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:

Mutational signatures impact the evolution of anti-EGFR antibody resistance in colorectal cancer

Abstract

Anti-EGFR antibodies such as cetuximab are active against KRAS/NRAS wild-type colorectal cancers (CRCs), but acquired resistance invariably evolves. It is unknown which mutational mechanisms enable resistance evolution and whether adaptive mutagenesis (a transient cetuximab-induced increase in mutation generation) contributes in patients. Here, we investigate these questions in exome sequencing data from 42 baseline and progression biopsies from cetuximab-treated CRCs. Mutation loads did not increase from baseline to progression, and evidence for a contribution of adaptive mutagenesis was limited. However, the chemotherapy-induced mutational signature SBS17b was the main contributor of specific KRAS/NRAS and EGFR driver mutations that are enriched at acquired resistance. Detectable SBS17b activity before treatment predicted shorter progression-free survival and the evolution of these specific mutations during subsequent cetuximab treatment. This result suggests that chemotherapy mutagenesis can accelerate resistance evolution. Mutational signatures may be a new class of cancer evolution predictor.

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: Cetuximab resistance models and analysis of mutation loads in 21 tumours treated with single-agent cetuximab.
Fig. 2: Mutational signatures in tumours treated with cetuximab.
Fig. 3: Relationship of mutational signatures to specific KRAS/NRAS and EGFR mutations.
Fig. 4: Association of detected SBS17b at BL with specific KRAS/NRAS and EGFR mutation evolution at the time of acquired resistance and with PFS.

Similar content being viewed by others

Data availability

All analyses were performed on previously published datasets3,5,20,26,27,28. The datasets can be accessed as described in the primary publications. The DNA sequencing data from the Prospect-C trial are deposited in the European Genome-phenome Archive with the accession code EGAS00001003367. As they include exome sequencing data that could permit the re-identification of trial participants, a data sharing agreement is required as stated in the primary publication3.

Code availability

The custom code to reproduce the mutational signature modelling is freely available on Github (https://github.com/AWoolston/Evolution-of-anti-EGFR-antibody-resistance).

References

  1. Karapetis, C. S. et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N. Engl. J. Med. 359, 1757–1765 (2008).

    Article  CAS  PubMed  Google Scholar 

  2. Van Cutsem, E. et al. Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. J. Clin. Oncol. 33, 692–700 (2015).

    Article  PubMed  CAS  Google Scholar 

  3. Woolston, A. et al. Genomic and transcriptomic determinants of therapy resistance and immune landscape evolution during anti-EGFR treatment in colorectal cancer. Cancer Cell 36, 35–50.e9 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Misale, S. et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 486, 532–536 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bettegowda, C. et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 6, 224ra24 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Montagut, C. et al. Identification of a mutation in the extracellular domain of the Epidermal Growth Factor Receptor conferring cetuximab resistance in colorectal cancer. Nat. Med. 18, 221–223 (2012).

    Article  CAS  PubMed  Google Scholar 

  7. Lipinski, K. A. et al. Cancer evolution and the limits of predictability in precision cancer medicine. Trends Cancer 2, 49–63 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Maley, C. C. et al. Classifying the evolutionary and ecological features of neoplasms. Nat. Rev. Cancer 17, 605–619 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gerlinger, M. & Swanton, C. How Darwinian models inform therapeutic failure initiated by clonal heterogeneity in cancer medicine. Br. J. Cancer 103, 1139–1143 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Diaz, L. A. Jr. et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 486, 537–540 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Russo, M. et al. Adaptive mutability of colorectal cancers in response to targeted therapies. Science 366, 1473–1480 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Gerlinger, M. Targeted drugs ramp up cancer mutability. Science 366, 1452–1453 (2019).

    Article  CAS  PubMed  Google Scholar 

  14. Kim, T. M., Laird, P. W. & Park, P. J. The landscape of microsatellite instability in colorectal and endometrial cancer genomes. Cell 155, 858–868 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature 578, 94–101 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Maura, F. et al. A practical guide for mutational signature analysis in hematological malignancies. Nat. Commun. 10, 2969 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Alexandrov, L. B. et al. Clock-like mutational processes in human somatic cells. Nat. Genet. 47, 1402–1407 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sveen, A. et al. Multilevel genomics of colorectal cancers with microsatellite instability—clinical impact of JAK1 mutations and consensus molecular subtype 1. Genome Med. 9, 46 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Christensen, S. et al. 5-fluorouracil treatment induces characteristic T>G mutations in human cancer. Nat. Commun. 10, 4571 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Pich, O. et al. The mutational footprints of cancer therapies. Nat. Genet. 51, 1732–1740 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tomkova, M., Tomek, J., Kriaucionis, S. & Schuster-Bockler, B. Mutational signature distribution varies with DNA replication timing and strand asymmetry. Genome Biol. 19, 129 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Meier, B. et al. Mutational signatures of DNA mismatch repair deficiency in C. elegans and human cancers. Genome Res. 28, 666–675 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Blokzijl, F., Janssen, R., van Boxtel, R. & Cuppen, E. MutationalPatterns: comprehensive genome-wide analysis of mutational processes. Genome Med. 10, 33 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Rosenthal, R., McGranahan, N., Herrero, J., Taylor, B. S. & Swanton, C. DeconstructSigs: delineating mutational processes in single tumors distinguishes DNA repair deficiencies and patterns of carcinoma evolution. Genome Biol. 17, 31 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Arena, S. et al. Emergence of multiple EGFR extracellular mutations during cetuximab treatment in colorectal cancer. Clin. Cancer Res. 21, 2157–2166 (2015).

