KRAS is the most frequently mutated oncogene, harboring mutations in approximately one in seven cancers. Allele-specific KRASG12C inhibitors are currently changing the treatment paradigm for patients with KRASG12C-mutated non–small cell lung cancer and colorectal cancer. The success of addressing a previously elusive KRAS allele has fueled drug discovery efforts for all KRAS mutants. Pan-KRAS drugs have the potential to address broad patient populations, including KRASG12D-, KRASG12V-, KRASG13D-, KRASG12R-, and KRASG12A-mutant or KRAS wild-type–amplified cancers, as well as cancers with acquired resistance to KRASG12C inhibitors. Here, we review actively pursued allele-specific and pan-KRAS inhibition strategies and their potential utility.

Significance:

Mutant-selective KRASG12C inhibitors target a fraction (approximately 13.6%) of all KRAS-driven cancers. A broad arsenal of KRAS drugs is needed to comprehensively conquer KRAS-driven cancers. Conceptually, we foresee two future classes of KRAS medicines: mutant-selective KRAS drugs targeting individual variant alleles and pan-KRAS therapeutics targeting a broad range of KRAS alterations.

In 2020, an estimated 19.3 million new cancer cases occurred worldwide, including 1.8 million new cases in the United States alone (1, 2). Recent analyses have found that approximately one in seven of all human cancers harbor KRAS (Kirsten rat sarcoma virus) alterations, making it one of the top oncogenic drivers of human cancer (3–5). The KRAS protein is a small membrane-bound GTPase (GTP hydrolase), acting as a switch for a multitude of cellular sig-naling functions (Fig. 1A). The balance between nucleotide hydrolysis and exchange determines the levels of active KRAS in cells. Bound to GDP, KRAS is in an “OFF” state. Upon GDP to GTP exchange, usually in response to growth factors and facilitated by guanine-nucleotide exchange factors (GEF) such as SOS1/SOS2, KRAS cycles to its activated “ON” state. In this form, KRAS activates effector pathways, including the MAPK and PI3K pathways, to promote cellular proliferation and survival. KRAS returns to the OFF state when GTP is hydrolyzed to GDP, a process that is catalyzed by GTPase activating proteins (GAP) such as NF1 (6). In its oncogenic form, KRAS remains predominantly in the active ON state as GTP hydrolysis, by its intrinsic GTPase function and enzymes, such as GAPs, is impaired (7, 8). Previous assumptions of the constitutive activity of KRAS oncoproteins were reevaluated after evidence illustrating that KRASG12C is not permanently GTP bound and maintains a dependency on upstream receptor tyrosine kinase (RTK) signaling for GEF-mediated GTP reloading (9, 10). Recent studies using SHP2 and SOS1 inhibitors in KRAS-driven cancer cell lines, as well as biochemical studies of KRAS mutants in otherwise RAS-less mouse embryo fibroblasts, have shown that a range of KRAS oncoproteins cycle between their active and inactive states and remain dependent on nucleotide exchange for activation (11–13).

Figure 1.

Overview of the RAS/MAPK signaling pathway and patient numbers/overall cohort prevalence for distinct KRAS alleles/amplification in seven cancer types. A, Schematic representation of KRAS cycling and signaling highlighting selected drug targets and inhibitors. B, Distribution of KRAS alleles/amplification and patient numbers in selected tumor types. Mutation and amplification rates for KRAS have been derived from the AACR GENIE 9.0 public database, whereas patient numbers for the respective tumor types have been extracted from the Cancer Facts & Figures 2000 report published by the American Cancer Society (2). The number of cases for lung adenocarcinoma was set to 40% of all lung cancers. In total, 81,996 distinct samples with mutation and copy number profiles were collapsed into unique patient samples and filtered for distinct alleles and amplification of KRAS. The top seven alleles/amplifications with the highest overall prevalence across tumor types are shown, whereas other mutations are grouped into the class “Other.” The grouping “Multiple” contains all cases, for which different KRAS alterations have been observed in a single patient, for example, two different mutations or a mutation coupled with a KRAS amplification. The “Total” subpanel summarizes the patient numbers for the seven cancer types depicted and ranks the alterations based on overall numbers. Similarly, patient numbers are highlighted for each tumor type and each alteration. The percentages in parentheses reflect the proportion in relation to the full cohort (e.g., 13.6% of all patients with lung adenocarcinoma carry a KRASG12C mutation). AMP, amplification; CRC, colorectal cancer; EAC/GEJC, esophageal adenocarcinoma/gastroesophageal junction cancer; IDC, invasive ductal carcinoma; LUAD, lung adenocarcinoma; PDAC, pancreatic ductal adenocarcinoma; STAD, stomach adenocarcinoma; UEC, undifferentiated endometrial carcinoma.

Figure 1.

Overview of the RAS/MAPK signaling pathway and patient numbers/overall cohort prevalence for distinct KRAS alleles/amplification in seven cancer types. A, Schematic representation of KRAS cycling and signaling highlighting selected drug targets and inhibitors. B, Distribution of KRAS alleles/amplification and patient numbers in selected tumor types. Mutation and amplification rates for KRAS have been derived from the AACR GENIE 9.0 public database, whereas patient numbers for the respective tumor types have been extracted from the Cancer Facts & Figures 2000 report published by the American Cancer Society (2). The number of cases for lung adenocarcinoma was set to 40% of all lung cancers. In total, 81,996 distinct samples with mutation and copy number profiles were collapsed into unique patient samples and filtered for distinct alleles and amplification of KRAS. The top seven alleles/amplifications with the highest overall prevalence across tumor types are shown, whereas other mutations are grouped into the class “Other.” The grouping “Multiple” contains all cases, for which different KRAS alterations have been observed in a single patient, for example, two different mutations or a mutation coupled with a KRAS amplification. The “Total” subpanel summarizes the patient numbers for the seven cancer types depicted and ranks the alterations based on overall numbers. Similarly, patient numbers are highlighted for each tumor type and each alteration. The percentages in parentheses reflect the proportion in relation to the full cohort (e.g., 13.6% of all patients with lung adenocarcinoma carry a KRASG12C mutation). AMP, amplification; CRC, colorectal cancer; EAC/GEJC, esophageal adenocarcinoma/gastroesophageal junction cancer; IDC, invasive ductal carcinoma; LUAD, lung adenocarcinoma; PDAC, pancreatic ductal adenocarcinoma; STAD, stomach adenocarcinoma; UEC, undifferentiated endometrial carcinoma.

Close modal

Targeting KRAS in cancer has been a central goal during the past four decades, and research and development efforts have intensified over the past 10 years, largely sparked by the seminal discovery by J. Ostrem, K. Shokat and colleagues (14) of compounds tethered to the cysteine of KRASG12C. The recent accelerated approval of the KRASG12C mutant-selective inhibitor sotorasib (AMG 510) for the treatment of patients with second-line KRASG12C mutation-positive non–small cell lung cancer (NSCLC) by the FDA on May 28, 2021, marks the first approved targeted therapy for tumors with any KRAS mutation (https://www.fda.gov/news-events/press-announcements/fda-approves-first-targeted-therapy-lung-cancer-mutation-previously-considered-resistant-drug). A second KRASG12C inhibitor, adagrasib (MRTX849), has recently received breakthrough therapy designation, and eight additional inhibitors have entered clinical studies. However, despite the success of KRASG12C mutant-selective inhibitors for G12C-driven NSCLCs, more than 85% of all KRAS-mutated cancers still lack effective therapies. The scope of this review is to highlight the unmet need for patients harboring KRAS mutations and the challenging goal of drugging all oncogenic KRAS variants across mutation and cancer types. This review also provides an update and outlook on the most promising therapeutic approaches toward generating pan-KRAS concepts aiming at bringing precision therapy options to a broad range of KRAS-driven cancers.

KRAS mutations and/or KRAS wild-type amplifications are frequently found in colorectal cancer (United States ∼45%, China ∼49% of cases), pancreatic cancer (United States ∼90%, China ∼87% of cases), and NSCLC (subtype adenocarcinoma: United States ∼35%, China ∼13% of cases; see Fig. 1B for US data), with some differences across ethnicities/countries, especially in lung cancer, as previously described (ref. 15; https://doi.org/10.21203/rs.3.rs-916644/v1). In seven selected KRAS-driven cancer types in the United States (Fig. 1B), around 166,000 newly diagnosed tumors harbor a KRAS driver mutation, around 9,000 carry a KRAS wild-type amplification, and 4,000 patients show multiple KRAS alterations. Based on the analysis of the American Association for Cancer Research (AACR) project GENIE (release 9.0-public; ref. 5) data, we estimate up to 210,000 new annual diagnoses/patients showing alterations in KRAS in the United States across all nonhematologic cancers, weighted by their respective baseline incidence rates. The KRAS allelic distribution varies across tumor types, as shown in Fig. 1B, with G12C mutated in 13.6% of all lung adenocarcinomas, whereas KRASG12D and KRASG12V are the two most common alleles in colorectal and pancreatic cancer. KRASG12D, KRASG12V, and KRASG12C also represent the top three most frequently mutated alleles overall, found in over 51,000, 39,000, and 19,000 new cases in the selected seven tumor types (Fig. 1B), respectively, and together amount to 109,000 annual new diagnoses. Notably, other KRAS alleles such as G12R are mainly restricted to pancreatic cancer, whereas colorectal cancer also shows a significant fraction of non-G12D/V/C KRAS mutant alleles. Although KRAS mutations represent the most predominant aberration in many cancer types, others, such as breast invasive ductal carcinomas, stomach adenocarcinoma, and esophageal and gastroesophageal junction cancer, show recurrent KRAS wild-type amplifications. A subset of patients also harbors multiple KRAS alterations, including co-occurring multiallelic variants, such as KRASG12C and KRASG13D, as well as co-occurring KRAS mutations and amplifications.

The presence of KRAS mutations, especially the most common mutations at codon 12, is strongly associated with cellular KRAS dependency, as shown by functional genomic approaches across a large panel of human cell lines from different cancer types (16), suggesting KRAS mutations act as bona fide oncogenic driver events. It is apparent that mutations at other sites in KRAS, including codon 12, 13, 59, 61, 117, and 146 mutations, plus some atypical variants not currently included in standard guidelines in KRAS testing, also enhance its oncogenic potential (17).

