Review article
The Pharmacologic Inhibition of KRAS Mutants as a Treatment for Cancer
Therapeutic Principles and Clinical Results
;
Background: Mutations of the KRAS oncogene are found in up to 20% of all cancers, and particularly in non–small-cell lung cancer (NSCLC) (20–40%) and colorectal cancer (CRC) (30–50%). Inhibitors of specific KRAS mutants have recently become available and are now a part of routine care.
Methods: This review is based on articles published in the past 5 years that were retrieved by a selective search in PubMed for clinical trials of the pharmacological inhibition of KRAS.
Results: Sotorasib and adagrasib have already been approved, on the basis of two randomized phase III trials, as specific inhibitors of the KRASG12C mutant for palliative second-line treatment. Compared to standard chemotherapy with docetaxel, both drugs significantly prolonged progression-free survival (PFS): 5.6 months (95% confidence interval [4.3; 7.8]) for sotorasib versus 4.5 [3.0; 5.7] for docetaxel, and 5.5 months [4.5; 6.7] for adagrasib versus 3.8 [2.7; 4.7] for docetaxel. Sotorasib was also found to cause fewer severe adverse drug events (33%, versus 40% with docetaxel). The most common ones were diarrhea and elevated liver enzymes. For already treated CRC, sotorasib combined with the anti-epidermal growth factor receptor (anti-EGFR) antibody panitumumab was found, in a randomized phase III trial, to prolong progression-free survival significantly compared to standard therapy with triflurdin/tipiracil or regorafenib (5.6 months [4.2; 6.3] versus 2.2 months [1.9; 3.9]), while also improving patients’ quality of life. Approval by the European Medicines Agency is pending. Further KRAS and pan-RAS inhibitors are now in early clinical development.
Conclusion: Pharmacological KRAS inhibition is a promising new approach to the treatment of many kinds of cancer.
Cite this as: Kasper S, Sebastian M: The pharmacologic inhibition of KRAS mutants as a treatment for cancer: Therapeutic principles and clinical results. Dtsch Arztebl Int 2024; 121: 163–7. DOI: 10.3238/arztebl.m2025.0002


The KRAS (Kirsten rat sarcoma virus) gene is one of several oncogenes that play a key role in the development of many types of cancer. Activating mutations of this gene lead to the activation of downstream signaling pathways; they promote proliferation, inhibit programmed cell death, and mediate resistance to anticancer treatment (see Figure). KRAS is one of the most commonly mutated oncogenes in solid tumors, with KRAS mutations being present in as many as 20% of such tumors. Their prevalence varies widely depending on the tumor type: for example, they are present in 85–90% of pancreatic ductal adenocarcinomas (PDAC), in 30–50% of colorectal carcinomas (CRC), and in 20–40% of non-small cell lung carcinomas (NSCLC) (1). The localization of the mutation within the KRAS gene varies widely as well. The G12D mutation is found most often in PDAC, the G12D and G12V mutations in CRC, and the G12C mutation in NSCLC (2). The various KRAS mutations appear to have different prognostic implications depending on the underlying tumor disease.
For example, patients with KRASG12C-mutated CRC in metastatic disease have a shorter progression-free (PFS) and overall survival (OS) under palliative chemotherapy than those with other KRAS mutations (9.4 versus 10.8 months and 21.1 months versus 27.3 months, respectively). In contrast, the KRASG12C mutation in NSCLC seems not to lead to worse outcomes (3, 4, 5).
The pharmacologic inhibition of KRAS
The direct pharmacological inhibition of the KRAS protein with targeted small-molecule drugs was found to be exceedingly difficult, because suitable drugs for the molecule’s complex binding pockets were hard to find (6). Recently, however, specific orally administered inhibitors of certain KRAS mutations, such as KRASG12C, have been developed and successfully tested clinically. In addition to the specific inhibitors of the KRAS isoforms, pan-KRAS and pan-RAS inhibitors are now available that inhibit not only KRAS, but also the other members of the RAS family (HRAS, NRAS) (7, 8, 9, 10).
