KRAS G12C Inhibitors Breaking the 40-Year Drug Development Barrier
Check out our extensive video library (see channel for our latest videos)
Introduction
KRAS G12C inhibitors represent a historic milestone in the field of oncology, marking the first successful attempt to therapeutically target a protein once considered “undruggable.” For more than four decades, KRAS remained one of the most challenging oncogenic drivers to inhibit pharmacologically despite being one of the most frequently mutated genes in human cancers. Mutations in KRAS are implicated in approximately 96 percent of pancreatic ductal adenocarcinomas, 52 percent of colorectal cancers, and 32 percent of lung carcinomas. This high prevalence underscores its critical role in tumor initiation and progression, as well as the long-standing unmet need for effective KRAS-directed therapies.
The KRAS G12C mutation is characterized by a cysteine substitution at codon 12, resulting in an altered protein conformation that enables continuous activation of downstream signaling pathways, including MAPK and PI3K-AKT. This mutation occurs in approximately 12 to 14 percent of non-small cell lung cancers (NSCLCs) and 3 to 4 percent of colorectal cancers (CRCs). Notably, it is strongly associated with tobacco exposure, being detected in 85 percent of current or former smokers compared to 56 percent of non-smokers. Among KRAS mutations, the G12C variant represents the fourth most common substitution and is most frequently identified in NSCLC, where it accounts for nearly half of all KRAS G12 substitutions.
For decades, efforts to directly target KRAS proved unsuccessful due to its high affinity for GTP/GDP and the absence of suitable binding pockets for small molecules. The breakthrough came with the discovery of a unique allosteric pocket that could be exploited to covalently bind the cysteine residue introduced by the G12C mutation. This discovery paved the way for the development of covalent inhibitors that irreversibly lock KRAS G12C in its inactive GDP-bound state, thereby disrupting downstream oncogenic signaling and inhibiting tumor growth.
A landmark moment occurred in May 2021 when the United States Food and Drug Administration (FDA) granted accelerated approval to sotorasib (Lumakras™), the first KRAS G12C inhibitor approved for clinical use. Sotorasib was authorized for the treatment of adults with KRAS G12C-mutated NSCLC who had received at least one prior systemic therapy. Clinical data from the CodeBreaK 100 trial demonstrated a confirmed objective response rate of 37.1 percent, with a median duration of response of 11.1 months. These results established proof of concept for direct KRAS inhibition and signaled a paradigm shift in the management of KRAS-driven malignancies.
Following this breakthrough, research in KRAS-targeted therapy has advanced rapidly. By August 2021, at least 20 clinical trials were registered, investigating nine different direct KRAS G12C inhibitors, including adagrasib, JNJ-74699157, and GDC-6036, among others. Early studies suggest that while monotherapy with KRAS G12C inhibitors achieves meaningful tumor regression, resistance frequently develops through multiple mechanisms, including secondary KRAS mutations, feedback activation of receptor tyrosine kinases, and adaptive signaling through parallel pathways. These challenges have prompted exploration of rational combination strategies, pairing KRAS inhibitors with agents targeting EGFR, SHP2, or MEK to enhance efficacy and delay resistance.
This review explores the scientific journey leading to the clinical success of KRAS G12C inhibitors, detailing the structural and biological underpinnings of the mutation, the mechanism of action of current therapeutic agents, and the evolving understanding of resistance mechanisms. It also examines ongoing clinical trials and emerging therapeutic approaches that aim to extend the benefits of KRAS-targeted therapy to a broader range of patients.
The development of KRAS G12C inhibitors not only represents a triumph of modern drug discovery but also exemplifies the power of structure-based design and translational research in addressing previously insurmountable oncologic targets. As ongoing studies continue to refine combination strategies and investigate resistance pathways, the future of KRAS-targeted therapy holds promise for improving survival outcomes in patients with KRAS-driven cancers across multiple tumor types.
Why KRAS Was Considered Undruggable for 40 Years
For nearly four decades, KRAS remained elusive to pharmaceutical intervention, earning its reputation as the “Holy Grail of drug discovery” [1]. The formidable challenges in developing direct KRAS inhibitors stemmed from multiple structural and biochemical barriers that frustrated researchers across academia and industry.
Lack of allosteric binding pockets in KRAS structure
The molecular architecture of KRAS presents fundamental obstacles for drug development. Unlike many druggable proteins, KRAS possesses a relatively smooth surface with no obvious deep pockets beyond its nucleotide-binding site [2]. The protein structure consists of a globular GTPase domain (residues 1-166) and a hypervariable region (167-188) [3], arranged into six beta strands and five alpha helices forming two major domains: the G domain and the hypervariable region [1].
The absence of well-defined, hydrophobic pockets viable for small molecule binding made KRAS poorly suited for traditional drug discovery approaches [2]. Moreover, the switch I/II pocket (P1 site) on KRAS is approximately seven times smaller in volume compared to the druggable acetyl lysine-binding pocket on bromodomain-containing protein BRD4 [3]. This minimal binding surface represents a daunting challenge for medicinal chemistry, as optimizing ligands for such a small, shallow, and polar pocket requires innovative approaches beyond conventional drug design paradigms.
Consequently, despite tens of thousands of scientific publications and more than 300 published structures of KRAS [4], the protein remained resistant to direct therapeutic intervention until recently. The lack of comprehensive allosteric maps further hampered progress, as such information could greatly accelerate drug development for proteins functioning via difficult-to-inhibit protein-protein interaction interfaces [4].