To characterize the medical need and opportunity for drugging all KRAS variant oncoproteins, we compared the number of KRAS-mutant cancer cases with the patient population that can be matched with FDA-approved drugs (approved before April 1, 2021), based on genomic tumor mutations. Our calculation of prevalence of fully actionable mutations, based on AACR project GENIE (release 9.0-public), suggests that a total of 256,000 (14.1%) of patients with newly diagnosed cancer per year in the United States are eligible for treatment by FDA-approved genome-driven therapies (ref. 18; Supplementary Table S1). The top five patient populations in terms of size include cancers with alterations in PIK3CA, BRCA1/2, BRAF, ERBB2, and FGFR1/2/3 (Fig. 2). A similar number of 14.6% of US patients are eligible for level 1 drugs based on the OncoKB precision oncology knowledge base (19). These data suggest that the number of KRAS-mutant/amplified cancer cases is almost comparable to the number of all cancers that are currently actionable based on a genome-driven precision medicine approach.

Figure 2.

Percentage of patients who are eligible for FDA-approved precision medicine drugs out of all yearly 1.8 million new cancer cases in the United States. The top five target genes plus KRAS/KRASG12C with respect to patient cohort sizes and approved drugs are shown. Drugs against some of these target genes are approved in multiple indications (Supplementary Table S1). Of note, a small percentage of the KRAS segment is currently addressable by the recently approved KRASG12C inhibitor sotorasib (yellow); however, the larger portion of KRAS-driven cancer remains unserved (brown). The cohort size of patients benefiting from KRAS-targeting therapies has been derived from non-hematologic cancers mapping major cancer type cohort sizes with the fractions of KRAS-altered (mutated, amplified, multiple) patients in the respective types.

Figure 2.

Percentage of patients who are eligible for FDA-approved precision medicine drugs out of all yearly 1.8 million new cancer cases in the United States. The top five target genes plus KRAS/KRASG12C with respect to patient cohort sizes and approved drugs are shown. Drugs against some of these target genes are approved in multiple indications (Supplementary Table S1). Of note, a small percentage of the KRAS segment is currently addressable by the recently approved KRASG12C inhibitor sotorasib (yellow); however, the larger portion of KRAS-driven cancer remains unserved (brown). The cohort size of patients benefiting from KRAS-targeting therapies has been derived from non-hematologic cancers mapping major cancer type cohort sizes with the fractions of KRAS-altered (mutated, amplified, multiple) patients in the respective types.

Close modal

The development of compounds that covalently bind to cysteine 12 in GDP-KRASG12C, acting as inactive state-selective KRAS drugs, has rejuvenated interest in drugging this elusive key cancer driver (14, 20–22). In patients with advanced NSCLC harboring KRASG12C mutations, sotorasib (AMG 510) and adagrasib (MRTX849) have demonstrated robust efficacy with objective response rates of 37% and 45% and disease control rates of 81% and 96%, respectively (20, 23–25). Sotorasib has recently received FDA approval for the treatment of patients with advanced KRASG12C-mutant NSCLC following at least one prior systemic therapy, based on data from the phase I/II CodeBreaK 100 study (24). Importantly, initial monotherapy response rates for both drugs in colorectal cancer are lower than in NSCLC. In colorectal cancer, RTK dependency and signaling rebound kinetics have been identified as potential mechanisms underlying resistance to KRASG12C inhibition (23, 26, 27). These data set an initial benchmark for response rates and response duration in monotherapy across cancer types, and learnings might be transferable to other KRAS inhibitors targeting other mutant variants. Multiple ongoing trials seek to augment responses to KRASG12C inhibitors in NSCLC, colorectal cancer, and other cancer types through rational combination strategies (28, 29), as further detailed below. Here again, data from combination trials will be instructive for patient populations beyond KRASG12C-mutated cancers.

All current clinical KRAS inhibitors target the GDP-bound OFF form. Revolution Medicines has described macrocyclic molecules that bind to the active state ON form of KRASG12C by acting as a molecular glue with cyclophilin A, a highly abundant immunophilin. The assembled tricomplex prevents KRASG12C (ON) from signaling via steric blockade of RAS effector signaling. An advanced compound, RMC-6291, shows sustained pathway inhibition following RTK activation, consistent with targeting the active form of KRASG12C. This activity is associated with profound antitumor activity and evidence of superior activity to KRASG12C (OFF) inhibitors in KRASG12C-driven preclinical models (30).

The next research frontier will be to discover effective therapeutic opportunities for all KRAS mutants. The druggability advantage of covalently targeting the nucleophilic cysteine residue has thus far not materialized for other amino acids (31) and hence it is questionable whether this strategy can be successfully applied to KRAS-mutant oncoproteins beyond KRASG12C. Noncovalent inhibitors may be a more promising path forward. Indeed, recently cyclic peptide scaffolds have been reported to bind to KRASG12D and inhibit its interaction with CRAF at high nanomolar concentrations, albeit currently lacking cellular activity (32). Recent reports at scientific meetings by Mirati Therapeutics and Boehringer Ingelheim have highlighted KRASG12D mutant-selective inhibitors. MRTX1133 and BI-KRASG12D1–3 can reversibly bind to KRASG12D and demonstrate selective inhibition of cell viability of KRASG12D-mutant, but not KRAS wild-type, tumor cells (ref. 18; https://ir.mirati.com/press-releases/press-release-details/2020/Mirati-Therapeutics-Reports-Investigational-Adagrasib-MRTX849-Preliminary-Data-Demonstrating-Tolerability-and-Durable-Anti-Tumor-Activity-as-well-as-Initial-MRTX1133-Preclinical-Data/default.aspx). Importantly, MRTX1133, BI-KRASG12D2 and BI-KRASG12D3 were also reported to be active against in KRASG12D-driven xenograft models (https://ir.mirati.com/press-releases/press-release-details/2020/Mirati-Therapeutics-Reports-Investigational-Adagrasib-MRTX849-Preliminary-Data-Demonstrating-Tolerability-and-Durable-Anti-Tumor-Activity-as-well-as-Initial-MRTX1133-Preclinical-Data/default.aspx and https://ras-drugdevelopment.com/speaker/marco-hofmann-2/). KRASG12D-mutated cancers represent a significant unmet need, with more than 2.5-fold the annual patient numbers of KRASG12C-mutated cancers, thus providing a strong impetus to drive further research efforts toward this target profile. Efforts based on the tricomplex technology for targeting either KRASG12D or KRASG13C oncoproteins are ongoing (Revolution Medicines, Corporate Overview, Q1–2021, https://ir.revmed.com/static-files/5ee2ba3e-dcdf-4780-94a7-ef35fe3df9f2). The feasibility to move beyond KRASG12C, KRASG12D, and KRASG13C with mutant-selective inhibitors or degraders remains to be determined.

The global disease burden associated with KRAS mutations for different cancer types has spurred intense efforts to identify therapeutic concepts that can address a broad spectrum of KRAS-mutant cancers.

Several indirect pan-KRAS drugs are currently being developed by pharmaceutical companies that interfere with KRAS nucleotide exchange and activation by inhibiting SHP2 or the GEF SOS1 (Table 1). The rationale for pursuing SHP2 and SOS1 inhibitors as pan-KRAS inhibitors is grounded in the evidence that several (K)RAS oncoproteins, including KRASG12C, still cycle between an inactive and active state and rely on upstream activation and nucleotide exchange to exhibit their full transforming potential (9–13, 33, 34).

Table 1.

Mutant-specific KRAS inhibitors, pan-(K)RAS inhibitors, SHP2 inhibitors, and SOS1 inhibitors described in ClinicalTrials.gov or currently in advanced preclinical stages

Mutant-specific KRAS inhibitors, pan-(K)RAS inhibitors, SHP2 inhibitors, and SOS1 inhibitors described in ClinicalTrials.gov or currently in advanced preclinical stages
Mutant-specific KRAS inhibitors, pan-(K)RAS inhibitors, SHP2 inhibitors, and SOS1 inhibitors described in ClinicalTrials.gov or currently in advanced preclinical stages

SHP2 inhibitors stabilize the auto-inhibited conformation of the enzyme and thereby disrupt SOS1-mediated nucleotide exchange of KRAS (11, 35, 36). In addition, SHP2 inhibition may also yield immunomodulatory effects in T cells and macrophages to elicit antitumor immune responses (37). Nine SHP2 inhibitors are currently being tested in the clinics as single agents as well as in combination with other pathway inhibitors. Three SHP2 inhibitors, RMC-4630, TNO155, and JAB-3068, have progressed to phase II clinical trials. In a phase I/II study (NCT03634982) with RMC-4630 that included patients with tumors harboring RAS node alterations (KRASG12mut, KRASamp, NF1LOF, BRAFclass3), first clinical data demonstrated a disease control rate of 71% (5/7) with reduction in tumor volume reported in three (43%) and one confirmed objective response in patients with KRASG12C-mutant NSCLC (38). First clinical data in a phase I clinical study (NCT03114319) with the SHP2 inhibitor TNO155 showed sensitivity of some KRASG12-mutant tumors, especially KRASG12C-mutant NSCLC and BRAF/NRAS wild-type melanoma, but in the absence of partial responses (39).

Inhibitors of the GEF SOS1 block the interaction of SOS1 with KRAS-GDP, preventing nucleotide exchange and GTP loading of KRAS (12). BI 1701963 is currently the only SOS1 inhibitor being investigated in clinical trials. First preliminary clinical data from a dose escalation trial employing continuous dosing of BI 1701963, a pan-KRAS SOS1 inhibitor, as monotherapy (NCT04111458) have been reported. BI 1701963 was generally well tolerated, with the maximum tolerated dose reached at 800 mg, and stable disease up to 18 weeks in 7 of 31 patients with solid tumors harboring KRAS mutations was demonstrated (40). The primary objective of this trial is to determine the monotherapy tolerability of BI 1701963, in order to define the recommended phase II dose for combination trials (40). Revolution Medicines (RMC-5845), Schrödinger (SDGR5), Genhouse Bio (GH52), Erasca (ERAS-9), and Mirati Therapeutics announced SOS1 inhibitors in their preclinical pipelines (Table 1).

