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:

Sotorasib with panitumumab in chemotherapy-refractory KRASG12C-mutated colorectal cancer: a phase 1b trial

Abstract

The current third-line (and beyond) treatment options for RAS-mutant metastatic colorectal cancer have yielded limited efficacy. At the time of study start, the combination of sotorasib, a KRAS (Kirsten rat sarcoma viral oncogene homolog)-G12C inhibitor, and panitumumab, an epidermal growth factor receptor (EGFR) inhibitor, was hypothesized to overcome treatment-induced resistance. This phase 1b substudy of the CodeBreaK 101 master protocol evaluated sotorasib plus panitumumab in patients with chemotherapy-refractory KRASG12C-mutated metastatic colorectal cancer. Here, we report the results in a dose-exploration cohort and a dose-expansion cohort. Patients received sotorasib (960 mg, once daily) plus panitumumab (6 mg kg−1, once every 2 weeks). The primary endpoints were safety and tolerability. Secondary endpoints included efficacy and pharmacokinetics. Exploratory biomarkers at baseline were assessed. Forty-eight patients (dose-exploration cohort, n = 8; dose-expansion cohort, n = 40) were treated. Treatment-related adverse events of any grade and grade ≥3 occurred in 45 (94%) and 13 (27%) patients, respectively. In the dose-expansion cohort, the confirmed objective response rate was 30.0% (95% confidence interval (CI) 16.6%, 46.5%). Median progression-free survival was 5.7 months (95% CI 4.2, 7.7 months). Median overall survival was 15.2 months (95% CI 12.5 months, not estimable). Prevalent genomic coalterations included APC (84%), TP53 (74%), SMAD4 (33%), PIK3CA (28%) and EGFR (26%). Sotorasib–panitumumab demonstrated acceptable safety with promising efficacy in chemotherapy-refractory KRASG12C-mutated metastatic colorectal cancer. ClinicalTrials.gov identifier: NCT04185883.

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: Study schema.
Fig. 2: Patient disposition.
Fig. 3: Activity of sotorasib therapy in the dose-expansion cohort.

Similar content being viewed by others

Data availability

Qualified researchers may request data from Amgen clinical studies. Complete details are available at https://www.amgen.com/science/clinical-trials/clinical-data-transparency-practices/clinical-trial-data-sharing-request. Source data are provided with this paper.

References

  1. Sung, H. et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209–249 (2021).

    Article  PubMed  Google Scholar 

  2. Siegel, R. L., Miller, K. D., Wagle, N. S. & Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 73, 17–48 (2023).

    Article  PubMed  Google Scholar 

  3. Benson, A. B. et al. Colon cancer, version 2.2021, NCCN clinical practice guidelines in oncology. J. Natl. Compr. Canc. Netw. 19, 329–359 (2021).

    Article  PubMed  Google Scholar 

  4. Grothey, A. et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 381, 303–312 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Mayer, R. J. et al. Randomized trial of TAS-102 for refractory metastatic colorectal cancer. N. Engl. J. Med. 372, 1909–1919 (2015).

    Article  PubMed  Google Scholar 

  6. Prager, G. W. et al. Trifluridine–tipiracil and bevacizumab in refractory metastatic colorectal cancer. N. Engl. J. Med. 388, 1657–1667 (2023).

    Article  CAS  PubMed  Google Scholar 

  7. Ji, J., Wang, C. & Fakih, M. Targeting KRASG12C-mutated advanced colorectal cancer: research and clinical developments. OncoTargets Ther. 15, 747–756 (2022).

    Article  Google Scholar 

  8. Schirripa, M. et al. KRAS G12C metastatic colorectal cancer: specific features of a new emerging target population. Clin. Colorectal Cancer 19, 219–225 (2020).

    Article  PubMed  Google Scholar 

  9. Lee, J. K. et al. Comprehensive pan-cancer genomic landscape of KRAS altered cancers and real-world outcomes in solid tumors. NPJ Precis. Oncol. 6, 91 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Henry, J. T. et al. Comprehensive clinical and molecular characterization of KRASG12C-mutant colorectal cancer. JCO Precis. Oncol. 5, 613–621 (2021).

    Article  Google Scholar 

  11. Fakih, 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 27, 663–674 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Alawawdeh, A. et al. Prognostic differences of RAS mutations: results from the South Australian Metastatic Colorectal Registry. Target. Oncol. 17, 35–41 (2022).