High GTP affinity and intracellular concentration
A second major obstacle involves KRAS’s biochemistry and its relationship with guanine nucleotides. Under physiological conditions, KRAS binds GTP with extraordinarily high affinity in the picomolar range [2]. This exceptional binding strength, coupled with relatively high cellular GTP concentrations (approximately 500 μM) [2], created an insurmountable hurdle for competitive inhibition strategies.
In normal cellular function, KRAS acts as a molecular switch, cycling between active (GTP-bound) and inactive (GDP-bound) conformations [1]. However, oncogenic KRAS mutations at positions G12 and Q61 impair GTP hydrolysis, resulting in persistently active GTP-bound KRAS and enhanced downstream signaling [3]. Additionally, these mutations lower the protein’s affinity for GTPase-activating proteins (GAPs), further reducing GTPase activity [3].
The design of competitive inhibitors capable of displacing GTP from the binding pocket remained elusive due to these biochemical properties [3]. Essentially, any potential competitive inhibitor would need to achieve adequate intracellular concentrations to effectively compete with the abundant GTP while matching or exceeding KRAS’s affinity for its natural ligand—requirements that conventional small molecules simply could not meet.
Failed attempts with farnesyltransferase inhibitors
Early therapeutic efforts focused on indirect approaches to target KRAS, with farnesyltransferase inhibitors (FTIs) representing one of the first comprehensive attempts [5]. These compounds aimed to block the post-translational lipid modification necessary for KRAS membrane localization, thereby preventing its signaling activity.
Initially, FTIs showed promise in preclinical experiments, demonstrating good pharmacological profiles and tolerability [6]. However, they ultimately failed to achieve clinically relevant antitumor effects in unselected patient populations [6]. The fundamental reason for this disappointment was subsequently identified: unlike HRAS, both KRAS and NRAS undergo alternative prenylation by geranylgeranyltransferase-I (GGTase-I) when farnesyltransferase is blocked [5].
This alternative lipid modification pathway provided KRAS with a cellular bypass mechanism that allowed for proper processing, membrane localization, and continued signaling despite FTI treatment [6]. Furthermore, KRAS was found to bind farnesyltransferase more strongly than HRAS, necessitating higher drug concentrations to effectively block KRAS farnesylation [6].
The failure of FTIs cemented KRAS’s reputation as undruggable and temporarily dampened enthusiasm for direct targeting approaches [5]. Nevertheless, recent analysis reveals that certain KRAS mutants, particularly G12C, may actually exhibit enhanced sensitivity to FTIs, suggesting potential for combination therapy strategies with modern KRAS G12C inhibitors [6].
The KRAS G12C Mutation: A Unique Therapeutic Opportunity 
Among the landscape of oncogenic mutations, KRAS G12C emerged as a breakthrough target after decades of futile attempts to inhibit KRAS directly. This specific mutation possesses unique properties that finally unlocked pharmaceutical intervention possibilities where other KRAS variants remained resistant to targeted approaches.
What is KRAS G12C and how it differs from other KRAS mutations
The KRAS G12C mutation involves a single-nucleotide variation causing glycine-to-cysteine substitution at codon 12 of the KRAS protein [5]. This alteration creates a nucleophilic cysteine residue that can form covalent bonds with specifically designed inhibitors [4]. In contrast to other common KRAS mutations like G12D and G12V, KRAS G12C exhibits a distinctive biochemical profile that allows for selective targeting.
G12C mutations display unique vulnerability because, unlike other KRAS mutations, they maintain an active cycle between GDP-bound and GTP-bound states [4]. This cycling behavior presents a critical therapeutic window absent in other variants. KRAS G12C shows strong association with tobacco exposure, appearing primarily in current or former smokers (83.8% vs. 59.5% for non-G12C mutations) [3]. The mutation burden is likewise elevated, with patients harboring G12C mutations demonstrating markedly higher tobacco use (median 36.42 pack-years vs. 21.46 pack-years) [3].
Prevalence in NSCLC, CRC, and PDAC
KRAS G12C exhibits varying prevalence across cancer types:
- NSCLC: G12C represents the most common KRAS mutation in lung adenocarcinoma, accounting for 40-46% of all KRAS-mutant NSCLC [3][5]. Overall, approximately 13-16% of lung adenocarcinoma patients harbor this specific mutation [5][7].
- Colorectal cancer: The mutation appears in 3.2-4% of colorectal cancers, representing roughly 7-9% of KRAS-mutated cases in this cancer type [8][8].
- Pancreatic cancer: Although KRAS mutations are extremely common in pancreatic ductal adenocarcinoma (PDAC), G12C variants are relatively rare at approximately 1.3% [8][4].
KRAS G12C mutations frequently co-occur with other genomic alterations, most commonly TP53 (25%), STK11 (10.2%), and ATM (10.2%) [3]. These mutated tumors show elevated tumor mutational burden (TMB), with 32% exhibiting ≥10 mutations/Mb compared to 12% for non-G12C KRAS mutations [1]. G12C-mutated tumors also demonstrate higher programmed death-ligand 1 (PD-L1) expression (37% vs. 14% for other KRAS mutations) [1], a finding with implications for immunotherapy response.