As upstream activators of KRAS, both SOS1 inhibitors and SHP2 inhibitors hold particular promise in the combination setting, and they are being explored preclinically and clinically in combination studies aimed to interrogate whether vertical pathway blockade can “tighten the grip” on the KRAS/MAPK pathway and thereby increase the rates and duration of responses. SHP2 inhibitors and BI 1701963 are being combined with MEK inhibitors [NCT04294160, NCT03989115 (41), NCT04720976, NCT04111458, and NCT048357] to enhance MAPK pathway modulation and to suppress pathway reactivation elicited by the alleviation of negative feedback control (12, 42). SHP2 inhibitors are also being combined with ERK inhibitors (NCT04916236) and EGFR tyrosine kinase inhibitors (NCT03989115 and NCT03114319). Although initial reports described tumor reduction in 3 of 8 patients with KRAS-mutant colorectal cancer in the RMC4630 and the MEK inhibitor cobimetinib combination trial (ref. 41; NCT03989115), Revolution Medicines has subsequently announced the termination of this combination trial due to lack of efficacy (Revolution Medicines, Financial Updates, Q2–2021, https://ir.revmed.com/news-releases/news-release-details/revolution-medicines-reports-second-quarter-financial-results). Ongoing SHP2/MEK inhibitor trials (Erasca, Jacobio Pharmaceuticals, and Novartis) and SOS1 inhibitor and MEK inhibitor combination trials (Boehringer Ingelheim) will provide more insights about the efficacy and tolerability of this combination in different patient populations.

As SOS1 inhibitors and SHP2 inhibitors shift the equilibrium of KRAS to the GDP-bound state, a strong rationale emerges to combine these indirect KRAS modulators with mutant-specific KRAS inhibitors, such as covalent KRASG12C inhibitors that bind KRAS in its GDP-bound state. Preclinical data have demonstrated synergistic antiproliferative effects and enhanced antitumor efficacy for both the SHP2 inhibitor/KRASG12C inhibitor combination (43) and the SOS1 inhibitor/KRASG12C inhibitor combination (44, 45). In heterozygous KRAS-mutant tumor cells, RAS signaling can be supplemented and cross-regulated by RTK-WT RAS signaling. A combination of a mutant-specific KRAS inhibitor with either a SHP2 inhibitor or a SOS1 inhibitor could effectively block mutant KRAS and wild-type RAS signaling (12, 43, 45, 46). Based on the strong underlying scientific rationale and supportive preclinical data, SHP2 inhibitor and SOS1 inhibitor combinations with allele-specific KRASG12C inhibitors are being tested in several ongoing clinical trials in patients with KRASG12C mutation-positive NSCLC and colorectal cancer (NCT04330664, NCT04185883, NCT04699188, NCT04973163, and NCT04975256). The allele-selective nature of KRASG12C inhibitors is anticipated to lead to improved tolerability for SHP2 and SOS1 inhibitor combinations compared with studies combining with MEK inhibitors.

Besides addressing KRAS-mutant tumors, the SHP2 inhibitor RMC-4630 is also being tested in a clinical study including patients with tumors carrying KRAS wild-type amplifications (NCT03989115). This positioning is supported by preclinical data demonstrating that targeting SOS1 or SHP2 can enhance the sensitivity of wild-type KRAS-amplified models to MEK inhibition (47). Based on their mode of action in suppressing nucleotide exchange, both SHP2 and SOS1 inhibitors are well poised to deliver therapeutic benefit in cancer settings that depend on RAS wild-type activity, including KRAS wild-type amplifications, BRAF class 3 mutations, and NF1 loss-of-function mutations.

Combinations of pan-KRAS concepts with approved standard-of-care therapies, such as chemotherapy, are not yet being extensively studied but could be impactful in case of synergistic activities. In preclinical experiments, a combination of the topoisomerase I inhibitor irinotecan with a SOS1 inhibitor enhances formation of DNA double-strand breaks in a KRAS-mutated tumor model (45, 48). The mechanistic basis for this effect has not been elucidated yet but could be associated with the impact of SOS1 inhibition on cell cycle progression and DNA repair pathway choice. The combination of SOS1 inhibition and irinotecan is now being studied in a phase I clinical trial recruiting patients with KRAS-mutant colorectal cancer in China (NCT04627142).

Besides its role in cancer cell intrinsic mitogenic signaling, SHP2 mediates signaling through the immune checkpoint receptor PD-1 in T cells (49). As the PD-1/SHP2 signaling axis mediates the immune-suppressive effect of PD-1, several clinical trials are enrolling patients studying the combination of SHP2 inhibitors with PD-1 blocking antibodies (NCT04000529, NCT04720976, NCT04721223, and NCT04418661).

Using KRAS fragment screening and structure-based drug design, Boehringer Ingelheim has recently reported the discovery of direct pan-KRAS inhibitors and direct pan-KRAS proteolysis targeting chimeras (PROTAC), which are able to spare NRAS and HRAS (18). In a panel of cell lines, pan-KRAS inhibitors are active against a broad range of KRAS-driven cell lines, including KRASG12C-, KRASG12D-, KRASG12V-, and KRASG13D-driven cells, whereas HRAS- and NRAS-mutated cell lines did not exhibit sensitivity (18). Importantly, pan-KRAS inhibitors show selective activity also on a panel of cell lines that harbor KRAS amplifications, whereas KRAS wild-type cells with normal copy number remain unaffected (18).

A pan-KRAS probe compound BI-panKRAS3 demonstrates dose-dependent inhibition of KRAS-dependent signaling and yields antitumor efficacy in KRASG12D- and KRASG13D-mutated colorectal cancer models (18).

An emerging new class of medicines are PROTACs, which specifically degrade proteins via the cellular protein degradation machinery (50). These bifunctional molecules simultaneously engage a protein of interest and an E3 ligase, forming a ternary complex, enabling the E3 ligase to ubiquitinate and subsequently induce degradation of the target protein (51). Covalent inhibitors that target KRASG12C in its GDP-bound OFF state have been successfully converted into PROTACs, but such covalent PROTACs are not catalytic, and it is unclear what advantages they possess beyond covalent KRASG12C inhibitors (52, 53). Although it may be technically more difficult to generate KRAS degraders based on the required physicochemical properties, degraders are in general more versatile in terms of target scope, as any functional binding site can be targeted. In principle, PROTACs also have the potential for a more rapid and sustained pathway blockade. In addition, by means of selecting a suitable E3-ligase, tissue selectivity can be built into the PROTAC design. Cellularly potent and selective KRAS PROTACs such as BI-KRASdegrader1 were reported, which can degrade all major cycling KRAS mutants while sparing NRAS and HRAS (18). Mutant alleles that are strongly GTP hydrolysis impaired, including G12R and Q61R/K/L mutants, are less potently degraded by KRAS PROTACs targeting the GDP-bound form. Importantly, the role SOS1 plays in feedback reactivation can be translated across the KRAS inhibitor space with evidence of combinatorial activity with mutant-selective as well as pan-KRAS compounds (18). An open question is which of the drug classes will be less prone to developing acquired resistance, as also PROTACs are likely susceptible to resistance mechanisms of their own (51).

Gaining a broad KRAS-mutant coverage also appears feasible by means of different approaches of generating pan-RAS inhibitors that block all three RAS isoforms KRAS, NRAS, and HRAS. The pan-RAS strategy adopted by Boehringer Ingelheim focuses on switch I/II pocket inhibitors exemplified by the compound BI-2852 (54). Revolution Medicines has applied a cyclophilin-dependent molecular glue approach to discover RMC-6236, which has been described as a potent, orally available tricomplex RASMULTI (ON) inhibitor. RMC-6236 displays activity across a diverse spectrum of RAS-driven cell lines, whereas no activity is reported in KRAS-independent cell lines that are driven by a BRAF mutation. Single-agent antitumor activity has been reported in a range of KRASG12D- and KRASG12V-driven colorectal cancer and pancreatic ductal adenocarcinoma (PDAC) tumor models (55). It remains to be established whether concepts targeting all three RAS isoforms simultaneously will be compatible with achieving a therapeutic window in patients.

Inhibitors targeting the MAPK pathway components downstream of RAS, such as RAF, MEK, and ERK, are not addressed in this review in detail but can serve as combination partners of RAS inhibitors given supportive tolerability. Guo and colleagues (56) reported a phase Ib study of the MEK inhibitor VS-6766 (previously known as CH5126766 or RO5126766), which showed promising antitumor activity across a spectrum of KRAS-mutant tumors, including KRASG12V mutation-positive NSCLC. VS-6766 is an allosteric MEK inhibitor that also blocks RAF phosphorylation of MEK, leading to a reduction of feedback-driven pathway rebound, thus potentially conferring enhanced therapeutic activity compared with other MEK inhibitors (57). VS-6766 is currently studied in combination with the FAK inhibitor defactinib in KRAS-mutated NSCLC, yielding promising early responses with partial responses being observed in 2 patients with NSCLC with KRASG12V-mutated cancers (58).

Conceptually, we expect the emergence of two major classes of KRAS medicines: KRAS drugs selectively targeting individual mutant variants and pan-KRAS concepts targeting a broad and diverse spectrum of KRAS alterations, covering mutations and amplification. Based on feasibility to generate small-molecule mutant-selective and pan-KRAS drugs, we could also see a range of spectrum-selective KRAS drugs, which balance the opportunities and risks associated with covering a narrower set of KRAS mutants.

