International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer-related mortalities globally and accounts for ~85% of all lung cancer cases (1). Among the molecular subtypes of NSCLC, Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations are one of the most commonly identified oncogenic drivers. These mutations are commonly prevalent in lung adenocarcinoma (LUAD) and are strongly linked with smoking and environmental carcinogens (2). KRAS is a member of the rat sarcoma viral (RAS) oncogene family and accounts for the majority of RAS-related mutations encountered in human cancer. KRAS alterations account for ~85% of all RAS mutations (3). The prevalence of KRAS mutations varies across cancer types, with the highest frequencies found in pancreatic (88%), colorectal (45–50%) and lung cancer (31–35%) (4). KRAS mutations are present in ~30% of LUAD cases in Western populations and ~10% of in Asian populations (5,6).
The KRAS proto-oncogene encodes a small guanine triphosphate (GTPase) that acts as a binary switch in signal transductions of multiple receptor tyrosine kinases, such as mesenchymal-epithelial transition (MET), epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK), thus regulating cellular functions such as proliferation, differentiation and survival. KRAS is a clonal transforming oncogene and is a key oncogenic driver across multiple solid tumors (7–9).
Structurally, KRAS is a 21-kDa GTPase composed of a conserved G-domain and two dynamic regulatory regions, Switch I and Switch II, which control its active (GTP-bound) and inactive (GDP-bound) states. The Switch II pocket (SII-P) forms the allosteric site targeted by KRAS G12C inhibitors, while C-terminal farnesylation enables its anchoring to the plasma membrane (10,11).
The prevailing outlook on the poor prognosis and limited treatment options for NSCLC has significantly shifted over the last 2 decades due to the development of targeted therapy, molecular profiling and immunotherapy (12,13). Although several preclinical and clinical studies have investigated RAS inhibitors, no authorized treatments directly inhibit mutant variants of KRAS or its downstream signaling (14,15). These novel KRAS inhibitors are currently being investigated in combination with other therapies, which may significantly improve the treatment of KRAS-mutated NSCLC. Targeted therapy and immunotherapy have shown promising results in treating NSCLC, therefore the present review explored novel treatment strategies and their efficacy in targeting the KRAS mutation in NSCLC.
KRAS is a key proto-oncogene that encodes a small GTPase that controls cell survival, differentiation and proliferation by acting as a molecular switch in important cellular signaling cascades (3). The GTPase cycles between an inactive GDP-bound and an active GTP-bound state. Activation is promoted by guanine nucleotide exchange factors (GEFs), which stimulate GDP-GTP exchange, such as SOS1 and the GTPase-activating proteins (GAPs); for example, neurofibromin 1 is a GAP that accelerates GTP hydrolysis to inactivate KRAS (14,16).
Multiple downstream effectors of KRAS, primarily, are part of the RAF-MEK-ERK signaling pathway. Activated KRAS-GTP recruits RAF kinases to the membrane, which promotes RAF dimerization and activation. RAF phosphorylates MEK1/2, leading to activation of ERK1/2. Upon activation, nuclear translocation of ERK occurs and targets transcription factors, including E26 transformation-specific family, serum response factor and ribosomal S6 kinase, to influence cell proliferation, differentiation and migration. Furthermore, KRAS stimulates phosphoinositide 3-kinase (PI3K), leading to AKT activation that promotes cell survival, metabolism and growth. KRAS also activates RalGEFs, which regulate cytoskeleton remodeling, membrane dynamics and vesicle trafficking (Fig. 1) (14,16,17). In its wild-type state, KRAS participates in normal cell signaling but does not directly suppress immune responses. KRAS mutations; however, it inhibits T-cell activity by stimulating an immunosuppressive tumor microenvironment (TME) through the induction of cytokine-(such as IL-6) and chemokine-(such as IL-8) driven inflammation and immune evasion, and increase programmed cell death-1 ligand (PD-L1) expression levels via activation of the MAPK and PI3K-AKT pathways, resulting in immune escape (14,18).
KRAS mutations are clinically significant in NSCLC due to their frequency, prognostic implications and impact on treatment decisions (19,20). Point mutations commonly deregulate the KRAS gene, resulting in an inherently active GTP-bound phase and activating downstream oncogenic signaling pathways (21–23). KRAS mutation frequency is lower (~5%) in squamous NSCLC and higher (20–40%) in LUAD (24). Furthermore, these mutations are more prevalent in Western populations compared with Asian populations (26 vs. 11%) and in smokers compared with non-smokers (30 vs. 11%); the presence of KRAS mutations in NSCLC is closely associated with smoking (25). Furthermore, KRAS mutations are more common in female and younger patients, according to a pooled study of resected NSCLC tumors; however, only the association with younger patients was significant in the multivariate analysis (P=0.044) conducted (26,27). The previous study by Shepherd et al (27) on the frequency of KRAS mutations did not include any analysis specific to histology or race. Smoking is suggested to leave a particular molecular mark on KRAS gene since point mutations (G12C and G12V) are often detected in former or current smokers; by contrast, transitional mutations (G12D) are more common in non-smokers (28,29). Furthermore, compared with individuals who have never smoked, smokers often have substantially more complicated KRAS-mutant tumors with a more considerable mutational burden and a higher probability of significant co-occurring mutations in tumor protein P53 (TP53) or serine/threonine kinase 11 (STK11) (30).
Co-mutations, including STK11/LKB1, may influence the prognosis of KRAS-mutant NSCLC. For example, ERK1/2 phosphorylation is more significant in tumors with KRAS G12C mutations compared with those with KRAS G12D mutations, and this enhanced MAPK signaling can be further amplified in the presence of STK11/LKB1 loss, contributing to more aggressive tumor biology. Supporting this, a previous study employing animal models driven by KRAS mutations showed that the MEK inhibitor selumetinib was more effective in KRAS G12C tumors compared with KRAS G12D tumors (31). As a result, various KRAS mutations may induce signal transduction cascades differently, resulting in unique drug sensitivity profiles (32). Regarding co-occurring mutations, LUAD is typically associated with KRAS single-driver mutations, accounting for 95–99% of KRAS-mutant cases, while double mutants (EGFR/ALK/BRAF and KRAS) are rare, occurring in <1% of cases (33–35).
Approximately one-third of patients with LUAD have a KRAS mutation; despite significant advancements in the discovery and development of targeted therapeutics for molecular subtypes of LUAD (36), no authorized medication currently targets any KRAS mutation directly (37). For the past 40 years, significant efforts have been made to develop medicines for KRAS. These investigations have targeted the KRAS protein and its membrane location, interactions between proteins, post-translational modifications, and downstream signaling pathways; however, these methods have not shown promising outcomes in clinical trials (38,39).
Since KRAS has a restricted number of active binding sites, complicated biochemistry (40) and a high affinity for GTP (41), direct targeting of the protein was thought to be complex until ~5 years ago (42). Developments in crystallography and computer modeling (43) have identified small compounds that directly bind to the inactive GDP-bound conformation of KRAS G12C within the SII-P, thereby locking the protein in its inactive state and preventing downstream signaling (44). However, the binding affinity of these early-stage drugs would need to be substantially increased before they can be used as effective treatments. These early compounds demonstrated that RAS is principally druggable; however, they lacked sufficient potency and pharmacological properties to achieve clinical efficacy and often lacked the precision required to target mutant KRAS effectively (45)
Prior to the advent of direct KRAS inhibitors, indirect strategies were investigated, such as by blocking KRAS post-translational modifications, preventing its anchoring to the cell membrane or disrupting the downstream signaling pathways it controls (46–48). Early analyses focused on farnesyl transferase inhibitors, salirasib and tipifarnib being tested to prevent membrane anchoring of KRAS. Although the preclinical activity was promising, the clinical efficacy of salirasib and tipifarnib in NSCLC was limited due to compensatory prenylation with geranylgeranyl transferase. In a phase II trial of salirasib in KRAS-mutant LUAD, no objective responses were observed, although 30–40% of patients achieved temporary disease stabilization at 10 weeks (49,50).
Blockage of the downstream KRAS signaling has also been investigated. Small molecules that target ERK and MEK decrease effector phosphorylation and block RAF interactions in preclinical models (45,51). Despite this, MEK inhibitor clinical trials as monotherapy have been ineffective. In a phase II study of previously treated patients with KRAS-mutant NSCLC, FAK inhibition with defactinib alone had limited clinical activity [median progression-free survival (PFS), 45 days], and activity was not associated with TP53 or CDKN2A status (52).
These studies highlight the primary limitations of indirect KRAS targeting: Biochemical redundancy, adaptive feedback loops and toxicity due to effects on essential signaling pathways (53). Post-prenylation processing enzymes such as Ras-converting enzyme 1 and isoprenylcysteine carboxyl methyltransferase remain potential targets, but inhibitors lack specificity and risk broad systemic toxicity (54). However, these limitations are being overcome through the development of pragmatic strategies. Combined inhibition of farnesyl- and geranylgeranyl transferase has been reported as a promising approach in models of pancreatic cancer, although it has not been tested in LUAD (55). Epidemiological studies suggest that oral bisphosphonate use is associated with a decreased risk of lung cancer in non-smoker postmenopausal women (56) and a case report described regression of hepatic metastases in primary lung adenocarcinoma following zoledronic acid monotherapy (57). Preclinical models indicate that these vulnerabilities are subtype-specific: Mesenchymal-like KRAS-mutant NSCLC is sensitive to combined FGFR-MEK inhibition, whereas epithelial-like tumors preferentially respond to ERBB-MEK blockade (58). Such biomarker-led combination strategies provide novel avenues for indirect targeting. Collectively, these indirect approaches illustrate the innovative nature of previous efforts to target KRAS and the associated limitations, and demonstrate the importance of investigations on direct interventions in the current therapeutic landscape.
