al. Comprehensive characterization of cancer driver genes and mutations. Cell 173, 371–385 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ciriello, G. et al. Emerging landscape of oncogenic signatures across human cancers. Nat. Genet. 45, 1127–1133 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Nichols, C. A. et al. Loss of heterozygosity of essential genes represents a widespread class of potential cancer vulnerabilities. Nat. Commun. 11, 2517 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. McFarland, J. M. et al. Improved estimation of cancer dependencies from large-scale RNAi screens using model-based normalization and data integration. Nat. Commun. 9, 4610 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  32. Dempster, J. M. et al. Chronos: a cell population dynamics model of CRISPR experiments that improves inference of gene fitness effects. Genome Biol. 22, 343 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Davoli, T. et al. Cumulative haploinsufficiency and triplosensitivity drive aneuploidy patterns and shape the cancer genome. Cell 155, 948–962 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dewhurst, S. M. et al. Tolerance of whole-genome doubling propagates chromosomal instability and accelerates cancer genome evolution. Cancer Discov. 4, 175–185 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. López, S. et al. Interplay between whole-genome doubling and the accumulation of deleterious alterations in cancer evolution. Nat. Genet. 52, 283–293 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  36. 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 

  37. Liu, Y. et al. Systematic proteome and proteostasis profiling in human Trisomy 21 fibroblast cells. Nat. Commun. 8, 1212 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  38. Hose, J. et al. Dosage compensation can buffer copy-number variation in wild yeast. Elife 4, e05462 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Stenberg, P. et al. Buffering of segmental and chromosomal aneuploidies in Drosophila melanogaster. PLoS Genet. 5, e1000465 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Muenzner, J. et al. The natural diversity of the yeast proteome reveals chromosome-wide dosage compensation in aneuploids. Preprint at bioRxiv https://doi.org/10.1101/2022.04.06.487392 (2022).

  41. Dumont, M. et al. Human chromosome-specific aneuploidy is influenced by DNA-dependent centromeric features. EMBO J. 39, e102924 (2020).

    Article  CAS  PubMed  Google Scholar 

  42. Klaasen, S. J. et al. Nuclear chromosome locations dictate segregation error frequencies. Nature 607, 604–609 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bersani, F. et al. Pericentromeric satellite repeat expansions through RNA-derived DNA intermediates in cancer. Proc. Natl Acad. Sci. USA 112, 15148–15153 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  44. Quinodoz, S. A. et al. Higher-order inter-chromosomal hubs shape 3D genome organization in the nucleus. Cell 174, 744–757 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kitagawa, K. & Hieter, P. Evolutionary conservation between budding yeast and human kinetochores. Nat. Rev. Mol. Cell Biol. 2, 678–687 (2001).

    Article  CAS  PubMed  Google Scholar 

  46. Barra, V. & Fachinetti, D. The dark side of centromeres: types, causes and consequences of structural abnormalities implicating centromeric DNA. Nat. Commun. 9, 4340 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  47. Knutsen, T. et al. Definitive molecular cytogenetic characterization of 15 colorectal cancer cell lines. Genes Chromosomes Cancer 49, 204–223 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Nurk, S. et al. The complete sequence of a human genome. Science 376, 44–53 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  49. Altemose, N. et al. Complete genomic and epigenetic maps of human centromeres. Science 376, eabl4178 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Gregan, J., Polakova, S., Zhang, L., Tolić-Nørrelykke, I. M. & Cimini, D. Merotelic kinetochore attachment: causes and effects. Trends Cell Biol. 21, 374–381 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Gisselsson, D. et al. Telomere dysfunction triggers extensive DNA fragmentation and evolution of complex chromosome abnormalities in human malignant tumors. Proc. Natl Acad. Sci. USA 98, 12683–12688 (2001).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wise, J. L. et al. Human telomere length correlates to the size of the associated chromosome arm. PLoS ONE 4, e6013 (2009).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  53. McCarroll, S. A. et al. Integrated detection and population-genetic analysis of SNPs and copy number variation. Nat. Genet. 40, 1166–1174 (2008).

    Article  CAS  PubMed  Google Scholar 

  54. Korn, J. M. et al. Integrated genotype calling and association analysis of SNPs, common copy number polymorphisms and rare CNVs. Nat. Genet. 40, 1253–1260 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Cancer Genome Atlas Research, Network. Integrated genomic analyses of ovarian carcinoma. Nature 474, 609–615 (2011).

