Abstract
Aneuploidy, a hallmark of cancer characterized by chromosome imbalances, drives tumorigenesis and facilitates cancer immune evasion. While high tumor aneuploidy is linked to immune checkpoint blockade (ICB) resistance and poor prognosis, evidence suggests that this resistance can be overcome through treatment intensification, for example, with the addition of ablative radiotherapy to ICB. In this Perspective, we argue that the predictive value of aneuploidy complements established biomarkers, such as tumor mutational burden (TMB) or programmed death ligand 1 (PD-L1) expression. We review contemporary methods for quantifying aneuploidy, explore novel approaches that target mitotic vulnerabilities in aneuploid tumors and highlight potential areas where aneuploidy-based stratification could be incorporated into ICB-based treatment paradigms across early-stage, locally advanced and metastatic cancers. Prospective trials incorporating aneuploidy-based stratification will be essential to validate its role in personalized cancer therapy.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout

Similar content being viewed by others
References
Weaver, B. A. & Cleveland, D. W. Does aneuploidy cause cancer? Curr. Opin. Cell Biol. 18, 658–667 (2006).
Bakhoum, S. F. & Compton, D. A. Chromosomal instability and cancer: a complex relationship with therapeutic potential. J. Clin. Invest. 122, 1138–1143 (2012).
Bakhoum, S. F. & Cantley, L. C. The multifaceted role of chromosomal instability in cancer and its microenvironment. Cell 174, 1347–1360 (2018).
Hassold, T. et al. Human aneuploidy: incidence, origin, and etiology. Environ. Mol. Mutagen. 28, 167–175 (1996).
Sheltzer, J. M. & Amon, A. The aneuploidy paradox: costs and benefits of an incorrect karyotype. Trends Genet. 27, 446–453 (2011).
Taylor, A. M. et al. Genomic and functional approaches to understanding cancer aneuploidy. Cancer Cell 33, 676–689 (2018).
Ben-David, U. et al. Patient-derived xenografts undergo mouse-specific tumor evolution. Nat. Genet. 49, 1567–1575 (2017).
Zack, T. I. et al. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 45, 1134–1140 (2013).
Sack, L. M. et al. Profound tissue specificity in proliferation control underlies cancer drivers and aneuploidy patterns. Cell 173, 499–514 (2018).
Li, Y. et al. Patterns of somatic structural variation in human cancer genomes. Nature 578, 112–121 (2020).
Gordon, D. J., Resio, B. & Pellman, D. Causes and consequences of aneuploidy in cancer. Nat. Rev. Genet. 13, 189–203 (2012).
Replogle, J. M. et al. Aneuploidy increases resistance to chemotherapeutics by antagonizing cell division. Proc. Natl Acad. Sci. USA 117, 30566–30576 (2020).
Haslam, A., Olivier, T. & Prasad, V. How many people in the US are eligible for and respond to checkpoint inhibitors: an empirical analysis. Int. J. Cancer 156, 2352–2359 (2025).
Davoli, T., Uno, H., Wooten, E. C. & Elledge, S. J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 355, eaaf8399 (2017).
Roh, W. et al. Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance. Sci. Transl. Med. 9, eaah3560 (2017).
Ock, C.-Y. et al. Genomic landscape associated with potential response to anti-CTLA-4 treatment in cancers. Nat. Commun. 8, 1050 (2017).
Alessi, J. V. et al. Multi-institutional analysis of aneuploidy and outcomes to chemoradiation and durvalumab in stage III non-small cell lung cancer. J. Immunother. Cancer 11, e007618 (2023).
Alessi, J. V. et al. Impact of aneuploidy and chromosome 9p loss on tumor immune microenvironment and immune checkpoint inhibitor efficacy in NSCLC. J. Thorac. Oncol. 18, 1524–1537 (2023).
Spurr, L. F. et al. Highly aneuploid non-small cell lung cancer shows enhanced responsiveness to concurrent radiation and immune checkpoint blockade. Nat. Cancer 3, 1498–1512 (2022).
Spurr, L. F., Weichselbaum, R. R. & Pitroda, S. P. Tumor aneuploidy predicts survival following immunotherapy across multiple cancers. Nat. Genet. 54, 1782–1785 (2022).
Spurr, L. F. & Pitroda, S. P. Clinical and molecular correlates of tumor aneuploidy in metastatic non-small cell lung cancer. Sci. Rep. 14, 19375 (2024).
