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:

Clinical Studies

Dynamic nature of BRAF or KRAS p.G12C mutations in second-line therapy for advanced colorectal cancer patients: do early and late effects exist?

Abstract

Introduction

The mitogen-activated protein kinase (MAPK) signalling network aberrations in metastatic colorectal cancer (mCRC) generate intrinsic dynamic effects and temporal variations that are crucial but often overlooked in clinical trial populations. Here, we investigate the time-varying impact of MAPK pathway mutation genotype on each treatment line’s contribution to the overall clinical course.

Methods

The PROMETEO study focused on mCRC patients undergoing second-line treatment at 20 hospitals. We evaluated genotypes and employed flexible models to analyse the dynamic effect of each mutation.

Results

We examined data derived from 1160 patients. The effects of KRAS G12C or G12V, and BRAF V600E are clearly time-varying, with unexpected consequences such as the deleterious effect of BRAF V600E vs other genotypes dissipating over time when subjects receive antiangiogenics, or KRAS G12V and G12C showing increasing aggressiveness over time. Thus, contrary to expectations, the 12-month survival rate from the second line for those who survived >6 months was 49.9% (95% CI, 32.7–67.3) for KRAS G12C and 59% (95% CI, 38.5–80.6) for BRAF V600E.

Conclusions

The dynamic perspective is essential for understanding the behaviour of tumours with specific genotypes, especially from the second line onward. This may be relevant in patient monitoring and treatment decision-making, particularly in cases with distinct mutations.

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
Fig. 2: Kaplan–Meier plots for progression-free survival and overall survival from second-line treatment, stratified by genetic variant.
Fig. 3: Partitioned survival analysis.
Fig. 4: Time-varying hazard ratios for overall survival from second-line treatment (OS-2).

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Ottaiano A, Normanno N, Facchini S, Cassata A, Nappi A, Romano C, et al. Study of Ras mutations’ prognostic value in metastatic colorectal cancer: STORIA analysis. Cancers. 2020;12:1919.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Bylsma LC, Gillezeau C, Garawin TA, Kelsh MA, Fryzek JP, Sangaré L, et al. Prevalence of RAS and BRAF mutations in metastatic colorectal cancer patients by tumor sidedness: a systematic review and meta‐analysis. Cancer Med. 2020;9:1044–57.

    PubMed  Google Scholar 

  3. Bazan V, Migliavacca M, Zanna I, Tubiolo C, Grassi N, Latteri MA, et al. Specific codon 13 K-ras mutations are predictive of clinical outcome in colorectal cancer patients, whereas codon 12 K-ras mutations are associated with mucinous histotype. Ann Oncol. 2002;13:1438–46.

    CAS  PubMed  Google Scholar 

  4. Zhu G, Pei L, Xia H, Tang Q, Bi F. Role of oncogenic KRAS in the prognosis, diagnosis and treatment of colorectal cancer. Mol Cancer. 2021;20:1–17.

    Google Scholar 

  5. Patelli G, Tosi F, Amatu A, Mauri G, Curaba A, Patane DA, et al. Strategies to tackle RAS-mutated metastatic colorectal cancer. ESMO Open. 2021;6:100156.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Goebel L, Müller MP, Goody RS, Rauh D. KRasG12C inhibitors in clinical trials: a short historical perspective. RSC Med Chem. 2020;11:760–70.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-Ras (G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013;503:548–51.

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Henry JT, Coker O, Chowdhury S, Shen JP, Morris VK, Dasari A, et al. Comprehensive clinical and molecular characterization of KRAS G12C-mutant colorectal cancer. JCO Precis Oncol. 2021;5:PO.20.00256.

  9. Dienstmann R, Connor K, Byrne AT, Fridman WH, Lambrechts D, Sadanandam A, et al. Precision therapy in RAS mutant colorectal cancer. Gastroenterology. 2020;158:806–11.