The mutant KRAS G12C protein shows decreased GAP-stimulated GTPase activity from biochemical analyses, consistent with earlier G12X mutants (excluding G12P). In contrast with other mutated KRAS proteins, the mutation KRAS G12C maintains periodic intrinsic GTPase function and alternates among the active KRAS-GTP and inactive KRAS-GDP states (59,60). Structural analysis demonstrated that the KRAS Cys12 mutation is proximal to a newly identified allosteric site, the SII-P, which is transiently accessible in the GDP-bound state. This finding allowed the design of covalent inhibitors to bind Cys12 irreversibly, trapping KRAS G12C in its inactive state and ablating downstream signaling (61,62). The first therapies to target this SII-P were sotorasib, adagrasib and, more recently, garsorasib (D-1553). These KRAS G12C inhibitors all bind covalently to the SII-P but differ in their binding kinetics and molecular interactions (63). Adagrasib exhibits higher conformational mobility, allowing more sustained binding to KRAS-G12C, while garsorasib has shown promising clinical activity in NSCLC and colorectal cancer (64,65). Structural scaffold differences between KRAS G12C inhibitors, such as sotorasib and adagrasib, are likely responsible for the different pharmacodynamic profiles and tissue penetration.
Although the SII-P is the most-validated and extensively targeted allosteric site, it is not the sole structural determinant of KRAS inhibitor binding; other regions of KRAS also influence drug design and activity. KRAS contains key structural regions, including the Switch I and Switch II domains and a hypervariable region that regulates membrane attachment. The switch I region can be indirectly modulated by inhibiting SOS1-KRAS interactions using SOS1 inhibitors (such as BI-3406 and BI-1701963). The nucleotide binding pocket has been the focus of investigation with nucleotide competitive analogs and membrane localization disrupted with FTase inhibitors through the hypervariable region (66–68). These findings underscore that KRAS contains several structural features beyond the SII-P that may be exploited therapeutically.
The first KRAS G12C inhibitor studied in a clinical trial for an advanced solid tumor was sotorasib. The initial investigation was a CodeBreaK 100 phase I/II basket trial to determine the safety and beneficial effects of sotorasib in patients with advanced tumors with a KRAS G12C mutation, particularly those who previously received standard treatment. Based on the clinical trial outcome, sotorasib received approval from the Food and Drug Administration (FDA) in 2021 to treat advanced-stage NSCLC (69,70). In a cohort of 126 patients with KRAS G12C mutation NSCLC, the objective response rate (ORR) was 37.1%, with 3.2% exhibiting a complete response (CR). The median overall survival (OS) was 12.5 months, median PFS was 6.8 months, and adverse events were seen in 69.8% of patients (71). Another study found a 42.9% confirmed ORR for 116 patients with KRAS G12C mutated NSCLC. the median OS was 12.6 months and the median PFS and response duration were 6.5 and 8.5 months, respectively (72). In a subsequent phase III trial comparing sotorasib and docetaxel, sotorasib increased PFS [PFS, 5.6 months vs. 4.5 months for docetaxel, hazard ratio (HR)=0.66, P=0.0017] in patients with NSCLC who had previously received chemotherapy (73). The IFCT-2102 Lung KG12Ci study, a nationwide study conducted in France, included 458 patients with metastatic or advanced KRAS G12C-mutated non-squamous NSCLC across 76 centers. The median age was 65.8 years and 43.4% of the patients were female. The majority (95.4%) were current or former smokers and 38.0% had brain metastases at the initiation of sotorasib therapy. The key efficacy endpoints were a median real-world PFS of 3.5 months and a median real-world OS of 8.3 months (median follow-up was 15.8 and 16.4 months, respectively). The real-world ORR was 35.5% and the disease control rate (DCR) was 63.7%. In the subgroup of patients with brain metastases, the intracranial real-world ORR was 20.1% and the intracranial real-world DCR was 66.9%. From a safety perspective, sotorasib was considered to be tolerable. However, 16.5% of patients discontinued their therapy because of adverse events, while 5.2% of patients experienced grade 3–4 hepatotoxicity (74).
Sotorasib changed the treatment landscape for KRAS G12C-mutant NSCLC by providing improved clinical outcomes compared with standard second-line therapies, such as docetaxel, docetaxel-ramucirumab and immune checkpoint inhibitor (ICI) monotherapy. Nevertheless, the intracranial efficacy of sotorasib is low, particularly in patients with brain metastases, and survival benefits are limited due to the acquisition of resistance; thus, there is an urgent need for mechanism-based combination approaches and next-generation inhibitors. The clinical decision to use sotorasib should be made with caution, particularly for patients with KRAS G12C-mutant NSCLC with central nervous system (CNS) involvement or a high disease burden.
Another KRAS G12C inhibitor with a positive clinical record is adagrasib. The phase I/IB KRYSTAL-1 study of adagrasib demonstrated a favorable safety profile and a median PFS of 11.1 months (75). KRYSTAL-12 (trial no. NCT04685135), a phase III study of adagrasib vs. docetaxel in patients with previously treated KRAS G12C-mutated NSCLC, demonstrated that 94.0% of patients who received adagrasib and 86.4% of patients who received chemotherapy had treatment-related adverse events (TRAEs). Grade ≥3 TRAEs occurred in 47.0% of patients who received adagrasib and 45.7% of patients who received docetaxel. These patients would have received immunotherapy before receiving adagrasib (76).
The KRAS G12C inhibitor garsorasib showed strong efficacy and tolerable safety in a phase II trial (trial no. NCT05383898) of 123 Chinese patients with pre-treated KRAS G12C-mutated NSCLC. All participants (median age, 64; 88% male) were administered 600 mg garsorasib twice daily and achieved an ORR of 50% (1 CR and 60 partial responses) and a DCR of 89%. With a median follow-up of 12.3 months, the median PFS was 9.1 months and the median OS was 14.1 months, the longest reported in commercialized KRAS G12C inhibitors. TRAEs were seen in 95% of patients, with grade ≥3 events (50%) mainly comprising hepatic enzymes (AST elevation, 17%; ALT, 15%) and gastrointestinal symptoms (nausea/vomiting, 2%); most toxicities were dose-modifiable (77,78). Garsorasib has demonstrated promising clinical activity with high response rates, durable responses and prolonged OS in patients with previously treated KRAS G12C-mutant NSCLC, and has shown superior outcomes compared with existing inhibitors, in certain contexts as aforementioned. However, widespread use and future studies with a larger sample size are necessary to collect more data on CNS activity, long-term survival and comparative effectiveness. Presently, the use of garsorasib may be considered post-chemotherapy and ICI therapy, particularly in patients with healthy liver function who can be closely monitored. Further to the aforementioned studies, numerous other clinical trials (Table I) are currently under investigation to identify therapies with increased efficacy of KRAS G12C inhibitors.
Clinical resistance continues to present a notable barrier and commonly involves allosteric sites such as the SII-P. Second-site mutations on or around the SII-P change pocket geometry and/or local dynamic structures that directly disrupt inhibitor binding (79). For example, Y96D eliminates hydrogen bonding in the pocket, A59T, and G13D disrupts the conformational state essential for drug interaction (80). Furthermore, bypass mechanisms of resistance, including upstream mechanisms by amplifying the HER2 gene, can induce KRAS activation and reduce dependence on SII-P inhibition (81). Taken together, these findings show that the structural confirmation of the SII-P influences inhibitor binding affinity, and demonstrates how mutations in this region can lead to resistance. This close association between the structure and function of KRAS underscores the importance of combining strategies or developing next-generation inhibitors to overcome these challenges, including resistance.
Combining KRAS G12C inhibitors with ICIs is supported by synergistic immunogenicity: Blocking KRAS can increase tumor antigen presentation and T-cell infiltration, while ICIs counteract immune suppression (62,82). The KRYSTAL-7 trial (trial no. NCT04613596) is a phase III study assessing first-line adagrasib in combination with pembrolizumab compared with monotherapy pembrolizumab in patients with advanced NSCLC with a KRAS G12C mutation. In patients with a PD-L1 tumor proportion score ≥50%, the confirmed ORR was 63% [32/51 patients; 95% confidence interval (CI), 48–76%]. The DCR in the same subgroup was 84% (43/51 patients; 95% CI, 71–93%). The median PFS at a median follow-up of 10.1 months was not achieved (95% CI, 8.2 months to not evaluable). The TRAEs reported among the 148 patients evaluated for safety led to permanent discontinuation of adagrasib in 6% (n=9) and pembrolizumab in 11% (n=16) of patients with TRAEs; 4% (n=6) of patients discontinued both agents due to TRAEs (83). The phase Ib 100/101 CodeBreaK study tested sotorasib in combination with pembrolizumab or atezolizumab in patients with metastatic KRAS G12C NSCLC. Although the combination demonstrated clinical activity (ORR, ~29%), numerous patients experienced severe grade 3–4 hepatotoxicity when starting both drugs concomitantly. A ‘sotorasib lead-in’ dose introduction strategy markedly ameliorated severe liver toxicity and reduced discontinuation of treatment, underscoring the relevance of dosage sequence to enhance tolerability (84). Presently, the ongoing Krascendo-170 trial (trial no. NCT05789082) is evaluating divarasib in combination with pembrolizumab sequentially, to determine whether the treatment dose can be maximized without losing an optimal balance between efficacy and toxicity in frontline KRAS G12C-mutated NSCLC.
Combining KRAS G12C inhibitors with ICIs may potentially improve efficacy, particularly in patients with PD-L1-high tumors. Adagrasib has potential as a frontline combination therapy, requiring clarification of the optimal order and timing for administration. Future clinical plans should individualize treatment according to tumor biology and immune status, guided by direct comparison trials initiated through biomarker-informed pathways.
The toxicities of KRAS G12C inhibitors (sotorasib and adagrasib) are clinically relevant, although frequently manageable with individualized approaches. In a prospective biomarker study, 75% of patients with a history of immune-related hepatitis from prior immunotherapy subsequently developed severe hepatotoxicity with sotorasib (85). Nevertheless, attempts at dose reduction and rechallenge were made in over half of cases, and long-term rechallenge was successful in some patients using corticosteroids. Similarly, gastrointestinal AEs (such as diarrhea, nausea and vomiting) may be controlled with supportive medications (such as antiemetics, antidiarrheals, hydration and dietary changes), and therapy is typically held or dose adjusted for grade ≥3 events in the case of adagrasib. Liver toxicity may be managed with regular liver enzyme monitoring and prompt dose modification. These results highlight that while toxicities may restrict dosing, personalized immunosuppression and dose adjustment interventions can sustain therapeutic benefit in appropriate patients (86,87).