Jang, H., Hur, Y. & Lee, H. Identification of cancer-driver genes in focal genomic alterations from whole genome sequencing data. Sci. Rep. 6, 25582 (2016).
Lukow, D. A. et al. Chromosomal instability accelerates the evolution of resistance to anti-cancer therapies. Dev. Cell 56, 2427–2439 (2021).
Carter, S. L. et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 30, 413–421 (2012).
Van Loo, P. et al. Allele-specific copy number analysis of tumors. Proc. Natl Acad. Sci. USA 107, 16910–16915 (2010).
Spurr, L. F. et al. Quantification of aneuploidy in targeted sequencing data using ASCETS. Bioinformatics 37, 2461–2463 (2021).
Nik-Zainal, S. et al. The life history of 21 breast cancers. Cell 149, 994–1007 (2012).
Talevich, E., Shain, A. H., Botton, T. & Bastian, B. C. CNVkit: genome-wide copy number detection and visualization from targeted DNA sequencing. PLoS Comput. Biol. 12, e1004873 (2016).
Boeva, V. et al. Control-FREEC: a tool for assessing copy number and allelic content using next-generation sequencing data. Bioinformatics 28, 423–425 (2012).
Shen, R. & Seshan, V. E. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 44, e131 (2016).
Babadi, M. et al. GATK-gCNV enables the discovery of rare copy number variants from exome sequencing data. Nat. Genet. 55, 1589–1597 (2023).
Adalsteinsson, V. A. et al. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat. Commun. 8, 1324 (2017).
Favero, F. et al. Sequenza: allele-specific copy number and mutation profiles from tumor sequencing data. Ann. Oncol. 26, 64–70 (2015).
Ha, G. et al. TITAN: inference of copy number architectures in clonal cell populations from tumor whole-genome sequence data. Genome Res. 24, 1881–1893 (2014).
Ballman, K. V. Biomarker: predictive or prognostic? J. Clin. Oncol. 33, 3968–3971 (2015).
Samstein, R. M. et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 51, 202–206 (2019).
Marabelle, A. et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 21, 1353–1365 (2020).
Larkin, J. et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373, 23–34 (2015).
Borghaei, H. et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N. Engl. J. Med. 373, 1627–1639 (2015).
Motzer, R. J. et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N. Engl. J. Med. 373, 1803–1813 (2015).
Mansfield, A. S. et al. Heterogeneity of programmed cell death ligand 1 expression in multifocal lung cancer. Clin. Cancer Res. 22, 2177–2182 (2016).
Haragan, A. et al. Heterogeneity of PD-L1 expression in non-small cell lung cancer: implications for specimen sampling in predicting treatment response. Lung Cancer 134, 79–84 (2019).
Lu, S. et al. Comparison of biomarker modalities for predicting response to PD-1/PD-L1 checkpoint blockade: a systematic review and meta-analysis. JAMA Oncol. 5, 1195–1204 (2019).
Kim, H. S. et al. Genomic scoring to determine clinical benefit of immunotherapy by targeted sequencing. Eur. J. Cancer 120, 65–74 (2019).
Chang, T.-G. et al. Optimizing cancer immunotherapy response prediction by tumor aneuploidy score and fraction of copy number alterations. npj Precis. Oncol. 7, 54 (2023).
Liu, L. et al. Combination of TMB and CNA stratifies prognostic and predictive responses to immunotherapy across metastatic cancer. Clin. Cancer Res. 25, 7413–7423 (2019).
Asleh, K. & Ouellette, R. J. Tumor copy number alteration burden as a predictor for resistance to immune checkpoint blockade across different cancer types. Cancers 16, 732 (2024).
Antonia, S. J. et al. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N. Engl. J. Med. 379, 2342–2350 (2018).
Han, G. et al. 9p21 loss confers a cold tumor immune microenvironment and primary resistance to immune checkpoint therapy. Nat. Commun. 12, 5606 (2021).
William, W. N. et al. Immune evasion in HPV− head and neck precancer–cancer transition is driven by an aneuploid switch involving chromosome 9p loss. Proc. Natl Acad. Sci. USA 118, e2022655118 (2021).
Zhao, X. et al. Somatic 9p24.1 alterations in HPV– head and neck squamous cancer dictate immune microenvironment and anti-PD-1 checkpoint inhibitor activity. Proc. Natl Acad. Sci. USA 119, e2213835119 (2022).