    Article  Google Scholar 

  56. Gao, G. F. et al. Tangent normalization for somatic copy-number inference in cancer genome analysis. Bioinformatics 38, 4677–4686 (2022).

  57. Olshen, A. B., Venkatraman, E. S., Lucito, R. & Wigler, M. Circular binary segmentation for the analysis of array-based DNA copy number data. Biostatistics 5, 557–572 (2004).

    Article  PubMed  MATH  Google Scholar 

  58. 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 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  60. Gao, Q. et al. Driver fusions and their implications in the development and treatment of human cancers. Cell Rep. 23, 227–238 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Saghafinia, S., Mina, M., Riggi, N., Hanahan, D. & Ciriello, G. Pan-cancer landscape of aberrant DNA methylation across human tumors. Cell Rep. 25, 1066–1080 (2018).

    Article  CAS  PubMed  Google Scholar 

  62. Delignette-Muller, M. L. & Dutang, C. fitdistrplus: An R package for fitting distributions. J. Stat. Softw. 64, 1–34 (2015).

  63. Benjamini, Y. & Yekutieli, D. The control of the false discovery rate in multiple testing under dependency. Ann. Stat. 29, 1165–1188 (2001).

    Article  MathSciNet  MATH  Google Scholar 

  64. Miotto, B., Ji, Z. & Struhl, K. Selectivity of ORC binding sites and the relation to replication timing, fragile sites, and deletions in cancers. Proc. Natl Acad. Sci. USA 113, E4810–E4819 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  65. Sondka, Z. et al. The COSMIC Cancer Gene Census: describing genetic dysfunction across all human cancers. Nat. Rev. Cancer 18, 696–705 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Endres, D. M. & Schindelin, J. E. A new metric for probability distributions. IEEE Trans. Inf. Theory 49, 1858–1860 (2003).

    Article  MathSciNet  MATH  Google Scholar 

  67. Lin, J. Divergence measures based on the Shannon entropy. IEEE Trans. Inf. Theory 37, 145–151 (1991).

    Article  MathSciNet  MATH  Google Scholar 

  68. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Gel, B. et al. regioneR: an R/Bioconductor package for the association analysis of genomic regions based on permutation tests. Bioinformatics 32, 289–291 (2016).

    Article  CAS  PubMed  Google Scholar 

  70. Kim, S. Y., Jacob, L. & Speed, T. P. Combining calls from multiple somatic mutation-callers. BMC Bioinform. 15, 154 (2014).

    Article  Google Scholar 

  71. Kasar, S. et al. Whole-genome sequencing reveals activation-induced cytidine deaminase signatures during indolent chronic lymphocytic leukaemia evolution. Nat. Commun. 6, 8866 (2015).

    Article  ADS  CAS  PubMed  Google Scholar 

  72. Kim, J. et al. Somatic ERCC2 mutations are associated with a distinct genomic signature in urothelial tumors. Nat. Genet. 48, 600–606 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Knijnenburg, T. A. et al. Genomic and molecular landscape of DNA damage repair deficiency across The Cancer Genome Atlas. Cell Rep. 23, 239–254 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Lundberg, A. S. et al. Immortalization and transformation of primary human airway epithelial cells by gene transfer. Oncogene 21, 4577–4586 (2002).

    Article  CAS  PubMed  Google Scholar 

  75. Adalsteinsson, V. A. et al. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat. Commun. 8, 1324 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  76. DePristo, M. A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 43, 491–498 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Van der Auwera, G. & O’Connor, B. Genomics in the Cloud: Using Docker, GATK, and WDL in Terra (O’Reilly Media, 2020).

Download references

Acknowledgements

We thank the members of the laboratories of R.B., A.M.T. and M.M. for their support and helpful discussions, as well as C. Stewart, C.-Z. Zhang and S. Schumacher. We also thank G. Macintyre for suggesting the stick-breaking model to explain partial-SCNA length distributions, I. Martincorena for personal communications regarding negative selection in cancer, E. Chan for the WRN overexpression construct and M. Huang for the name of the main method. For flow cytometry analyses, we acknowledge National Institutes of Health P30CA013696 (HICCC flow cytometry core), and for RNA sequencing of cell lines we acknowledge the Sulzberger Genome Center. We also acknowledge the following sources of financial support: the National Institutes of Health and National Cancer Institute (R.B. and A.M.T.), National Institute for General Medical Sciences (A.M.T.), Fund for Innovative Cancer Informatics (R.B.), Gray Matters Brain Cancer Foundation (R.B.), Pediatric Brain Tumor Foundation (R.B.), Brain Tumour Charity (R.B.) and St. Baldrick’s Foundation (R.B.).