    PubMed  Google Scholar 

  10. Amodio V, Yaeger R, Arcella P, Cancelliere C, Lamba S, Lorenzato A, et al. EGFR blockade reverts resistance to KRASG12C inhibition in colorectal cancer overcoming KRASG12C inhibitor resistance in colorectal cancer. Cancer Discov. 2020;10:1129–39.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Hong DS, Fakih MG, Strickler JH, Desai J, Durm GA, Shapiro GI, et al. KRASG12C inhibition with sotorasib in advanced solid tumors. N Engl J Med. 2020;383:1207–17.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Yaeger R, Weiss J, Pelster MS, Spira AI, Barve M, Ou S-HI, et al. Adagrasib with or without cetuximab in colorectal cancer with mutated KRAS G12C. N Engl J Med. 2023;388:44–54.

    CAS  PubMed  Google Scholar 

  13. Hunter JC, Manandhar A, Carrasco MA, Gurbani D, Gondi S, Westover KD. Biochemical and structural analysis of common cancer-associated KRAS mutations. Mol Cancer Res. 2015;13:1325–35.

    CAS  Google Scholar 

  14. de la Chapelle A, Hampel H. Clinical relevance of microsatellite instability in colorectal cancer. J Clin Oncol. 2010;28:3380.

    PubMed  PubMed Central  Google Scholar 

  15. Kopetz S, Desai J, Chan E, Hecht JR, O’Dwyer PJ, Maru D, et al. Phase II pilot study of vemurafenib in patients with metastatic BRAF-mutated colorectal cancer. J Clin Oncol. 2015;33:4032.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Rowland A, Dias MM, Wiese MD, Kichenadasse G, McKinnon RA, Karapetis CS, et al. Meta-analysis of BRAF mutation as a predictive biomarker of benefit from anti-EGFR monoclonal antibody therapy for RAS wild-type metastatic colorectal cancer. Br J Cancer. 2015;112:1888–94.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Pietrantonio F, Petrelli F, Coinu A, Di Bartolomeo M, Borgonovo K, Maggi C, et al. Predictive role of BRAF mutations in patients with advanced colorectal cancer receiving cetuximab and panitumumab: a meta-analysis. Eur J Cancer. 2015;51:587–94.

    CAS  PubMed  Google Scholar 

  18. Kopetz S, Grothey A, Yaeger R, Van Cutsem E, Desai J, Yoshino T, et al. Encorafenib, binimetinib, and cetuximab in BRAF V600E–mutated colorectal cancer. N Engl J Med. 2019;381:1632–43.

    CAS  PubMed  Google Scholar 

  19. Seligmann JF, Fisher D, Smith CG, Richman SD, Elliott F, Brown S, et al. Investigating the poor outcomes ofBRAF-mutant advanced colorectal cancer: analysis from 2530 patients in randomised clinical trials. Ann Oncol. 2017;28:562–8.

    CAS  PubMed  Google Scholar 

  20. Ciardiello D, Chiarazzo C, Famiglietti V, Damato A, Pinto C, Zampino MG, et al. Clinical efficacy of sequential treatments in KRASG12C-mutant metastatic colorectal cancer: findings from a real-life multicenter Italian study (CRC-KR GOIM). ESMO Open. 2022;7:100567.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Hochster H, Hager S, Pipas JM, Tebbutt N, Laurent S, Gravalos C, et al. O-010 KRAS and BRAF gene subgroup analysis in the phase 3 RECOURSE trial of TAS-102 versus placebo in patients with metastatic colorectal cancer. Ann Oncol. 2015;26:iv111.

    Google Scholar 

  22. Cervantes A, Adam R, Roselló S, Arnold D, Normanno N, Taïeb J, et al. Metastatic colorectal cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up. Ann Oncol. 2023;34:10–32.

    CAS  PubMed  Google Scholar 

  23. Humbert M, Sitbon O, Yaïci A, Montani D, O’callaghan DS, Jaïs X, et al. Survival in incident and prevalent cohorts of patients with pulmonary arterial hypertension. Eur Respir J. 2010;36:549–55.

    CAS  PubMed  Google Scholar 

  24. Carmona-Bayonas A, Jimenez-Fonseca P, Garrido M, Custodio A, Hernandez R, Lacalle A, et al. Multistate models: accurate and dynamic methods to improve predictions of thrombotic risk in patients with cancer. Thromb Haemost. 2019;119:1849–59.