Ebot, E. M. et al. Deletions on 9p21 are associated with worse outcomes after anti-PD-1/PD-L1 monotherapy but not chemoimmunotherapy. npj Precis. Oncol. 6, 44 (2022).
Andor, N. et al. Pan-cancer analysis of the extent and consequences of intratumor heterogeneity. Nat. Med. 22, 105–113 (2016).
Birkbak, N. J. et al. Paradoxical relationship between chromosomal instability and survival outcome in cancer. Cancer Res. 71, 3447–3452 (2011).
Litchfield, K. et al. Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell 184, 596–614 (2021).
Lu, Z. et al. Tumor copy-number alterations predict response to immune-checkpoint-blockade in gastrointestinal cancer. J. Immunother. Cancer 8, e000374 (2020).
Sia, D. et al. Identification of an immune-specific class of hepatocellular carcinoma, based on molecular features. Gastroenterology 153, 812–826 (2017).
Shimada, S. et al. Comprehensive molecular and immunological characterization of hepatocellular carcinoma. EBioMedicine 40, 457–470 (2019).
Miao, D. et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science 359, 801–806 (2018).
Alcaraz, A. et al. Aneuploidy and aneusomy of chromosome 7 detected by fluorescence in situ hybridization are markers of poor prognosis in prostate cancer. Cancer Res. 54, 3998–4002 (1994).
Kallioniemi, O.-P. et al. Aneuploid DNA content and high S-phase fraction of tumour cells are related to poor prognosis in patients with primary breast cancer. Eur. J. Cancer Clin. Oncol. 23, 277–282 (1987).
Van Mackelenbergh, M. T. et al. Pathologic complete response and individual patient prognosis after neoadjuvant chemotherapy plus anti-human epidermal growth factor receptor 2 therapy of human epidermal growth factor receptor 2-positive early breast cancer. J. Clin. Oncol. 41, 2998–3008 (2023).
Forde, P. M. et al. Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. N. Engl. J. Med. 386, 1973–1985 (2022).
Senthi, S., Lagerwaard, F. J., Haasbeek, C. J., Slotman, B. J. & Senan, S. Patterns of disease recurrence after stereotactic ablative radiotherapy for early stage non-small-cell lung cancer: a retrospective analysis. Lancet Oncol. 13, 802–809 (2012).
Chang, J. Y. et al. Stereotactic ablative radiotherapy with or without immunotherapy for early-stage or isolated lung parenchymal recurrent node-negative non-small-cell lung cancer: an open-label, randomised, phase 2 trial. Lancet 402, 871–881 (2023).
Pircher, A. et al. 117O Stereotactic body radiotherapy (SBRT) with pembrolizumab (pembro) for unresected stage I/II non-small cell lung cancer (NSCLC): the randomized, double-blind, phase III KEYNOTE-867 study. Immuno-Oncol. Technol. 24, 100746 (2024).
Gross, N. D. et al. Neoadjuvant cemiplimab for stage II to IV cutaneous squamous-cell carcinoma. N. Engl. J. Med. 387, 1557–1568 (2022).
Blank, C. U. et al. Neoadjuvant nivolumab and ipilimumab in resectable stage III melanoma. N. Engl. J. Med. 391, 1696–1708 (2024).
Topalian, S. L. et al. Neoadjuvant nivolumab for patients with resectable merkel cell carcinoma in the CheckMate 358 trial. J. Clin. Oncol. 38, 2476–2487 (2020).
Cercek, A. et al. PD-1 blockade in mismatch repair-deficient, locally advanced rectal cancer. N. Engl. J. Med. 386, 2363–2376 (2022).
Cercek, A. et al. Mismatch repair-deficient rectal cancer and resistance to neoadjuvant chemotherapy. Clin. Cancer Res. 26, 3271–3279 (2020).
Altorki, N. K. et al. Neoadjuvant durvalumab with or without stereotactic body radiotherapy in patients with early-stage non-small-cell lung cancer: a single-centre, randomised phase 2 trial. Lancet Oncol. 22, 824–835 (2021).
Cosper, P. F., Copeland, S. E., Tucker, J. B. & Weaver, B. A. Chromosome missegregation as a modulator of radiation sensitivity. Semin. Radiat. Oncol. 32, 54–63 (2022).