GDP-bound state as a druggable conformation
The revolutionary breakthrough enabling KRAS G12C inhibition came with the discovery of an allosteric binding site behind the switch-II region, termed the switch-II pocket (S-IIP) [5]. This pocket, accessible only in GDP-bound KRAS G12C but not in other KRAS mutations, offered the first viable avenue for direct KRAS targeting [4].
The switch-II pocket creates a unique opportunity because:
- It exists exclusively in the GDP-bound (inactive) state of KRAS G12C [5]
- It allows for covalent binding to the reactive cysteine at position 12 [4]
- It permits the design of inhibitors that trap KRAS G12C in its inactive conformation [4]
In 2013, Ostrem and colleagues identified this critical allosteric site, catalyzing the development of covalent inhibitors that irreversibly target KRAS G12C [7]. These compounds operate through a unique mechanism – they bind to the GDP-bound form of KRAS G12C, locking it in an inactive conformation and preventing its activation [2]. Accordingly, this approach effectively halts downstream signaling through MAPK and PI3K pathways that normally promote cell proliferation and survival [2].
The reactive thiol group in cysteine provides an ideal nucleophilic target for electrophilic compounds, allowing for prolonged target engagement [4]. This mechanism differs fundamentally from traditional competitive inhibition approaches that previously failed against KRAS, making G12C the first clinically actionable KRAS mutation after four decades of research.
Discovery of the Switch-II Pocket and First Inhibitors
The groundbreaking discovery of the switch-II pocket (S-IIP) in KRAS G12C marked the pivotal moment that transformed the once “undruggable” protein into a viable therapeutic target. In 2013, Shokat and colleagues identified this cryptic allosteric site behind the switch-II region of KRAS, providing the first opportunity to develop direct KRAS inhibitors after decades of failed attempts.
Covalent binding mechanism in ARS-853 and ARS-1620
The first-generation KRAS G12C inhibitors exploited the nucleophilic properties of the mutant cysteine through a unique covalent binding mechanism. ARS-853, developed from the initial compound 12, demonstrated the first selective inhibition of KRAS in cells with potency in the drug candidate range [6]. The activity of ARS-853 and its successor ARS-1620 is primarily driven by KRAS-mediated catalysis of the chemical reaction with Cys12 in KRASG12C, rather than by high-affinity reversible binding [6]. Indeed, their reversible binding affinity remains weak—in the hundreds of micromolar range—yet they achieve remarkable potency through the covalent mechanism.
ARS-853 reduced KRAS-GTP levels by over 95% at 10 μM concentration and inhibited the proliferation of lung cancer H358 cells with an IC50 of 2.5 μM [6]. Nevertheless, poor metabolic stability in plasma (half-life <20 minutes) and minimal oral bioavailability in mice (F <2%) limited its clinical potential [6].
ARS-1620, a quinazoline-based second-generation inhibitor, markedly improved upon ARS-853’s foundation. This compound exhibited a 10-fold increase in potency for covalently modifying KRAS G12C with an observed rate of 1,100 ± 200 M^-1s^-1 [6]. Remarkably, the R-conformational atropisomer of ARS-1620 showed nearly 1,000-fold less potency (1.2 ± 0.6 M^-1s^-1), highlighting the extraordinary structural precision required for effective inhibition [6].
Structural targeting of cysteine-12 in GDP-bound KRAS
The structural basis for these inhibitors’ efficacy lies in their selective targeting of KRAS G12C in its GDP-bound state. Both ARS-853 and ARS-1620 bind irreversibly in the S-IIP, exploiting both the strong nucleophilicity of the acquired cysteine and the predominance of GDP-bound form in this specific mutant [9].
X-ray crystallography revealed that ARS-1620’s binding mode differs distinctly from earlier S-IIP inhibitors. The compound forms a direct hydrogen bond with His-95, creating a more rigidified and favorable conformation for covalent reaction [6]. The hydroxyl group on the active S-atropisomer occupies a solvated region and forms multiple water-mediated hydrogen bonds to KRAS residues, whereas the fluoro group occupies a hydrophobic region [6].
KRAS G12C covalently bound to these inhibitors loses SOS- and EDTA-mediated nucleotide exchange capacity, effectively trapping the protein in its GDP-bound inactive conformation [6]. This mechanism prevents reactivation through nucleotide exchange, thereby inhibiting downstream signaling pathways that drive cancer cell proliferation and survival.
Preclinical validation in 2D and 3D models
Preclinical testing validated the efficacy of these first KRAS G12C inhibitors across multiple experimental platforms. ARS-1620 displayed potent antiproliferative activity with IC50 values of approximately 150 nM in KRAS G12C-mutated cell lines, yet showed minimal effects on control cell lines [6]. This pronounced selectivity confirmed that activity stemmed specifically from covalent modification of Cys-12.
Notably, 3D cell culture models proved more predictive of in vivo sensitivity than traditional 2D monolayer cultures. Janes and colleagues observed that 2D-adherent cell cultures notably underestimated KRAS dependence in vivo, whereas 3D ultra-low adherent suspension spheroid cultures better predicted in vivo sensitivity of KRAS mutant cancer cells to ARS-1620 [10].