The KRASG12C mutation
The prevalence of the KRASG12C mutation varies widely across tumor types. In NSCLC, this specific isoform is the most common KRAS mutation, present in an estimated 8.9% of patients (11). It is much rarer in gastrointestinal tumors such as appendiceal carcinoma (3.9%), CRC (3.2%), CUP (“cancer of unknown primary”) syndrome (1.6%), PDAC (1.3%), small bowel carcinoma (1.4%), and hepatobiliary tumors (<1%) , and very rare in gynecological, urogenital, and hematological neoplasms (all <1%).
Clinical data on KRASG12C inhibitors in NSCLC
Sotorasib was the first orally administered KRASG12C inhibitor that could irreversibly block the mutant protein (12). In the single-arm Phase II CodeBreaK 100 trial in patients with previously treated NSCLC, a PFS of 6.7 months [5.3; 8.2] and an OS of 12.5 months [10.0; 17.8] were observed (13, 14). Although this trial was conducted without a control arm, these results led to the provisional approval of the drug by the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA), on condition that a confirmatory phase III study be conducted. In the subsequent randomized phase III CodeBreaK 200 trial, sotorasib was tested against standard second-line chemotherapy with docetaxel (15). The primary endpoint of this trial, PFS, was significantly longer with sotorasib (5.6 months [4.3; 7.8]) than with docetaxel (4.5 months [3.0; 5.7]), but longer OS was not found. Another noteworthy finding in this trial was the lower rate of ADR of grade 3 or above (33% in patients taking sotorasib and 40% in patients taking docetaxel). The main adverse drug reactions of sotorasib were diarrhea and elevated liver function tests; those of docetaxel were neutropenia, fatigue, and fever in neutropenia. The efficacy of sotorasib was also recently shown in an observational study involving a less stringently selected cohort, including patients with brain metastases (16). In this study, PFS was 4.8 months [3.9; 5.9] and OS was 9.8 months [6.5; not reached]. The cost of treatment with 960 mg qd sotorasib is approximately 4,800 euros. In addition to sotorasib, the KRASG12C -specific inhibitor adagrasib was also recently approved by the EMA and the FDA for the palliative second-line treatment of patients with advanced NSCLC who have a KRASG12C mutation. Approval was based on data from the randomized phase III KRYSTAL-12 trial of adagrasib versus docetaxel. The findings were presented at the American Society of Clinical Oncology (ASCO) in 2024 and have not yet been published in full (17). PFS was significantly longer under adagrasib (5.5 months [4.5; 6.7]) than under docetaxel [2.7; 4.7]. Adagrasib was also shown to be associated with a higher quality of life and fewer lung cancer symptoms than docetaxel (18). The rate of higher-grade adverse drug reactions (ADR) was similar with the two drugs: 47% under adagrasib (mainly diarrhea, nausea, and vomiting) and 45.7% under docetaxel (mainly anemia, asthenia, and diarrhea). Data on overall survival have not yet been released. Adagrasib is not yet available in Germany, but it is expected that the cost of treatment with 600 mg of adagrasib bid will be similar to that of sotorasib.
Other specific KRASG12C inhibitors, such as divarasib, are still in the early stages of clinical testing (19). Further trials now in progress involve KRASG12C inhibitors in combination with immunotherapeutic, cytostatic, or other targeted drugs.