KRAS G12C inhibitors have presented a new era for NSCLC treatment through direct targeting of KRAS, as both sotorasib and adagrasib have shown significant clinical activity, and as well as the more recent therapy, garsorasib. However, modest survival benefits, low intracranial activity and a resistance mechanism remain significant limitations. Continued work with rational combinations, tailored sequencing with immunotherapy, toxicity-mitigation approaches and the emergence of next-generation inhibitors are necessary to sustain long-term responses and provide further treatment options for patients with KRAS G12C tumors.
The lack of effective targeted treatment for patients with non-G12C KRAS mutations in NSCLC is a notable clinical issue, particularly considering the recent FDA approval of KRAS G12C inhibitors. In contrast to G12C inhibitors, which covalently trap the G12C-specific cysteine residue, non-G12C mutations require alternative treatment strategies. Immune profiling studies show that non-G12C mutations are often associated with increased rates of PD-L1 negativity and STK11 co-mutations, which are both predictors of poor response to ICIs (88–90).
By targeting pan-KRAS inhibition, TME modulation and upstream and downstream signaling pathways, emerging strategies aim to overcome limitations such as allele-specific resistance, pathway redundancy, and adaptive signaling escape. Pan-RAS and RAS-GTP inhibitors are being further developed, as they have demonstrated activity against other KRAS mutations beyond G12C (88). The non-covalent inhibitors MRTX1133 and HRS-4642 disrupt KRAS G12D interactions with SOS1 or RAF1, resulting in downstream MEK-ERK inhibition. MRTX1133 (trial no. NCT05737706) has progressed to phase I clinical trials, and HRS-4642 is in phase I/II trials (91), showing preclinical tumor growth inhibition in vitro and in vivo (92).
Although tumor mutational burden (TMB) and PD-L1 expression levels are comparable in G12C and non-G12C subgroups, clinical responses in non-G12C variants are unsatisfactory (93). These tumors are frequently associated with co-mutations such as STK11, which shape an immune-cold microenvironment, contributing to poor response to ICIs (90). Despite novel targeted approaches, the therapeutic landscape for non-G12C mutations of KRAS remains limited.
Due to the constraints of targeted approaches, immunotherapy has become an essential alternative and complementary treatment option, especially for KRAS-mutant NSCLC. Furthermore, novel approaches such as combination regimens with KRAS pathway inhibitors and ICIs are being explored, based on data that KRAS mutations alter the immune microenvironment and response to ICIs (89). These strategies highlight the need for novel immunotherapeutic regimens and rational combinations to improve outcomes for patients with non-G12C KRAS mutations. Although emerging KRAS G12C inhibitors show potential, patients with non-G12C KRAS mutations in NSCLC have limited options for targeted therapy and respond poorly to immunotherapy because of co-mutations and a cold tumor immune microenvironment. Novel non-covalent KRAS inhibitors and combination approaches targeting signaling pathways and immune modulation are emerging as promising treatment options; however, further development of therapies targeted at specific molecular and immune mechanisms is necessary to improve outcomes for this complex cohort.
ICIs, combined with platinum-based chemotherapy or alone, are the conventional first-line treatment for patients with advanced KRAS mutation-positive NSCLC (94). Smoking has been associated with elevated TMB in addition to the immune-related characteristics of KRAS-mutant tumors. Increased TMB may correlate with an improved response to ICIs (95,96). Mazieres et al (97) retrospectively analyzed NSCLC cases with different oncogenic driver mutations and reported that increased PD-L1 expression levels were observed in KRAS-mutant tumors, which also showed superior responses to ICIs compared with that of other oncogenic driver mutations such as EGFR, ALK, ROS1 and RET alterations. However, most phase III ICI trials in NSCLC did not stratify patients by KRAS subtype, and only a few included post-hoc analyses (98,99). Although KRAS G12C inhibitors have shown benefit, other KRAS mutations still lack targeted options. Table II summarizes key immunotherapy trials in KRAS-mutant NSCLC.
The KEYNOTE-042 study is a phase III trial comparing pembrolizumab to platinum-based chemotherapy as a first-line treatment for patients with PD-L1-positive advanced NSCLC. KRAS mutations, including NRAS mutations, were found in 22.9% (9.6% KRAS G12C) of 301 patients; these patients exhibited increased levels of tissue TMB and PD-L1 expression. In KRAS-mutant cohorts, pembrolizumab treatment showed a significantly improved outcomes, with an OS hazard ratio of 0.42 (95% CI, 0.22–0.81) and a PFS hazard ratio of 0.51 (95% CI, 0.29–0.87) compared with KRAS wild-type tumors (100). The KEYNOTE-189 study, which examined the efficacies of first-line platinum-based chemotherapy alone or combined with pembrolizumab in advanced-stage NSCLC, reported that 12.8% of the 289 patients (95% CI, 10.1–15.6%) had a KRAS G12C mutation, while 30.8% (95% CI, 22.9–38.8%) reported any KRAS mutation. Furthermore, tumors with any KRAS mutation had elevated tissue TMB and PD-L1 levels. Pembrolizumab-based treatment showed higher clinical outcomes regardless of KRAS status than chemotherapy alone in PFS, OS and ORR (101). These trials indicate that KRAS-mutated NSCLC, particularly KRAS G12C, have an enhanced response to pembrolizumab and immunotherapy due to high-level expression of PD-L1 and TMB. Pembrolizumab, alone or combined with chemotherapy, could result in survival and progression-free benefits for these patients vs. chemotherapy alone.
A retrospective study conducted in China showed that immunotherapy-based treatment in patients with KRAS-mutant NSCLC showed improved results compared with that of chemotherapy alone. The immunotherapy regimens used in this cohort were PD-1/PD-L1 inhibitors either alone or in combination with chemotherapy. Overall, the median OS was 22.9 months (95% CI, 14.1–31.7) and PFS was 9.4 months (95% CI, 6.6–12.1). Immunotherapy regimens had a median OS of 45.2 vs. 11.3 months (P=1.81E-5) and a median PFS of 10.5 vs. 8.2 months (P=0.706) compared with chemotherapy. The first line therapy cohort demonstrated a median OS and median PFS of 33.5 vs. 16.1 months (P=0.010) and 32.2 vs. 6.9 months (P=0.00038), respectively. In the second line therapy cohort, median OS was not reached vs. 9.23 months (P=0.026) and median PFS was 10.8 vs. 5.5 months (P=0.010). Immunotherapy improved the median OS compared with that of chemotherapy for both patients with KRAS G12C tumors and those with non-KRAS G12C tumors, according to subgroup analysis (G12C, 25.2 vs. 9.1 months, P=0.0037; non-G12C, not reached vs. 25.7 months, P=0.020). In addition, concurrent KRAS/TP53 mutants also showed an improved median OS with immunotherapy-based regimens compared to those who received chemotherapy (33.5 vs. 11.8 months; P=0.036). These findings suggest that immunotherapy is beneficial for OS relative to chemotherapy in patients with late-stage KRAS-mutant NSCLC, regardless of the treatment line or KRAS mutation subtype (102).
Recent studies have provided novel insights into the role of immunotherapy in KRAS-mutant NSCLC. A retrospective analysis was conducted in Italy with a cohort of 143 patients with advanced NSCLC and KRAS mutations received ICIs, with or without chemotherapy. The most frequent KRAS mutation was G12C (41%), followed by G12V (23.7%) and G12D (11.8%); ≥50% of the patients received ICIs as monotherapy, while the rest had it in combination with chemotherapy, mainly as a first-line therapy. The OS or PFS among the KRAS subgroups showed no significant differences. By contrast, patients with KRAS Q61, 13X and G12C mutations had a relatively longer OS (46.5, 31.8 and 28.7 months, respectively) of the KRAS subgroups analyzed. The KRAS G12D cohort showed the most improved response to treatment with an (ORR) of 73%, largely driven by patients with stable disease (40%), which was greater for chemoimmunotherapy. The median duration of response (DOR) was 7.4 months overall, with the longest DOR at 10.2 months seen in G12V. Co-mutations were also assessed: STK11 was present in 24% of cases and TP53 in 29%. Patients with STK11 co-mutations tended towards more prolonged OS (39.7 months) compared with those without STK11 co-mutations (26.1 months). By contrast, TP53 co-mutations were associated with a shorter OS (19.1 months), although the analysis did not reach statistical significance. Notably, bone metastases were associated with lower survival and almost doubled the risk of death (HR, 2.81; P<0.001) (103). However, the analysis was retrospective, single institution-based, underpowered and the treatment strategies were heterogeneous, which limited the strength and external validity of the results. Furthermore, the negative association between STK11 co-mutations and improved survival is in contrast to numerous previous findings (104,105) and may be a cohort-specific artifact. Therefore, the aforementioned study requires further validation in larger, homogeneously treated cohorts.
A multicenter retrospective study compared first-line programmed cell death protein 1 (PD-1)/PD-L1 inhibitors (n=78) vs. platinum-based chemotherapy (n=29) in metastatic KRAS-mutant NSCLC. The PFS was significantly longer in the immunotherapy group (7.9 vs. 6.0 months; P=0.030), although there were no differences in the median OS (16.2 vs. 19.2 months; P>0.05). Positive PD-L1 status was associated with an improved PFS, whereas increased CRP, CEA and NLR levels were associated with poor outcomes. It was also suggested that PD-1/PD-L1 inhibitors may achieve more robust disease control, particularly in PD-L1-positive patients, using inflammatory parameters such as C-reactive protein (CRP), Carcinoembryonic antigen (CEA), and neutrophil-to-lymphocyte ratio (NLR) as potential prognostic values (106).