    Article  CAS  PubMed  Google Scholar 

  26. Khan, K. H. et al. Longitudinal liquid biopsy and mathematical modeling of clonal evolution forecast time to treatment failure in the PROSPECT-C phase II colorectal cancer clinical trial. Cancer Discov. 8, 1270–1285 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. The Cancer Genome Atlas Research Network et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat. Genet. 45, 1113–1120 (2013).

  28. Tabernero, J. et al. Analysis of circulating DNA and protein biomarkers to predict the clinical activity of regorafenib and assess prognosis in patients with metastatic colorectal cancer: a retrospective, exploratory analysis of the CORRECT trial. Lancet Oncol. 16, 937–948 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Gerstung, M. et al. The evolutionary history of 2,658 cancers. Nature 578, 122–128 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Price, T. et al. Frequency of S492R mutations in the epidermal growth factor receptor: analysis of plasma DNA from patients with metastatic colorectal cancer treated with panitumumab or cetuximab monotherapy. Cancer Biol. Ther. 21, 891–898 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Priestley, P. et al. Pan-cancer whole-genome analyses of metastatic solid tumours. Nature 575, 210–216 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Cannataro, V. L., Gaffney, S. G. & Townsend, J. P. Effect sizes of somatic mutations in cancer. J. Natl Cancer Inst. 110, 1171–1177 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Ali, M. et al. Codon bias imposes a targetable limitation on KRAS-driven therapeutic resistance. Nat. Commun. 8, 15617 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Montagut, C. et al. Efficacy of Sym004 in patients with metastatic colorectal cancer with acquired resistance to anti-EGFR therapy and molecularly selected by circulating tumor DNA analyses: a phase 2 randomized clinical trial. JAMA Oncol. 4, e175245 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Poulos, R. C., Wong, Y. T., Ryan, R., Pang, H. & Wong, J. W. H. Analysis of 7,815 cancer exomes reveals associations between mutational processes and somatic driver mutations. PLoS Genet. 14, e1007779 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Temko, D., Tomlinson, I. P. M., Severini, S., Schuster-Bockler, B. & Graham, T. A. The effects of mutational processes and selection on driver mutations across cancer types. Nat. Commun. 9, 1857 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Poetsch, A. R. The genomics of oxidative DNA damage, repair, and resulting mutagenesis. Comput. Struct. Biotechnol. J. 18, 207–219 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kruger, S. & Piro, R. M. decompTumor2Sig: identification of mutational signatures active in individual tumors. BMC Bioinform. 20, 152 (2019).

    Article  Google Scholar 

  39. Niu, B. et al. MSIsensor: microsatellite instability detection using paired tumor-normal sequence data. Bioinformatics 30, 1015–1016 (2014).

    Article  CAS  PubMed  Google Scholar 

  40. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  PubMed  Google Scholar 

  41. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal 6, pl1 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Schumann, F. et al. SigsPack, a package for cancer mutational signatures. BMC Bioinform. 20, 450 (2019).

    Article  CAS  Google Scholar 

  43. Alexandrov, L. B., Nik-Zainal, S., Wedge, D. C., Campbell, P. J. & Stratton, M. R. Deciphering signatures of mutational processes operative in human cancer. Cell Rep. 3, 246–259 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. R Core Team R: A Language and Environment for Statistical Computing v.3.5.0 (R Foundation for Statistical Computing, 2018).

Download references

Acknowledgements

D.C. received funding from the NIHR Biomedical Research Centre for Cancer at the Institute of Cancer Research and the Royal Marsden Hospital. M.G., A.W. and L.J.B. received funding from the European Research Council under the European Union’s Horizon 2020 research and innovation programme (grant agreement no. 820137). The paper is dedicated to the memory of Tim Morgan, who supported this work with a generous donation.

Author information

Authors and Affiliations

Authors

Contributions

M.G. conceived, funded and supervised the molecular analysis. D.C. is the chief investigator of the Prospect-C trial and funded the trial. N.S., I.C., S.R. and D.W. recruited the trial patients. B.G. prepared the trial samples, and N.M. supervised the sequencing. L.J.B. performed the ctDNA sequencing and analysis. A.W. performed the bioinformatics analysis. O.P. and N.L.-B. provided the analysis of metastatic CRC samples from the Hartwig Medical Foundation. A.W. and M.G. performed the statistical analysis. A.W. and M.G. wrote the manuscript. L.J.B., O.P. and N.L.-B. provided feedback. All authors approved the final manuscript.