Author information

Authors and Affiliations

Authors

Contributions

J.S., A.D.C., A.M.T. and R.B. conceived of this study. J.S., G.F.G., L.F.S., A.C.B., A.D.C. and R.B. developed analytic methods. J.S. and S.S. implemented analytic methods. J.S., S.S., S.Z., S.H.H., N.Z.-K., Y.G., H.S. and A.M.T. carried out computational analyses. N.Z.-K., Y.G., M.S.C. and A.M.T. developed and carried out in vitro experiments. G.H., V.R. and M.M. provided feedback and advice on analyses. A.D.C., A.M.T. and R.B. supervised the work. J.S., N.Z.-K., S.S., S.Z., A.M.T. and R.B. wrote the manuscript with input from all coauthors.

Corresponding authors

Correspondence to Alison M. Taylor or Rameen Beroukhim.

Ethics declarations

Competing interests

G.F.G., A.C.B., A.D.C. and M.M. receive or received research support from Bayer AG. M.M. and A.M.T. received research support from Ono Pharmaceutical. M.M. is an equity holder of, consultant for, and Scientific Advisory Board chair for OrigiMed. M.M. additionally receives research support from Novo Nordisk and Janssen Pharmaceuticals, consults for Interline Therapeutics, and is an inventor of a patent for EGFR mutation diagnosis in lung cancer, licensed to Labcorp. R.B. consults for and owns equity in Scorpion Therapeutics and receives research support from Novartis. J.S., S.S., S.Z., N.Z.-K., Y.G., S.H.H., M.S.C., L.F.S., G.H., V.R. and H.S. declare no competing interests.

Peer review

Peer review information

Nature thanks Geoff Macintyre and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Additional information on different types of SCNA and the BISCUT method.

(a) Empirical examples of low centromeric mechanical bias (1q telomere-bounded deletions, for which the ratio of breakpoints occurring in the centromere over those occurring in the arm is less than 1), and high centromeric mechanical bias (5p telomere-bounded amplifications, for which the centromere/arm breakpoint ratio is much greater than 1). Within the chromosome arm, bins are 1 Mb large. (b) Mean amplification and deletion breakpoint density within chromosome arms, aggregated across all tumors and all chromosome arms (n = 67; binned by Mb), versus breakpoint density within all centromeres (values in breakpoints per megabase). Error bars represent the 95% confidence interval for the mean. C/A Ratio represents centromeric breaks over arm breaks. (c) Comparison of length distributions of telomere-bounded, centromere-bounded, and interstitial amplifications and deletions, aggregated across all chromosome arms. (d) Example depicting BISCUT’s recursion steps. From top to bottom: BISCUT detects peaks iteratively, walking both left and right if a significant peak is detected, with the new boundaries including the detected peak. If a peak is not detected, overlaps with a previous peak, or there are fewer than 4 samples, the analysis is stopped. See Fig. 2c and Methods for details.

Extended Data Fig. 2 Pan-cancer BISCUT analysis.

(a) Summary statistics of the four types of BISCUT peaks in pan-cancer. (b) Sizes of peaks (in bases) from the pan-cancer BISCUT analysis. From left to right, peaks are categorized by direction of selection (n = 90 and 103 for positive and negative selection respectively), direction of copy number imbalance (n = 80 and 113 for amplifications and deletions respectively), and origin of partial-SCNA (n = 163 and 30 for telomere-bounded and centromere-bounded respectively). Two-tailed p-value was calculated using a Mann-Whitney U test. (c) Overlap between genes in BISCUT peaks and Tier 1 COSMIC cancer genes. The numbers of peaks containing these genes are depicted in green. A one-tailed p-value was calculated using a permutation test as outlined in the Methods. (d) Negative selection peaks from the pan-cancer BISCUT analysis, sorted from highest to lowest by fraction of samples subject to these fitness effects that also possessed overlapping focal SCNAs in the opposite direction. Peaks that overlap with GISTIC 2.0 peaks are denoted in dark red and dark blue. (e) BISCUT analysis detecting two positive selection peaks (top: 9p telomere-bounded deletions, overlapping with CDKN2A focal deletions; bottom: 8q telomere-bounded amplifications, overlapping with MYC focal amplifications) with focal SCNAs removed (left) and with focal SCNAs included (right). (f) BISCUT analysis detecting two negative selection peaks (top: 8q telomere-bounded deletions, overlapping with MYC focal amplifications; bottom: 11q telomere-bounded deletions, overlapping with YAP1/BIRC3 focal amplifications) with focal SCNAs removed (left) and with focal SCNAs included (right).