Immunotherapy, particularly in combination with chemotherapy, markedly prolongs survival for advanced KRAS-mutant NSCLC over chemotherapy alone; this is greater among patients with PD-L1-positive tumors or high tumor mutational burden. KRAS G12C is the predominant subtype and exhibits notable responsiveness to ICIs, providing evidence for ICIs-based chemoimmunotherapy as the optimal first-line treatment.
Co-mutations in STK11, TP53 and KEAP1 determine the characteristic phenotype of the TME and clinical prognosis for KRAS-mutant NSCLC. KRAS/TP53 tumors have an immune-inflamed profile with high TMB (10–12 mutations/Mb), high PD-L1 expression (≥50%) and dense CD8+ T-cell infiltration, which supports an improved response to ICIs (107). In a pooled analysis of 713 patients, KRAS/TP53 cases had significantly higher benefit from ICIs compared with chemotherapy, with an OS HR of 0.71 (95% CI, 0.55–0.92) and interaction HR of 0.53–0.56 favoring immunotherapy (108,109). Thus, KRAS/TP53 cases had significantly higher benefit from ICIs when compared with that of chemotherapy.
By contrast, KRAS/STK11 tumors are more consistently characterized by non-inflamed immune-cold with low PD-L1 expression (<10%), minimal CD8+ cell infiltration and a TMB of 4–6 mutations/Mb. Poor responses to ICIs and chemoimmunotherapy suggest the clinical manifestation of this (110). Numerous studies have shown that an STK11 co-mutation is associated with worse PFS and OS relative to KRAS-only or KRAS/TP53 disease, which often translates to the level of benefit derived from chemotherapy alone (111–113). These mutations also significantly correlated with worse PFS (4.2 months vs. 11.0 months; HR, 2.2; P < 0.01) and OS (9.8 months vs. NR; HR 2.6; P<0.01). Although there are conflicting data as to their effects on KRAS G12C inhibitor efficacy, with some studies showing reduced responses and others reporting similar outcomes regardless of STK11 status, the prognosis for patients with KRAS/STK11 co-mutations remains poor (114–116). This highlights the necessity for novel single and combination strategies in KRAS/STK11 disease, as standard chemo-immuno-oncology regimens offer limited benefit.
KRAS/KEAP1 tumors are the most aggressive subtype and possess infrequent broad resistance to chemotherapy, ICIs and KRAS-targeting agents. KEAP1/NFE2L2 mutations are associated with an inferior prognosis to platinum-pemetrexed and ICI regimens, as a pooled analysis reported an HR of 1.96 (95% CI, 1.33–2.92) for shorter survival (117). In the KRYSTAL-1 study, patients with KEAP1 co-mutation had a significantly shorter median PFS (4.1 months) and OS (5.4 months) when treated with adagrasib in comparison with their wild-type counterparts (9.9 and 19.0 months, respectively) (114). Functional validation also demonstrates that KEAP1 deletion significantly elevates IC50 of KRAS G12C inhibitors in H358 cells (118). Since KRAS/KEAP1 tumors are resistant to multiple drugs, KRAS/KEAP1 tumors warrant prioritization for clinical trials and experimental strategies beyond current standard therapies.
Together, this emphasizes the need for co-mutation profiling in treatment algorithms, as patients with KRAS/TP53 tumors may be more responsive to frontline ICI therapy; by contrast, patients with KRAS/STK11 and KRAS/KEAP1 tumors represent aggressive, immune-refractory subsets warranting alternative or combination strategies.
Although targeted medicines provide a notable response after treatment initiation, a large proportion of tumors eventually resist targeted medications because of intertumoral heterogeneity, where pre-existing resistance subclones survive treatment and expand, driving tumor evolution toward resistance (119). Secondary mutations in the KRAS gene also cause resistance to specific inhibitors of the G12C mutation. Mutations such as Y96D, A59T and G13D result in changes to the KRAS protein structure that prevent inhibitors, including sotorasib and adagrasib, from engaging with their target. Such mutations frequently occur in the SII-P, a key area for drug binding (120,121).
Concurrent mutations or alterations in driver oncogenes, including TP53 and STK11/LKB1, are also involved in resistance. It has been reported that tumors in which KRAS and STK11 are mutant are less sensitive to immune checkpoint blockade and KRAS-targeted agents, potentially due to the activation of compensatory survival signaling pathways (122,123). Second, distant activation of alternative signaling cascades exists that may mediate escape from KRAS inhibition. MET amplification is the most extensively known bypass mechanism, allowing downstream activation of the PI3K-AKT-mTOR signaling in the presence of KRAS inhibition (79,124). Other reported bypass routes include RET fusions and wild-type RAS activation (121,123). These alterations enable tumor cells to maintain proliferative signaling despite KRAS blockade. Aside from developing secondary mutations, reactivation of wild-type KRAS or other RAS family members can resurrect signaling downstream of the pathway. This reactivation undermines the efficacy of KRAS G12C inhibitors and leads to therapeutic failure (121).
Tumor cells can also acquire resistance via nongenetic means unrelated to new mutations. Cancer cells are kept alive under drug pressure by cellular plasticity, stress-response signaling and reprogramming of signal networks. For example, activation of integrin β4 and paxillin pathways, among others, can stimulate AKT signaling, contributing to a drug-tolerant persistent cell phenotype (125,126). Finally, modifications in the TME are significantly associated with therapy resistance. As such, hypoxia and TME nutrient deprivation can induce adaptive stress responses in cancer cells, leading to improved survival and resistance to targeted therapies (127). Patients receiving KRAS inhibitors also had worse prognoses when they had oxidative stress response-related alterations, such as KEAP1 mutations (71). Table III summarizes the various resistance mechanisms to KRAS-targeted therapies in NSCLC, including genetic factors, signaling pathways and TME-associated modifications associated with treatment resistance.
Multiplex screening via next-generation sequencing (NGS) is recommended for adequate patient selection in the European Society for Medical Oncology clinical practice guidelines (4,128). KRAS G12C inhibitors are a key treatment option for patients who are not eligible for standard first-line therapies. For example, co-mutations, including STK11 and KRAS G12C, are associated with inferior responses to ICIs, indicating that KRAS G12C inhibitors may be preferential as first-line treatment in those settings. Another potential strategy is to use chemotherapy or ICIs in combination with KRAS G12C inhibitors (129–131).
The development of resistance to conventional therapies is a notable problem that has led to a paradigm shift toward multitargeted therapies and improvements in drug delivery systems for improved patient prognostic results. KRAS mutations are considered actionable, particularly for G12C subtype for which direct inhibitors are now available. Personalization of therapy is essential because KRAS mutations are heterogeneous and exert diverse effects on treatment response (132–134). Additional investigation is warranted for KRAS G12D and G12V mutations, which represents 38% of KRAS-mutant LUAD cases, as these variants currently lack effective targeted therapies. Furthermore, emerging pan-KRAS inhibitors should be further explored as potential treatment strategies (135).
Emerging strategies for KRAS-mutant NSCLC are increasingly shaped by advanced bioinformatic and genomic tools that enable personalized therapy. Artificial intelligence (AI) is increasingly shaping the drug discovery process, notably with its rapid expansion induced by drug repurposing strategies. Machine learning model coupled with molecular docking has demonstrated that the FDA-approved drugs, including afatinib, neratinib and zanubrutinib, are likely to be effective against KRAS G12C. This method demonstrates how AI can identify novel therapeutic opportunities and is particularly useful for hard-to-target alleles such as KRAS G12D, where direct inhibitors remain limited (89).
Neoantigen prediction algorithms represent another avenue of research. Mutant KRAS peptides have demonstrated potent T-cell responses, which is relevant for STK11 co-mutated or high TMB tumors. Such subsets are frequently resistant to ICIs monotherapy, and the options of personalizing a vaccine or establishing adoptive T-cell therapy constitute a rational approach for these populations (136,137).
Single-cell RNA sequencing (scRNA-seq) has also been applied to KRAS-induced tumors, and a tumor-associated epidermal subpopulation that depended on the oncogene BMI-1 has been identified, which contributes to tumor growth and treatment resistance. Treatment approaches targeting this subpopulation with small-molecule inhibitor PTC596 inhibited tumor growth in preclinical models, demonstrating how scRNA-seq could detect resistant subclones and guide targeted therapy (138).
Finally, CRISPR-based therapies are also broadening treatment choices. High-fidelity Cas9 editing has shown that KRAS G12C and G12D mutations can be corrected in organoid or xenograft models with reduced tumor growth (139). In addition, genome-wide CRISPR screens have identified synthetic lethal relationships that might be exploited for rational combination strategies or resistance to KRAS inhibitors by targeting TEA domain transcription factor, CDK4 and Wee1-like protein kinase dependencies (140). Together, these approaches represent the innovative customized therapy for KRAS-mutation lung cancer; however, these CRISPR-based and synthetic-lethal strategies still face key limitations, such as off-target effects and uncertain clinical translation, additional research is required to overcome these limitations and improve patient outcomes.
KRAS has emerged as a viable target for therapeutic inhibition in lung cancer, overcoming the previous challenges associated with a limited understanding of KRAS biology and protein biochemistry. Technological progress has deepened understanding of RAS biology, including the discovery of novel druggable pockets and the usefulness of molecular subtyping. Among the current investigational treatments, small-molecule inhibitors targeting the KRAS G12C mutation have demonstrated promising results. However, molecular heterogeneity and resistance mechanisms remain concerns, thus requiring additional translational research. Combination strategies, such as immuno-targeted therapy, are increasingly popular to maximize KRAS inhibition. The current challenges and strategies in KRAS-mutated NSCLC research are overcoming resistance to ICIs and KRAS inhibitors, discovering predictive biomarkers, expanding targeted therapies to additional KRAS mutations and optimizing diagnostic accuracy using NGS analysis of patient plasma and tissue samples. Furthermore, worldwide accessibility to novel agents remains essential. These efforts will help to improve the quality of life and survival of patients with NSCLC.