In vivo, ARS-1620 achieved over 75% target occupancy for extended periods, which proved necessary and sufficient for therapeutic efficacy [6]. At 200 mg/kg daily dosing, ARS-1620 demonstrated tumor growth inhibition exceeding 70% in mice bearing xenografts derived from five types of NSCLC cells [6]. In Mia.paca-2 xenograft models, it completely inhibited tumor growth while significantly downregulating RAS-GTP, phospho-ERK, phospho-AKT, and phospho-S6 levels [11].
These foundational discoveries and compounds paved the way for more advanced clinical candidates including sotorasib (AMG 510) and adagrasib (MRTX849), which would ultimately demonstrate clinical efficacy against KRAS G12C-mutant tumors and receive regulatory approval.
Clinical Breakthroughs: Sotorasib and Adagrasib 
The clinical development of KRAS G12C inhibitors reached a watershed moment with the approval of two targeted therapies—sotorasib and adagrasib—marking the culmination of four decades of research efforts.
FDA approval timeline and CodeBreaK trials
The U.S. Food and Drug Administration granted accelerated approval to sotorasib (Lumakras) on May 28, 2021, establishing it as the first direct KRAS G12C inhibitor available to patients [12]. Subsequently, adagrasib (Krazati) received accelerated approval on December 12, 2022 [13]. Both approvals were specifically for adults with locally advanced or metastatic non-small cell lung cancer (NSCLC) harboring the KRAS G12C mutation who had received at least one prior systemic therapy.
Sotorasib’s approval stemmed from the CodeBreaK 100 trial, wherein patients received 960 mg orally once daily. The pivotal phase 2 results demonstrated an objective response rate (ORR) of 37.1% with a median response duration of 10 months [12]. Long-term analysis revealed deeper clinical benefits: disease control rate (DCR) of 84%, median progression-free survival (PFS) of 6.3 months, and overall survival (OS) of 12.5 months [5].
The confirmatory phase 3 CodeBreaK 200 trial compared sotorasib with docetaxel in previously treated patients. Results showed improved median PFS with sotorasib (5.6 months versus 4.5 months with docetaxel) [14]. Though statistically superior, this modest PFS improvement prompted some regulatory scrutiny regarding the absence of OS benefit [5].
KRYSTAL-1 and KRYSTAL-12 trial outcomes
Adagrasib’s clinical development followed a similar trajectory. The phase 1/2 KRYSTAL-1 trial evaluated adagrasib at 600 mg twice daily in patients whose disease progressed after platinum-based chemotherapy and immunotherapy. The trial yielded an impressive 43% ORR with median duration of response reaching 8.5 months [15]. Extended follow-up data showed median OS of 14.1 months and median PFS of 6.9 months, with approximately one-third of patients maintaining durable efficacy at two years [5].
The phase 3 KRYSTAL-12 trial directly compared adagrasib with docetaxel. With a median follow-up of 9.4 months, adagrasib demonstrated superior PFS (5.49 versus 3.84 months, hazard ratio 0.58) [16]. ORR was substantially higher with adagrasib (31.9% versus 9.2% for docetaxel) [16]. Importantly, adagrasib showed greater efficacy for brain metastases, achieving a 40% intracranial ORR compared to 11% with docetaxel [14].
Efficacy in NSCLC vs CRC
Both inhibitors demonstrated more robust activity in NSCLC than in colorectal cancer (CRC). For sotorasib, early studies revealed a stark contrast—37.1% ORR in NSCLC versus merely 7.1% in CRC [3]. Similarly, adagrasib showed 45% ORR in NSCLC but only 17% in CRC patients [3].
Recognizing the limited efficacy in CRC, researchers explored combination approaches. Henceforth, on June 21, 2024, adagrasib received accelerated approval in combination with cetuximab for KRAS G12C-mutated metastatic CRC after prior chemotherapy. In 94 patients, this combination achieved 34% ORR with median response duration of 5.8 months [17]. Concurrently, sotorasib combined with panitumumab showed promise in the CodeBreaK 300 trial, with 30.2% ORR and potential 30% reduction in mortality risk versus standard care [18].
The pharmacokinetic profiles of these agents differ substantially—adagrasib features a longer half-life (23 hours versus 5 hours for sotorasib) and demonstrated superior blood-brain barrier penetration [19], potentially explaining its better performance in patients with brain metastases.
Despite these advances, neither drug provides curative treatment, primarily offering disease control for approximately 6-7 months before resistance typically emerges.

Mechanisms of Resistance to KRAS G12C Inhibitors
Despite the revolutionary advances in targeting KRAS G12C, resistance inevitably emerges during treatment, limiting the durability of clinical response. Both intrinsic and acquired resistance mechanisms have been observed in preclinical models and patient samples, affecting the efficacy of sotorasib and adagrasib alike.
On-target mutations: Y96C, G13D, Q61H
Secondary mutations in KRAS represent a major mechanism of resistance to G12C inhibitors. These on-target alterations primarily function by either interfering with covalent binding to cysteine-12 or maintaining KRAS in its active GTP-bound state. Among these, the Y96D/S mutations confer exceptionally high resistance to both sotorasib and adagrasib (resistance index >100 for sotorasib) [20]. This critical residue lies within the switch-II pocket where inhibitors bind.