    Article  PubMed  Google Scholar 

  13. Osterlund, E. et al. KRAS-G12C mutation in one real-life and three population-based Nordic cohorts of metastatic colorectal cancer. Front. Oncol. 12, 826073 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Canon, J. et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 575, 217–223 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Fakih, M. G. et al. Sotorasib for previously treated colorectal cancers with KRASG12C mutation (CodeBreaK100): a prespecified analysis of a single-arm, phase 2 trial. Lancet Oncol. 23, 115–124 (2022).

    Article  CAS  PubMed  Google Scholar 

  16. Ryan, M. B. et al. KRASG12C-independent feedback activation of wild-type RAS constrains KRASG12C inhibitor efficacy. Cell Rep. 39, 110993 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Amodio, V. et al. EGFR blockade reverts resistance to KRASG12C inhibition in colorectal cancer. Cancer Discov. 10, 1129–1139 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Yaeger, R. et al. Adagrasib with or without cetuximab in colorectal cancer with mutated KRAS G12C. N. Engl. J. Med. 388, 44–54 (2023).

    Article  CAS  PubMed  Google Scholar 

  19. Fakih, M. G. et al. Sotorasib plus panitumumab in refractory colorectal cancer with mutated KRAS G12C. N. Engl. J. Med. https://doi.org/10.1056/nejmoa2308795 (2023).

    Article  PubMed  Google Scholar 

  20. Tabernero, J. 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. 39, 273–284 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Fakih, M. & Vincent, M. Adverse events associated with anti-EGFR therapies for the treatment of metastatic colorectal cancer. Curr. Oncol. 17, S18–S30 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Tabernero, J. et al. Trifluridine/tipiracil plus bevacizumab for third-line treatment of refractory metastatic colorectal cancer: the phase 3 randomized SUNLIGHT study. J. Clin. Oncol. 41, 4-4 (2023).

    Article  Google Scholar 

  23. Ahronian, L. G. et al. Clinical acquired resistance to RAF inhibitor combinations in BRAF-mutant colorectal cancer through MAPK pathway alterations. Cancer Discov. 5, 358–367 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Corcoran, R. B. et al. BRAF gene amplification can promote acquired resistance to MEK inhibitors in cancer cells harboring the BRAF V600E mutation. Sci. Signal. 3, ra84 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. De Roock, W. et al. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol. 11, 753–762 (2010).

    Article  PubMed  Google Scholar 

  26. Johnson, R. M. et al. ARID1A mutations confer intrinsic and acquired resistance to cetuximab treatment in colorectal cancer. Nat. Commun. 13, 5478 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Oddo, D. et al. Molecular landscape of acquired resistance to targeted therapy combinations in BRAF-mutant colorectal cancer. Cancer Res. 76, 4504–4515 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Topham, J. T. et al. Circulating tumor DNA identifies diverse landscape of acquired resistance to anti-epidermal growth factor receptor therapy in metastatic colorectal cancer. J. Clin. Oncol. 41, 485–496 (2023).

    Article  CAS  PubMed  Google Scholar 

  29. Kato, S. et al. Genomic assessment of blood-derived circulating tumor DNA in patients with colorectal cancers: correlation with tissue sequencing, therapeutic response, and survival. JCO Precis. Oncol. 3, PO.18.00158 (2019).

    PubMed  PubMed Central  Google Scholar 

  30. Gupta, R. et al. Guardant360 circulating tumor DNA assay is concordant with FoundationOne next-generation sequencing in detecting actionable driver mutations in anti-EGFR naive metastatic colorectal cancer. Oncologist 25, 235–243 (2020).

    Article  CAS  PubMed  Google Scholar 

  31. Liao, S. et al. A genetic interaction analysis identifies cancer drivers that modify EGFR dependency. Genes Dev. 31, 184–196 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Guo, W., Wang, S. J., Yang, S., Lynn, H. & Ji, Y. A Bayesian interval dose-finding design addressingOckham’s razor: mTPI-2. Contemp. Clin. Trials 58, 23–33 (2017).

    Article  PubMed  Google Scholar 

  33. Clopper, C. J. & Pearson, E. S. The use of confidence or fiducial limits illustrated in the case of the binomial. Biometrika 26, 404–413 (1934).