Not applicable.
Funding: No funding was received.
Not applicable.
US prepared the original draft. HK reviewed the manuscript. JS, YZ and ZD reviewed and edited the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
El Osta B, Behera M, Kim S, Berry LD, Sica G, Pillai RN, Owonikoko TK, Kris MG, Johnson BE, Kwiatkowski DJ, et al: Characteristics and outcomes of patients with metastatic KRAS-mutant lung adenocarcinomas: The lung cancer mutation consortium experience. J Thorac Oncol. 14:876–889. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Moore AR, Rosenberg SC, McCormick F and Malek S: RAS-targeted therapies: is the undruggable drugged? Nat Rev Drug Discov. 19:533–552. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Simanshu DK, Nissley DV and McCormick F: RAS proteins and their regulators in human disease. Cell. 170:17–33. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Prior IA, Hood FE and Hartley JL: The frequency of ras mutations in cancer. Cancer Res. 80:2669–2974. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Dearden S, Stevens J, Wu YL and Blowers D: Mutation incidence and coincidence in non small-cell lung cancer: Meta-analyses by ethnicity and histology (mutMap). Ann Oncol. 24:2371–2376. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Yang H, Liang SQ, Schmid RA and Peng RW: New horizons in KRAS-mutant lung cancer: Dawn after darkness. Front Oncol. 9:9532019. View Article : Google Scholar : PubMed/NCBI | |
|
Naim MJ, Alam MJ, Ahmad S, Nawaz F, Shrivastava N, Sahu M and Alam O: Therapeutic journey of 2,4-thiazolidinediones as a versatile scaffold: An insight into structure activity relationship. Eur J Med Chem. 129:218–250. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Bos JL: Ras oncogenes in human cancer: A review. Cancer Res. 49:4682–4689. 1989.PubMed/NCBI | |
|
Prior IA, Lewis PD and Mattos C: A comprehensive survey of Ras mutations in cancer. Cancer Res. 72:2457–2467. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Yin G, Huang J, Petela J, Jiang H, Zhang Y, Gong S, Wu J, Liu B, Shi J and Gao Y: Targeting small GTPases: Emerging grasps on previously untamable targets, pioneered by KRAS. Signal Transduct Target Ther. 8:2122023. View Article : Google Scholar : PubMed/NCBI | |
|
Michalak DJ, Unger B, Lorimer E, Grishaev A, Williams CL, Heinrich F and Lösche M: Structural and biophysical properties of farnesylated KRas interacting with the chaperone SmgGDS-558. Biophys J. 121:3684–3697. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Herbst RS, Morgensztern D and Boshoff C: The biology and management of non-small cell lung cancer. Nature. 553:446–454. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Jeon H, Wang S, Song J, Gill H and Cheng H: Update 2025: Management of non-small-cell lung cancer. Lung. 203:532025. View Article : Google Scholar : PubMed/NCBI | |
|
Huang L, Guo Z, Wang F and Fu L: KRAS mutation: From undruggable to druggable in cancer. Signal Transduct Target Ther. 6:3862021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang F, Wang B, Wu M, Zhang L and Ji M: Current status of KRAS G12C inhibitors in NSCLC and the potential for combination with anti-PD-(L)1 therapy: A systematic review. Front Immunol. 16:15091732025. View Article : Google Scholar : PubMed/NCBI | |
|
Uniyal P, Kashyap VK, Behl T, Parashar D and Rawat R: KRAS mutations in cancer: Understanding signaling pathways to immune regulation and the potential of immunotherapy. Cancers (Basel). 17:7852025. View Article : Google Scholar : PubMed/NCBI | |
|
Parikh K, Banna G, Liu SV, Friedlaender A, Desai A, Subbiah V and Addeo A: Drugging KRAS: Current perspectives and state-of-art review. J Hematol Oncol. 15:1522022. View Article : Google Scholar : PubMed/NCBI | |
|
Hamarsheh S, Groß O, Brummer T and Zeiser R: Immune modulatory effects of oncogenic KRAS in cancer. Nat Commun. 11:54392020. View Article : Google Scholar : PubMed/NCBI | |
|
Ghimessy A, Radeczky P, Laszlo V, Hegedus B, Renyi-Vamos F, Fillinger J, Klepetko W, Lang C, Dome B and Megyesfalvi Z: Current therapy of KRAS-mutant lung cancer. Cancer Metastasis Rev. 39:1159–1177. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Eser M, Hekimoglu G, Yarar MH, Canbek S and Ozcelik M: KRAS G12C mutation in NSCLC in a small genetic center: Insights into sotorasib therapy response potential. Sci Rep. 14:265812024. View Article : Google Scholar : PubMed/NCBI | |
|
Bourne HR, Sanders DA and McCormick F: The GTPase superfamily: A conserved switch for diverse cell functions. Nature. 348:125–132. 1990. View Article : Google Scholar : PubMed/NCBI | |
|
Bourne HR, Sanders DA and McCormick F: The GTPase superfamily: Conserved structure and molecular mechanism. Nature. 349:117–127. 1991. View Article : Google Scholar : PubMed/NCBI | |
|
Scheffzek K, Ahmadian MR, Kabsch W, Wiesmüller L, Lautwein A, Schmitz F and Wittinghofer A: The Ras-RasGAP complex: Structural basis for GTPase activation and its loss in oncogenic Ras mutants. Science. 277:333–338. 1997. View Article : Google Scholar : PubMed/NCBI | |
|
Martin P, Leighl NB, Tsao MS and Shepherd FA: KRAS mutations as prognostic and predictive markers in non-small cell lung cancer. J Thorac Oncol. 8:530–542. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Adderley H, Blackhall FH and Lindsay CR: KRAS-mutant non-small cell lung cancer: Converging small molecules and immune checkpoint inhibition. EBioMedicine. 41:711–716. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Matikas A, Mistriotis D, Georgoulias V and Kotsakis A: Targeting KRAS mutated non-small cell lung cancer: A history of failures and a future of hope for a diverse entity. Crit Rev Oncol Hematol. 110:1–12. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Shepherd FA, Domerg C, Hainaut P, Jänne PA, Pignon JP, Graziano S, Douillard JY, Brambilla E, Le Chevalier T, Seymour L, et al: Pooled analysis of the prognostic and predictive effects of KRAS mutation status and KRAS mutation subtype in early-stage resected non-small-cell lung cancer in four trials of adjuvant chemotherapy. J Clin Oncol. 31:2173–2181. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Riely GJ, Kris MG, Rosenbaum D, Marks J, Li A, Chitale DA, Nafa K, Riedel ER, Hsu M, Pao W, et al: Frequency and distinctive spectrum of KRAS mutations in never smokers with lung adenocarcinoma. Clin Cancer Res. 14:5731–5734. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Redig AJ, Chambers ES, Lydon CA, Dahlberg SE, Alden RS and Janne PA: Genomic complexity in KRAS mutant non-small cell lung cancer (NSCLC) from never/light-smokers v smokers. J Clin Oncol. 34 (15 Suppl):S90872016. View Article : Google Scholar | |
|
Ferrer I, Zugazagoitia J, Herbertz S, John W, Paz-Ares L and Schmid-Bindert G: KRAS-mutant non-small cell lung cancer: From biology to therapy. Lung Cancer. 124:53–64. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Li S, Liu S, Deng J, Akbay EA, Hai J, Ambrogio C, Zhang L, Zhou F, Jenkins RW, Adeegbe DO, et al: Assessing therapeutic efficacy of MEK inhibition in a KRASG12C-driven mouse model of lung cancer. Clin Cancer Res. 24:4854–4864. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Garassino MC, Marabese M, Rusconi P, Rulli E, Martelli O, Farina G, Scanni A and Broggini M: Different types of K-Ras mutations could affect drug sensitivity and tumour behaviour in non-small-cell lung cancer. Ann Oncol. 22:235–237. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Imielinski M, Berger AH, Hammerman PS, Hernandez B, Pugh TJ, Hodis E, Cho J, Suh J, Capelletti M, Sivachenko A, et al: Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell. 150:1107–1120. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Lee B, Lee T, Lee SH, Choi YL and Han J: Clinicopathologic characteristics of EGFR, KRAS, and ALK alterations in 6,595 lung cancers. Oncotarget. 7:23874–23884. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Li S, Li L, Zhu Y, Huang C, Qin Y, Liu H, Ren-Heidenreich L, Shi B, Ren H, Chu X, et al: Coexistence of EGFR with KRAS, or BRAF, or PIK3CA somatic mutations in lung cancer: A comprehensive mutation profiling from 5125 Chinese cohorts. Br J Cancer. 110:2812–2820. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ Jr, Wu YL and Paz-Ares L: Lung cancer: Current therapies and new targeted treatments. Lancet. 389:299–311. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Skoulidis F and Heymach JV: Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat Rev Cancer. 19:495–509. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Christensen JG, Olson P, Briere T, Wiel C and Bergo MO: Targeting Krasg12c-mutant cancer with a mutation-specific inhibitor. J Intern Med. 288:183–191. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Haidar M and Jacquemin P: Past and future strategies to inhibit membrane localization of the KRAS oncogene. Int J Mol Sci. 22:131932021. View Article : Google Scholar : PubMed/NCBI | |
|