Clinical data on KRASG12C inhibitors in colorectal cancer
In CRC, the specific KRASG12C mutation is rarer than in NSCLC, affecting only about 3% of patients. Yet in CRC, unlike in NSCLC, KRASG12C is associated with a poorer outcome than other KRAS mutations. Retrospective analyses showed that patients with KRASG12C had significantly shorter PFS (9.4 versus 10.8 months) and OS (21.1 versus 27.3 months) than patients with other KRAS mutations when given systemic chemotherapy for palliative treatment. New therapeutic strategies are urgently needed for these patients in particular (3). In early clinical trials, patients with CRC had lower response rates to the specific KRASG12C inhibitors sotorasib, adagrasib, and divarasib than patients with NSCLC, and combination therapies with other targeted drugs were therefore investigated directly (19, 20, 21). Thus, patients with chemotherapy-refractory CRC underwent trials of KRASG12C inhibitors combined with the epithelial growth factor receptor (EGFR) antibodies cetuximab or panitumumab. This type of combination therapy achieved higher response rates and longer median PFS times than monotherapy. In the phase II CodeBreaK 101 trial, the combination of sotorasib and panitumumab yielded a response rate of 33% with a median PFS and OS of 5.7 months [4.2; 7.7] and 15.2 months [12.5; not reached], respectively (22). The combination of adagrasib and cetuximab was investigated in the phase I/II KRYSTAL-1 trial (21): there was a 46% response rate with a median PFS of 6.9 months [5.4; 8.1]. In a phase Ib trial, the combination of divarasib and cetuximab yielded a 62.5% response rate with a median PFS of 8.1 months [5.5; 12.3] (23) (eTable).
On the basis of these data, the randomized phase III CodeBreaK 300 trial was set up to compare the combination of sotorasib and panitumumab with standard treatment [either trifluridine/tipiracil (oral chemotherapy) or regorafenib (a multitarget tyrosine kinase inhibitor)] in patients with previously treated metastatic CRC who had a KRASG12C mutation (2 4).
In this trial, sotorasib was given in two different doses (960 mg and 240 mg). 160 patients were included, and the trial achieved its primary endpoint (PFS). A significant prolongation of median PFS was achieved with sotorasib (960 mg) and panitumumab (5.6 months; [4.2; 6.3]) compared to standard treatment with either trifluridine/tipiracil or regorafenib (2.2 months; [1.9; 3.9]), with a higher response rate (26% versus 0%) (Table) (24). The rate of higher-grade ADR was 35.8% in the combination arm with sotorasib and panitumumab (mainly skin side effects, hypomagnesemia and diarrhea), and 43.1% in the standard treatment arm (mainly hematotoxicity, nausea and diarrhea). It was shown with standardized questionnaires that patients who received sotorasib and panitumumab had a better quality of life as well (25). These data led to FDA approval of the combination of sotorasib and panitumumab in the USA. The potential utility of KRASG12C inhibitors in earlier lines of therapy is now being investigated. In the ongoing randomized phase III KRYSTAL-10 trial, adagrasib combined with cetuximab is being compared with standard FOLFIRI (irinotecan, folinic acid and 5-fluorouracil) or FOLFOX (oxaliplatin, folinic acid and 5-fluorouracil) chemotherapy, each optionally in combination with a VEGF antibody, in patients with metastatic CRC who had progressive disease after first-line chemotherapy. Sotorasib together with panitumumab in combination with the FOLFIRI protocol in first-line treatment is now being tested in the randomized phase III CodeBreaK 301 study against the standard FOLFIRI chemotherapy in combination with bevacizumab-awwb. Safety data and initial efficacy data for this type of combination therapy are already available for patients with chemotherapy-refractory disease (eTable) (26).
Clinical data on KRASG12C inhibitors in pancreatic cancer and other gastrointestinal tumors
KRAS mutations are present in 85–90% of pancreatic carcinomas; the specific KRASG12C mutation is present in 1–2%. The KRASG12C mutation is also rarely present in other, less common gastrointestinal tumors, such as biliary tract carcinoma (BTC), appendiceal carcinoma, or small bowel carcinoma. Early screening is nevertheless worthwhile because of the availability of specific KRASG12C inhibitors and the often limited therapeutic options for these rare diseases. Patients with PDAC or other types of tumor were also included in the early clinical trials of sotorasib, adagrasib, and divarasib (eTable). In patients with previously treated pancreatic cancer, the response rates were 21% for sotorasib and 33% for adagrasib (27, 28). No entity-specific response rates have been reported for divarasib to date; the objective response rate (ORR) in the Basket study was 36%, but many of these patients had pancreatic or biliary tract cancer (19). Adagrasib yielded a 41.7% response rate, with a progression-free survival of 8.6 [2.7; 11.3] months and an overall survival of 15.1 [8.6; not reached] months, in patients with previously treated biliary tract cancer.