The promise of mutant-selective KRAS drugs is manifold. Foremost, deep and durable target inhibition in a clearly defined patient population, a low risk of KRAS wild-type–mediated toxicity, and good combinability with other agents can be expected. Based on a wide therapeutic index, as seen for many of the current KRASG12C inhibitors, this drug class seems to be an obvious choice for early line settings in future clinical practice, alone or in combination with other targeted or immune-directed therapies. An open issue is clearly the feasibility of generating mutant-selective drugs beyond KRASG12C and perhaps KRASG12D. Importantly, there will be critical limitations of this approach in heterogeneous and drug-resistant tumors, as elaborated below. In contrast, pan-KRAS drugs and pan-RAS drugs face the still open issue of tolerability based on inhibition of wild-type (K)RAS. Data generated in the laboratory of Mariano Barbacid (CNIO, Spain), obtained in genetically modified murine models, indicate that ablation of the Kras locus in young mice does not result in immediate toxic effects but resulted in decreased viability starting at around 8 months of age (59). The effects on normal tissues of inhibiting or degrading wild-type KRAS in humans are yet to be interrogated. An important point in this context is the distinction between the likely toxicities associated with isoform-specific pan-KRAS inhibition compared with isoform-agnostic pan-RAS drugs that inhibit all three RAS isoforms, KRAS, HRAS, and NRAS. Barbacid's studies in mice indicate that in contrast to the late- and slow-onset effects obtained with genetic KRAS inactivation, the ablation of all RAS isoform expression causes death of adult mice within a few weeks of induction (60, 61). Therefore, it is highly likely that pan-RAS inhibitors will show a markedly higher level of toxicity than KRAS isoform-specific inhibitors. Pan-KRAS drugs have the potential to address a broad patient population; in particular, they could be positioned in early line settings where we lack mutant-selective KRAS drugs. Pan-KRAS inhibitors and degraders could also become the prime choice for cancers driven by wild-type KRAS, such as KRAS wild-type–amplified or cancers driven by loss of the NF1 tumor suppressor, as well as neurofibromatosis type 1 or its associated malignancies (34, 62, 63). Pan-KRAS drugs would have advantages versus mutant-selective KRAS drugs in heterogeneous cancers driven by multiple KRAS alterations at baseline or upon development of acquired resistance due to secondary KRAS on-target alterations (Fig. 3), with first examples emerging in the clinic (64–66). In the following section, these opportunities are further highlighted.

Figure 3.

Distribution of KRAS alterations across all KRAS-driven tumors with a focus on putative benefits (green text) and drawbacks (red text) for mutant-selective and pan-KRAS drugs. The left pie chart shows KRAS alleles that are currently addressable or worked on in non-transparent colors (G12D, G12C, G13C), whereas transparent colors visualize mutated KRAS alleles, which remain elusive to targeted therapy so far. Alleles are color-coded as in Fig. 1, with a long tail of other alleles shown in gray. In total, around 200 distinct KRAS alleles/alterations are reported in the AACR GENIE database. The right pie chart shows all KRAS alterations putatively targetable by pan-KRAS drugs. NCE, New Chemical Entity; WT, wild-type.

Figure 3.

Distribution of KRAS alterations across all KRAS-driven tumors with a focus on putative benefits (green text) and drawbacks (red text) for mutant-selective and pan-KRAS drugs. The left pie chart shows KRAS alleles that are currently addressable or worked on in non-transparent colors (G12D, G12C, G13C), whereas transparent colors visualize mutated KRAS alleles, which remain elusive to targeted therapy so far. Alleles are color-coded as in Fig. 1, with a long tail of other alleles shown in gray. In total, around 200 distinct KRAS alleles/alterations are reported in the AACR GENIE database. The right pie chart shows all KRAS alterations putatively targetable by pan-KRAS drugs. NCE, New Chemical Entity; WT, wild-type.

Close modal

NSCLC

KRAS is the most prevalent cancer driver in lung adenocarcinoma, with more than 35% of patients harboring a KRAS mutation. KRAS mutations other than KRASG12C represent more than half of all KRAS mutations in lung adenocarcinoma, with high prevalence of G12V, G12D, and G12A mutations. These mutant alleles all appear amenable to the pan-KRAS approaches described above, including pan-KRAS inhibitors and degraders, as well as SOS1 inhibitor/SHP2 inhibitor plus MEK inhibitor combinations. Similar to the KRASG12C mutation, these mutations also rarely overlap with other actionable driver mutations, such as EGFR mutations and ALK rearrangements (5, 67). Non-G12C KRAS-mutant NSCLC tumors are distinct from KRASG12C-mutated lung cancers in terms of comutation of tumor suppressor genes. Although the frequency of TP53 co-occurring mutations is comparable in non-G12C KRAS-mutated NSCLC (32%–35%), differences can be observed for the tumor suppressors STK11/LKB1 (comutation rates: G12C, 23%; G12D, 14%) and KEAP1 (comutation rates: G12C, 8%; G12D, 4%). Immune checkpoint inhibitors have been established as standard-of-care treatment for patients with NSCLC whose tumors express PD-L1 and lack EGFR mutations or ALK rearrangements, as a single agent or in combination with chemotherapy (68–70). However, response rates to single-agent immune checkpoint inhibitors overall are modest. Initial data suggest that a higher tumor mutational burden (TMB) occurs in the overall KRAS-mutant population, potentially resulting in improved response to immune checkpoint inhibition (71). This retrospective study showed that patients with NSCLC harboring a KRAS gene mutation accompanied by high expression levels of PD-L1 lived longer when they received immunotherapy alone, compared with patients without the mutation, whereas the survival difference was not seen in patients who received both chemotherapy and immunotherapy. Although these data are intriguing, validation in prospective randomized trials is pending (71). In KRAS-mutated NSCLC, the tumor microenvironment (TME) is frequently characterized by a lack or dysfunction of tumor-infiltrating lymphocytes (TIL), especially in the presence of co-occurring mutations in STK11/LKB1 (refs. 72, 73). Preclinical studies suggest that sotorasib and adagrasib can induce a more proinflammatory and TIL-infiltrated TME in mouse models, translating into durable complete responses in combination with anti–PD-1 therapy (21, 74). Exploratory correlative analyses from the KRYSTAL-1 and CodeBreaK 100 trials in this context suggest higher response rates for single-agent adagrasib (64% vs. 45%) and sotorasib (50% vs. 39%) among patients whose tumors harbored an STK11/LKB1 comutation versus the entire treatment cohort (24, 25). Although these early findings will need to be confirmed in larger combination trials with anti–PD-1 antibodies, they may offer an attractive approach for KRAS-targeted agents for this historically difficult-to-treat patient subgroup in a first-line setting (NCT04933695). Further studies are required to completely establish tumor mutational burden and immune checkpoint inhibitor sensitivity among non-G12C KRAS-mutant subtypes. Early data suggest that the presence of KRAS alleles other than G12C mutations (75), especially in conjunction with a TP53 comutation, may represent a negative predictive biomarkers for anti–PD-1/PD-L1 immune checkpoint inhibitors in patients with NSCLC (76). We expect the coming years in the field of KRAS-mutant NSCLC to be very insightful in particular because of the translation of learnings from the KRASG12C inhibitor field on combinations and comutations to non-G12C KRAS-mutated cancers.

Colorectal Cancer

KRAS mutations occur in approximately 45% of colorectal cancers, led in frequency by the G12D (13%), G12V (9%), and G13D (7%) alleles. Non-G12/13-type KRAS alleles occur with appreciable frequency at the A146 and Q61 position (3% and 2%, respectively). Besides its negative predictive role for response to anti-EGFR therapy, KRAS mutation status also has a negative prognostic impact in colorectal cancer and is associated with right-sidedness of tumor location (77). Emergence of KRAS amplification and acquisition of secondary KRAS mutations (G12C, G12D, G13D, and Q61H) was detected as one mechanism of acquired resistance in patients with metastatic colorectal cancer who relapsed on cetuximab or panitumumab treatment (78, 79). This finding suggests that a combination of an anti-EGFR therapy with a pan-KRAS inhibitor or a mutant-selective KRAS drug could be an effective second-line treatment to reverse KRAS-driven drug resistance. Learnings from the efficacy data obtained in colorectal cancer with sotorasib and adagrasib may be impactful also for non-G12C-type KRAS inhibitors. In KRASG12C-mutated colorectal cancer, sotorasib showed an initial objective response rate (ORR) of 7% (3 of 42 patients), whereas treatment with adagrasib yielded a preliminary response rate of 17% (3 of 18 patients; refs. 23, 26). This is reminiscent of the limited sensitivity of BRAFV600E mutant colorectal cancer to single-agent vemurafenib, as a selective BRAF inhibitor, in comparison to other tumor types and could be a result of a rapid adaptive response that occurs in colorectal cancer. Recent studies showed that, similar to BRAFV600E, KRASG12C-mutated colorectal cancers maintain sensitivity to upstream RTK signaling, particularly EGFR, and that EGFR reactivation restricts the efficacy of KRASG12C inhibition in colorectal cancer. The combination of KRASG12C and EGFR blockade overcomes this adaptive resistance, both in cell lines and in patient-derived xenograft models (27). Recent data suggest that the rationale to combine KRAS inhibitors with anti-EGFR agents in colorectal cancer translates into enhanced clinical efficacy. The combination of adagrasib plus cetuximab demonstrated a meaningful improvement over adagrasib monotherapy in a heavily pretreated patient population with KRASG12C-mutated colorectal cancer, resulting in a 43% ORR versus an ORR of 22% for the monotherapy (80). Data presented for the combination of sotorasib plus panitumumab versus sotorasib monotherapy in colorectal cancer point into a similar direction (81), although the data set for this combination is less mature. These data fuel future rational clinical combinations of KRAS-directed therapies in colorectal cancer, including a phase III study called KRYSTAL-10 combining adagrasib and cetuximab in second-line KRASG12C-mutated colorectal cancer (NCT04793958), and further emphasize the relevance of the underlying histologic and molecular context of each tumor type when developing KRAS blocking agents.

Other Gastrointestinal Cancers

KRAS alterations are the most frequently occurring somatic change in PDACs, with approximately 90% of cases. Learnings from KRASG12C inhibitors have been limited so far, with only case reports of responses, because KRASG12D and KRASG12V represent the majority of KRAS mutations occurring in about 65% of all PDAC cases, whereas G12C mutations are very rare with 1.1% (23, 26). Very recently, Mirati Therapeutics presented initial data on 10 KRASG12C mutant pancreatic cancer patients showing partial responses in 5 patients and a disease control rate of 100% (https://mirati.com/wp-content/uploads/ENA_Oct-2021_MRTX1133_vF.pdf). In addition to small molecule inhibitors, several strategies of KRAS inhibition have been investigated in PDAC, most notably using small interfering RNAs and inhibitory exosomes (82, 83), both in ongoing clinical trials. Emerging evidence suggests KRAS mutations constitute attractive targets for immune-based treatments in pancreatic cancer and beyond. One example is the mRNA vaccine mRNA-5671 that encodes neoepitopes for common KRAS mutations (G12C, G12D, G12V, and G13D; NCT03948763).