Other commonly identified on-target resistance mutations include:
- Switch-II pocket mutations (14% of resistant patients) with Y96C being most frequent [8]
- G13D mutations that decrease GTP hydrolysis [20]
- A59S/T mutations with resistance indices >100 against sotorasib [20]
- R68S/M mutations that disrupt non-covalent binding interactions [20]
- Q61H mutations that maintain KRAS in its GTP-bound active state [3]
In addition to point mutations, KRAS G12C gene amplification occurs in approximately 22% of patients with acquired resistance [8]. This amplification increases production of mutant KRAS protein, overwhelming the inhibitor’s capacity to maintain suppression.
Off-target resistance: EGFR, MET, PI3K pathway activation
Bypass signaling represents a prevalent resistance strategy wherein alternate pathways become activated to circumvent KRAS inhibition. After exposure to KRAS G12C inhibitors, feedback activation of receptor tyrosine kinases (RTKs) can reactivate downstream signaling through wild-type RAS proteins or parallel pathways [21].
MET amplification emerged as an important mechanism in several studies. In KRAS G12C NSCLC cells with acquired sotorasib resistance, MET amplification reinforced RAS cycling from inactive to active forms while simultaneously activating AKT independent of RAS [22]. Equally important, upstream RTKs including EGFR, FGFR2, and RET demonstrated activating mutations or amplifications in patient samples after progression on sotorasib or adagrasib [23].
Other off-target resistance mechanisms include:
- Activating mutations in NRAS and BRAF [23]
- MAP2K1 alterations [8]
- PI3K pathway activation through multiple mechanisms [1]
- Loss-of-function mutations in KEAP1 and NF1 [20]
- Histologic transformation from adenocarcinoma to squamous cell carcinoma [3]
Notably, colorectal cancers treated with KRAS G12C inhibitors display more complex resistance patterns than lung cancers, often developing multiple simultaneous bypass mechanisms [3].
Role of epithelial-to-mesenchymal transition (EMT)
EMT represents a fundamental process wherein epithelial cells acquire mesenchymal characteristics, and this cellular transformation consistently correlates with both intrinsic and acquired resistance to KRAS G12C inhibitors [24]. In experimental models, TGFβ-induced EMT rendered previously sensitive cell lines resistant to ARS-1620 and sotorasib treatment [2].
The molecular basis for EMT-mediated resistance involves sustained activation of the PI3K pathway even during KRAS G12C inhibition [25]. In mesenchymal-like tumor cells, this pathway remains predominantly regulated by IGFR-IRS1 signaling rather than KRAS [25]. In contrast to the PI3K pathway, SHP2 plays a minimal role in PI3K activation yet remains critical for MAPK pathway stimulation [25].
Gene set enrichment analyzes confirmed that EMT induction leads to upregulated PI3K and ERK pathway activation in a cell type-dependent manner [26]. First, resistant cells demonstrate higher EMT scores, and second, artificially inducing EMT through TGFβ treatment increases KRAS-GTP levels, thereby limiting inhibitor efficacy [2].
Combination Therapy Strategies to Overcome Resistance
As resistance to KRAS G12C inhibitor monotherapy inevitably develops, research has pivoted toward rational combination strategies that target multiple signaling pathways simultaneously. These approaches aim to extend treatment duration and enhance clinical outcomes for patients with KRAS G12C-mutated cancers.
EGFR inhibitors: cetuximab and panitumumab
Epidermal growth factor receptor (EGFR) reactivation represents a key resistance mechanism in colorectal cancer (CRC). Upon exposure to KRAS G12C inhibitors, feedback activation of EGFR can reactivate downstream signaling pathways. To overcome this obstacle, researchers have explored combinations of anti-EGFR antibodies with KRAS G12C inhibitors.
In the KRYSTAL-1 trial, adagrasib plus cetuximab demonstrated an objective response rate (ORR) of 46% and median progression-free survival (PFS) of 6.9 months in pretreated KRAS G12C-mutated CRC [27]. This remarkable improvement over adagrasib monotherapy (19% ORR) led to FDA approval of this combination.
Alternatively, panitumumab combined with sotorasib in the CodeBreaK 101 trial achieved 30% ORR with median PFS of 5.7 months [28]. The phase III CodeBreaK 300 trial subsequently compared sotorasib (960 mg) plus panitumumab against standard care, showing superior PFS (5.6 vs 2.2 months, hazard ratio 0.49) [4].
SHP2 inhibitors: RMC-4550 and TNO155
SHP2 inhibition offers a promising strategy by blocking feedback-mediated RTK signaling that contributes to KRAS G12C inhibitor resistance. SHP2 serves as a critical mediator between receptor tyrosine kinases and RAS activation.
Clinical trials evaluating TNO155 (a SHP2 inhibitor) with JDQ443 (a KRAS G12C inhibitor) in the KontRASt-01 trial demonstrated 33% confirmed responses among KRAS G12C inhibitor-naïve NSCLC patients [7]. Correspondingly, RMC-4550 combined with sotorasib achieved 50% ORR in treatment-naïve NSCLC patients [7].
Beyond cancer cell effects, SHP2 inhibitors modify the tumor microenvironment and enhance anti-tumor immune responses. Combining RMC-4998 (a KRAS G12C-ON inhibitor) with RMC-4550 prevented tumor relapse after treatment withdrawal in preclinical models [29], indicating potential for durable responses.
PI3K/AKT/mTOR and CDK4/6 co-targeting
The PI3K/AKT/mTOR pathway functions as a parallel signaling cascade that can maintain cancer cell survival during KRAS G12C inhibition. Preclinical studies demonstrate that combined inhibition of PI3K or mTOR alongside KRAS G12C produces more pronounced antitumor effects than either approach alone [30].