    Article  Google Scholar 

Download references

Acknowledgements

We thank the patients and their families for participating in this trial. We thank T. Harrison of Amgen Inc. and J. Martucci of Amgen Inc. for operational planning assistance, K. Price (all while employed by Amgen Inc.) and A. Joshi of Cactus Life Sciences (part of Cactus Communications) for medical writing support, S. Dastidar of Cactus Life Sciences (part of Cactus Communications) for editorial assistance and R. Dawson of Cactus Life Sciences (part of Cactus Communications) for graphics assistance. This study received support from the following grants to D.S.H.: MD Anderson Cancer Center Support Grant (National Institutes of Health and National Cancer Institute P30 CA016672), Clinical Translational Science Award 1UL1 TR003167, Cancer Prevention Research Institute of Texas (CPRIT) Precision Oncology Decision Support Core (RP150535) and Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy. This study was sponsored and funded by Amgen Inc.

Author information

Authors and Affiliations

Authors

Contributions

E.C. contributed to the design and conception of the study. Y.K., M.F., J.S., R.Y., T.M., E.J.K., C.M.B., S.K., G.S.F., C.L., J.K., S.P. and D.S.H. contributed to patient data collection. All authors contributed to data analysis and interpretation. All authors contributed to the writing and editing of the manuscript and approved the final version for submission to the journal. All authors are accountable for all aspects of the work.

Corresponding author

Correspondence to David S. Hong.