    PubMed  Google Scholar 

  25. Royston P, Parmar MKB. Flexible parametric proportional‐hazards and proportional‐odds models for censored survival data, with application to prognostic modelling and estimation of treatment effects. Stat Med. 2002;21:2175–97.

    PubMed  Google Scholar 

  26. Harrell F. Regression modeling strategies: with applications to linear models, logistic and ordinal regression, and survival analysis. 2nd ed. New York: Springer; 2015.

  27. Woods BS, Sideris E, Palmer S, Latimer N, Soares M. Partitioned survival and state transition models for healthcare decision making in oncology: where are we now? Value Health. 2020;23:1613–21.

    PubMed  Google Scholar 

  28. Cranmer H, Shields GE, Bullement A. A comparison of partitioned survival analysis and state transition multi-state modelling approaches using a case study in oncology. J Med Econ. 2020;23:1176–85.

    PubMed  Google Scholar 

  29. R Core Team. R: a language and environment for statistical computing. Vienna: R Foundation for Statistical Computing; 2014.

  30. De Wreede LC, Fiocco M, Putter H. The mstate package for estimation and prediction in non-and semi-parametric multi-state and competing risks models. Comput Methods Prog Biomed. 2010;99:261–74.

    Google Scholar 

  31. Therneau T, Crowson C, Atkinson E. Multi-state models and competing risks. 2016. https://cran.r-project.org/web/packages/survival/vignettes/compete.pdf.

  32. Allaire JJ, Ellis P, Gandrud C, Kuo K, Lewis BW, Owen J, et al. Package ‘networkD3.’ D3 JavaScript Netw graphs from R. 2017. https://CRAN.R-project.org/package=networkD3.

  33. Incerti D, Jeroen G, Jansen P. hesim: Health Economic Simulation Modeling and Decision Analysis. 2021 Feb 18 [cited 2024 Jan 3]; Available from: https://arxiv.org/abs/2102.09437v2.

  34. Jones RP, Sutton PA, Evans JP, Clifford R, McAvoy A, Lewis J, et al. Specific mutations in KRAS codon 12 are associated with worse overall survival in patients with advanced and recurrent colorectal cancer. Br J Cancer. 2017;116:923–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Imamura Y, Morikawa T, Liao X, Lochhead P, Kuchiba A, Yamauchi M, et al. Specific mutations in KRAS codons 12 and 13, and patient prognosis in 1075 BRAF wild-type colorectal cancers. Clin Cancer Res. 2012;18:4753–63.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Modest DP, Ricard I, Heinemann V, Hegewisch-Becker S, Schmiegel W, Porschen R, et al. Outcome according to KRAS-, NRAS-and BRAF-mutation as well as KRAS mutation variants: pooled analysis of five randomized trials in metastatic colorectal cancer by the AIO Colorectal Cancer Study Group. Ann Oncol. 2016;27:1746–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Castañon E, Sanchez-Arraez Á, Jimenez-Fonseca P, Alvarez-Manceñido F, Martínez-Martínez I, Gongora LM, et al. Bayesian interpretation of immunotherapy trials with dynamic treatment effects. Eur J Cancer. 2022;161:79–89.

    PubMed  Google Scholar 

  38. Castañon E, Sanchez-Arraez A, Alvarez-Manceñido F, Jimenez-Fonseca P, Carmona-Bayonas A. Critical reappraisal of phase III trials with immune checkpoint inhibitors in non-proportional hazards settings. Eur J Cancer. 2020;136:159–68.

    PubMed  Google Scholar 

  39. Mitani S, Taniguchi H, Honda K, Masuishi T, Narita Y, Kadowaki S, et al. Analysis of efficacy and prognostic factors in second-line chemotherapy for BRAF V600E mutant metastatic colorectal cancer. Ann Oncol. 2017;28:v180.

    Google Scholar 

  40. Tabernero J, Van Cutsem E, Yoshino T, Yaeger R, Wasan H, Desai J, et al. SO-29 Clinical characteristics, efficacy, and safety in patients receiving second-or third-line encorafenib plus cetuximab (E+ C) vs control for metastatic colorectal cancer (mCRC): BEACON CRC post hoc analysis. Ann Oncol. 2023;34:S174.