Development of resistance to KRASG12C inhibitors
Primary and acquired resistance are common problems with targeted therapies. For example, resistance-conferring genetic alterations have been identified by sequential analysis of tumor tissue or circulating DNA (cfDNA) for all three KRASG12C inhibitors mentioned above, either as monotherapy or in combination with EGFR antibodies (23, 29, 30). In addition to other acquired KRAS or NRAS mutations, genetic alterations in downstream signaling pathways and in receptor tyrosine kinases such as EGFR or fibroblast growth factor receptor (FGFR) were also frequently detected. In order to overcome these resistance mechanisms, various drug combinations are now under investigation in phase I trials.
Clinical data on pan-RAS inhibitors
The pan-RAS inhibitor RMC-6236 blocks KRAS, NRAS, and HRAS in their active conformation and is thus what is called a RAS-on inhibitor. It has been tested in a phase I trial in various tumor diseases with evidence of a KRAS mutation (RMC-6236–001/NCT05379985) (9, 31). The response rate was 38% in patients with previously treated NSCLC (N = 40) and 20% in patients with pancreatic cancer (N = 46).
Overview
The pharmacological inhibition of KRAS mutants is a new therapeutic approach for solid tumors. Because of the special conformation of this protein molecule, suitable inhibitors block it effectively could not be developed for many years. Recently, however, it has been possible to synthesize specific inhibitors for certain subtypes, such as the KRASG12C mutation, and test them clinically. Some of these inhibitors have already been approved and have rapidly become a part of routine oncological practice. As with many other targeted therapies, acquired resistance mediated by new genetic changes is a problem, and combination therapies are now being studied as a way to overcome it.
Conflict of interest statement
SK states that he and his institution have received financial support from Amgen, Roche, Lilly, GSK, MerckHealthcare, Novartis, J&J, Daiihi-Sankyo, BMS, MSD, and AstraZeneca. He has also served as a paid consultant for Amgen, BMS, MSD, Roche, AstraZeneca, Oncowissen.de, Daiichi-Sankyo, Takeda, Lilly, Beigene, and Novartis and has received payment from these companies for continuing medical education events. He has received reimbursement of scientific meeting participation fees from Amgen, BMS, MSD, Takeda, and Beigene. He is a member of the trial steering committee at Amgen and the CRC task force of the German Cancer Society’s Medical Oncology Working Group (Arbeitsgemeinschaft Internistische Onkologie, AIO).
MS states that his institution has received funding from AstraZeneca. He has personally served as a paid consultant for, and has received payment for continuing medical education events and the like from, the following companies: AstraZeneca, Sanofi, Merck, Novartis, Boehringer-Ingelheim, Pfizer, Takeda, Pierre-Fabre, Daiichi-Sankyo, Regeneron, and Gilead. He has received reimbursement of scientific meeting participation fees and/or travel expenses from Takeda, Merck, and Pfizer.
Submitted on 7 August 2024, revised version accepted on 6 January 2025
Translated from the original German by Ethan Taub, M.D.