Extended Data Fig. 3 Lineage-specific divergence of breakpoint distributions from the background distribution.

Heatmaps of lineage divergence scores for each tumor type (x-axis) and chromosome arm (y-axis). Amplifications are on top (in red) and deletions are on the bottom (in blue). Darker color represents a higher divergence score.

Extended Data Fig. 4 Patterns of chromosome 3p deletions are highly lineage-specific.

(a) Lineage-specificity scores across chromosomes. The left chromatid is shaded in red and represents amplifications, whereas the right chromatid is shaded in blue and represents deletions. Darker colors indicate greater lineage-specificity. (b) BISCUT analysis of telomere-bounded deletions on chromosome 3p in three different cohorts. The top panels display telomere-bounded deletions, sorted by length. The bottom panels show the vertical distance of each tel-SCNA from the background distribution; the maximum deviation is denoted by the solid vertical line. The dashed lines represent the peak regions determined to be under significant positive selection (i.e. conferring survival advantage in this cohort). (c) Genomic locations and corresponding significance score of positive selection deletion BISCUT peaks on chromosome 3p across lineages. See Supplementary Table 2a for tumor type abbreviations.

Extended Data Fig. 5 Hierarchical clustering of BISCUT peaks across lineages.

Matrix of significantly recurring BISCUT peaks across 33 independent tumor types. Peaks are sorted by genomic location (vertical axis), with four distinct classes of peaks in dark red (positive selection in amplifications), light red (negative selection in deletions), light blue (negative selection in amplifications), and dark blue (positive selection in deletions). Tumor types are sorted and color-coded (k = 5) according to hierarchical clustering by Ward’s method (horizontal axis).

Extended Data Fig. 6 Cells engineered with chr8p deletion for validation of genes in BISCUT selection peaks.

(a) Schematic for 8p deletion approach. Cells were transfected with a CRISPR targeting 8p just outside the centromere and with a linearized plasmid containing an artificial telomere, puromycin selection cassette, and 1 kilobase of sequence homologous to the 8p pericentromeric sequence. Puromycin selection was used to isolate cells with 8p replaced by the artificial telomere. (b) ichorCNA output of ultra-low-pass whole genome sequencing data from five AALE cell clones with 8p disomy or 8p monosomy. Horizontal axis is chromosome number, vertical axis is log copy number ratio. Green denotes copy number loss, red denotes copy number gain. (c) Caspase-glo for cells with 8p deletion compared to cells with 8p disomy (n = 3 for both). Each point represents one biological replicate from a representative experiment. One-tailed p-values from two different experiments were combined using Fisher’s method. (d) Flow cytometry analysis of cells with 8p deletion compared to cells with 8p disomy. Bar graphs represent the percentage of apoptotic cells dually stained for Annexin V and PI. One representative experiment is shown. One-tailed p-values from three independent experiments were combined using Fisher’s method. (e) Vertical axis represents normalized read counts from RNA sequencing of cells with 8p disomy or 8p deletion. Each point is an individual clone (n = 8 for all columns). Two-tailed p-values are reported. (f) Relative COSMIC SBS39 mutational signature activity (vertical axis) of engineered cells with 8p disomy versus 8p monosomy. Two-tailed p-values are calculated from a Mann-Whitney U test. (g) WRN qPCR for cell clones with 8p disomy after siRNA treatment. Cells were treated with either control siRNA or siRNA against WRN for 3 days prior to qPCR (n = 2 for each condition). Each point represents the average value across technical replicates in an individual biological replicate. h) Percentages of apoptotic cells detected by flow cytometry for Annexin V and propidium iodide (PI) across three 8p wild-type cell lines, on day three after transfection with WRN versus control siRNAs. This is representative data from one of four experiments. A ratio paired t-test was used to calculate one-sided p-values for all four experiments, which were combined using Fisher’s method. (i) Log-fold changes in apoptotic cells detected by trypan blue across these three 8p wild-type cell lines (n = 3 for all cell lines), on day three after transfection with WRN vs control siRNAs. Each point represents a different experiment. One-tailed p-values from all experiments were combined using Fisher’s method. (j) WRN qPCR in 8p disomic cell clones with overexpression of WRN or GFP (n = 3 for both). A two-tailed p-value is reported. (k) Cell viability is significantly lower when genes in del-neg peaks are knocked down by RNAi (left, DEMETER2 score) or knocked out by CRISPR (right, Chronos score) in Dependency Map screens31,32, compared to all other genes. The reported p-value is two-tailed. Box plots center on median values and extend to the first and third quartiles; the whiskers extend to 1.5 times the interquartile range. (l) KAT6A qPCR for cell clones three days after siRNA-mediated knockdown (n = 3 for both conditions). A two-tailed p-value is reported. (m) EPN2 qPCR for cell clones three days after siRNA-mediated knockdown (n = 3 for both conditions). The reported p-value is two-tailed. All p-values in this figure were calculated using Student’s t-test except as otherwise noted; no adjustments were made for multiple comparisons.