Similarly, CDK4/6 inhibitors have shown promise in combination with KRAS G12C inhibitors. Multiple ongoing clinical trials are evaluating these combinations, including palbociclib with MRTX849 (adagrasib) for advanced KRAS G12C-mutant solid tumors [31]. Co-targeting CDK4/6 may help overcome resistance arising from cell cycle dysregulation.
Through these strategic combinations, clinicians now have expanded options for treating KRAS G12C-mutated cancers, potentially extending therapeutic benefit beyond the limitations of monotherapy.

Emerging KRAS G12C Inhibitors and RAS-ON Approaches
The landscape of KRAS G12C targeted therapy continues to evolve rapidly with next-generation inhibitors that surpass first-generation compounds in potency and versatility.
Divarasib, Garsorasib, and MK-1084 pharmacokinetics
Next-generation KRAS G12C selective inhibitors offer improved pharmacological profiles over sotorasib and adagrasib. Divarasib (GDC-6036) demonstrates great efficacy with an objective response rate (ORR) of 53.4% and median progression-free survival (mPFS) of 13.1 months in previously treated metastatic non-small cell lung cancer (NSCLC) [32]. This compound exhibits fewer grade ≥3 treatment-related adverse events (11%) alongside higher response rates than first-generation inhibitors [32].
Garsorasib (D-1553) represents another promising advancement, achieving 50% ORR with disease control in 88.6% of patients [5]. Its distinctive 12.78-month median duration of response underscores its clinical value [5]. Besides its effectiveness, garsorasib possesses high oral bioavailability and distribution to central nervous system tissues [33].
MK-1084, currently in phase 1 trials, displays versatility across multiple tumor types. In colorectal cancer (CRC), it achieves 38% ORR as monotherapy, increasing to 46% when combined with cetuximab [34]. For NSCLC, MK-1084 combined with pembrolizumab produces an impressive 77% response rate [34].
RAS-ON inhibitors like RMC-6236
A groundbreaking approach targets active GTP-bound KRAS through tri-complex inhibitors. Traditional KRAS G12C inhibitors target only inactive GDP-bound forms, yet RAS-ON inhibitors overcome this limitation through an innovative mechanism.
Daraxonrasib (RMC-6236), a pan-RAS inhibitor, forms a tri-complex with cyclophilin A (CypA) and RAS-GTP, effectively blocking RAS-effector interactions [35]. This noncovalent macrocyclic molecule occupies a composite binding pocket comprising CypA and SWI/SWII regions [35]. In clinical trials, RMC-6236 achieves 38% ORR in KRAS G12X NSCLC patients with 9.8-month mPFS [36].
Similarly, RMC-6291 binds to active GTP-bound KRAS G12C and CypA, creating an inactive tri-complex that halts KRAS signaling [32]. Intriguingly, this approach shows activity in patients who developed resistance to first-generation inhibitors [36].
Targeted protein degradation (TPD) and PROTACs
Proteolysis-targeting chimeras (PROTACs) represent an exciting alternative approach to KRAS G12C inhibition. These bifunctional molecules consist of a ligand targeting KRAS G12C linked to an E3 ubiquitin ligase recruiter, triggering protein degradation rather than mere inhibition [37].
Several KRAS G12C-PROTACs have been synthesized using sotorasib (AMG-510) as a foundation [9]. These compounds demonstrate both binding capacity and degradation ability toward KRAS G12C [9]. Particularly, compounds III-2 and IV-1 exhibit enhanced potency over sotorasib in downstream pathway inhibition [9].
This expanding therapeutic arsenal promises to address both primary and acquired resistance mechanisms, potentially extending survival benefits for patients with KRAS G12C-mutated cancers.
Biomarkers and Patient Selection for KRAS G12C Therapy
Effective selection of patients for KRAS G12C inhibitor therapy relies heavily on biomarker analysis that extends beyond mere detection of the G12C mutation itself. Comprehensive molecular profiling has emerged as a critical component for optimizing treatment outcomes.
KEAP1, STK11, and TP53 co-mutations
Co-occurring mutations substantially impact response to KRAS G12C inhibitors. KEAP1-mutated patients demonstrate markedly reduced response rates compared to wild-type counterparts (P = 0.009) [38] alongside significantly shorter overall survival (6-month OS P < 0.0001) [38]. In experimental validation, KEAP1 knockout in NCI-H358 cells increased IC50 values for KRAS G12C inhibitors from 21.72 nM to 141.4 nM [38].
STK11 mutations similarly predict poor outcomes, with patients harboring this alteration experiencing shorter survival despite minimal differences in objective response rates [38]. STK11-mutated cell lines show increased resistance to AMG510 [38].
TP53 co-mutations affect various KRAS mutant subtypes differently—reducing median overall survival by more than 30% in KRAS-only mutated lung adenocarcinoma [39], yet potentially exerting protective effects when occurring with KEAP1 mutations [39].
ctDNA monitoring for early resistance detection
Circulating tumor DNA offers valuable prognostic information through non-invasive monitoring. Patients achieving ctDNA clearance showed dramatically improved objective response rates (80% versus 8%, P < 0.001) [6] and disease control rates (100% versus 42%, P = 0.003) [6]. Clearance correlated with substantially longer progression-free survival (7.9 versus 2.8 months, P < 0.001) [6] and overall survival (16.8 versus 6.4 months, P = 0.001) [6].