A second promising immunotherapeutic approach directed at KRAS mutations involves adoptive T-cell therapies and is extensively reviewed elsewhere (84). Recent preclinical studies have identified bispecific T-cell engager protein molecules that specifically bind to mutant RAS peptide neoepitopes (G12V, Q61H/L/R) complexed with HLA, inducing T-cell–mediated killing of target cancer cells expressing endogenous levels of the mutant RAS proteins and cognate HLA alleles (85). Although clinical translation of these immune-based treatments is in its early stage for epithelial cancers, a multipronged approach of cancer- and immune cell–directed therapies targeting multiple KRAS mutant alleles may become a reality in the foreseeable future.

Mutant-selective KRASG12D inhibitors as well as pan-KRAS concepts hold promise for affecting large segments of KRAS-driven PDAC. In addition to the initial promising data reported by Mirati on adagrasib in KRASG12C -mutated PDAC, a targeted approach against NRG1 fusion– and NTRK fusion–positive PDAC has yielded early response rates of roughly 40% (86) and 75% (87), respectively, hinting at the possibility that also targeting KRAS-driven PDAC could be broadly effective as a monotherapy.

Based on their distinct structural properties that impinge on signaling, particular attention needs to be given to special KRAS mutants, such as KRASG12R (the third most predominant alteration in PDAC), KRASA146T (especially in colorectal cancer), and KRASQ61H mutations (88–90). The implication of these unusual variants is that specific KRAS variants can have tissue-specific oncogenic effects and exhibit allele-specific therapeutic vulnerabilities in downstream effector pathways. Although A146T and Q61H mutations in KRAS remain sensitive to pan-KRAS inhibitors and degraders targeting the OFF form of KRAS, this appears not the case for the KRASG12R mutant, which could require KRAS ON drugs.

The role of KRAS, when activated through canonical mutations, has been well established, as summarized above. A secondary means of KRAS activation in cancer is the focal high-level amplification of the KRAS gene, which is the fourth most prevalent KRAS alteration in cancer comparable in frequency to around 60% of the KRASG12C-mutated patient population. KRAS wild-type amplifications have been prominently observed in esophageal (12%), stomach (5%), and breast invasive ductal carcinomas (2%; Fig. 1B). Importantly, KRAS, ERBB2, and FGFR2 amplifications are mutually exclusive in stomach cancer (91), with the latter two types of amplification subtypes already representing actionable alterations for precision medicine approaches (92, 93). The aggressive nature of these cancers, the paucity of current therapeutic strategies, and the fact that wild-type KRAS in an amplified state may reside preferentially in the GDP-bound, OFF form, with corresponding high susceptibility to inhibition by KRAS OFF drugs, render these tumors an attractive segment for developing pan-KRAS inhibitors and degraders.

Collectively, we see the potential to establish KRAS drugs as precision oncology approaches for a broad range of cancers and to ultimately replace chemotherapy as a standard of care in these KRAS-driven cancer types. As there is a lack of genetic overlap of KRAS aberrations with RTK-driven subtypes [e.g., in NSCLC (EGFR, ALK, MET, ROS1, RET, BRAF), colorectal cancer (BRAF), PDAC (NRG1), and gastric cancer (ERBB2, FGFR2)], KRAS-targeting drugs could be uniquely positioned in areas of particularly high unmet need.

Cancers Driven by Co-occurring KRAS Aberrations

Deep analysis of the AACR GENIE data revealed a subset of patients (approximately 2.8% of all KRAS-driven cancers) who harbor multiple KRAS alterations in a single biopsy specimen (Fig. 1B). These multiple alterations are less frequent in colorectal cancer and lung adenocarcinoma (about 1%) than in PDAC (2.1%), but given limited sequencing read coverage and putative low allelic frequencies, these numbers might be higher in patients. These heterogenous cancers carry either multiple forms of mutated KRAS or a mutated variant of KRAS in conjunction with a KRAS amplification. Allele-selective inhibitors would only target a portion of the tumor driven by a specific allele, whereas pan-KRAS inhibitors could be used to cover all distinct KRAS alterations, especially if these co-occur in the same tumor lesion.

Acquired Resistance to Mutant-Selective KRASG12C Inhibitors

Although KRASG12C inhibitors deliver clinical benefit, all patients who achieved an objective response ultimately progressed on treatment with sotorasib in a phase II study (24). Thus, acquired resistance limits the impact of KRASG12C inhibitors, a conclusion that is likely to apply to other KRAS inhibitors. Two recent reports provide much-awaited first insights into the mechanisms underlying acquired resistance to the KRASG12C inhibitors in the clinic (64, 65). The larger study interrogated genomic and histologic alterations using circulating free tumor DNA and/or tissue samples in 38 patients with disease progression on treatment with adagrasib after an initial clinical response (64). The analyzed cohort included 27 patients with NSCLC, 10 with colorectal cancer, and 1 with appendiceal cancer. One or more putative resistance mechanisms were identified in 17 patients (45%). Most of these patients (15 of 17) acquired molecular alterations leading to reactivation of RAS/MAPK signaling, albeit with the majority at low allelic frequencies while in most cases maintaining the KRASG12C-mutated allele. This striking convergence suggests that RAS/MAPK signaling restoration drives therapeutic resistance to KRASG12C inhibitors in the clinic and highlights the strong addiction of KRASG12C-mutated tumors to RAS/MAPK signaling. Identified putative resistance mechanisms include on-target and beyond-target alterations (Fig. 4) and fall into six different classes: i) KRAS mutations in the switch II pocket that block inhibitor binding (secondary mutations in KRAS residues R68, H95, or Y96), ii) activating KRAS mutations (G12D, G12R, G12V, G13D, or Q61H) likely in a trans allele configuration, iii) cis allele conversion of the KRASG12C codon mutation (KRASG12CKRASG12W), iv) KRASG12C amplification, v) KRASG12C bypass mutations in the RAS/MAPK/PI3K pathway (amplifications, mutations, or activating fusions of the genes encoding oncogenic RTKs MET, EGFR, RET, ALK, and FGFR3; activating mutations or fusions of the downstream effector kinases BRAF, CRAF, and MEK1; NRAS and PI3K mutations; and PTEN or NF1 tumor suppressor mutations), and vi) cell state transformation from adenocarcinoma to squamous cell carcinoma histology (64, 94, 95). Importantly, 9 of 17 patients (53%) with detected putative resistance mechanisms harbored a least one acquired KRAS alteration. Furthermore, 7 of 17 patients (41%) acquired more than one putative resistance mechanism, a finding that appeared more prevalent in patients with colorectal cancer than NSCLC.

Figure 4.

Putative acquired resistance mechanisms detected in KRASG12C inhibitor-resistant patients are shown in a schematic pathway diagram. The potential utility of direct and indirect pan-KRAS inhibitors in addressing resistance is indicated using dashed boxes.

Figure 4.

Putative acquired resistance mechanisms detected in KRASG12C inhibitor-resistant patients are shown in a schematic pathway diagram. The potential utility of direct and indirect pan-KRAS inhibitors in addressing resistance is indicated using dashed boxes.

Close modal

Differential preclinical profiling of sotorasib and adagrasib revealed specific secondary KRAS mutations that elicit resistance to one drug but maintain sensitivity to the other (64, 95); for example, KRASG12C harboring additional mutations in the position H95, such as H95D/Q/R, abrogated the activity of adagrasib but remained vulnerable to sotorasib. This suggests that the sequential use of KRASG12C inhibitors in patients, who are selected based on the appropriate KRAS resistance mutation, may be able to deliver clinical benefit. Alternatively, KRASG12C inhibitors with a different mode of action to adagrasib and sotorasib have the potential to address secondary KRAS resistance mutations in the switch II pocket that cause cross-resistance to off-state inhibitors. This has been shown preclinically with the tricomplex KRASG12C active state inhibitor RM-018 in cellular models expressing a KRASG12C Y96D double-mutant variant (65). In addition, KRASG12C active state inhibitors are expected to address adaptive changes of KRAS resulting in the production of new, active KRAS (10). Nevertheless, the acquisition of a diverse set of KRAS mutations in response to KRASG12C inhibitor treatment across the cohort, as well as the subclonal intratumor heterogeneity of resistance mechanisms within many patients, poses a challenge for the development of allele-specific next-generation KRASG12C inhibitors. In contrast, direct pan-KRAS inhibitors or degraders with potent activity against a set of activating KRAS mutations have the potential to address KRAS inhibitor resistance mechanisms more broadly (Fig. 4; ref. 95). Furthermore, direct pan-KRAS agents, especially those with potent activity against wild-type KRAS, might be able to suppress upstream bypass resistance events, such as activating RTK alterations and NF1 loss-of-function mutations. The efficacy of pan-KRAS inhibitors could be augmented by vertical pathway combination with indirect pan-KRAS inhibitors, such as SHP2 or SOS1 inhibitors, that reduce KRAS-GTP loading of different KRAS variants, address adaptive feedback mechanisms (e.g., mediated by diverse RTKs upstream), and potentially dampen RTK alteration-driven resistance (refs. 11, 12, 36, 42, 96; Fig. 4).

Preclinical studies with models carrying molecularly defined and clinically relevant resistance mechanisms will be crucial to identify and prioritize KRAS-targeted monotherapies and combinations for clinical exploration to treat and possibly prevent acquired resistance to KRAS inhibitors. We anticipate that larger clinical data sets characterizing acquired resistance to KRASG12C inhibitors in NSCLC and colorectal cancer will soon become available. This will help to define the most recurrent mechanisms of resistance and the alterations that are responsible for driving clinically manifested tumor progression. The latter point is pertinent given the low allele frequency of putative resistance alterations detected in circulating tumor DNA (64–66). Importantly, additional clinical data may be able to address what drives therapy resistance in more than half of the patients (21 of 38), who harbored no identifiable molecular or histologic alterations upon progression on adagrasib (64). Insights gleaned from the most advanced clinical KRASG12C inhibitors, sotorasib and adagrasib, regarding acquired resistance in patient tumors and how to address it therapeutically will likely be instructive for the next generation of KRAS inhibitors.