Tomasini P, Walia P, Labbe C, Jao K and Leighl NB: Targeting the KRAS pathway in non-small cell lung cancer. Oncologist. 21:1450–1460. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Gysin S, Salt M, Young A and McCormick F: Therapeutic strategies for targeting ras proteins. Genes Cancer. 2:359–372. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Spoerner M, Herrmann C, Vetter IR, Kalbitzer HR and Wittinghofer A: Dynamic properties of the Ras switch I region and its importance for binding to effectors. Proc Natl Acad Sci USA. 98:4944–4949. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Buhrman G, O'Connor C, Zerbe B, Kearney BM, Napoleon R, Kovrigina EA, Vajda S, Kozakov D, Kovrigin EL and Mattos C: Analysis of binding site hot spots on the surface of Ras GTPase. J Mol Biol. 413:773–789. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Ostrem JM, Peters U, Sos ML, Wells JA and Shokat KM: K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 503:548–551. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Shima F, Matsumoto S, Yoshikawa Y, Kawamura T, Isa M and Kataoka T: Current status of the development of Ras inhibitors. J Biochem. 158:91–99. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Berndt N, Hamilton AD and Sebti SM: Targeting protein prenylation for cancer therapy. Nat Rev Cancer. 11:775–791. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Roberts PJ and Der CJ: Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 26:3291–3310. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Zimmermann G, Papke B, Ismail S, Vartak N, Chandra A, Hoffmann M, Hahn SA, Triola G, Wittinghofer A, Bastiaens PI and Waldmann H: Small molecule inhibition of the KRAS-PDEδ interaction impairs oncogenic KRAS signalling. Nature. 497:638–642. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Riely GJ, Johnson ML, Medina C, Rizvi NA, Miller VA, Kris MG, Pietanza MC, Azzoli CG, Krug LM, Pao W and Ginsberg MS: A phase II trial of Salirasib in patients with lung adenocarcinomas with KRAS mutations. J Thorac Oncol. 6:1435–1437. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Whyte DB, Kirschmeier P, Hockenberry TN, Nunez-Oliva I, James L, Catino JJ, Bishop WR and Pai JK: K- and N-Ras are geranylgeranylated in cells treated with farnesyl protein transferase inhibitors. J Biol Chem. 272:14459–14464. 1997. View Article : Google Scholar : PubMed/NCBI | |
|
Shima F, Yoshikawa Y, Ye M, Araki M, Matsumoto S, Liao J, Hu L, Sugimoto T, Ijiri Y, Takeda A, et al: In silico discovery of small-molecule Ras inhibitors that display antitumor activity by blocking the Ras-effector interaction. Proc Natl Acad Sci USA. 110:8182–8187. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Gerber DE, Camidge DR, Morgensztern D, Cetnar J, Kelly RJ, Ramalingam SS, Spigel DR, Jeong W, Scaglioni PP, Zhang S, et al: Phase 2 study of the focal adhesion kinase inhibitor defactinib (VS-6063) in previously treated advanced KRAS mutant non-small cell lung cancer. Lung Cancer. 139:60–67. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Cox AD, Fesik SW, Kimmelman AC, Luo J and Der CJ: Drugging the undruggable RAS: Mission possible? Nat Rev Drug Discov. 13:828–851. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Wang M and Casey PJ: Protein prenylation: Unique fats make their mark on biology. Nat Rev Mol Cell Biol. 17:110–122. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Kazi A, Xiang S, Yang H, Chen L, Kennedy P, Ayaz M, Fletcher S, Cummings C, Lawrence HR, Beato F, et al: Dual farnesyl and geranylgeranyl transferase inhibitor thwarts mutant KRAS-driven patient-derived pancreatic tumors. Clin Cancer Res. 25:5984–5996. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Tao MH, Chen S, Freudenheim JL, Cauley JA, Johnson KC, Mai X, Sarto GE, Wakelee H, Boffetta P and Wactawski-Wende J: Oral bisphosphonate use and lung cancer incidence among postmenopausal women. Ann Oncol. 29:1476–1485. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Nagao S, Hattori N, Fujitaka K, Iwamoto H, Ohshimo S, Kanehara M, Ishikawa N, Haruta Y, Murai H and Kohno N: Regression of a primary pulmonary adenocarcinoma after zoledronic acid monotherapy. Hiroshima J Med Sci. 60:7–9. 2011.PubMed/NCBI | |
|
Kitai H, Ebi H, Tomida S, Floros KV, Kotani H, Adachi Y, Oizumi S, Nishimura M, Faber AC and Yano S: Epithelial-to-mesenchymal transition defines feedback activation of receptor tyrosine kinase signaling induced by MEK inhibition in KRAS-mutant lung cancer. Cancer Discov. 6:754–769. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Johnson C, Burkhart DL and Haigis KM: Classification of KRAS-activating mutations and the implications for therapeutic intervention. Cancer Discov. 12:913–923. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Hunter JC, Manandhar A, Carrasco MA, Gurbani D, Gondi S and Westover KD: Biochemical and structural analysis of common cancer-associated KRAS mutations. Mol Cancer Res. 13:1325–1335. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Ostrem JML and Shokat KM: Direct small-molecule inhibitors of KRAS: From structural insights to mechanism-based design. Nat Rev Drug Discov. 15:771–785. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Canon J, Rex K, Saiki AY, Mohr C, Cooke K, Bagal D, Gaida K, Holt T, Knutson CG, Koppada N, et al: The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 575:217–223. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Pandey D, Chauhan SC, Kashyap VK and Roy KK: Structural insights into small-molecule KRAS inhibitors for targeting KRAS mutant cancers. Eur J Med Chem. 277:1167712024. View Article : Google Scholar : PubMed/NCBI | |
|
Warnecke B and Nagasaka M: Adagrasib in the treatment of KRAS p.G12C positive advanced NSCLC: Design, development and place in therapy. Drug Des Devel Ther. 18:5673–5683. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Li Z, Song Z, Zhao Y, Wang P, Jiang L, Gong Y, Zhou J, Jian H, Dong X, Zhuang W, et al: D-1553 (Garsorasib), a potent and selective inhibitor of KRASG12C in patients with NSCLC: Phase 1 study results. J Thorac Oncol. 18:940–951. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
D'Alessio-Sands L, Gaynier J, Michel-Milian V, Agbowuro AA and Brackett CM: Current strategies and future dimensions in the development of KRAS inhibitors for targeted anticancer therapy. Drug Dev Res. 86:e700422025. View Article : Google Scholar : PubMed/NCBI | |
|
Deming DA: Development of KRAS inhibitors and their role for metastatic colorectal cancer. J Natl Compr Canc Netw. 23:e2470672025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao T and Liang SH: Novel tricyclic KRAS inhibitors for the treatment of cancer. ACS Med Chem Lett. 16:178–179. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Hong DS, Fakih MG, Strickler JH, Desai J, Durm GA, Shapiro GI, Falchook GS, Price TJ, Sacher A, Denlinger CS, et al: KRASG12C inhibition with sotorasib in advanced solid tumors. N Engl J Med. 383:1207–1217. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
No authors listed. FDA approves first KRAS inhibitor: Sotorasib. Cancer Discov. 11:OF42021. View Article : Google Scholar | |
|
Skoulidis F, Li BT, Dy GK, Price TJ, Falchook GS, Wolf J, Italiano A, Schuler M, Borghaei H, Barlesi F, et al: Sotorasib for lung cancers with KRAS p.G12C mutation. N Engl J Med. 384:2371–2381. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Jänne PA, Riely GJ, Gadgeel SM, Heist RS, Ou SI, Pacheco JM, Johnson ML, Sabari JK, Leventakos K, Yau E, et al: Adagrasib in non-small-cell lung cancer harboring a KRASG12C mutation. N Engl J Med. 387:120–131. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
de Langen AJ, Johnson ML, Mazieres J, Dingemans AC, Mountzios G, Pless M, Wolf J, Schuler M, Lena H, Skoulidis F, et al: Sotorasib versus docetaxel for previously treated non-small-cell lung cancer with KRASG12C mutation: A randomised, open-label, phase 3 trial. Lancet. 401:733–746. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wislez M, Mascaux C, Cadranel J, Thomas QD, Ricordel C, Swalduz A, Pichon E, Veillon R, Gounant V, Rousseau-Bussac G, et al: Real-world effectiveness and tolerability of sotorasib in patients with KRAS G12C-mutated metastatic non-small cell lung cancer: The IFCT-2102 lung KG12Ci study. Eur J Cancer. 219:1153012025. View Article : Google Scholar : PubMed/NCBI | |
|
Ou SHI, Jänne PA, Leal TA, Rybkin II, Sabari JK, Barve MA, Bazhenova L, Johnson ML, Velastegui KL, Cilliers C, et al: First-in-human phase I/IB dose-finding study of adagrasib (MRTX849) in patients with advanced KRASG12C solid tumors (KRYSTAL-1). J Clin Oncol. 40:2530–2538. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Mok TSK, Yao W, Duruisseaux M, Doucet L, Azkárate Martínez A, Gregorc V, Juan-Vidal O, Lu S, De Bondt C, de Marinis F, et al: KRYSTAL-12: Phase 3 study of adagrasib versus docetaxel in patients with previously treated advanced/metastatic non-small cell lung cancer (NSCLC) harboring a KRASG12C mutation. J Clin Oncol. 42 (Suppl 17):LBA85092024. View Article : Google Scholar | |
|
Li Z, Dang X, Huang D, Jin S, Li W, Shi J, Wang X, Zhang Y, Song Z, Zhang J, et al: Abstract CT246: Open-label, single-arm, multicenter, phase 2 trial of garsorasib in KRAS G12C-mutated non-small-cell lung cancer. Cancer Res. 84 (7 Suppl):CT2462024. View Article : Google Scholar | |
|
Li Z, Dang X, Huang D, Jin S, Li W, Shi J, Wang X, Zhang Y, Song Z, Zhang J, et al: Garsorasib in patients with KRASG12C-mutated non-small-cell lung cancer in China: An open-label, multicentre, single-arm, phase 2 trial. Lancet Respir Med. 12:589–598. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Awad MM, Liu S, Rybkin II, Arbour KC, Dilly J, Zhu VW, Johnson ML, Heist RS, Patil T, Riely GJ, et al: Acquired resistance to KRASG12C inhibition in cancer. N Engl J Med. 384:2382–2393. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Cox AD and Der CJ: ‘Undruggable KRAS’: Druggable after all. Genes Dev. 39:132–162. 2025.PubMed/NCBI | |