Importantly, sotorasib treatment induced faster reduction in KRAS G12C variant allele frequency than docetaxel, detectable merely one week after treatment initiation [40]. Among sotorasib-treated patients, 43% achieved ctDNA clearance by cycle 2/day 1 versus only 14% with docetaxel [40].
Transcriptional subtypes: KC, KL, KP
Three robust transcriptional subtypes characterize KRAS-mutant NSCLC, with distinct treatment implications. The KP subtype (42.3% of patients) shows higher mutational burden [11] and exhibits improved relapse-free survival [11]. KL tumors (35.0% of patients) [40] demonstrated superior progression-free survival with sotorasib versus docetaxel (5.85 versus 2.69 months) [40]. Conversely, KC tumors (22.6% of patients) [40] showed poor outcomes regardless of treatment type.
TTF-1 expression provides additional prognostic insight—patients with TTF1-low tumors showed dramatically worse outcomes with sotorasib (mPFS 2.76 versus 8.11 months, P = 2.9 × 10^-9) [40] and lower objective response rates (4.17% versus 42.1%, P = 0.000151) [40].
Conclusion

KRAS G12C inhibitors represent a watershed moment in precision oncology, breaking through a four-decade barrier of failed attempts to target the once “undruggable” KRAS protein. These therapeutic agents work through a unique covalent binding mechanism that locks KRAS G12C in its inactive GDP-bound state, effectively halting downstream oncogenic signaling. Sotorasib and adagrasib have demonstrated compelling clinical activity, primarily in non-small cell lung cancer, with objective response rates ranging from 37-43% and progression-free survival of approximately 6-7 months. Their modest efficacy in colorectal cancer, however, underscores the complex biology of KRAS-driven tumors across different tissue contexts.
Despite these advances, resistance inevitably emerges through diverse mechanisms. On-target KRAS mutations like Y96C and G13D prevent inhibitor binding, while off-target adaptations through EGFR, MET, and PI3K pathway activation enable cancer cells to circumvent KRAS blockade. Epithelial-to-mesenchymal transition likewise confers resistance through sustained PI3K signaling independent of KRAS control. Accordingly, combination strategies targeting these escape pathways have shown promise, with EGFR inhibitor combinations already achieving regulatory approval for colorectal cancer.
Next-generation KRAS G12C inhibitors such as divarasib, garsorasib, and MK-1084 offer improved pharmacokinetics and potentially greater efficacy than first-generation compounds. Perhaps most exciting, RAS-ON inhibitors like daraxonrasib (RMC-6236) and targeted protein degradation approaches represent paradigm shifts that may overcome fundamental limitations of current therapies. These novel agents can target active GTP-bound KRAS through innovative mechanisms, potentially addressing both primary and acquired resistance.
Patient selection undoubtedly plays a crucial role in treatment outcomes. Co-mutations in KEAP1, STK11, and TP53 substantially impact response to KRAS G12C inhibitors, while circulating tumor DNA monitoring provides early indicators of treatment efficacy or resistance. The identification of distinct transcriptional subtypes (KC, KL, and KP) further refines our ability to select patients most likely to benefit from these targeted therapies.
The decades-long pursuit of effective KRAS inhibitors has finally yielded clinically meaningful results. Though current therapies remain non-curative, they provide valuable disease control for patients with limited treatment options. Future directions will certainly include development of pan-KRAS inhibitors, exploration of immunotherapy combinations, and strategies targeting multiple nodes in KRAS signaling networks simultaneously. This remarkable scientific journey exemplifies how persistent investigation of fundamental cancer biology ultimately translates to therapeutic innovations capable of improving outcomes for patients with KRAS-driven malignancies.
Key Takeaways
After 40 years of failed attempts, scientists have finally cracked the code on targeting KRAS G12C, one of cancer’s most elusive proteins, leading to breakthrough treatments that are transforming patient outcomes.
- KRAS G12C’s unique cysteine mutation creates a druggable target – Unlike other KRAS mutations, G12C allows covalent binding in its inactive GDP-bound state through the switch-II pocket.
- Sotorasib and adagrasib achieve 37-43% response rates in lung cancer – These first-in-class inhibitors provide 6-7 months disease control, marking historic progress against previously untreatable tumors.
- Resistance emerges through on-target mutations and bypass signaling – Y96C mutations and EGFR/MET pathway activation limit durability, driving combination therapy development.
- Combination strategies significantly improve colorectal cancer outcomes – Adding EGFR inhibitors like cetuximab boosts response rates from 7% to 46% in resistant CRC patients.
- Next-generation inhibitors and RAS-ON approaches show superior efficacy – Compounds like divarasib achieve 53% response rates, while RMC-6236 targets active KRAS forms previously considered impossible to drug.
This breakthrough demonstrates how understanding fundamental cancer biology can eventually overcome seemingly insurmountable therapeutic challenges, offering hope for patients with KRAS-driven cancers and paving the way for targeting other “undruggable” proteins.

Frequently Asked Questions: 
FAQs
Q1. What is the significance of KRAS G12C inhibitors in cancer treatment? KRAS G12C inhibitors represent a major breakthrough in cancer therapy, targeting a protein that was considered “undruggable” for 40 years. They provide a new treatment option for patients with certain types of lung and colorectal cancers harboring this specific mutation.