Given that pan-KRAS concepts will address extensive unmet needs, a significant effort is now being applied within the pharmaceutical industry to move beyond KRASG12C inhibitors and discover new therapeutics with the ultimate goal to target all KRAS mutants. Exciting progress has already been reported for KRASG12D mutant-selective inhibitors as well as pan-KRAS inhibitors and degraders. We see allele-specific and pan-KRAS drugs as highly complementary therapeutic concepts that can be positioned to comprehensively conquer all KRAS cancers. It remains to be seen whether it will be possible to develop additional mutant-specific inhibitors, such as KRASG12V inhibitors or pan-mutant KRAS inhibitors, that spare wild-type KRAS. We are still at the beginning of drugging KRAS, and KRASG12C inhibitors represent the first chapter of the saga on cracking KRAS. Based on the intense efforts and rapid progress in the field, we see the beginning of the first “beyond KRASG12C” chapter becoming a reality. However, we expect many more chapters need to be written before we have sufficient medicines against KRAS, the Everest of oncogenes (97), for patients with cancer driven by KRAS.

M.H. Hofmann reports grants from Austrian Research Promotion Agency (FFG) during the conduct of the study and is listed as inventor on several patent applications for SOS1 inhibitors and is a full-time employee of Boehringer Ingelheim Regional Center Vienna GmbH & Co KG. D. Gerlach reports grants from Austrian Research Promotion Agency (FFG) during the conduct of the study and is a full-time employee of Boehringer Ingelheim Regional Center Vienna GmbH & Co KG. S. Misale reports other support from Boehringer Ingelheim outside the submitted work. M. Petronczki reports grants from Austrian Research Promotion Agency (FFG) during the conduct of the study and is a full-time employee of Boehringer Ingelheim Regional Center Vienna GmbH & Co KG. N. Kraut reports grants from Austrian Research Promotion Agency (FFG) during the conduct of the study and is a full-time employee of Boehringer Ingelheim Regional Center Vienna GmbH & Co KG.

The authors acknowledge support from Waltraud Pasteiner (Boehringer Ingelheim), Marcelo Marotti (Boehringer Ingelheim), Darryl B. McConnell (Boehringer Ingelheim), Mark Pearson (Boehringer Ingelheim), Markus Johann Bauer (Boehringer Ingelheim), and Mariano Barbacid (Centro Nacional de Investigaciones Oncológicas). Editorial assistance, funded by Boehringer Ingelheim, was provided by Caroline Perry (Ashfield MedComms) and Tracy South (Ashfield MedComms).

Note: Supplementary data for this article are available at Cancer Discovery Online (http://cancerdiscovery.aacrjournals.org/).