|
Maruyama K, Shimizu Y, Nomura Y, Oh-Hara T, Takahashi Y, Nagayama S, Fujita N and Katayama R: Mechanisms of KRAS inhibitor resistance in KRAS-mutant colorectal cancer harboring Her2 amplification and aberrant KRAS localization. NPJ Precis Oncol. 9:42025. View Article : Google Scholar : PubMed/NCBI | |
|
Mugarza E, van Maldegem F, Boumelha J, Moore C, Rana S, Llorian Sopena M, East P, Ambler R, Anastasiou P, Romero-Clavijo P, et al: Therapeutic KRASG12C inhibition drives effective interferon-mediated antitumor immunity in immunogenic lung cancers. Sci Adv. 8:eabm87802022. View Article : Google Scholar : PubMed/NCBI | |
|
Garassino MC, Jänne PA, Barlesi F, Spira AI, Mok TSK, Hochmair M, O'Byrne KJ, Felip E, Kim SW, Cappuzzo F, et al: 1394TiP KRYSTAL-7: A phase III study of first-line adagrasib plus pembrolizumab versus pembrolizumab alone in patients with advanced NSCLC with KRASG12C mutation mutation. Ann Oncol. 35 (Suppl 2):S872–S873. 2024. View Article : Google Scholar | |
|
Li BT, Falchook GS, Durm GA, Burns TF, Skoulidis F, Ramalingam SS, Spira A, Bestvina CM, Goldberg SB, Veluswamy R, et al: OA03.06 CodeBreaK 100/101: First report of safety/efficacy of sotorasib in combination with pembrolizumab or atezolizumab in advanced KRAS p.G12C NSCLC. J Thorac Oncol. 17 (Suppl):S10–S11. 2022. View Article : Google Scholar | |
|
Ernst SM, Hofman MM, van der Horst TE, Paats MS, Heijboer FWJ, Aerts JGJV, Dumoulin DW, Cornelissen R, von der Thüsen JH, de Bruijn P, et al: Hepatotoxicity in patients with non-small cell lung cancer treated with sotorasib after prior immunotherapy: A comprehensive clinical and pharmacokinetic analysis. EBioMedicine. 102:1050742024. View Article : Google Scholar : PubMed/NCBI | |
|
Chour A, Basse C, Lebossé F, Bonte PE, Girard N and Duruisseaux M: Management of sotorasib-related adverse events and hepatotoxicities following anti-PD-(L)1 therapy: Experience with sotorasib in two French anti-cancer centers and practical guidance proposal. Lung Cancer. 191:1077892024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Johnson M, Barve M, Bazhenova L, McCarthy M, Schwartz R, Horvath-Walsh E, Velastegui K, Qian C and Spira A: Practical guidance for the management of adverse events in patients with KRASG12C-mutated non-small cell lung cancer receiving adagrasib. Oncologist. 28:287–296. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Mina SA, Shanshal M, Leventakos K and Parikh K: Emerging targeted therapies in non-small-cell lung cancer (NSCLC). Cancers (Basel). 17:3532025. View Article : Google Scholar : PubMed/NCBI | |
|
Fancelli S, Petroni G, Pillozzi S and Antonuzzo L: Unconventional strategy could be the future: From target to KRAS broad range treatment. Heliyon. 10:e297392024. View Article : Google Scholar : PubMed/NCBI | |
|
Masfarre Pinto L, Parreira ASDFM, Castro Unanua N, Rocha P, Morilla Ruiz I, Teijeira Sanchez L, Clave S, Navarro Gorro N, Bach Mora R, Sánchez I, et al: 1408P Differences on immune biomarkers between KRAS G12C and KRAS non-G12C mutated non-small cell lung cancer. Ann Oncol. 34 (Suppl2):S805–S806. 2023. View Article : Google Scholar | |
|
Zhou C, Li C, Luo L, Li X, Jia K, He N, Mao S, Wang W, Shao C, Liu X, et al: Anti-tumor efficacy of HRS-4642 and its potential combination with proteasome inhibition in KRAS G12D-mutant cancer. Cancer Cell. 42:1286–1300.e8. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Zhao J and Li Y: New exploration of KRASG12D inhibitors and the mechanisms of resistance. Exp Hematol Oncol. 14:392025. View Article : Google Scholar : PubMed/NCBI | |
|
Arbour KC, Rizvi H, Plodkowski AJ, Hellmann MD, Knezevic A, Heller G, Yu HA, Ladanyi M, Kris MG, Arcila ME, et al: Treatment outcomes and clinical characteristics of patients with KRAS-G12C-mutant non-small cell lung cancer. Clin Cancer Res. 27:2209–2215. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Bironzo P, Cani M, Jacobs F, Napoli VM, Listì A, Passiglia F, Righi L, Di Maio M, Novello S and Scagliotti GV: Real-world retrospective study of KRAS mutations in advanced non-small cell lung cancer in the era of immunotherapy. Cancer. 129:1662–1671. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Rodenhuis S, van de Wetering ML, Mooi WJ, Evers SG, van Zandwijk N and Bos JL: Mutational activation of the K-ras oncogene. A possible pathogenetic factor in adenocarcinoma of the lung. N Engl J Med. 317:929–935. 1987. View Article : Google Scholar : PubMed/NCBI | |
|
Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, Stephens PJ, Daniels GA and Kurzrock R: Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 16:2598–2608. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Mazieres J, Drilon A, Lusque A, Mhanna L, Cortot AB, Mezquita L, Thai AA, Mascaux C, Couraud S, Veillon R, et al: Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: Results from the IMMUNOTARGET registry. Ann Oncol. 30:1321–1328. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Rothschild SI: KRAS and immune checkpoint inhibitors-serendipity raising expectations. J Thorac Oncol. 14:951–954. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Torralvo J, Friedlaender A, Achard V and Addeo A: The activity of immune checkpoint inhibition in KRAS mutated non-small cell lung cancer: A single centre experience. Cancer Genomics Proteomics. 16:577–582. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Mok TSK, Lopes G, Cho BC, Kowalski DM, Kasahara K, Wu YL, de Castro G Jr, Turna HZ, Cristescu R, Aurora-Garg D, et al: Associations of tissue tumor mutational burden and mutational status with clinical outcomes in KEYNOTE-042: Pembrolizumab versus chemotherapy for advanced PD-L1-positive NSCLC. Ann Oncol. 34:377–388. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Gadgeel S, Rodriguez-Abreu D, Felip E, Esteban E, Speranza G, Reck M, Hui R, Boyer M, Garon EB, Horinouchi H, et al: LBA5-KRAS mutational status and efficacy in KEYNOTE-189: Pembrolizumab (pembro) plus chemotherapy (chemo) vs placebo plus chemo as first-line therapy for metastatic non-squamous NSCLC. Ann Oncol. 30 (Suppl 11):xi64–xi65. 2019. View Article : Google Scholar | |
|
Peng L, Guo J, Kong L, Huang Y, Tang N, Zhang J, Wang M, He X, Li Z, Peng Y, et al: Efficacy of immunotherapy in KRAS-mutant advanced NSCLC: A real-world study in a Chinese population. Front Oncol. 12:10707612023. View Article : Google Scholar : PubMed/NCBI | |
|
Urtecho SB, Provenzano L, Spagnoletti A, Bottiglieri A, Pircher C, Massa G, Sposetti C, Proto C, Brambilla M, Occhipinti M, et al: Decoding KRAS mutation in non-small cell lung cancer patients receiving immunotherapy: A retrospective institutional comparison and literature review. Lung Cancer. 199:1080512025. View Article : Google Scholar : PubMed/NCBI | |
|
Koyama S, Akbay EA, Li YY, Aref AR, Skoulidis F, Herter-Sprie GS, Buczkowski KA, Liu Y, Awad MM, Denning WL, et al: STK11/LKB1 deficiency promotes neutrophil recruitment and proinflammatory cytokine production to suppress T-cell Activity in the lung tumor microenvironment. Cancer Res. 76:999–1008. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Knetki-Wróblewska M, Wojas-Krawczyk K, Krawczyk P and Krzakowski M: Emerging insights into STK11, KEAP1 and KRAS mutations: Implications for immunotherapy in patients with advanced non-small cell lung cancer. Transl Lung Cancer Res. 13:3718–3730. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wei K, Sun T, Feng X, Chen Y, Liu Q and Tang H: PD-1/L1 immune checkpoint inhibitors for KRAS-mutant non-small cell lung cancer: A multicenter retrospective real-world study. BMC Cancer. 25:4442025. View Article : Google Scholar : PubMed/NCBI | |
|
Dong ZY and Wu YL: What is the significance of TP53 and KRAS mutation for immunotherapy in non-small cell lung cancer? Transl Cancer Res. 6 (Suppl 6):S1115–S1117. 2017. View Article : Google Scholar | |
|
Budczies J, Romanovsky E, Kirchner M, Neumann O, Blasi M, Schnorbach J, Shah R, Bozorgmehr F, Savai R, Stiewe T, et al: KRAS and TP53 co-mutation predicts benefit of immune checkpoint blockade in lung adenocarcinoma. Br J Cancer. 131:524–533. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Bischoff P, Reck M, Overbeck T, Christopoulos P, Rittmeyer A, Lüders H, Kollmeier J, Kulhavy J, Kemper M, Reinmuth N, et al: Outcome of first-line treatment with pembrolizumab according to KRAS/TP53 mutational status for nonsquamous programmed death-ligand 1-high (≥50%) NSCLC in the German national network genomic medicine lung cancer. J Thorac Oncol. 19:803–817. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Skoulidis F, Goldberg ME, Greenawalt DM, Hellmann MD, Awad MM, Gainor JF, Schrock AB, Hartmaier RJ, Trabucco SE, Gay L, et al: STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov. 8:822–835. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Sun L, Handorf EA, Zhou Y, Borghaei H, Aggarwal C and Bauman J: Outcomes in patients treated with frontline immune checkpoint inhibition (ICI) for advanced NSCLC with KRAS mutations and STK11/KEAP1 comutations across PD-L1 levels. Lung Cancer. 190:1075102024. View Article : Google Scholar : PubMed/NCBI | |