Jia, Q. et al. Mutational burden and chromosomal aneuploidy synergistically predict survival from radiotherapy in non-small cell lung cancer. Commun. Biol. 4, 131 (2021).
Bakhoum, S. F. et al. Numerical chromosomal instability mediates susceptibility to radiation treatment. Nat. Commun. 6, 5990 (2015).
Wei, B. et al. Comprehensive analysis of aneuploidy status and its effect on the efficacy of EGFR-TKIs in lung cancer. J. Thorac. Dis. 14, 625–634 (2022).
Goodman, A. M. et al. Assessing CAR T-cell therapy response using genome-wide sequencing of cell-free DNA in patients with B-cell lymphomas. Transplant. Cell. Ther. 28, 30.e1–30.e7 (2022).
Girish, V. et al. Oncogene-like addiction to aneuploidy in human cancers. Science 381, eadg4521 (2023).
Boudhraa, Z. et al. NR1D1 regulation by Ran GTPase via miR4472 identifies an essential vulnerability linked to aneuploidy in ovarian cancer. Oncogene 41, 309–320 (2022).
Quinton, R. J. et al. Whole-genome doubling confers unique genetic vulnerabilities on tumour cells. Nature 590, 492–497 (2021).
Cohen-Sharir, Y. et al. Aneuploidy renders cancer cells vulnerable to mitotic checkpoint inhibition. Nature 590, 486–491 (2021).
Marquis, C. et al. Chromosomally unstable tumor cells specifically require KIF18A for proliferation. Nat. Commun. 12, 1213 (2021).
Phillips, A. F. et al. Targeting chromosomally unstable tumors with a selective KIF18A inhibitor. Nat. Commun. 16, 307 (2025).
Payton, M. et al. Small-molecule inhibition of kinesin KIF18A reveals a mitotic vulnerability enriched in chromosomally unstable cancers. Nat. Cancer 5, 66–84 (2023).
Agata, Y. et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int. Immunol. 8, 765–772 (1996).
Freeman, G. J. et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 192, 1027–1034 (2000).
Yokosuka, T. et al. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J. Exp. Med. 209, 1201–1217 (2012).
Dong, H., Zhu, G., Tamada, K. & Chen, L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat. Med. 5, 1365–1369 (1999).
Lau, J. et al. Tumour and host cell PD-L1 is required to mediate suppression of anti-tumour immunity in mice. Nat. Commun. 8, 14572 (2017).
Takahashi, T. et al. Immunologic self-tolerance maintained by CD25+CD4+ regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J. Exp. Med. 192, 303–310 (2000).
Read, S., Malmström, V. & Powrie, F. Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25+CD4+ regulatory cells that control intestinal inflammation. J. Exp. Med. 192, 295–302 (2000).
Linsley, P. S. et al. CTLA-4 is a second receptor for the B cell activation antigen B7. J. Exp. Med. 174, 561–569 (1991).
Van Der Merwe, P. A., Bodian, D. L., Daenke, S., Linsley, P. & Davis, S. J. CD80 (B7-1) binds both CD28 and CTLA-4 with a low affinity and very fast kinetics. J. Exp. Med. 185, 393–404 (1997).
Linsley, P. S. et al. Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity 1, 793–801 (1994).
Leach, D. R., Krummel, M. F. & Allison, J. P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 271, 1734–1736 (1996).
Hellmann, M. D. et al. Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N. Engl. J. Med. 381, 2020–2031 (2019).
Doroshow, D. B. et al. PD-L1 as a biomarker of response to immune-checkpoint inhibitors. Nat. Rev. Clin. Oncol. 18, 345–362 (2021).
André, T. et al. Pembrolizumab in microsatellite-instability-high advanced colorectal cancer. N. Engl. J. Med. 383, 2207–2218 (2020).
Ayers, M. et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 127, 2930–2940 (2017).
Ott, P. A. et al. T-cell-inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028. J. Clin. Oncol. 37, 318–327 (2019).
Dawson, N. V. & Weiss, R. Dichotomizing continuous variables in statistical analysis: a practice to avoid. Med. Decis. Making 32, 225–226 (2012).
Efron, B. Bootstrap methods: another look at the jackknife. Ann. Stat. 7, 1–26 (1979).
Garcia, E. P. et al. Validation of OncoPanel: a targeted next-generation sequencing assay for the detection of somatic variants in cancer. Arch. Pathol. Lab. Med. 141, 751–758 (2017).