Q2. How do KRAS G12C inhibitors work? These inhibitors work by binding to the KRAS G12C protein in its inactive GDP-bound state, locking it in this conformation and preventing its activation. This halts the downstream signaling pathways that drive cancer cell growth and survival.
Q3. What are the main challenges in using KRAS G12C inhibitors? The primary challenges include the development of resistance mechanisms, such as secondary KRAS mutations or activation of bypass signaling pathways. Additionally, these inhibitors show varying efficacy across different cancer types, with better responses observed in lung cancer compared to colorectal cancer.
Q4. Are there any combination therapies being explored with KRAS G12C inhibitors? Yes, several combination approaches are being investigated to overcome resistance and improve efficacy. These include combining KRAS G12C inhibitors with EGFR inhibitors, SHP2 inhibitors, and drugs targeting the PI3K/AKT/mTOR pathway.
Q5. What new developments are on the horizon for KRAS-targeted therapies? Emerging approaches include next-generation KRAS G12C inhibitors with improved pharmacokinetics, RAS-ON inhibitors that can target active GTP-bound KRAS, and targeted protein degradation strategies using PROTACs. These novel approaches aim to address limitations of current therapies and potentially overcome resistance mechanisms.
References: 
[1] – https://aacrjournals.org/cancerdiscovery/article/14/11
/2135/749207/Mechanisms-of-Resistance-to-Oncogenic-KRAS
[2] – https://pmc.ncbi.nlm.nih.gov/articles/PMC8774282/
[3] – https://www.nejm.org/doi/full/10.1056/NEJMoa2105281
[4] – https://pmc.ncbi.nlm.nih.gov/articles/PMC11535806/
[5] – https://www.frontiersin.org/journals/immunology/articles/
10.3389/fimmu.2025.1509173/full
[6] – https://acsjournals.onlinelibrary.wiley.com/doi/full/10.1002/cncr.35917
[7] – https://www.frontiersin.org/journals/oncology/articles/
10.3389/fonc.2024.1380584/full
[8] – https://www.annalsofoncology.org/article/S0923-7534(25)00052-3/fulltext
[9] – https://pubmed.ncbi.nlm.nih.gov/36621179/
[10] – https://jeccr.biomedcentral.com/articles/10.1186/s13046-021-02225-w
[11] – https://aacrjournals.org/cancerdiscovery/article/5/8/860/5019/Co-occurring-Genomic-Alterations-Define-Major
[12] – https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-sotorasib-kras-g12c-mutated-nsclc
[13] – https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-adagrasib-kras-g12c-mutated-nsclc
[14] – https://pmc.ncbi.nlm.nih.gov/articles/PMC11534526/
[15] – https://www.ons.org/publications-research/voice/news-views/12-2022/fda-grants-accelerated-approval-adagrasib-kras-g12c
[16] – https://ascopubs.org/doi/10.1200/JCO.2024.42.17_suppl.LBA8509
[17] – https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-adagrasib-cetuximab-kras-g12c-mutated-colorectal-cancer
[18] – https://ascopubs.org/doi/10.1200/JCO-24-02026
[19] – https://www.nejm.org/doi/full/10.1056/NEJMoa2204619
[20] – https://ehoonline.biomedcentral.com/articles/10.1186/s40164-023-00453-8
[21] – https://pmc.ncbi.nlm.nih.gov/articles/PMC8178176/
[22] – https://aacrjournals.org/clincancerres/article/27/20/5697/671727/KRAS-Inhibitor-Resistance-in-MET-Amplified
[23] – https://ascopubs.org/doi/10.1200/EDBK_351333
[24] – https://pubmed.ncbi.nlm.nih.gov/32900796/
[25] – https://aacrjournals.org/clincancerres/article/26/22/5962/82840/Epithelial-to-Mesenchymal-Transition-is-a-Cause-of
[26] – https://www.nature.com/articles/s41417-021-00383-9
[27] – https://www.nejm.org/doi/full/10.1056/NEJMoa2212419
[28] – https://pmc.ncbi.nlm.nih.gov/articles/PMC11340593/
[29] – https://www.nature.com/articles/s41467-024-52324-3
[30] – https://pmc.ncbi.nlm.nih.gov/articles/PMC7669705/
[31] – https://www.mdpi.com/2072-6694/17/12/1941
[32] – https://pmc.ncbi.nlm.nih.gov/articles/PMC11963745/
[33] – https://www.nature.com/articles/s41392-025-02274-z
[34] – https://www.merck.com/news/merck-announces-mk-1084-an-investigational-kras-g12c-inhibitor-shows-antitumor-activity-in-phase-1-trial-of-patients-with-advanced-colorectal-cancer-and-non-small-cell-lung-cancer-whose-tumors-harbo/
[35] – https://pubs.acs.org/doi/10.1021/acs.jmedchem.4c02314
[36] – https://actr.amegroups.org/article/view/11484/html
[37] – https://www.sciencedirect.com/science/article/abs/pii/S0223523423008243
[38] – https://translational-medicine.biomedcentral.com/articles/10.1186/s12967-025-06089-y
[39] – https://pmc.ncbi.nlm.nih.gov/articles/PMC11327858/
[40] – https://www.nature.com/articles/s41591-025-03732-5
Video Section
Recent Articles