Extended Data Fig. 7 Quantitative assessment of selective and mechanical pressures driving aneuploidy.

(a) Calculation of peak-specific relative fitness (RF), arm-level RF, telomeric mechanical coefficients, and chromosome-level centromeric mechanical coefficients. See Methods for further details. (b) Centromeric mechanical coefficients (log) plotted against centromere length (in bases). (c) Centromeric mechanical coefficients (log) plotted against total frequency of arm-SCNAs affecting a specific chromosome (i.e. amplifications and deletions of the p and q arms in aggregate). Acrocentric chromosomes are excluded from analysis. (d) From the original BISCUT analysis: telomeric mechanical coefficients (log) plotted against telomere length, in RTLU, for amplifications (left; in red) and deletions (right; in blue). (e) From the original BISCUT analysis: telomeric mechanical coefficients (log) plotted against frequency of arm-level amplifications (left; in red) and deletions (right; in blue). For all panels, two-tailed p-values and rho correlation coefficients were calculated using Spearman’s rank correlation. No adjustments were made for multiple comparisons.

Extended Data Fig. 8 Telomeric mechanical pressures are better reflected when using baseline ploidies of 2 or 4.

(a) Relative fitness (log) plotted against frequency of arm-SCNAs. From left to right: positive selection in amplifications (dark red), negative selection in amplifications (light blue), positive selection in deletions (dark blue), and negative selection in deletions (light red). (b) Missegregation probability in percentage (determined by single-cell sequencing of RPE1-hTERT non-transformed cells42) plotted against frequency of all arm-SCNAs affecting each chromosome, averaged across arms. The horizontal black line at 4.3% reflects the expected random chance of missegregation of each chromosome. (c) Relative fitness (log) plotted against arm length. (d) Chromosome arm length plotted against frequency of arm-SCNAs. (e) Strength of correlation (β; vertical axis) between various coefficients (horizontal axis) and arm-SCNA rates from a multivariate Generalized Linear Model (GLM), with p-values above each predictor (significant values in bold). Amplifications are in red, and deletions are in blue. All p-values in this figure were calculated using Spearman’s correlation except as otherwise noted; no adjustments were made for multiple comparisons.

Supplementary information

Supplementary Information

This file contains Supplementary Figs. 1–4, Notes 1–5 and References that extend and support the data and discussion presented in the main text.

Reporting Summary

Supplementary Table 1

Most common somatic alterations in cancer.

Supplementary Table 2

Cohort information and statistics of different types of SCNA.

Supplementary Table 3

Pan-cancer regions of selection.

Supplementary Table 4

Cohort-specific regions of selection and lineage-specificity analysis.

Supplementary Table 5

Validation of peak regions.

Supplementary Table 6

RF values and mechanical coefficients.

Peer Review File

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) 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

Shih, J., Sarmashghi, S., Zhakula-Kostadinova, N. et al. Cancer aneuploidies are shaped primarily by effects on tumour fitness. Nature 619, 793–800 (2023). https://doi.org/10.1038/s41586-023-06266-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-06266-3

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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