Teer, J. K. et al. Evaluating somatic tumor mutation detection without matched normal samples. Hum. Genomics 11, 22 (2017).
Chowell, D. et al. Improved prediction of immune checkpoint blockade efficacy across multiple cancer types. Nat. Biotechnol. 40, 499–506 (2022).
Chang, T.-G. et al. LORIS robustly predicts patient outcomes with immune checkpoint blockade therapy using common clinical, pathologic and genomic features. Nat. Cancer 5, 1158–1175 (2024).
Sdeor, E., Okada, H., Saad, R., Ben-Yishay, T. & Ben-David, U. Aneuploidy as a driver of human cancer. Nat. Genet. 56, 2014–2026 (2024).
Schmid, P. et al. Pembrolizumab for early triple-negative breast cancer. N. Engl. J. Med. 382, 810–821 (2020).
Cheng, Y. et al. Durvalumab after chemoradiotherapy in limited-stage small-cell lung cancer. N. Engl. J. Med. 391, 1313–1327 (2024).
Lee, N. Y. et al. Avelumab plus standard-of-care chemoradiotherapy versus chemoradiotherapy alone in patients with locally advanced squamous cell carcinoma of the head and neck: a randomised, double-blind, placebo-controlled, multicentre, phase 3 trial. Lancet Oncol. 22, 450–462 (2021).
Machiels, J.-P. et al. Pembrolizumab plus concurrent chemoradiotherapy versus placebo plus concurrent chemoradiotherapy in patients with locally advanced squamous cell carcinoma of the head and neck (KEYNOTE-412): a randomised, double-blind, phase 3 trial. Lancet Oncol. 25, 572–587 (2024).
Liu, X. et al. Induction-concurrent chemoradiotherapy with or without sintilimab in patients with locoregionally advanced nasopharyngeal carcinoma in China (CONTINUUM): a multicentre, open-label, parallel-group, randomised, controlled, phase 3 trial. Lancet 403, 2720–2731 (2024).
Liu, S.-L. et al. Neoadjuvant and adjuvant toripalimab for locoregionally advanced nasopharyngeal carcinoma: a randomised, single-centre, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 25, 1563–1575 (2024).
Lorusso, D. et al. Pembrolizumab or placebo with chemoradiotherapy followed by pembrolizumab or placebo for newly diagnosed, high-risk, locally advanced cervical cancer (ENGOT-cx11/GOG-3047/KEYNOTE-A18): a randomised, double-blind, phase 3 clinical trial. Lancet 403, 1341–1350 (2024).
Acknowledgements
S.P.P. is supported by the LUNGevity Foundation, the Falk Medical Research Trust and the American Lung Association. S.P.P. and R.R.W. receive funding from the National Institutes of Health (U54-CA274291 ROBIN grant) and the Ludwig Cancer Research Foundation.
Author information
Authors and Affiliations
Contributions
D.H. wrote the initial draft of the manuscript and designed the figure with input from all authors. L.F.S. contributed his technical expertise on the quantification of tumor aneuploidy. D.H., L.F.S., R.R.W. and S.P.P. provided critical revisions to the manuscript.
Corresponding author
Ethics declarations
Competing interests
R.R.W. has stock and other ownership interests with Boost Therapeutics, ImmVira, RefleXion Pharmaceuticals, Coordination Pharmaceuticals, Magi Therapeutics and Oncosenescence. He has served in a consulting or advisory role for Aettis, AstraZeneca, Coordination Pharmaceuticals, Genus, Merck Serono, Nanoproteagen, NKMax America and Shuttle Pharmaceuticals. He has received research grant funding from Varian and Regeneron. He has received compensation including travel, accommodations or expense reimbursement from AstraZeneca, Boehringer Ingelheim and Merck Serono. S.P.P. and R.R.W. own a patent related to this work (PCT/US23/81165). S.P.P. and R.R.W. are cofounders of PersonaDx. The other authors declare no competing interests.
Peer review
Peer review information
Nature Genetics thanks Teresa Davoli 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.
Supplementary information
Supplementary Information
Supplementary Table 1
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.
About this article
Cite this article
Huang, D., Spurr, L.F., Weichselbaum, R.R. et al. Tumor aneuploidy as a prognostic and predictive biomarker in immune checkpoint blockade. Nat Genet 57, 1802–1811 (2025). https://doi.org/10.1038/s41588-025-02226-x
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41588-025-02226-x