|
Ricciuti B and Garassino MC: Precision immunotherapy for STK11/KEAP1-mutant NSCLC. J Thorac Oncol. 19:877–882. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
De Giglio A, De Biase D, Favorito V, Maloberti T, Di Federico A, Zacchini F, Venturi G, Parisi C, Gustavo Dall'Olio F, Ricciotti I, et al: STK11 mutations correlate with poor prognosis for advanced NSCLC treated with first-line immunotherapy or chemo-immunotherapy according to KRAS, TP53, KEAP1, and SMARCA4 status. Lung Cancer. 199:1080582025. View Article : Google Scholar : PubMed/NCBI | |
|
Negrao MV, Paula AG, Molkentine D, Hover L, Nilsson M, Vokes N, Engstrom L, Calinisan A, Briere DM, Waters L, et al: Impact of co-mutations and transcriptional signatures in non-small cell lung cancer patients treated with adagrasib in the KRYSTAL-1 trial. Clin Cancer Res. 31:1069–1081. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Olsen A, Lebedeva A, Nosova P, Nikulin V, Sharova M, Ignatova E, Mileyko V and Ivanov M: Impact of the STK11/KRAS co-mutation on the response to immunotherapy in a real-world pan-cancer cohort. Tumori. 110:146–152. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Sisca L, Cascetta P, Aijaz A, Catania C, Facchinetti F, Naqash AR, Ricciuti B and Cortellini A: KRAS and STK11 co-mutations in resectable non-small cell lung cancer: Enduring prognostic value and impaired immunotherapy response. Transl Lung Cancer Res. 14:2374–2382. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Hellyer JA, Padda SK, Diehn M and Wakelee HA: Clinical implications of KEAP1-NFE2L2 mutations in NSCLC. J Thorac Oncol. 16:395–403. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Tian L, Liu C, Zheng S, Shi H, Wei F, Jiang W, Dong Y, Xu H, Yin E, Sun N, et al: KEAP1 mutations as key crucial prognostic biomarkers for resistance to KRAS-G12C inhibitors. J Transl Med. 23:822025. View Article : Google Scholar : PubMed/NCBI | |
|
Jänne PA, van den Heuvel MM, Barlesi F, Cobo M, Mazieres J, Crinò L, Orlov S, Blackhall F, Wolf J, Garrido P, et al: Selumetinib plus docetaxel compared with docetaxel alone and progression-free survival in patients with KRAS-mutant advanced non-small cell lung cancer. JAMA. 317:1844–1853. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Sreter KB, Catarata MJ, von Laffert M and Frille A: Resistance to KRAS inhibition in advanced non-small cell lung cancer. Front Oncol. 14:13578982024. View Article : Google Scholar : PubMed/NCBI | |
|
Blaquier JB, Cardona AF and Recondo G: Resistance to KRASG12C inhibitors in non-small cell lung cancer. Front Oncol. 11:7875852021. View Article : Google Scholar : PubMed/NCBI | |
|
Cascetta P, Marinello A, Lazzari C, Gregorc V, Planchard D, Bianco R, Normanno N and Morabito A: KRAS in NSCLC: State of the art and future perspectives. Cancers (Basel). 14:54302022. View Article : Google Scholar : PubMed/NCBI | |
|
Chour A, Toffart AC, Berton E and Duruisseaux M: Mechanisms of resistance to KRASG12C inhibitors in KRASG12C-mutated non-small cell lung cancer. Front Oncol. 14:13287282024. View Article : Google Scholar : PubMed/NCBI | |
|
Tanaka N, Lin JJ, Li C, Ryan MB, Zhang J, Kiedrowski LA, Michel AG, Syed MU, Fella KA, Sakhi M, et al: Clinical acquired resistance to KRASG12C inhibition through a novel KRAS switch-II pocket mutation and polyclonal alterations converging on RAS-MAPK reactivation. Cancer Discov. 11:1913–1922. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Mohanty A, Nam A, Srivastava S, Jones J, Lomenick B, Singhal SS, Guo L, Cho H, Li A, Behal A, et al: Acquired resistance to KRAS G12C small-molecule inhibitors via genetic/nongenetic mechanisms in lung cancer. Sci Adv. 9:eade38162023. View Article : Google Scholar : PubMed/NCBI | |
|
Akhave NS, Biter AB and Hong DS: Mechanisms of resistance to KRASG12C-targeted therapy. Cancer Discov. 11:1345–1352. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Labrie M, Brugge JS, Mills GB and Zervantonakis IK: Therapy resistance: Opportunities created by adaptive responses to targeted therapies in cancer. Nat Rev Cancer. 22:323–339. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Planchard D, Popat S, Kerr K, Novello S, Smit EF, Faivre-Finn C, Mok TS, Reck M, Van Schil PE, Hellmann MD, et al: Metastatic non-small cell lung cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 29 (Suppl 4):iv192–iv237. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Seo KY, Jelinsky SA and Loechler EL: Factors that influence the mutagenic patterns of DNA adducts from chemical carcinogens. Mutat Res. 463:215–246. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Goulding RE, Chenoweth M, Carter GC, Boye ME, Sheffield KM, John WJ, Leusch MS, Muehlenbein CE, Li L, Jen MH, et al: KRAS mutation as a prognostic factor and predictive factor in advanced/metastatic non-small cell lung cancer: A systematic literature review and meta-analysis. Cancer Treat Res Commun. 24:1002002020.PubMed/NCBI | |
|
Awad MM, Gadgeel SM, Borghaei H, Patnaik A, Yang JC, Powell SF, Gentzler RD, Martins RG, Stevenson JP, Altan M, et al: Long-term overall survival from KEYNOTE-021 cohort G: Pemetrexed and carboplatin with or without pembrolizumab as first-line therapy for advanced nonsquamous NSCLC. J Thorac Oncol. 16:162–168. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Feng W: Advancements in targeted therapies based on the EGFR, and KRAS genetic mutations in lung cancer. Theor Nat Sci. 67:22–29. 2024. View Article : Google Scholar | |
|
Harris E and Thawani R: Current perspectives of KRAS in non-small cell lung cancer. Curr Probl Cancer. 51:1011062024. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang Z: A review of KRAS inhibitors and potential improvement in the future. Transact Mater Biotechnol Life Sci. 7:322–327. 2024. | |
|
Ricciuti B, Leonardi GC, Metro G, Grignani F, Paglialunga L, Bellezza G, Baglivo S, Mencaroni C, Baldi A, Zicari D and Crinò L: Targeting the KRAS variant for treatment of non-small cell lung cancer: Potential therapeutic applications. Expert Rev Respir Med. 10:53–68. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Rosner S, Connor S, Sanber K, Zahurak M, Zhang T, Gurumurthy I, Zeng Z, Presson B, Singh D, Rayes R, et al: Divergent clinical and immunologic outcomes based on STK11 co-mutation status in resectable KRAS-mutant lung cancers following neoadjuvant immune checkpoint blockade. Clin Cancer Res. 31:339–351. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Choi J, Goulding SP, Conn BP, McGann CD, Dietze JL, Kohler J, Lenkala D, Boudot A, Rothenberg DA, Turcott PJ, et al: Systematic discovery and validation of T cell targets directed against oncogenic KRAS mutations. Cell Rep Methods. 1:1000842021. View Article : Google Scholar : PubMed/NCBI | |
|
Gondal MN, Cieslik M and Chinnaiyan AM: Integrated cancer cell-specific single-cell RNA-seq datasets of immune checkpoint blockade-treated patients. Sci Data. 12:1392025. View Article : Google Scholar : PubMed/NCBI | |
|
Álvarez-Pérez JC, Sanjuán-Hidalgo J, Arenas AM, Hernández-Navas I, Benitez-Cantos MS, Andrades A, Calabuig-Fariñas S, Jantus-Lewintre E, Paz-Ares L, Ferrer I and Medina PP: High-fidelity Cas9-mediated targeting of KRAS driver mutations restrains lung cancer in preclinical models. Nat Commun. 16:70802025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Bui TA, Yang X, Hutvagner G and Deng W: Advancements in gene therapies targeting mutant KRAS in cancers. Cancer Metastasis Rev. 44:242025. View Article : Google Scholar : PubMed/NCBI | |
|
Mok TSK, Wu YL, Kudaba I, Kowalski DM, Cho BC, Turna HZ, Castro G Jr, Srimuninnimit V, Laktionov KK, Bondarenko I, et al: Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): A randomised, open-label, controlled, phase 3 trial. Lancet. 393:1819–1830. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, Chow LQ, Vokes EE, Felip E, Holgado E, et al: Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 373:1627–1639. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Rodríguez-Abreu D, Powell SF, Hochmair MJ, Gadgeel S, Esteban E, Felip E, Speranza G, De Angelis F, Dómine M, Cheng SY, et al: Pemetrexed plus platinum with or without pembrolizumab in patients with previously untreated metastatic nonsquamous NSCLC: Protocol-specified final analysis from KEYNOTE-189. Ann Oncol. 32:881–895. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, Rodríguez-Abreu D, Moro-Sibilot D, Thomas CA, Barlesi F, et al: Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 378:2288–2301. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Garassino MC, Theelen WSME, Jänne PA, Boom LN, De Marinis F, Dómine Gómez M, Nadal E, Paz-Ares L, Gu W, Felip E, et al: 5MO: First-line adagrasib (ADA) with pembrolizumab (PEMBRO) in patients (pts) with advanced/metastatic KRASG12C-mutated non-small cell lung cancer (NSCLC) and PD-L1 ≥50% from the phase II portion of KRYSTAL-7. J Thorac Oncol. 20 (Suppl 1):S8–S10. 2025. View Article : Google Scholar | |
|
Neal J, Pavlakis N, Kim SW, Goto Y, Lim SM, Mountzios G, Fountzilas E, Mochalova A, Christoph DC, Bearz A, et al: CONTACT-01: A randomized phase III trial of atezolizumab + cabozantinib versus docetaxel for metastatic non-small cell lung cancer after a checkpoint inhibitor and chemotherapy. J Clin Oncol. 42:2393–2403. 2024. View Article : Google Scholar : PubMed/NCBI |