Corresponding author
Prof. Dr. med. Stefan Kasper
stefan.kasper-virchow@uk-essen.de
Information on CME
This article has been certified by the North Rhine Academy for Continuing Medical Education. The questions on this article may be found at http://daebl.de/RY95. The closing date for entries is 20 March 2026. Participation is possible at cme.aerzteblatt.de
Department of Medicine III, Hematology/Oncology, Rheumatology, Infectious Diseases, HIV, University Hospital Frankfurt: Dr. med. Martin Sebastian
1. | Cook JH, Melloni GEM, Gulhan DC, Park PJ, Haigis KM: The origins and genetic interactions of KRAS mutations are allele- and tissue-specific. Nat Commun 2021; 12: 1808 CrossRef MEDLINE PubMed Central |
2. | Biernacka A, Tsongalis PD, Peterson JD, et al.: The potential utility of re-mining results of somatic mutation testing: KRAS status in lung adenocarcinoma. Cancer Genet 2016; 209: 195–8 CrossRef MEDLINE PubMed Central |
3. | Wiesweg M, Kasper S, Worm K, et al.: Impact of RAS mutation subtype on clinical outcome—a cross-entity comparison of patients with advanced non-small cell lung cancer and colorectal cancer. Oncogene 2019; 38: 2953–66 CrossRef MEDLINE |
4. | Sebastian M, Eberhardt WEE, Hoffknecht P, et al.: KRAS G12C-mutated advanced non-small cell lung cancer: a real-world cohort from the German prospective, observational, nation-wide CRISP registry (AIO-TRK-0315). Lung Cancer 2021; 154: 51–61 CrossRef MEDLINE |
5. | Chida K, Kotani D, Masuishi T, 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 CrossRef MEDLINE PubMed Central |
6. | Cox AD, Fesik SW, Kimmelman AC, Luo J, Der CJ: Drugging the undruggable RAS: mission possible? Nat Rev Drug Discov 2014; 13: 828–51 CrossRef CrossRef MEDLINE PubMed Central |
7. | Schulze CJ, Seamon KJ, Zhao Y, et al.: Chemical remodeling of a cellular chaperone to target the active state of mutant KRAS. Science 2023; 381: 794–9. |
8. | Kim D, Herdeis L, Rudolph D, et al.: Pan-KRAS inhibitor disables oncogenic signalling and tumour growth. Nature 2023; 619: 160–6. |
9. | Jiang J, Jiang L, Maldonato BJ, et al.: Translational and therapeutic evaluation of RAS-GTP inhibition by RMC-6236 in RAS-driven cancers. Cancer Discov 2024; 14: 994–1017 CrossRef MEDLINE PubMed Central |
10. | Holderfield M, Lee BJ, Jiang J, et al.: Concurrent inhibition of oncogenic and wild-type RAS-GTP for cancer therapy. Nature 2024; 629: 919–26 CrossRef MEDLINE PubMed Central |
11. | Salem ME, El-Refai SM, Sha W, et al.: Landscape of KRAS(G12C), associated genomic alterations, and interrelation with immuno-oncology biomarkers in KRAS-mutated cancers. JCO Precis Oncol 2022; 6: e2100245 CrossRef MEDLINE PubMed Central |
12. | Canon J, Rex K, Saiki AY, et al.: The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 2019; 575: 217–23 CrossRef MEDLINE |
13. | Skoulidis F, Li BT, Dy GK, et al.: Sotorasib for lung cancers with KRAS p.G12C mutation. N Engl J Med 2021; 384: 2371–81 CrossRef MEDLINE PubMed Central |
14. | Dy GK, Govindan R, Velcheti V, et al.: Long-term outcomes and molecular correlates of sotorasib efficacy in patients with pretreated KRAS G12C-mutated non-small-cell lung cancer: 2-year analysis of CodeBreaK 100. J Clin Oncol 2023; 41: 3311–7 CrossRef MEDLINE PubMed Central |
15. | de Langen AJ, Johnson ML, Mazieres J, et al.: Sotorasib versus docetaxel for previously treated non-small-cell lung cancer with KRAS(G12C) mutation: a randomised, open-label, phase 3 trial. Lancet 2023; 401: 733–46 CrossRef MEDLINE |