    Google Scholar 

  41. Tabernero J, Grothey A, Van Cutsem E, Yaeger R, Wasan H, Yoshino T, et al. Encorafenib plus cetuximab as a new standard of care for previously treated BRAF V600E–mutant metastatic colorectal cancer: updated survival results and subgroup analyses from the BEACON study. J Clin Oncol. 2021;39:273.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Wirapati P, Pomella V, Vandenbosch B, Kerr P, Maiello E, Jeffery GM, et al. Velour trial biomarkers update: Impact of RAS, BRAF, and sidedness on aflibercept activity. J Clin Oncol. 2017;35:3538.

  43. Loupakis F, Cremolini C, Masi G, Lonardi S, Zagonel V, Salvatore L, et al. Initial therapy with FOLFOXIRI and bevacizumab for metastatic colorectal cancer. N Engl J Med. 2014;371:1609–18.

    PubMed  Google Scholar 

  44. Yoshino T, Portnoy DC, Obermannova R, Bodoky G, Prausová J, Garcia-Carbonero R, et al. Biomarker analysis beyond angiogenesis: RAS/RAF mutation status, tumour sidedness, and second-line ramucirumab efficacy in patients with metastatic colorectal carcinoma from RAISE—a global phase III study. Ann Oncol. 2019;30:124–31.

    CAS  PubMed  Google Scholar 

  45. Chida K, Kotani D, Masuishi T, Kawakami T, Kawamoto Y, Kato K, et al. The prognostic impact of KRAS G12C mutation in patients with metastatic colorectal cancer: a multicenter retrospective observational study. Oncologist. 2021;26:845–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Fakih M, Tu H, Hsu H, Aggarwal S, Chan E, Rehn M, et al. Real-world study of characteristics and treatment outcomes among patients with KRAS p. G12C-mutated or other KRAS mutated metastatic colorectal cancer. Oncologist. 2022;27:663–74.

    PubMed  PubMed Central  Google Scholar 

  47. Cremolini C, Antoniotti C, Lonardi S, Bergamo F, Cortesi E, Tomasello G, et al. Primary tumor sidedness and benefit from FOLFOXIRI plus bevacizumab as initial therapy for metastatic colorectal cancer. Retrospective analysis of the TRIBE trial by GONO. Ann Oncol. 2018;29:1528–34.

    CAS  PubMed  Google Scholar 

  48. Sunakawa Y, Satake H, Usher J, Jaimes Y, Miyamoto Y, Nakamura M, et al. Dynamic changes in RAS gene status in circulating tumour DNA: a phase II trial of first-line FOLFOXIRI plus bevacizumab for RAS-mutant metastatic colorectal cancer (JACCRO CC-11). ESMO Open. 2022;7:100512.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Mok TS, Wu Y-L, Thongprasert S, Yang C-H, Chu D-T, Saijo N, et al. Gefitinib or carboplatin–paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947–57.

    CAS  PubMed  Google Scholar 

  50. Siena S, Sartore-Bianchi A, Garcia-Carbonero R, Karthaus M, Smith D, Tabernero J, et al. Dynamic molecular analysis and clinical correlates of tumor evolution within a phase II trial of panitumumab-based therapy in metastatic colorectal cancer. Ann Oncol. 2018;29:119–26.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank PROMETEO study researchers for their contribution to this study and Miguel Vaquero, Natalia Cateriano and the IRICOM S.L. team for the support of the website registry.

Funding

The study has been funded by Roche and Sanofi Spain.

Author information

Authors and Affiliations

Authors

Contributions

DC-T, PJ-F, AFM, CLL, and ACB conceived the project, monitored variables, analysed data, and wrote the initial draft of the manuscript. The remaining authors were involved in patient recruitment, supplying clinical information, and offering comments and suggestions for manuscript improvement. All authors contributed to data interpretation, discussion, and critical review of the manuscript.

Corresponding authors

Correspondence to Débora Contreras-Toledo or Alberto Carmona-Bayonas.