1.
Sung
H
,
Ferlay
J
,
Siegel
RL
,
Laversanne
M
,
Soerjomataram
I
,
Jemal
A
et al
.
Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries
.
CA Cancer J Clin
2021
;
71
:
209
49
.
2.
Viale
PH
.
The American Cancer Society's facts & figures: 2020 edition
.
J Adv Pract Oncol
2020
;
11
:
135
6
.
3.
Prior
IA
,
Hood
FE
,
Hartley
JL
.
The frequency of Ras mutations in cancer
.
Cancer Res
2020
;
80
:
2969
74
.
4.
Cerami
E
,
Gao
J
,
Dogrusoz
U
,
Gross
BE
,
Sumer
SO
,
Aksoy
BA
et al
.
The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data
.
Cancer Discov
2012
;
2
:
401
4
.
5.
AACR Project GENIE Consortium
.
AACR Project GENIE: powering precision medicine through an international consortium
.
Cancer Discov
2017
;
7
:
818
31
.
6.
Ratner
N
,
Miller
SJ
.
A RASopathy gene commonly mutated in cancer: the neurofibromatosis type 1 tumour suppressor
.
Nat Rev Cancer
2015
;
15
:
290
301
.
7.
Kim
D
,
Xue
JY
,
Lito
P
.
Targeting KRAS(G12C): from inhibitory mechanism to modulation of antitumor effects in patients
.
Cell
2020
;
183
:
850
9
.
8.
Moore
AR
,
Rosenberg
SC
,
McCormick
F
,
Malek
S
.
RAS-targeted therapies: is the undruggable drugged?
Nat Rev Drug Discov
2020
;
19
:
533
52
.
9.
Lito
P
,
Solomon
M
,
Li
LS
,
Hansen
R
,
Rosen
N
.
Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism
.
Science
2016
;
351
:
604
8
.
10.
Xue
JY
,
Zhao
Y
,
Aronowitz
J
,
Mai
TT
,
Vides
A
,
Qeriqi
B
et al
.
Rapid non-uniform adaptation to conformation-specific KRAS(G12C) inhibition
.
Nature
2020
;
577
:
421
5
.
11.
Nichols
RJ
,
Haderk
F
,
Stahlhut
C
,
Schulze
CJ
,
Hemmati
G
,
Wildes
D
et al
.
RAS nucleotide cycling underlies the SHP2 phosphatase dependence of mutant BRAF-, NF1- and RAS-driven cancers
.
Nat Cell Biol
2018
;
20
:
1064
73
.
12.
Hofmann
MH
,
Gmachl
M
,
Ramharter
J
,
Savarese
F
,
Gerlach
D
,
Marszalek
JR
et al
.
BI-3406, a potent and selective SOS1-KRAS interaction inhibitor, is effective in KRAS-driven cancers through combined MEK inhibition
.
Cancer Discov
2021
;
11
:
142
57
.
13.
Zhao
Y
,
Xue
JY
,
Lito
P
.
Suppressing nucleotide exchange to inhibit KRAS-mutant tumors
.
Cancer Discov
2021
;
11
:
17
9
.
14.
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
.
15.
Dearden
S
,
Stevens
J
,
Wu
YL
,
Blowers
D
.
Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology (mutMap)
.
Ann Oncol
2013
;
24
:
2371
6
.
16.
McDonald
ER
III
,
de Weck
A
,
Schlabach
MR
,
Billy
E
,
Mavrakis
KJ
,
Hoffman
GR
et al
.
Project DRIVE: a compendium of cancer dependencies and synthetic lethal relationships uncovered by large-scale, deep RNAi screening
.
Cell
2017
;
170
:
577
92
.
17.
Loree
JM
,
Wang
Y
,
Syed
MA
,
Sorokin
AV
,
Coker
O
,
Xiu
J
et al
.
Clinical and functional characterization of atypical KRAS/NRAS mutations in metastatic colorectal cancer
.
Clin Cancer Res
2021
;
27
:
4587
98
.
18.
Kraut
N
.
Expanding the reach of precision oncology by drugging all KRAS mutants
. In:
Proceedings of the 112th Annual Meeting of the American Association for Cancer Research; 2021 May 17–21
.
Philadelphia (PA)
:
AACR
;
2021
.
19.
Suehnholz
SP
,
Kundra
R
,
Zhang
H
,
Smith
S
,
Nissan
M
,
Yao
Y
et al
.
Evolution of OncoKB, a precision oncology knowledgebase
.
Cancer Res
2021
;
81
:
abstr99
.
20.
Fell
JB
,
Fischer
JP
,
Baer
BR
,
Blake
JF
,
Bouhana
K
,
Briere
DM
et al
.
Identification of the clinical development candidate MRTX849, a covalent KRAS(G12C) inhibitor for the treatment of cancer
.
J Med Chem
2020
;
63
:
6679
93
.
21.
Canon
J
,
Rex
K
,
Saiki
AY
,
Mohr
C
,
Cooke
K
,
Bagal
D
et al
.
The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity
.
Nature
2019
;
575
:
217
23
.
22.
Lanman
BA
,
Allen
JR
,
Allen
JG
,
Amegadzie
AK
,
Ashton
KS
,
Booker
SK
et al
.
Discovery of a covalent inhibitor of KRAS(G12C) (AMG 510) for the treatment of solid tumors
.
J Med Chem
2020
;
63
:
52
65
.
23.
Hong
DS
,
Fakih
MG
,
Strickler
JH
,
Desai
J
,
Durm
GA
,
Shapiro
GI
et al
.
KRAS(G12C) inhibition with sotorasib in advanced solid tumors
.
N Engl J Med
2020
;
383
:
1207
17
.
24.
Skoulidis
F
,
Li
BT
,
Dy
GK
,
Price
TJ
,
Falchook
GS
,
Wolf
J
et al
.
Sotorasib for lung cancers with KRAS p.G12C mutation
.
N Engl J Med
2021
;
384
:
2371
81
.
25.
Jänne
PA
,
Rybkin
II
,
Spira
AI
,
Riely
GJ
,
Papadopoulos
KP
,
Sabari
JK
et al
.
3LBA KRYSTAL24 1: activity and safety of adagrasib (MRTX849) in advanced/metastatic non–small-cell lung cancer (NSCLC) harboring KRAS G12C mutation
.
Eur J Cancer
2020
;
138
:
S1
S2
.
26.
Johnson
ML
,
Ou
S-HI
,
Barve
M
,
Rybkin
II
,
Papadopoulos
KP
,
Leal
TA
et al
.
4LBA KRYSTAL-1: activity and safety of adagrasib (MRTX849) in patients with colorectal cancer (CRC) and other solid tumors harboring a KRAS G12C mutation
.
Eur J Cancer
2020
;
138
:
S2
.
27.
Amodio
V
,
Yaeger
R
,
Arcella
P
,
Cancelliere
C
,
Lamba
S
,
Lorenzato
A
et al
.
EGFR blockade reverts resistance to KRAS(G12C) inhibition in colorectal cancer
.
Cancer Discov
2020
;
10
:
1129
39
.
28.
Chakraborty
A
.
KRASG12C inhibitor: combing for combination
.
Biochem Soc Trans
2020
;
48
:
2691
701
.
29.
Merz
V
,
Gaule
M
,
Zecchetto
C
,
Cavaliere
A
,
Casalino
S
,
Pesoni
C
et al
.
Targeting KRAS: the elephant in the room of epithelial cancers
.
Front Oncol
2021
;
11
:
638360
.
30.
Nichols
RJ
,
Cregg
J
,
Schulze
CJ
,
Whang
Z
,
Yang
K
,
Whalen
DM
et al
.
A next generation tri-complex KRASG12C(ON) inhibitor directly targets the active, GTP-bound state of mutant RAS and may overcome resistance to KRASG12C(OFF) inhibition
.
Cancer Res
2021
;
81
:
abstr1261
.
31.
Mukherjee
H
,
Grimster
NP
.
Beyond cysteine: recent developments in the area of targeted covalent inhibition
.
Curr Opin Chem Biol
2018
;
44
:
30
8
.
32.
Zhang
Z
,
Gao
R
,
Hu
Q
,
Peacock
H
,
Peacock
DM
,
Dai
S
et al
.
GTP-state-selective cyclic peptide ligands of K-Ras(G12D) block its interaction with raf
.
ACS Cent Sci
2020
;
6
:
1753
61
.
33.
Valencia-Sama
I
,
Ladumor
Y
,
Kee
L
,
Adderley
T
,
Christopher
G
,
Robinson
CM
et al
.
NRAS status determines sensitivity to SHP2 inhibitor combination therapies targeting the RAS-MAPK pathway in neuroblastoma
.
Cancer Res
2020
;
80
:
3413
23
.
34.
Sheffels
E
,
Kortum
RL
.
The role of wild-type RAS in oncogenic RAS transformation
.
Genes
2021
;
12
:
662
.
35.
Garcia Fortanet
J
,
Chen
CH
,
Chen
YN
,
Chen
Z
,
Deng
Z
,
Firestone
B
et al
.
Allosteric inhibition of SHP2: identification of a potent, selective, and orally efficacious phosphatase inhibitor
.
J Med Chem
2016
;
59
:
7773
82
.
36.
Chen
YN
,
LaMarche
MJ
,
Chan
HM
,
Fekkes
P
,
Garcia-Fortanet
J
,
Acker
MG
et al
.
Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases
.
Nature
2016
;
535
:
148
52
.
37.
Kerr
DL
,
Haderk
F
,
Bivona
TG
.
Allosteric SHP2 inhibitors in cancer: targeting the intersection of RAS, resistance, and the immune microenvironment
.
Curr Opin Chem Biol
2021
;
62
:
1
12
.
38.
Ou
SI
,
Koczywas
M
,
Ulahannan
S
,
Janne
P
,
Pacheco
J
,
Burris
H
et al
.
The SHP2 inhibitor RMC-4630 in patients with KRAS-mutant non-small cell lung cancer: preliminary evaluation of a first-in-man phase 1 clinical trial
.
J Thorac Oncol
2020
;
15
:
abstrA12
.
39.
Brana
I
,
Shapiro
G
,
Johnson
ML
,
Yu
HA
,
Robbrecht
D
,
Tan
DS-W
et al
.
Initial results from a dose finding study of TNO155, a SHP2 inhibitor, in adults with advanced solid tumors
.
J Clin Oncol
2021
;
39
:
abstr3005
.
40.
Johnson
M
,
Gort
E
,
Pant
S
,
Lolkema
M
,
Sebastian
M
,
Scheffler
M
et al
.
A phase I, open-label, dose-escalation trial of BI 1701963 (SOS1::KRAS inhibitor) in patients with KRAS mutated solid tumours: a snapshot analysis
.
Poster 542. Presented at the European Society for Medical Oncology (ESMO), Virtual Format, September 16–21
,
2021
.
41.
Bendell
J
,
Ulahannan
S
,
Koczywas
M
,
Brahmer
J
,
Capasso
A
,
Eckhardt
SG
et al
.
Intermittent dosing of RMC-4630, a potent, selective inhibitor of SHP2, combined with the MEK inhibitor cobimetinib, in a phase 1b/2 clinical trial for advanced solid tumors with activating mutations of RAS signaling
.
Eur J Cancer
2020
;
138
:
S8
S9
.
42.
Lu
H
,
Liu
C
,
Velazquez
R
,
Wang
H
,
Dunkl
LM
,
Kazic-Legueux
M
et al
.
SHP2 inhibition overcomes RTK-mediated pathway reactivation in KRAS-mutant tumors treated with MEK inhibitors
.
Mol Cancer Ther
2019
;
18
:
1323
34
.
43.
Liu
C
,
Lu
H
,
Wang
H
,
Loo
A
,
Zhang
X
,
Yang
G
et al
.
Combinations with allosteric SHP2 inhibitor TNO155 to block receptor tyrosine kinase signaling
.
Clin Cancer Res
2021
;
27
:
342
54
.
44.
Theard
PL
,
Sheffels
E
,
Sealover
NE
,
Linke
AJ
,
Pratico
DJ
,
Kortum
RL
.
Marked synergy by vertical inhibition of EGFR signaling in NSCLC spheroids shows SOS1 is a therapeutic target in EGFR-mutated cancer
.
eLife
2020
;
9
:
e58204
.
45.
Hofmann
MH
,
Lu
H
,
Duenzinger
U
,
Gerlach
D
,
Trapani
F
,
Machado
AA
et al
.
Trial in process: phase 1 studies of BI 1701963, a SOS1::KRAS Inhibitor, in combination with MEK inhibitors, irreversible KRASG12C inhibitors or irinotecan
.
Cancer Res
2021
;
81
:
abstrCT210
.
46.
Hayes
TK
,
Der
CJ
.
Mutant and wild-type Ras: co-conspirators in cancer
.
Cancer Discov
2013
;
3
:
24
6
.
47.
Wong
GS
,
Zhou
J
,
Liu
JB
,
Wu
Z
,
Xu
X
,
Li
T
et al
.
Targeting wild-type KRAS-amplified gastroesophageal cancer through combined MEK and SHP2 inhibition
.
Nat Med
2018
;
24
:
968
77
.
48.
Bhonde
MR
,
Hanski
ML
,
Notter
M
,
Gillissen
BF
,
Daniel
PT
,
Zeitz
M
et al
.
Equivalent effect of DNA damage-induced apoptotic cell death or long-term cell cycle arrest on colon carcinoma cell proliferation and tumour growth
.
Oncogene
2006
;
25
:
165
75
.
49.
Wang
Y
,
Mohseni
M
,
Grauel
A
,
Diez
JE
,
Guan
W
,
Liang
S
et al
.
SHP2 blockade enhances anti-tumor immunity via tumor cell intrinsic and extrinsic mechanisms
.
Sci Rep
2021
;
11
:
1399
.
50.
Qi
SM
,
Dong
J
,
Xu
ZY
,
Cheng
XD
,
Zhang
WD
,
Qin
JJ
.
PROTAC: an effective targeted protein degradation strategy for cancer therapy
.
Front Pharmacol
2021
;
12
:
692574
.
51.
Farnaby
W
,
Koegl
M
,
McConnell
DB
,
Ciulli
A
.
Transforming targeted cancer therapy with PROTACs: a forward-looking perspective
.