16. | Stratmann JA, Althoff FC, Doebel P, et al.: Sotorasib in KRAS G12C-mutated non-small cell lung cancer: a multicenter real-world experience from the compassionate use program in Germany. Eur J Cancer 2024; 201: 113911 CrossRef MEDLINE |
17. | Mok TSK, Yao W, Duruisseaux M, et al.: KRYSTAL-12: phase 3 study of adagrasib versus docetaxel in patients with previously treated advanced/metastatic non-small cell lung cancer (NSCLC) harboring a KRASG12C mutation. J Clin Oncol 2024; 42: LBA8509. (Abstract ASCO 2024 CrossRef |
18. | Felip E, Mok TSK, Lu S, et al.: OA09.06 Health-related quality of life (HRQoL) outcomes with adagrasib vs docetaxel in KRASG12C-mutated advanced NSCLC in KRYSTAL-12. J Thorac Oncol 2024; 19: S30. (Abstract WCLC 2024 CrossRef |
19. | Sacher A, LoRusso P, Patel MR, et al.: Single-agent divarasib (GDC-6036) in solid tumors with a KRASG12C mutation. N Engl J Med 2023; 389: 710–21 CrossRef MEDLINE |
20. | Fakih MG, Kopetz S, Kuboki Y, et al.: Sotorasib for previously treated colorectal cancers with KRASG12C mutation (CodeBreaK100): a prespecified analysis of a single-arm, phase 2 trial. Lancet Oncol 2022; 23: 115–24 CrossRef MEDLINE |
21. | Yaeger R, Weiss J, Pelster MS, et al.: Adagrasib with or without cetuximab in colorectal cancer with mutated KRAS G12C. N Engl J Med 2023; 388: 44–54 CrossRef MEDLINE PubMed Central |
22. | Kuboki Y, Fakih M, Strickler J, et al.: Sotorasib with panitumumab in chemotherapy-refractory KRAS(G12C)-mutated colorectal cancer: a phase 1b trial. Nat Med 2024; 30: 265–70 CrossRef MEDLINE PubMed Central |
23. | Desai J, Alonso G, Kim SH, et al.: Divarasib plus cetuximab in KRAS G12C-positive colorectal cancer: a phase 1b trial. Nat Med 2024; 30: 271–8 CrossRef MEDLINE PubMed Central |
24. | Fakih MG, Salvatore L, Esaki T, et al.: Sotorasib plus panitumumab in refractory colorectal cancer with mutated KRAS G12C. N Engl J Med 2023; 389: 2125–39 CrossRef MEDLINE |
25. | Modest DP, Fakih M, Salvatore L, et al.: Health-related quality of life (HRQoL) in patients with metastatic colorectal cancer (mCRC) treated with sotorasib and panitumumab (pmab) versus trifluridine/tipiracil (T/T) or regorafenib (rego) in CodeBreaK 300. J Clin Oncol 2024; 42: 10. (Abstract ASCO-GI 2024 CrossRef |
26. | Hong DS, Kuboki Y, Strickler JH, et al.: Sotorasib (Soto) plus panitumumab (Pmab) and FOLFIRI for previously treated KRAS G12C-mutated metastatic colorectal cancer (mCRC): CodeBreaK 101 phase 1b safety and efficacy. J Clin Oncol 2023; 41: 3513. (Abstract ASCO 2024) |
27. | Strickler JH, Satake H, George TJ, et al.: Sotorasib in KRAS p.G12C-mutated advanced pancreatic cancer. N Engl J Med 2023; 388: 33–43 CrossRef MEDLINE PubMed Central |
28. | Bekaii-Saab TS, Yaeger R, Spira AI, et al.: Adagrasib in advanced solid tumors harboring a KRASG12C mutation. J Clin Oncol 2023; 41: 4097–106 CrossRef MEDLINE PubMed Central |
29. | Awad MM, Liu S, Rybkin II, et al.: Acquired resistance to KRASG12C inhibition in cancer. N Engl J Med 2021; 384: 2382–93 CrossRef MEDLINE PubMed Central |
30. | Hong DS, Kuboki Y, Yaeger R, et al.: Abstract 2308: biomarkers of acquired resistance to sotorasib (soto) plus panitumumab (pani) in chemorefractory KRAS G12C-mutated metastatic colorectal cancer (mCRC). Cancer Research 2023; 83: 2308 CrossRef |
31. | Arbour KC, Punekar S, Garrido-Laguna I, et al.: 652O preliminary clinical activity of RMC-6236, a first-in-class, RAS-selective, tri-complex RAS-MULTI(ON) inhibitor in patients with KRAS mutant pancreatic ductal adenocarcinoma (PDAC) and non-small cell lung cancer (NSCLC). Ann Oncol 2023; 34: S458. (Abstract ESMO 2023 CrossRef |