Corresponding author

Correspondence to Marco Gerlinger.

Ethics declarations

Competing interests

I.C. has consultant/advisory roles with Eli-Lilly, BMS, MSD, Merck KG, Roche, Bayer and Five Prime Therapeutics. D.C. receives research funding from Amgen, Sanofi, Merrimack, Astra Zeneca, Celegene, MedImmune, Bayer, 4SC, Clovis, Eli-Lilly, Janssen and Merck KG. M.G. and N.S. receive research funding from Merck KG and BMS. The other authors declare no competing interests.

Additional information

Peer review information Nature Ecology & Evolution thanks Christos Karapetis, Peter Campbell 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.

Extended data

Extended Data Fig. 1 Plots of cancer cell content, sequencing depth and mutation load for the paired BL/PD biopsies from 21 patients in the Prospect-C trial.

a, Estimated cancer cell contents of paired BL and PD samples. A 1:1 ratio line has been added for reference. b, Mutation load vs. mean sequencing depth for all BL and PD samples. p-value from Spearman’s test. A linear regression line has been added for reference. c, Mutation load vs. cancer cell content for all BL and PD samples. p-value from Spearman’s test. A linear regression line has been added for reference.

Extended Data Fig. 2 Clonal mutation trees for 21 tumors from the Prospect-C trial.

Grouped into cases with prolonged benefit and primary progression. The numbers next to the trunk or the branches indicate clonal somatic mutations.

Extended Data Fig. 3 Number of unique mutations detected for each of 21 paired biopsies from the Prospect-C trial vs. time lapse between BL and PD biopsies.

p-value from Spearman’s test. A linear regression line has been added for reference.

Extended Data Fig. 4 Proportion of SBS mutations attributed to each mutational signature.

Signatures were selected using the ‘ColoRect-AdenoCa’ samples from the SigProfiler TCGA whole exome cohort (n = 496) (syn11801497.7). All signatures in the cohort with a non-zero mutation attribution were considered along with all MMR-deficiency signatures and platinum treatment signatures. Plots show the cohort wide signature attribution among (a) all 21 Prospect-C samples and (b) only in the PD tumors of the 12 patients with prolonged benefit. The red horizontal dashed line illustrates the 3% threshold used to define signatures as ‘active’ and the red box shows the signatures retained for subsequent analysis. SBS17a and SBS17b are described as ‘connected’ signatures15. SBS17a was retained due to the inclusion of SBS17b despite not reaching the threshold.

Extended Data Fig. 5 Signature attributions based on 21 paired BL/PD biopsies from the Prospect-C trial using MutationalPatterns and deconstructSigs.

a, Mutation signature attribution using independent decomposition methods (deconstructSigs and MutationalPatterns). b, Fig. 2 repeated with the ‘fit_to_sigs’ function in MutationalPatterns to assess the variability of estimates between methods.

Extended Data Fig. 6 Mutation frequency profiles of treatment naïve CRCs from the TCGA Pan-Cancer study vs. the KRAS hotspot mutations identified in ref. 28.

The TCGA profile has been adjusted to only consider KRAS mutations that were assessed in the CORRECT trial.

Extended Data Fig. 7 Modelling the impact of mutational signatures on the likelihood of acquired hotspot mutations.

a, Modelled mutational profile of a BL tumor with prolonged benefit. Exome normalised reference signatures have been scaled by the observed signature exposures of the 12 BL tumors with prolonged benefit to represent a mutation probability at each trinucleotide mutation context. b, Observed mutation frequencies of KRAS/NRAS Q61H vs. all other KRAS/NRAS hotspot mutations identified in CRCs with acquired EGFR-AB resistance3,5,26. c, Modelled mutation accumulation of the permanent signatures. A varying acceleration parameter of x1, x5, x10 is applied to the tumor growth period. d, Impact of SBS17b and SBS35 on the likelihood of generating KRAS/NRAS Q61H mutations vs. all other detected KRAS/NRAS hotspot mutations.

Extended Data Fig. 8 Analysis of an independent cohort of 239 patients with metastatic colorectal cancer and a KRAS/NRAS G12/G13 or Q61H mutation.

a, Total mutations attributed to SBS17b. Statistical significance was assessed with the Fisher’s exact test. b, Proportion of tumors with a detectable SBS17b signature activity. Statistical significance was assessed with the Mann-Whitney U test.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Woolston, A., Barber, L.J., Griffiths, B. et al. Mutational signatures impact the evolution of anti-EGFR antibody resistance in colorectal cancer. Nat Ecol Evol 5, 1024–1032 (2021). https://doi.org/10.1038/s41559-021-01470-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41559-021-01470-8

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