Curr Opin Pharmacol
2021
;
57
:
175
83
.
52.
Bond
MJ
,
Chu
L
,
Nalawansha
DA
,
Li
K
,
Crews
CM
.
Targeted degradation of oncogenic KRAS(G12C) by VHL-recruiting PROTACs
.
ACS Cent Sci
2020
;
6
:
1367
75
.
53.
Kargbo
R
.
PROTAC-mediated degradation of KRAS protein for anticancer therapeutics
.
ACS Med Chem Lett
2020
;
11
:
5
6
.
54.
Kessler
D
,
Gmachl
M
,
Mantoulidis
A
,
Martin
LJ
,
Zoephel
A
,
Mayer
M
et al
.
Drugging an undruggable pocket on KRAS
.
Proc Natl Acad Sci U S A
2019
;
116
:
15823
9
.
55.
Koltun
E
,
Cregg
J
,
Rice
MA
,
Whalen
DM
,
Freilich
R
,
Jiang
J
et al
.
First-in-class, orally bioavailable KRASG12V(ON) tri-complex inhibitors, as single agents and in combinations, drive profound anti-tumor activity in preclinical models of KRASG12V mutant cancers
.
Cancer Res
2021
;
81
:
abstr1260
.
56.
Guo
C
,
Chenard-Poirier
M
,
Roda
D
,
de Miguel
M
,
Harris
SJ
,
Candilejo
IM
et al
.
Intermittent schedules of the oral RAF-MEK inhibitor CH5126766/VS-6766 in patients with RAS/RAF-mutant solid tumours and multiple myeloma: a single-centre, open-label, phase 1 dose-escalation and basket dose-expansion study
.
Lancet Oncol
2020
;
21
:
1478
88
.
57.
Ishii
N
,
Harada
N
,
Joseph
EW
,
Ohara
K
,
Miura
T
,
Sakamoto
H
et al
.
Enhanced inhibition of ERK signaling by a novel allosteric MEK inhibitor, CH5126766, that suppresses feedback reactivation of RAF activity
.
Cancer Res
2013
;
73
:
4050
60
.
58.
Krebs
MG
,
Shinde
R
,
Rahman
RA
,
Grochot
R
,
Little
M
,
King
J
et al
.
A phase I trial of the combination of the dual RAF-MEK inhibitor VS-6766 and the FAK inhibitor defactinib: evaluation of efficacy in KRAS mutated NSCLC
.
Cancer Res
2021
;
81
:
abstrCT019
.
59.
Scala
AD
.
Generation and characterization of triple knockout mice for H, N and K-ras genes [dissertation
].
Madrid (Spain)
:
University of Madrid
;
2008
.
60.
Drosten
M
,
Dhawahir
A
,
Sum
EY
,
Urosevic
J
,
Lechuga
CG
,
Esteban
LM
et al
.
Genetic analysis of Ras signalling pathways in cell proliferation, migration and survival
.
EMBO J
2010
;
29
:
1091
104
.
61.
Drosten
M
,
Lechuga
CG
,
Barbacid
M
.
Ras signaling is essential for skin development
.
Oncogene
2014
;
33
:
2857
65
.
62.
Ferner
RE
.
Neurofibromatosis 1 and neurofibromatosis 2: a twenty first century perspective
.
Lancet Neurol
2007
;
6
:
340
51
.
63.
Gross
AM
,
Dombi
E
,
Widemann
BC
.
Current status of MEK inhibitors in the treatment of plexiform neurofibromas
.
Childs Nerv Syst
2020
;
36
:
2443
52
.
64.
Awad
MM
,
Liu
S
,
Rybkin
II
,
Arbour
KC
,
Dilly
J
,
Zhu
VW
et al
.
Acquired resistance to KRAS(G12C) inhibition in cancer
.
N Engl J Med
2021
;
384
:
2382
93
.
65.
Tanaka
N
,
Lin
JJ
,
Li
C
,
Ryan
MB
,
Zhang
J
,
Kiedrowski
LA
et al
.
Clinical acquired resistance to KRAS(G12C) inhibition through a novel KRAS switch-II pocket mutation and polyclonal alterations converging on RAS-MAPK reactivation
.
Cancer Discov
2021
;
11
:
1913
22
.
66.
Zhao
Y
,
Murciano-Goroff
YR
,
Xue
JY
,
Ang
A
,
Lucas
J
,
Mai
TT
et al
.
Diverse alterations associated with resistance to KRAS(G12C) inhibition
.
Nature
2021
;
599
:
679
83
.
67.
Aggarwal
S
,
Whipple
S
,
Hsu
H
,
Tu
H
,
Carrigan
G
,
Wang
X
et al
.
Clinicopathological characteristics and treatment patterns observed in real-world care in patients with advanced non-small cell lung cancer (NSCLC) and KRAS G12C mutations in the Flatiron Health (FH)-Foundation Medicine (FMI) Clinico-Genomic Database (CGDB)
.
Ann Oncol
2020
;
31
:
S754
840
.
68.
Gandhi
L
,
Rodriguez-Abreu
D
,
Gadgeel
S
,
Esteban
E
,
Felip
E
,
De Angelis
F
et al
.
Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer
.
N Engl J Med
2018
;
378
:
2078
92
.
69.
Reck
M
,
Rodriguez-Abreu
D
,
Robinson
AG
,
Hui
R
,
Csoszi
T
,
Fulop
A
et al
.
Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer
.
N Engl J Med
2016
;
375
:
1823
33
.
70.
Socinski
MA
,
Jotte
RM
,
Cappuzzo
F
,
Orlandi
F
,
Stroyakovskiy
D
,
Nogami
N
et al
.
Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC
.
N Engl J Med
2018
;
378
:
2288
301
.
71.
Sun
L
,
Hsu
M
,
Cohen
RB
,
Langer
CJ
,
Mamtani
R
,
Aggarwal
C
.
Association between KRAS variant status and outcomes with first-line immune checkpoint inhibitor-based therapy in patients with advanced non-small-cell lung cancer
.
JAMA Oncol
2021
;
7
:
937
9
.
72.
Skoulidis
F
,
Byers
LA
,
Diao
L
,
Papadimitrakopoulou
VA
,
Tong
P
,
Izzo
J
et al
.
Co-occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities
.
Cancer Discov
2015
;
5
:
860
77
.
73.
Skoulidis
F
,
Goldberg
ME
,
Greenawalt
DM
,
Hellmann
MD
,
Awad
MM
,
Gainor
JF
et al
.
STK11/LKB1 mutations and PD-1 inhibitor resis-tance in KRAS-mutant lung adenocarcinoma
.
Cancer Discov
2018
;
8
:
822
35
.
74.
Briere
DM
,
Li
S
,
Calinisan
A
,
Sudhakar
N
,
Aranda
R
,
Hargis
L
et al
.
The KRAS(G12C) inhibitor MRTX849 reconditions the tumor immune microenvironment and sensitizes tumors to checkpoint inhibitor therapy
.
Mol Cancer Ther
2021
;
20
:
975
85
.
75.
Frost
N
,
Kollmeier
J
,
Vollbrecht
C
,
Grah
C
,
Matthes
B
,
Pultermann
D
et al
.
KRAS(G12C)/TP53 co-mutations identify long-term responders to first line palliative treatment with pembrolizumab monotherapy in PD-L1 high (≥50%) lung adenocarcinoma
.
Transl Lung Cancer Res
2021
;
10
:
737
52
.
76.
Gao
G
,
Liao
W
,
Ma
Q
,
Zhang
B
,
Chen
Y
,
Wang
Y
.
KRAS G12D mutation predicts lower TMB and drives immune suppression in lung adenocarcinoma
.
Lung Cancer
2020
;
149
:
41
5
.
77.
Giopanou
I
,
Pintzas
A
.
RAS and BRAF in the foreground for non-small cell lung cancer and colorectal cancer: similarities and main differences for prognosis and therapies
.
Crit Rev Oncol Hematol
2020
;
146
:
102859
.
78.
Misale
S
,
Yaeger
R
,
Hobor
S
,
Scala
E
,
Janakiraman
M
,
Liska
D
et al
.
Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer
.
Nature
2012
;
486
:
532
6
.
79.
Misale
S
,
Arena
S
,
Lamba
S
,
Siravegna
G
,
Lallo
A
,
Hobor
S
et al
.
Blockade of EGFR and MEK intercepts heterogeneous mechanisms of acquired resistance to anti-EGFR therapies in colorectal cancer
.
Sci Transl Med
2014
;
6
:
224ra26
.
80.
Weiss
J
,
Yaeger
RD
,
Johnson
ML
,
Spira
A
,
Klempner
SJ
,
Barve
MA
et al
.
KRYSTAL-1: adagrasib (MRTX849) as monotherapy or combined with cetuximab (Cetux) in patients (Pts) with colorectal cancer (CRC) harboring a KRASG12C mutation
.
Ann Oncol
2021
;
32
:
abstrLBA6
.
81.
Fakih
M
,
Falchook
GS
,
Hong
DS
,
Yaeger
RD
,
Chan
E
,
Mather
O
et al
.
CodeBreaK 101 subprotocol H: Phase Ib study evaluating combination of sotorasib (Soto), a KRASG12C inhibitor, and panitumumab (PMab), an EGFR inhibitor, in advanced KRAS p.G12C-mutated colorectal cancer (CRC)
.
Ann Oncol
2021
;
32
:
abstr434P
.
82.
Golan
T
,
Khvalevsky
EZ
,
Hubert
A
,
Gabai
RM
,
Hen
N
,
Segal
A
et al
.
RNAi therapy targeting KRAS in combination with chemotherapy for locally advanced pancreatic cancer patients
.
Oncotarget
2015
;
6
:
24560
70
.
83.
Kamerkar
S
,
LeBleu
VS
,
Sugimoto
H
,
Yang
S
,
Ruivo
CF
,
Melo
SA
et al
.
Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer
.
Nature
2017
;
546
:
498
503
.
84.
Chatani
PD
,
Yang
JC
.
Mutated RAS: targeting the “untargetable” with T cells
.
Clin Cancer Res
2020
;
26
:
537
44
.
85.
Douglass
J
,
Hsiue
EH
,
Mog
BJ
,
Hwang
MS
,
DiNapoli
SR
,
Pearlman
AH
et al
.
Bispecific antibodies targeting mutant RAS neoantigens
.
Sci Immunol
2021
;
6
:
eabd5515
.
86.
Schram
AM
,
O'Reilly
EM
,
O'Kane
GM
,
Goto
K
,
Kim
D-W
,
Neuzillet
C
et al
.
Efficacy and safety of zenocutuzumab in advanced pancreas cancer and other solid tumors harboring NRG1 fusions
.
J Clin Oncol
2021
;
39
:
abstr3003
.
87.
Bazhenova
L
,
Liu
SV
,
Lin
JJ
,
Lu
S
,
Drilon
A
,
Chawla
SP
et al
.
Efficacy and safety of entrectinib in patients with locally advanced/metastatic NTRK fusion-positive (NTRK-fp) solid tumours
.
Ann Oncol
2021
;
32
:
abstr533P
.
88.
Hobbs
GA
,
Baker
NM
,
Miermont
AM
,
Thurman
RD
,
Pierobon
M
,
Tran
TH
et al
.
Atypical KRAS(G12R) mutant is impaired in PI3K signaling and macropinocytosis in pancreatic cancer
.
Cancer Discov
2020
;
10
:
104
23
.
89.
Zhou
ZW
,
Ambrogio
C
,
Bera
AK
,
Li
Q
,
Li
XX
,
Li
L
et al
.
KRAS(Q61H) preferentially signals through MAPK in a RAF dimer-dependent manner in non-small cell lung cancer
.
Cancer Res
2020
;
80
:
3719
31
.
90.
Poulin
EJ
,
Bera
AK
,
Lu
J
,
Lin
YJ
,
Strasser
SD
,
Paulo
JA
et al
.
Tissue-specific oncogenic activity of KRAS(A146T)
.
Cancer Discov
2019
;
9
:
738
55
.
91.
Deng
N
,
Goh
LK
,
Wang
H
,
Das
K
,
Tao
J
,
Tan
IB
et al
.
A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets
.
Gut
2012
;
61
:
673
84
.
92.
Shitara
K
,
Bang
YJ
,
Iwasa
S
,
Sugimoto
N
,
Ryu
MH
,
Sakai
D
et al
.
Trastuzumab deruxtecan in previously treated HER2-positive gastric cancer
.
N Engl J Med
2020
;
382
:
2419
30
.
93.
Wainberg
ZA
,
Enzinger
PC
,
Kang
Y-K
,
Yamaguchi
K
,
Qin
S
,
Lee
K-W
et al
.
Randomized double-blind placebo-controlled phase 2 study of bemarituzumab combined with modified FOLFOX6 (mFOLFOX6) in first-line (1L) treatment of advanced gastric/gastroesophageal junction adenocarcinoma (FIGHT)
.
J Clin Oncol
2021
;
39
:
abstr160
.
94.
Adachi
Y
,
Ito
K
,
Hayashi
Y
,
Kimura
R
,
Tan
TZ
,
Yamaguchi
R
et al
.
Epithelial-to-mesenchymal transition is a cause of both intrinsic and acquired resistance to KRAS G12C inhibitor in KRAS G12C-mutant non-small cell lung cancer
.
Clin Cancer Res
2020
;
26
:
5962
73
.
95.
Koga
T
,
Suda
K
,
Fujino
T
,
Ohara
S
,
Hamada
A
,
Nishino
M
et al
.
KRAS secondary mutations that confer acquired resistance to KRAS G12C inhibitors, sotorasib and adagrasib, and overcoming strategies: insights from in vitro experiments
.
J Thorac Oncol
2021
;
16
:
1321
32
.
96.
Ryan
MB
,
Fece de la Cruz
F
,
Phat
S
,
Myers
DT
,
Wong
E
,
Shahzade
HA
et al
.
Vertical pathway inhibition overcomes adaptive feedback resistance to KRAS(G12C) inhibition
.
Clin Cancer Res
2020
;
26
:
1633
43
.
97.
Russo
M
,
Di Nicolantonio
F
,
Bardelli
A
.
Climbing RAS, the Everest of oncogenes
.
Cancer Discov
2014
;
4
:
19
21
.
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.

Supplementary data