Ethics declarations

Competing interests

Y.K.: honoraria—Bristol-Myers Squibb Japan, Lilly Japan, Taiho Pharmaceutical; consulting or advisory role—Amgen, Boehringer Ingelheim, Takeda; research funding—AbbVie (institution), Amgen (institution), Astellas Pharma (institution), AstraZeneca (institution), Boehringer Ingelheim (institution), Chugai Pharma (institution), Genmab (institution), GlaxoSmithKline (institution), Incyte (institution), Janssen Oncology (institution), Lilly (institution), Merck Serono (institution), Ono Pharmaceutical (institution), Taiho Pharmaceutical (institution), Takeda (institution), Hengrui (institution), Novartis Pharma (institution); travel, accommodations, expenses—Amgen. M.F.: grants or contracts—Amgen (institution), Agenus Bio (institution), Verastem (institution), Genentech/imCORE (institution); consulting fees (payments made to self)—AstraZeneca, Bristol-Myers Squibb, Bayer, Incyte, Pfizer, Taiho Oncology; participation in a data safety monitoring board or an advisory board (payments made to self)—Bayer, Eisai Oncology, Entos, Merck, Mirati Therapeutics, Nouscom, Roche/Genentech, Seattle Genetics, Xenthera Inc.; receipt of equipment, materials, drugs, medical writing, gifts or other services—Bristol-Myers Squibb (study-related drugs provided to institution). J.S.: consultant or advisory role—AbbVie, Amgen, AstraZeneca, Bayer, BeiGene, Daiichi-Sankyo, Eli Lilly, GlaxoSmithKline, Janssen, Natera, Pfizer, Pionyr Immunotherapeutics, Roche/Genentech, Seagen, Silverback Therapeutics, Taiho Pharmaceutical, Takeda, Viatris, Zentalis; research funding or contracted research—AbbVie (institution), Amgen (institution), AStar D3 (institution), Bayer (institution), BeiGene (institution), Curegenix (institution), Daiichi-Sankyo (institution), Eli Lilly (institution), Erasca (institution), Leap Therapeutics (institution), Roche/Genentech (institution), Seagen (institution), Silverback Therapeutics (institution). R.Y.: honoraria—Zai Lab; consulting or advisory role—Mirati Therapeutics, Amgen Inc.; research funding—Array BioPharma (institution), Boehringer Ingelheim (institution), Mirati Therapeutics (institution), Pfizer (institution), Daiichi-Sankyo (institution). T.M.: honoraria—Bayer Yakuhin, Bristol-Myers Squibb, Chugai Pharma, Daiichi-Sankyo, Lilly, Merck Serono, Ono Pharmaceutical, Sanofi, Taiho Pharmaceutical, Takeda, Yakult Honsha, MSD, Nippon Kayaku; research funding—Amgen (institution), Boehringer Ingelheim (institution), CMIC (institution), Daiichi-Sankyo (institution), Lilly Japan (institution), MSD (institution), Novartis (institution), Ono Pharmaceutical (institution), Pfizer (institution), Syneos Health (institution). E.J.K.: grants or contracts—NGM (institution), Erytech (institution), Fibrogen (institution), Eureka (institution), Merck (institution), Bristol-Myers Squibb (institution), Astellas (institution), Boston Biomedical (institution), AstraZeneca (institution), Celgene (institution), Halozyme (institution), Samumed (institution), Epicentrx (institution), MedImmune (institution), BeiGene (institution), Theriva Biologics (institution); consulting fees—Eisai (self), Taiho (self), Lilly (self), Relay (self); payment or honoraria for lectures, presentations, speakers’ bureaus, manuscript writing or educational events—Eisai (speaker), Seagen (speaker); monitoring board or advisory board—Lilly (self), Relay (self); stock or stock options—ROMTech. C.M.B.: consulting or advisory role—AbbVie (institution), AstraZeneca, Bristol-Myers Squibb/Celgene, Clinical Care Options, Creative Educational Concepts, Curio Science, CVS, Daiichi-Sankyo, EMD Serono, Genentech, Janssen, Jazz Pharmaceuticals, Medical Learning Institute, MJH Holdings, Novartis, Novocure, OncLive Clinical Congress Consultants, Sanofi/Regeneron, Seagen, Takeda, Targeted Oncology, Tempus; speakers’ bureau—Merck; research funding—Amgen, AstraZeneca/MedImmune, Bristol-Myers Squibb. S.K.: stock and other ownership interests—Frontier Medicines, Iylon, Lutris, Navire, Xilis; consulting or advisory role—AbbVie, Accademia Nazionale di Medicina (ACCMED), Amal Therapeutics, Amgen, AstraZeneca/MedImmune, Bayer Health, Bicara Therapeutics, Black Diamond Therapeutics, Boehringer Ingelheim, Bristol-Myers Squibb/Medarex, Cardiff Oncology, Carina Biotech, CureTeq, EMD Serono, Endeavor BioMedicines, Flame Biosciences, Foundation Medicine, Frontier Medicines, Genentech, Genomic Health, Gilead Sciences, GlaxoSmithKline, HalioDx, Harbinger Oncology Inc., Holy Stone Healthcare, Inivata, Ipsen, Iylon, Jacobio, Jazz Pharmaceuticals, Johnson & Johnson/Janssen, Lilly, Lutris, Merck, Mirati Therapeutics, NeoGenomics Laboratories, Novartis, Numab, Ono Pharmaceutical, Pfizer, Redx Pharma, Repare Therapeutics, Replimune, Servier, Taiho Pharmaceutical, Takeda, Tempus, Xilis, Zentalis; research funding—Amgen, Array BioPharma, Biocartis, Daiichi-Sankyo, EMD Serono, Genentech/Roche, Guardant Health, Lilly, MedImmune, Novartis, Sanofi. G.S.F.: royalties (self)—Wolters Kluwer (2014–present); advisory role (to institution)—AbbVie (2022), Fujifilm (2018), Silicon (2020, 2021), Navire (2021), Turning Point (2021), Predicine (2021), Inspirna (2021), Regeneron (2021), Jubilant (2022), BostonGene (2022), Teon (2022), Merck (2022), Sanofi (2023), BridgeBio (2023); advisory role (self)—EMD Serono (2010, 2011); speaker’s honorarium for continuing medical education—Total Health Conferencing (2019), Rocky Mountain Oncology Society (2020); travel (self, for work and/or research related to institution)—Amgen (2022), Bristol-Myers Squibb (2015), EMD Serono (2011, 2012, 2013), Fujifilm (2018), Millennium (2013), Sarah Cannon Research Institute (employer, at least once yearly), Synthorx/Sanofi (2022); research funding (to institution, for any trial for which G.S.F. has been the primary investigator (ever) or subinvestigator (minimum last 4 years))—3-V Biosciences, Abbisko, AbbVie, ABL Bio, ADC Therapeutics, Accutar, Agenus, Aileron, American Society of Clinical Oncology, Amgen, ARMO/Eli Lilly, Artios, AstraZeneca, Bayer, BeiGene, Bioatla, Bioinvent, Biothera, Bicycle, Black Diamond, Boehringer Ingelheim, Celgene, Celldex, Ciclomed, Curegenix, Curis, Cyteir, Daiichi, DelMar, eFFECTOR, Eli Lilly, EMD Serono, Epizyme, Erasca, Exelixis, Freenome, Fujifilm, Genmab, GlaxoSmithKline, Hutchison MediPharma, IGM Biosciences, Ignyta, Immunitas, ImmunoGen/MacroGenics, Incyte, Jacobio, Jazz, Jounce, Jubilant, Kineta, Kolltan, Loxo/Bayer, MedImmune, Merck, Metabomed, Millennium, Mirati, miRNA Therapeutics, Molecular Templates, National Institutes of Health, Navire/BridgeBio, NGM Bio, NiKang, Novartis, OncoMed, Oncorus, Oncothyreon, Poseida, Precision Oncology, Prelude, PureTech, Pyramid, Pyxis, RasCal, Regeneron, Relay, Rgenix, Ribon, Roche, Samumed, Sapience, Seagen, Silicon, Simcha, Sirnaomics, Strategia, Syndax, Synthorx/Sanofi, Taiho, Takeda, Tallac, Tarus, Tarveda, Teneobio, Tesaro, Tocagen, Turning Point, University of Texas MD Anderson Cancer Center, Vegenics, Xencor, Zhuhai Yufan. C.L.: honoraria—AstraZeneca, Genentech/Roche, Lilly/ImClone, Merck, Takeda Science Foundation; consulting or advisory role—Abbott Biotherapeutics, ARIAD, AstraZeneca, Bayer/Onyx, Bristol-Myers Squibb, Cancer Support Community, Celgene, Clarient, Clovis Oncology, Genentech/Roche, Gilead Sciences, Lilly/ImClone, Merck, Novocure, Pfizer, Regeneron, Takeda; research funding—Advantagene (institution), Amgen, ARIAD (institution), AstraZeneca, Celgene (institution), Clovis Oncology (institution), Genentech/Roche (institution), GlaxoSmithKline (institution), Incyte, Inovio Pharmaceuticals (institution), Johnson & Johnson/Janssen, Lilly, Merck (institution), Stem CentRx (institution), Trizell (institution). J.K.: Bristol-Myers Squibb, Hutchison Medipharma/Syneos Health, Sumitomo Dainippon/Paraexel, Tempest Therapeutics, Merck/NSABP (National Surgical Adjuvant Breast and Bowel Project) Foundation, Ignyta/Qintiles, Tempest Therapeutics/ICON, AstraZeneca/NSABP Foundation, AbbVie, Bristol-Myers Squibb/Mayo Clinic, MedImmune, Pfizer, Daiichi-Sankyo/Syneos Health, Xencor, Amgen Inc., Merck, Gilead Sciences, Janssen. S.P.: honoraria—Aptitude Health (institution), IntegrityCE; consulting or advisory role—AstraZeneca (institution), G1 Therapeutics, Jazz Pharmaceuticals (institution), Pfizer; travel, accommodations, expenses—Dava Oncology. P.C.: employee and stockholder—Amgen Inc.; patents, royalties, other intellectual property—Amgen. E.C.: employee and stockholder—Amgen Inc. T.V.: employee and stockholder—Amgen Inc. L.M.: employee and stockholder—Amgen Inc. A.A.: employee and stockholder—Amgen Inc. Q.T.: employee and stockholder—Amgen Inc. D.S.H.: stock and other ownership interests—MolecularMatch, OncoResponse, Telperian; consulting or advisory role—Acuta, Adaptimmune, Alkermes, Alphasights, Amgen, AUM Biosciences, Axiom Biotechnologies, Baxter, Bayer, Boxer Capital, Bridgebio, COG, Cor2Ed, Cowen, EcoR1 Capital, Genentech, Gennao Bio, Gerson Lehrman Group, Gilead Sciences, GroupH, Guidepoint Global, HCW Precision, Immunogen, Infinity Pharmaceuticals, Janssen, Liberum, MedaCorp, Medscape, Numab, Oncologia Brasil, Pfizer, Pharma Intelligence, Precision Oncology Experimental Therapeutics, Prime Oncology, RAIN, STCube, Takeda, Tavistock Life Sciences, Trieza Therapeutics, Turning Point Therapeutics, WebMD, YingLing Pharma, ZIOPHARM Oncology; research funding—AbbVie (institution), Adaptimmune (institution), Adlai Nortye (institution), Amgen (institution), AstraZeneca (institution), Bayer (institution), Bristol-Myers Squibb (institution), Daiichi-Sankyo (institution), Deciphera (institution), Eisai (institution), Endeavor BioMedicines (institution), Erasca Inc. (institution), F. Hoffmann LaRoche (institution), Fate Therapeutics (institution), Genentech (institution), Genmab (institution), Ignyta (institution), Infinity Pharmaceuticals (institution), Kite, a Gilead Company (institution), Kyowa (institution), Lilly (institution), Loxo (institution), MedImmune (institution), Merck (institution), Mirati Therapeutics (institution), Mologen (institution), National Cancer Institute (institution), Navire (institution), Novartis (institution), Numab (institution), Pfizer (institution), Pyramid Biosciences (institution), Seagen (institution), Takeda (institution), TCR2 Therapeutics (institution), Teckro (institution), Turning Point Therapeutics (institution), VM Pharma (institution); travel, accommodations, expenses—American Association for Cancer Research, American Society of Clinical Oncology, Bayer Schering Pharma, Genmab, Society for Immunotherapy of Cancer, Telperian.