Ethics declarations

Competing interests

PJ-F discloses honoraria as speaker from Astellas, Astra Zeneca, BMS, Lilly, MSD, Novartis, Roche. CLL discloses consulting/advisory role for Amgen, Roche, Sanofi, MSD, Merck, Servier, Bayer; research grants from Amgen, Roche, MSD, Merck, Servier and travel grants and/or congress support from Servier, Sanofi, Amgen, Roche, MSD, Merck. AFM discloses honoraria as speaker from Amgen, Astra Zeneca, Astra Zeneca, Eisai, Lilly, MSD, Pierre Fabre, Servier. AMLM discloses consulting/advisory role for Amgen, Bayer, Roche, Eisai; honoraria as speaker from Eisai, Lilly, Amgen, Bayer, Sanofi, Merck Serono, Roche, Bristol, Servier, Pierre Fabre; travel grants and/or congress support from Amgen, Roche, Servier. FVR discloses consulting/advisory role for MSD; honoraria from Roche, Servier, Lilly, Merck, Astra-Zeneca; travel grants and/or congress support from Merck, Roche, Servier. VA discloses advisory role for Amgen, Roche, Merck, Servier, Ipsen pharma, Novartis; travel grants and/or congress support from Merck. JA discloses consulting/advisory role for Merck; honoraria as speaker from Amgen, Merck and Servier; travel grants and/or congress support from Roche, Amgen, Merck, Servier and Sanofi. FS discloses consulting/advisory role for Merck Serono, Amgen; honoraria and travel grants and/or congress support from Hoffman La-Roche, Sanofi Aventis, Amgen, Merck Serono, Servier, Bristol-Myers Squibb and Terumo corporation. MG discloses travel grants and/or congress support from Roche, Amgen and Servier. AMC discloses consulting/advisory role for BMS, Sanofi and Novartis; honoraria as speaker from Roche, BMS, MSD, Novartis, Merck and Servier; travel grants and/or congress support from BMS, Novartis, Merck and Servier. RJM discloses honoraria as speaker from Servier, Sanofi, Pfizer and Pierre Fabre; travel grants and/or congress support from Merck. SCG discloses travel grants and/or congress support from Bristol Myers Squibb, MSD, Rovi, Amgem. BGA discloses consulting/advisory role for Sanofi; honoraria as speaker from Roche, BMS, Amgen MSD, Merck and Servier; travel grants and/or congress support from Amgen, Merck and Servier. PGV discloses consulting/advisory role for Servier, Amgen; travel grants and/or congress support from Merck, Sanofi, Amgen. ACVM discloses travel grants and/or congress support from Amgen, Merck, MSD, Roche, Sanofi, Servier. MMS discloses consulting/advisory role: Merck, Servier; honoraria as speaker from Amgen, Merck, Servier and Bayer; travel grants and/or congress support from Roche, Amgen, Merck, Servier. CCL discloses consulting/advisory role for Merck, honoraria as speaker from Amgen, Bayer, Sanofi, Merck Serono, Roche, Servier; travel grants and/or congress support from: Amgen, Bayer, Sanofi, Merck Serono, Roche, Servier. JAp discloses consulting/advisory role for Amgen, Bayer, Merck, Merck Sharp & Dohme, Pierre Fabre and Servier. AC-B discloses honoraria as speaker from Amgen, Astellas, Bayer, BMS, Eisai, Lilly, MSD, Merck, Novartis, Roche, Servier. DC-T, MCR, EAM, ABF-D, BA, and ACC have no conflict of interest, financial or otherwise, in relation to the scope of this work.

Ethics approval and consent to participate

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.

Consent for publication

Informed consent was obtained from all individual participants included in the study.

Additional information

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

Supplementary information

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

Contreras-Toledo, D., Jiménez-Fonseca, P., López, C.L. et al. Dynamic nature of BRAF or KRAS p.G12C mutations in second-line therapy for advanced colorectal cancer patients: do early and late effects exist?. Br J Cancer 130, 777–787 (2024). https://doi.org/10.1038/s41416-023-02563-w

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41416-023-02563-w

Search

Quick links