Peer review

Peer review information

Nature Medicine thanks Thierry André, Jorge Barriuso, Andres Cervantes and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Ulrike Harjes, in collaboration with the Nature Medicine team.

Additional information

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

Extended data

Extended Data Fig. 1 Cmax and AUC0–24 on Day 1 and Day 8 following oral administration of 960 mg sotorasib QD in Japanese vs US patients.

The lower and upper hinges represent the first and third quartiles (the 25th and 75th percentiles). The upper whisker extends from the hinge to the largest value no further than 1.5 * IQR (inter-quartile range) from the hinge. The lower whisker extends from the hinge to the smallest value at most 1.5 * IQR of the hinge. Data beyond the end of the whiskers represents outliers. Day 1: Japan, n=9; United States, n=37. Day 8: Japan, n=8, United States, n=34–35. AUC0–24h, area under the concentration-time curve from time 0 to 24 h after dose; Cmax, maximum observed drug concentration.

Extended Data Fig. 2 Mean (+SD) plasma concentration on Day 1 and Day 8 following oral administration of 960 mg sotorasib QD.

The black line represents mean (+SD) average sotorasib plasma concentration for the overall population. Red and blue lines represent mean (+SD) average sotorasib plasma concentration for Japanese and US patients, respectively. The inset shows results for Japanese vs United States sites. Day 1: Japan, n=9; United States, n=36–37. Day 8: Japan, n=8; United States, n=32–37. SD, standard deviation.

Extended Data Fig. 3 Survival according to tumor location.

Kaplan-Meier curves of (A) progression-free survival and (B) overall survival, according to right-sided vs left-sided tumor. Vertical lines indicate censoring. For the PFS curve, disease progression or death (whichever occurs earlier) is an event. For the OS curve, death is an event. CI, confidence interval; CRC, colorectal cancer; NE, not estimable; OS, overall survival; PFS, progression-free survival.

Extended Data Fig. 4 Survival according to baseline genomic co-alterations.

Kaplan-Meier curves of (A) PFS according to BRAF alterations, and (B) PFS according to ARID1A mutations. Vertical lines indicate censoring. PFS, progression-free survival.

Supplementary information

Supplementary Information

Supplementary Methods, list of sites and their laboratories and imaging facilities that screened and/or enrolled patients into this study, additional information on patients enrolled in this study and Supplementary Tables 1–8.

Reporting Summary

Supplementary Data

Statistical data for Supplementary Tables 2–6.

Source data

Source Data

Statistical source data for Tables 1–3 and Figs. 2 and 3.

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

Kuboki, Y., Fakih, M., Strickler, J. et al. Sotorasib with panitumumab in chemotherapy-refractory KRASG12C-mutated colorectal cancer: a phase 1b trial. Nat Med 30, 265–270 (2024). https://doi.org/10.1038/s41591-023-02717-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-023-02717-6

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing