Next Article in Journal
Online Crowdfunding for Urologic Cancer Care
Next Article in Special Issue
In Silico Strategies for Designing of Peptide Inhibitors of Oncogenic K-Ras G12V Mutant: Inhibiting Cancer Growth and Proliferation
Previous Article in Journal
Lynch Syndrome: From Carcinogenesis to Prevention Interventions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Biology and Therapeutic Perspectives for K-Ras Mutant Non-Small Cell Lung Cancers

1
Oncology Institute of Southern Switzerland (IOSI), Ospedale Regionale di Bellinzona e Valli, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland
2
Laboratory of Molecular Pathology, Institute of Pathology, Ente Ospedaliero Cantonale (EOC), 6601 Locarno, Switzerland
3
Oncology Institute of Southern Switzerland (IOSI), Ospedale Regionale di Lugano—Civico e Italiano, Ente Ospedaliero Cantonale (EOC), 6900 Lugano, Switzerland
4
Oncology Institute of Southern Switzerland (IOSI), Ospedale Regionale di Locarno, Ente Ospedaliero Cantonale (EOC), 6600 Locarno, Switzerland
*
Author to whom correspondence should be addressed.
These authors contributed equally to the work.
These authors contributed equally to the work.
Cancers 2022, 14(17), 4103; https://doi.org/10.3390/cancers14174103
Submission received: 15 July 2022 / Revised: 22 August 2022 / Accepted: 22 August 2022 / Published: 24 August 2022

Abstract

:

Simple Summary

Mutations in the Kirsten rat sarcoma viral oncogene homolog (KRAS) gene are the most common alterations in non-small cell lung cancer (NSCLC); they are generally linked to smoking history and are typical of the lung adenocarcinoma (AC) subtype. The clinical relevance of KRAS mutations in NSCLC was generally low or null until a few years ago. What is now emerging is that KRAS-mutant lung AC patients are generally associated with poorer survival. However, the approval of Sotorasib, a KRAS inhibitor, improved the survival of a previously untargetable KRAS-mutant lung AC group. Furthermore, new approaches targeting KRAS are under development. Starting from the description of the biology of KRAS-mutant NSCLC, the present review focuses on the clinical aspects of KRAS mutations in NSCLC. Finally, the interaction between KRAS mutations and immune checkpoint inhibitors will also be discussed.

Abstract

In non-small cell lung cancer (NSCLC) the most common alterations are identified in the Kirsten rat sarcoma viral oncogene homolog (KRAS) gene, accounting for approximately 30% of cases in Caucasian patients. The majority of mutations are located in exon 2, with the c.34G > T (p.G12C) change being the most prevalent. The clinical relevance of KRAS mutations in NSCLC was not recognized until a few years ago. What is now emerging is a dual key role played by KRAS mutations in the management of NSCLC patients. First, recent data report that KRAS-mutant lung AC patients generally have poorer overall survival (OS). Second, a KRAS inhibitor specifically targeting the c.34G > T (p.G12C) variant, Sotorasib, has been approved by the U.S. Food and Drug Administration (FDA) and by the European Medicines Agency. Another KRAS inhibitor targeting c.34G > T (p.G12C), Adagrasib, is currently being reviewed by the FDA for accelerated approval. From the description of the biology of KRAS-mutant NSCLC, the present review will focus on the clinical aspects of KRAS mutations in NSCLC, in particular on the emerging efficacy data of Sotorasib and other KRAS inhibitors, including mechanisms of resistance. Finally, the interaction between KRAS mutations and immune checkpoint inhibitors will be discussed.

1. Introduction

The Rat Sarcoma virus (RAS) gene family is characterized by three genes (KRAS, NRAS and HRAS) encoding for closely related GTPase proteins responsible for the signal transduction within the mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3 kinase (PI3K) pathways. This group of genes is associated with the control of cell growth, survival and differentiation [1,2], and it is characterized by the most frequently observed genomic alterations in human cancers, being detected in approximately 19% of tumors [3,4].
KRAS mutations are the most important and frequent alterations in this family, accounting for 85% of RAS mutations observed in the oncologic population [5]. More specifically, KRAS exon 2, exon 3 and exon 4 mutations are present in 13.98%, 1.17% and 0.62% of all cancer types [2,6], respectively, especially in pancreatic ductal adenocarcinoma (PDAC) (88% of patients), in colorectal cancer (CRC) (45–50% of cases) and in lung adenocarcinoma (AC) (30–35% of patients) (Table 1) [5,6,7,8].
Despite the fact that KRAS is altered in a consistent number of cases, the clinical interest in assessing KRAS mutations in lung was generally low or null until a few years ago; it has gained great relevance in the last few years [9,10], with the development of several targeted approaches and the recent introduction of sotorasib into clinical practice. This review will therefore summarize the current state-of-the-art of the clinical relevance of KRAS mutations as well as the most recent advances in KRAS-targeted anti-cancer strategies, focusing in particular in lung AC. Furthermore, the main mechanisms of acquired resistance to KRAS G12C inhibitors will be outlined and the attempts to overcome this resistance will be reported.

2. KRAS Mutations in Lung Cancer

The protein encoded by KRAS is a GTPase regulated by tyrosine kinase receptors that in turn activate the RAF/MEK/MAPK and PI3K/AKT signaling pathways, leading to a strong promotion of cell growth and replication that determines cell survival. The presence of mutations in KRAS blocks this protein in the active GTP-bound conformation, thereby activating constitutively downstream signaling pathways, finally resulting in uncontrolled cell growth [11]. Specifically, this continuous activation confers to cancer cells the ability to grow in lower glucose concentrations than those required for the growth of normal cells or cancer cells that do not have KRAS mutations [12,13]. As for other cancer types, KRAS mutations in lung cancers typically occur in hot-spot codons of exons 2, 3 and 4.

2.1. KRAS Mutations in NSCLC

In general, lung cancer has a complex and heterogeneous background. Indeed, it can be histologically subdivided into non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC), accounting for 85% and 15% of all lung neoplasia, respectively [14]. NSCLCs are further subdivided into three histologic types: AC, squamous-cell carcinoma (SCC) and large-cell carcinoma (LCC), which represent 40%, 25–30% and 5–10% of all lung cancers, respectively [8]. The occurrence of KRAS mutations differs based on the specific lung cancer type/subtype, being higher in NSCLC than in SCLC, and within NSCLC, KRAS mutations are more frequent in lung AC than SCC (Table 2). Interestingly, a recent study established two different groups of KRAS-mutant NSCLC, KRAS-dependent and KRAS-independent according to their requirement for KRAS mutations to maintain oncogenicity. However, to date, these data are preliminary and need to be confirmed in larger cohorts [11].

2.1.1. KRAS Mutations in Lung Adenocarcinoma

In the AC subtype, almost 30–35% of cases are driven by a KRAS mutation [5,6,7,8], mainly in exon 2, with the c.34G > T (p.G12C) change being the most prevalent (Table 3) [15].

2.1.2. KRAS Mutations in Squamous-Cell Carcinoma

The percentage of SCC-harboring KRAS mutations is lower than in lung AC cases, corresponding to only 3–4% of patients [7,16,17]; again, the c.34G > T (p.G12C) change as the most prevalent one, identified in up to 41% of KRAS-mutant SCC patients (Table 2) [18].

3. KRAS Mutations in Population-Based Cohorts of Lung Adenocarcinomas

The frequency of KRAS mutation in patients affected by lung AC differs among ethnicities and countries of origin, being higher in Caucasian (where they are detected in up to 30% of patients) than in Asians or in Africans (occurring in 16% and 9% of cases, respectively) [19,20,21,22,23,24,25].

3.1. KRAS Mutations in Caucasians Affected by Lung AC

In Europe, the most frequent KRAS mutations in lung AC are found in exon 2, with a general mutational rate close to 30% [19,20,21]. Differences can also be observed among European countries (28% in Germany, 29–31% in France, 27% in Italy, 19–27% in Greece and 43.3% in Spain) [19,20,21,26,27,28,29] and even among regions of the same country: for example, the island of Sardinia in Italy has the lowest frequency among all Italian regions [30]. This difference could be due to the physical separation of the island from the country, reflecting different lifestyle or environmental behaviors.
In Europe, 86% of mutations in exon 2 are located in codon 12, with the c.34G > T (p.G12C) mutation being the most common one (observed in 34% of cases) [19] (Table 4).

3.2. KRAS Mutations in Asians Affected by Lung AC

Several studies show that Asian populations have significant lower KRAS mutation incidence than that in the Caucasian and Afro-American populations [37,38]. As for Europe, even in Asia there is a wide range in the mutation frequencies among countries, with the highest values in Vietnam (21%, close to European populations) and the lowest in China (8.4–13%), Japan (10%) and Indonesia (7%) [33,39,40,41,42]. As usual, the most affected region is KRAS exon 2, but the most common KRAS exon 2 mutational subtype is the c.35G > A (p.G12D) change, with a rate of 25.5%, followed by c.34G > T (p.G12C) (24.5%) [22,32,43] (Table 4).
The frequency and type of mutations in exons 3 and 4 in Asian populations mirror those observed in European countries [43] (Table 4).

3.3. KRAS Mutations in Africans Affected by Lung AC

Central and West Africa are the regions with the lowest incidence rates of lung cancer for both sexes (0.8%) worldwide [44], while in North Africa, the KRAS mutations is observed in 6–9% of patients [31,36,45]. Specific studies concerning molecular characterization in African populations are extremely rare, and the data are sporadic. In Morocco, the c.34G > T (p.G12C) change is the most prevalent one (detected in 73% of KRAS-mutant cases) [31], while in Tunisia, c.38G > A (p.G13D) followed by c.35G > A (p.G12D) are predominant (42.8% and 28.5% of cases, respectively) [36] (Table 4). However, the size cohort is often too small to derive any conclusion.

3.4. KRAS Mutations in the Heterogeneous Lung AC American Population

In the USA, KRAS mutations are observed in a significantly high proportion of cases, up to 47.5% of patients, especially in exon 2 (90.6%), with the c.34G > T (p.G12C) change being the most prevalent one (41.2%) [34]. Focusing on the different ethnicities, the prevalence of KRAS mutations in African-American patients was less than that observed in the Caucasian group (17% versus 26%) [46]. Interestingly, American Hispanic patients have better survival than non-Hispanic/non-African cases, despite the fact that this population is diagnosed for cancer at a later stage due to social factors. This situation, defined also as the “Hispanic paradox”, has suggested investigating Hispanic individuals’ biological, pathological and molecular factors [35]. First, researchers observed a lower frequency of KRAS mutations compared with the Caucasian population (15.7% and 20–35% respectively) [35], and, second, researchers described that Hispanics have more KRAS changes in exon 2 codon 13 and in exon 3 codon 59 than non-Hispanics [47], possibly explaining the inferior cancer aggressiveness.
In Latin America, the frequency of NSCLC (in particular lung AC) with KRAS mutations corresponds to 14% [48], with little differences, ranging from 12.9% in Colombia to 15.9% in Mexico and 16.8% in Peru [48].
Two relevant observations come from analyses of Puerto Rican and Brazilian cohorts [49,50]. In Puerto Rico, lung ACs present with KRAS mutations in 18.7% of the cases, a rate that is significantly higher than in the aforementioned general Hispanic populations [48,50]; in Brazil, 14.6% of lung ACs harbour KRAS mutations, with a higher prevalence in non-Asian patients [49]: in both populations, c.34G > T (p.G12C) is the most diffused type of alteration.

4. Risk Factors and KRAS Mutations in Lung Adenocarcinomas

Recently, the number of lung cancer cases has increased with different trends in men versus women [51]. In women, lung cancer incidence rates began to increase significantly in 1973 and reached a plateau in the late 1990s, over a decade later than men [52,53]. Women’s lung cancer mortality stabilized for the first time in 2003, two decades later than men, and has yet to decline [53,54]. In 2021, the estimated new cases for males and females were nearly comparable (119,100 new patients versus 116,660, respectively) [51].
“Women’s emancipation” and the strategy of marketing cigarettes directly to women and young girls both contributed to increasing the percentages of female smokers and consequently to rising rates of lung cancer in women [55]. Hormonal influences, reduced DNA damage reparability and elevated amount of DNA adduct are also important factors in the high percentage of women who suffer from lung cancer [56].
However, among all the factors correlated with the development of lung tumors, tobacco smoking is sufficient to increase the risk of lung cancer compared with never smokers and represents the predominant cause, accounting for 80% of all lung cancer cases [57]. In the meta-analysis conducted by Chapman and his group [58], never-smokers present a distinct and separate mutational pattern of lung cancer from that observed in smokers. Smokers are characterized by more complex KRAS mutations, have a higher mutational burden and often present co-occurring mutations in TP53 or STK11 compared with cancers occurring in never smokers [59]. STK11 is a tumor suppressor encoding for the serine/threonine kinase STK11, also known as liver kinase B1 (LKB1), and it regulates cell polarity and energy metabolism [60]. In addition, it has been proposed that KRAS mutations might be more common in smoking females than in smoking males [61], a finding not confirmed by subsequent studies [62]. Smoking-associated KRAS mutations are mainly transversion mutations (G > T and G > C) and are more frequent in females than in males [61,63,64]: In fact, the two most common KRAS mutations in NSCLC are the c.34G > T (p.G12C) and the c.35G > T (p.G12V) changes [65,66], which are more common in females and in current or past smokers [62]. In contrast, KRAS transition mutations (G > A) are more common in non-smoker patients, suggesting different mechanisms of carcinogenesis [63,64].
Genetic factors are also associated with a higher risk of developing lung cancer. A meta-analysis involving the results of 32 studies demonstrated that most non-smokers with a familial history of lung cancer have a 2-fold increase probability of developing this type of malignancy [67]. In more detail, individuals with a first-degree relative affected by lung cancer have a 1.5 higher probability of developing this disease. This risk is even greater when a sibling is affected by lung cancer [68]. Nevertheless, no specific correlation between familiarity and KRAS mutations has been reported because KRAS mutations in lung AC are somatic mutations and not linked to an inherited predisposition [69].
Brenner [70] found that nonsmokers who suffer from previous respiratory diseases such as chronic obstructive pulmonary disease (COPD), asthma, pneumonia and tuberculosis are at higher risk of developing lung cancer. However, COPD is not associated with the presence of KRAS mutations in lung cancer [61].
Additional risk factors associated with lung cancer development are viral infections, improper lifestyle habits such as obesity, dietary factors, alcohol, consumption of marijuana and environmental air pollution. To the best of our knowledge, none of these factors has been associated with the occurrence of KRAS mutations [71,72,73,74,75,76,77,78,79,80].
In conclusion, only tobacco smoking is strictly related to the occurrence of KRAS mutations in lung cancer patients.

5. KRAS Biology in Lung Cancer

As was aforementioned, mutations in the KRAS gene primarily cause the constitutive activation of the downstream signaling pathways, leading to uncontrolled cell proliferation and abnormal cell survival [81,82]. Mutant allele specific imbalance (MASI) has been observed in combination with KRAS mutation in 58% of lung cancers and has been demonstrated to be mainly caused by uniparental disomy, which results from the complete loss of the wild-type (wt) allele without copy number gain [83]. Additionally, Soh et al. found a significant association between KRAS mutations and MASI with increased GTP-bound active RAS protein [83].
An in vitro analysis conducted on cell lines carrying different KRAS codon subtypes found that the c.35G > A (p.G12D) mutation was associated with PI3K and MEK phosphorylation, while KRAS c.34G > T (p.G12C) and KRAS c.35G > T (p.G12V) were correlated with Ral pathway activation and subsequently with AKT phosphorylation reduction [84].
As described before, cigarette smoking has been associated with KRAS mutations, but alterations in this gene were also related to abnormal metabolic parameters such as hypertension, dyslipidemia and diabetes mellitus [57,85]. This might indicate a relationship of the KRAS mutational status with metabolic parameters. However, this hypothesis was rejected by Yilmaz and colleagues [86].

6. KRAS and Co-Occurring Mutations in Lung AC

Much evidence suggests that KRAS-mutant cancers, including lung ACs, are a heterogeneous disease composed of different cellular clones and subclones carrying specific molecular patterns. These biological diversities can be ascribed to the presence of co-occurring genetic events, to the co-presence of different KRAS gene mutation subtypes and to the mutant KRAS allelic content [87]. Although the role of KRAS co-occurring mutations remains to be elucidated, it has been described that they may affect treatment responses [88].

6.1. Co-Occurring Genetic Events

6.1.1. KRAS and Other Molecular Markers Alterations

KRAS mutations are mutually exclusive with alterations in other molecular markers such as EGFR, ALK and ROS1, considered genetic drivers of lung cancer development. However, rare cases (<1%) harboring mutations in both KRAS and these markers have been reported [88]. In lung ACs, the rate of co-occurring mutations with KRAS alterations is 3.2%, and of the KRAS-mutant cases, 33% [88,89]. These frequencies differ based on the specific KRAS mutation: c.34G > T (p.G12C) presents reportable co-mutations in 6.1% of lung ACs, c.35G > A (p.G12D) in 2.5% and c.35G > T (p.G12V) in 1.6% (including co-existing alterations with EGFR, BRAF and NRAS) [87,88]. The most frequent KRAS co-mutations were found in TP53 (42%), STK11 (29%) and Kelch-like ECH-associated protein 1 (KEAP1)/Nuclear factor erythroid 2-related factor 2 (NFE2L2) genes (27%) [90]. This is relevant because Keap1, interacting with Nrf2, a transcription factor encoded by the NFE2L2 gene, regulates the antioxidant response that is involved in the amelioration of oxidative stress [91].
Recently, the presence of mutations in different genes as well as concomitant mutations in the same gene have been investigated as a factor influencing targeted therapy responses or patients’ prognosis [88]. Although they rarely occur in lung cancer, KRAS mutations combined with EGFR mutations or ALK rearrangement predict poorer response to tyrosine kinase inhibitors (TKIs) [88].
Concerning co-occurring genetic events, an integrative analysis of genomic, transcriptomic and proteomic data in lung ACs identified three subsets of KRAS-mutant cases. The first one was characterized by co-occurring alterations in KRAS/STK11/LKB1 (KL group), the second by mutations in KRAS/TP53 (KP group) and the third by mutations in KRAS plus CDKN2A/B inactivation coupled with low expression of the NKX2-1 (TTF1) transcription factor (KC group) [92]. These groups differ in biology, response to therapies and prognosis (please see chapter 15 for specific details).

6.1.2. KRAS and PD-L1 Expression

Preclinical evidence indicates that PD-L1 level is elevated through the activation of downstream KRAS-signaling pathways [93]. Consequently, many studies investigated the association between KRAS mutations and PD-L1 expression. Liu and coauthors described that KRAS-mutant tumors were more likely to be PD-L1 positive and showed more T-cell infiltration than KRAS wt tumors (p = 0.0002 and p = 0.003) [93,94,95]. In addition, a higher proportion of combined PD-L1 expression and CD8+ tumor infiltrating lymphocyte (TILs) presence was observed in the KRAS-mutant group compared with the wt (p = 0.008). The percentage of KRAS-mutant cases also harboring PD-L1 expression is equal to 49.5% considering PD-L1 ≥ 1% or to 21.2% considering PD-L1 ≥ 50% [94]. These high frequencies suggest how KRAS mutations lead to an inflammatory phenotype [93]. In deepening the investigation of the different KRAS mutations subtypes, a higher proportion of PD-L1-positive tumors (≥50%) has been found in patients with c.35G > A (p.G12D), accounting for 41.7%; c.35G > T (p.G12V) accounted for 33.3%; and c.37G > T (p.G13C) accounted for 40%, whereas on the lower end, the rates were c.34G > T (p.G12C) accounting for 9.8% and c.35G > C (p.G12A) accounting for 14.3% [94]. The concomitance between KRAS mutations and PD-L1 expression is also related to a longer survival after immunotherapies and as a consequence can be defined as a prognostic and predictive factor of benefit from anti-PD1/PD-L1 immunotherapies [93,95].
The relevance of KRAS compared with PD-L1 expression has been further evaluated in smokers comparing KRAS-mutant versus wt, with mutant showing higher PD-L1 expression and T-cell infiltration (p = 0.042 and p = 0.008) [93].

6.2. Co-Presence of Different KRAS Gene Mutation

In rare cases (<1%), patients with lung AC can present more than one co-occurring mutation in KRAS, usually occurring in two different KRAS exons (2 and 3), with the most frequent combination being c.34G > T (p.G12C) or c.35G > A (p.G12D) with c.182A > T (p.Q61L) or c.182A > G (p.Q61R) [88,89]. The double mutations in KRAS represent in general 11–26% of the co-occurring genetic mutations, with c.34G > T (p.G12C) accounting for the most diffused mutation associated with other alterations (mainly c.38G > A, p.G13D; c.34G > A, p.G12S; c.35G > T, p.G12V; c.182A > T, p.Q61L; c.182A > G, p.Q61R) in the same gene (nearly 20%) [89].

7. KRAS Influence on Prognosis

Multiple studies have shown conflicting results regarding the prognostic value of KRAS mutations in lung cancer [21,96,97]. Many researchers reported that KRAS mutations correlate with shorter survival and shorter time to relapse [98,99,100]. Aredo et al. [101] reported that the KRAS c.35G > A (p.G12D) mutation was significantly associated with poor OS, while Nadal and his colleagues [34] found a worse prognosis in individuals harboring the KRAS c.34G > T (p.G12C) mutation with resectable disease. In addition, Park [102] demonstrated a low response to the treatment with pemetrexed in patients with the KRAS c.34G > T (p.G12C) mutation. Furthermore, the KRAS c.35G > T (p.G12V) mutation was correlated with a shortened OS and time to recurrence [20], while Izar et al. [103] showed a superior disease-free survival in patients with resected stage I disease, characterized by KRAS c.34G > T (p.G12C) and c.35G > T (p.G12V) mutations. Finally, other studies did not find any correlation between KRAS mutations and survival [104,105].
The prognosis associated with KRAS mutations depends on co-alterations. A poorer prognosis is found in patients with co-occurring mutations of KRAS and STK11 or KEAP1 [106,107] or KRAS c.34G > T (p.G12C) and PD-L1 expression [105,108]. Regarding the correlation between KRAS mutations and PD-L1 expression, a recent contribution of our group reported the potential role played by the KRAS c.34G > T (p.G12C) variant in the prolonged survival found in NSCLC patients showing a PD-L1 overexpression [109].

8. Predictive Value of KRAS Mutations

Many attempts have been made to outline the predictive value of KRAS mutations in NSCLC patients treated with standard chemotherapy [110,111,112,113], targeted therapies [114,115] and immune check point inhibition [116].

8.1. Predictive Value of KRAS Mutation for Response to Chemotherapy

In spite of recent advances in the treatment of NSCLC, a significant percentage of patients with advanced-stage disease are still treated with platinum-based chemotherapy regimens. KRAS mutations as a predictive biomarker in NSCLC were investigated in the neo-adjuvant and perioperative setting [117], in the adjuvant setting [111], and in metastatic disease, with both exclusive conventional chemotherapy and the association of erlotinib or placebo [10,110]. In general, in the above-mentioned experiences, KRAS mutations were not predictors of response rate, progression-free survival (PFS) or OS in patients with lung cancer.
The type of chemotherapy regimen may also have to be considered. In a subgroup analysis from the JBR10 trial, the combination of vinorelbine and cisplatin in the adjuvant setting appeared to confer a statistically significant survival benefit in the KRAS wt subgroup, although RAS status did not appear to have a statistically significant effect on outcomes in the interaction analysis [118]. On the contrary, Sun et al. reported significantly poorer PFS and OS in patients with KRAS mutations when treated with pemetrexed or gemcitabine but not in those receiving taxanes [112].
The exact subtype of KRAS mutation should be considered when investigating its potential role as a predictive biomarker for chemotherapy in NSCLC patients. A potential detrimental effect of KRAS codon 13 mutations was suggested by Garassino et al. [66], and similar results have been reported by other authors who observed that PFS and OS were significantly shorter in patients harboring KRAS codon 13 mutations and treated with carboplatin-based adjuvant chemotherapy [97]. The role of the KRAS c.35G > T (p.G12V) mutation is controversial. One study showed poorer response to chemotherapy in terms of PFS and disease control rate (DCR) compared with wt patients and non-KRAS c.35G > T (p.G12V) metastatic mutated patients [119], while another project demonstrated better response to platinum-based chemotherapy compared with other codon 12 mutations [120].
Preclinical data from Garassino et al. add further complexity, suggesting an interaction between the KRAS mutation subtype and the type of chemotherapy used. Specifically, their data show that the c.34G > T (p.G12C) clones are associated with a reduced response to cisplatin and an increased sensitivity to taxol and pemetrexed, whereas the c.35G > A (p.G12D) variant results in resistance to taxol treatment and sensitivity to sorafenib. Furthermore, c.35G > T (p.G12V) mutant clones are associated with a better response to cis-platin compared with the wt clones. On the other hand, these clones appeared to be more resistant to pemetrexed. Compared with other chemotherapy regimens, the sensitivity of tumor cells to gemcitabine does not depend on different subtypes of KRAS mutations [66]

8.2. Predictive Value of KRAS Mutations for Response to Anti-Angiogenic Therapy

KRAS promotes the initiation of tumor growth and ensures tumor progression by stimulating tumor-associated angiogenesis [121,122] and activating different effector pathways through the upregulation of VEGF (vascular endothelial growth factor) and CXC chemokine interleukin-8 (IL-8) [121,123,124,125,126].
However, very few studies have investigated the influence of KRAS mutations on the efficacy of anti-angiogenic therapy [127,128,129].
A phase II trial assessed the efficacy of the addition of bevacizumab to neo-adjuvant chemotherapy in resectable, non-squamous NSCLC and found that no patients with KRAS mutation had pathological response to neo-adjuvant bevacizumab combined with chemotherapy, whereas 35% of patients with KRAS wt status showed a significant pathological response [114].
As retrospectively shown by Ghimessy et al., patients with KRAS mutations, and especially those with KRAS c.35G > A (p.G12D) mutant disease, had a significantly shorter OS and PFS compared with those with KRAS WT when treated with bevacizumab plus platinum-based chemotherapy [115]. Accordingly, c.35G > A (p.G12D) mutations may define a subset of KRAS types that might not be eligible for treatment with anti-VEGF drugs [115].

8.3. Predictive Value of KRAS Mutations for Response to EGFR Targeted Therapy

The majority of published data suggest that KRAS mutational status is a significant negative predictor of the efficacy of EGFR-directed TKI [10,128,129,130], probably because the KRAS mutation can counteract the therapeutic effect of EGFR inhibitors [130]. Indeed, when patients with KRAS-mutated disease are treated with EGFR TKIs, they tend toward worse objective response rates (ORR), PFS and OS compared with patients without a KRAS mutation [10,130,131].
However, controversies exist, likely in relation to a potential role of the involved KRAS mutation subtype since specific subtypes have been shown to induce differential sensitivity to EGFR TKIs [24,132]. Within the KRAS-mutant group, Metro and colleagues identified great variability in terms of sensitivity to treatment: KRAS codon 13 mutant patients displayed worse outcomes when compared with KRAS codon 12 mutant patients and KRAS wt patients [133]. Similarly, other researchers demonstrated poorer treatment efficacy in the case of KRAS c.34G > T (p.G12C) and c.35G > T (p.G12V) mutations and found promising response rates in c.35G > A (p.G12D) and c.34G > A (p.G12S) KRAS-mutant NSCLC treated with EGFR-targeted TKIs [134].
Fiala et al. reported that EGFR-directed TKIs improved PFS in patients with non- c.34G > T (p.G12C) KRAS-mutant tumors when compared with the c.34G > T (p.G12C) group [135]. In a meta-analysis, Mao and colleagues showed an ORR of 3% (6/210) in patients with mutant KRAS NSCLC and of 26% (287/1125) in KRAS wt lung cancers, fulfilling the hypothesis that KRAS mutations represent negative predictive biomarkers for tumor response in NSCLC patients treated with EGFR-TKIs [131]. However, due to the substantial mutually exclusive relationship between KRAS and EGFR mutations, the clinical usefulness of KRAS mutation as a selection marker for EGFR-TKIs sensitivity in NSCLC is limited [131].

8.4. Predictive Value of KRAS Mutations for Response to IMMUNOTHERAPY in NSCLC

PD-L1 expression has been shown to be in close relationship with KRAS status, and KRAS mutations were described as possible biomarkers for immune checkpoint inhibitor efficacy [114], in particular the c.34G > T (p.G12C) mutant variant [109,135,136,137,138,139]. Monotherapy of immune checkpoint inhibitors in advanced lines demonstrated a higher objective response rate (26%) in KRAS-mutant patients when compared with other oncogene-driven NSCLC patients (BRAF, ROS1, MET, EGFR, HER2, RET, ALK) as reported in immunotarget study [140]. The same clinical trial showed no differences within KRAS mutation subtypes in terms of PFS and response rate, although patients with KRAS c34G > T (p.G12C) seemed to have better PFS and response rates compared with non-KRAS c34 > T (p.G12C) patients. [139]. The positive correlation of KRAS mutations and ICI efficacy was confirmed in a comprehensive analysis [141] and in several studies including patients with PD-L1 overexpression (≥50%) [94,112,142].
Concerning KRAS mutation subtypes, in the retrospective study by Jeanson et al., PD-L1 positivity was significantly higher among patients with KRAS c.35G > A (p.G12D), c.35G > T (p.G12V) and c.37G > T (p.G13C) mutations, making these subgroups of patients potential candidates for immunotherapy; meanwhile, PD-L1 negativity was higher among those who had c.35G > C (p.G12A) and c.34G > T (p.G12C) mutations [139].
Overall, the correlation between KRAS mutations and efficacy of ICI seems to be quite robust.

9. Current Therapeutic Approaches and Perspectives in KRAS-Mutant NSCLC

Because of its high mutation frequency in NSCLC, KRAS is an appealing target. However, the development of targeted therapies for KRAS-mutant lung cancers has long been marked by failures [11,143,144]. In contrast to most of the oncogenic drivers, KRAS was for many years considered undruggable because of its extremely high affinity for GTP, the lack of well-defined hydrophobic pockets and the complexity of the downstream pathways [145,146,147]. KRAS inhibition strategies have included reducing the proportion of active RAS-GTP, disrupting protein–protein interactions, decreasing RAS presence at the plasma membrane, inhibiting downstream effector signaling and inducing synthetic lethality.
Initial development focused on preventing RAS from associating with the cell membrane, e.g., through the inhibition of the post-translational farnesylation of its C-terminal domain or through the competitive inhibition of RAS membrane binding; however, these strategies failed to demonstrate clinical efficacy due to the emergence of escape mechanisms such as adaptive KRAS prenylation by geranylgeranyl transferase [148,149,150,151]. Attention therefore turned to strategies for targeting KRAS indirectly by interfering with its upstream, downstream or parallel pathways; however, initial experiences with the inhibition of signaling proteins such as RAF, FAK, MEK, MET, mTOR and AKT met limited success [11,152,153,154].
Unsuccessful attempts were made with synthetic lethality approaches as well, probably due to the presence of either the feedback activation of RAS upstream effectors or tumor heterogeneity [149,155,156,157,158,159].

9.1. Direct Inhibition of KRAS

A breakthrough in the direct targeting of KRAS occurred in 2013, when a new allosteric pocket was discovered beneath the effector switch-II region of mutant KRAS c.34G > T (p.G12C) [160] and a small molecule (ARS-1620) was identified that could bind covalently to this pocket. Later, a second small molecule with the same activity was discovered (ARS-853) [161].
These studies demonstrated that the direct targeting of KRAS c.34G > T (p.G12C) is dependent on covalent binding to a switch-II pocket region when KRAS c.34G > T (p.G12C) is in its inactive GDP-bound state and to cysteine binding as well: therefore, these drugs do not recognize, and as a consequence are inactive against, other KRAS-mutant or KRAS wild-type proteins. Later preclinical studies allowed for better defining and identifying the mechanisms of action of inhibitors in KRAS c.34G > T (p.G12C) mutant cases [160]; [161,162,163]. Subsequently, AMG-510 and MRTX849 were the first inhibitors introduced in a preclinical setting to demonstrate increased KRAS c.34G > T (p.G12C) inhibition activity over previous inhibitors, mediated by the enhanced synergic activity of H95 residue in the a-3 helix of KRAS c.34G > T (p.G12C) [162,163,164,165].
These results led to the initiation of the first-in-human trial of AMG-510 (sotorasib) in 2018 (ClinicalTrials.gov NCT03600883). Today, several clinical studies are evaluating direct KRAS inhibition using similar drugs (Table 5).

9.2. Sotorasib, First KRAS c.34G > T (p.G12C) Inhibitor in Clinical Practice

Sotorasib successfully passed all testing phases, from in vitro KRAS c.34G > T (p.G12C) mutant cell lines to preclinical setting: its activity led to the inhibition of ERK phosphorylation and tumor cell growth, while the PI3K signaling was substantially unaffected [146,166]. Moreover, sotorasib was found to upregulate a pro-inflammatory microenvironment, in this way enhancing anti-tumor T cell activity [164,167] and as a consequence inhibiting a potential active promoter of tumor progression. The repercussion of these positive results was the introduction in a first phase I/II trial of sotorasib as the first drug against KRAS mutation in clinical development. In this first study, sotorasib showed an objective response rate of 32.2% in the subgroup of patients with NSCLC and disease control in 88.1% of patients with a mPFS of 6.3 months [168].
On the post hoc analysis of Codebreak 100 (phase II study), sotorasib demonstrated an ORR of 37.1%, DCR of 80.6% with a median duration of response (DOR) of 11 months, a PFS of 6.8 months and a median OS of 12 months. These outcomes led to an FDA priority review in February 2021 and to an ongoing phase III clinical trial comparing sotorasib with docetaxel for patients with KRAS c34G > T (p.G12C) mutant NSCLC (ClinicalTrials.gov NCT04303780).
The efficacy of sotorasib among patients with brain metastases was also evaluated in post hoc analysis by Ramingam et al. In this subgroup of 40 patients included in a phase I/II Codebreak 100 trial, sotorasib demonstrated clinical efficacy in terms of PFS (5.3 months) and OS (8.3 months). [169]. However, several side effects were also reported: grade 3–4 treatment-related adverse events were reported in nearly 20% of patients, diarrea in 31.7%, nausea in 19%, transaminitis (ALT and AST) in 15% and asthenia in 11.1% of cases. Regarding liver tolerability, it was reported that sotorasib might trigger immune-related hepatitis in patients previously treated with ICIs [170]. Overall, sotorasib maintained or improved global health status, physical functioning and the severity of the main lung-cancer-related symptoms, including cough, dyspnea and chest pain [171]. In late 2021, a randomized study of dose efficacy was prepared, focusing on the comparison of different doses of the drug (240 mg versus 960 mg, with the higher dose being the one that was used in the phase II and III studies) as part of a US FDA post-marketing requirement [172].
Furthermore, it has been demonstrated that cases with STK11 and TP53 co-occurring mutations responded better to sotorasib than patients harboring KEAP1 co-mutation [169,170,171,173,174,175]. Moreover, alterations in cell cycle effectors like in the WNT and MAPK pathways are more common in the late progressors group [176]. As recently reported by Zhao and colleagues, patients with a lower plasma KRAS c34G > T (pG12C) allele frequency and a lower tumor burden at baseline tend to be what are called exceptional responders (i.e., complete or partial response lasting more than 12 months) when treated with sotorasib [177]. Based on that clinical efficacy and safety profile, sotorasib was therefore recently approved by the European Commission, which granted it a conditional marketing authorization, underlining that confirmatory trials are awaited.

9.3. Adagrasib (MRTX849): Pre-Clinical and Clinical Development

The second small-molecule inhibitor of KRAS c.34G > T (p.G12C) that is under evaluation in clinical trials is adagrasib (formerly MRTX849) [178]. Similar to sotorasib, adagrasib impairs cell viability in pancreatic and lung AC monolayer and spheroid models harboring KRAS c.34G > T (p.G12C) [179]. Beyond KRAS c.34G > T (p.G12C), no single genomic co-alteration predicted the antitumor activity of adagrasib. A preliminary report on the clinical efficacy of adagrasib confirmed an objective response and a disease control rate in 45% and 96% of evaluable patients in the NSCLC cohort, respectively (n = 79) [178].
More recently, results from the KRYSTAL-1 phase II study enrolling 116 patients with NSCLC harboring a KRAS c.34G > T (p.G12C) mutation who received adagrasib following prior treatment with immunotherapy and chemotherapy were presented by Spira et al. at the 2022 ASCO annual meeting [179]. Adagrasib showed promising efficacy, with ORR of 42.9% and the DCR of 79.5%. Median DOR was 8.5 months, median PFS was 6.5 months and median OS was 12.6 months.
The most common side effects reported on adagrasib are those of gastrointestinal pertinence (nausea, vomiting and diarrhea), fatigue and asthenia, a pattern similar to that observed after sotorasib administration. Discrete hepatic functional impairment, associated with alanine aminotransferase increase, was also registered among patients treated with adagrasib. Concomitant mutations like in STK11 gene did not exclude adagrasib efficacy as reported, regardless of limited subgroup numerosity [180,181]. Adagrasib is the second-most selective KRAS inhibitor currently being reviewed by the FDA for accelerated approval for patients with lung cancer who harbor the KRAS c.34G > T (p.G12C) mutation. A phase III trial (KRYSTAL-12) evaluating adagrasib monotherapy versus docetaxel in pretreated patients with KRAS c.34G > T (p.G12C)-mutated NSCLC is ongoing (ClinicalTrials.gov NCT04685135).

9.4. Other KRAS-Targeted Molecules

Several other direct KRAS c.34G > T (p.G12C) allosteric inhibitors are being tested in the clinical setting as monotherapy and in combination with other therapies. These KRAS c.34G > T (p.G12C) inhibitors include GDC-6036 (phase I; ClinicalTrials.gov NCT04449874), D-1553 (phase I/II; ClinicalTrials.gov NCT04585035) JDQ443 (phase I/II; ClinicalTrials.gov. NCT04699188), JAB-21822 (phase I/II: ClinicalTrials.gov NCT 05002270), and LY3499446, and ARS-3248/JNJ-74699157, which are next-generation ARS-1620 inhibitors (ClinicalTrials.gov NCT04006301, and ClinicalTrials.gov NCT04585035) with inhibitory activity at least 10 times higher than that of sotorasib and adagrasib (ClinicalTrials.gov NCT04006301).
Compared with direct inhibition, degradation could be a more potent strategy that affects cell proliferation and downstream signaling responses [182,183], which is why recently, proteolysis-targeting chimaeras (PROTACs) targeting KRAS c.34G > T (p.G12C) proteins are being explored.
Based on covalent inhibitors targeting KRAS c.34G > T (p.G12C), PROTACs are designed to target KRAS c.34G > T (p.G12C). Trials based on PROTAC molecules demonstrated the first clinical proof-of-concept against two of the most relevant cancer targets: the estrogen receptor (ER) and androgen receptor (AR). Clinical trials of oral PROTACs (ARV-110 and ARV-471) have manifested uplifting outcomes for prostate and breast cancer treatment [184,185]. Encouraged by such results, the targeted protein degradation field (TPD) is now assured to reach undrugged targets.
Finally, a nanoparticle-formulated mRNA based cancer vaccine (V941) that targets four of the most prevalent occurring KRAS mutations (c.35G > A, p.G12D; c.35G > T, p.G12V; c.38G > A, p.G13D and c.34G > T, p.G12C) is in developing stages as monotherapy or in combination with pembrolizumab in NSCLC, CRC or PDAC patients (ClinicalTrials.gov NCT03948763).

10. Biological Mechanism of Acquired Resistance

10.1. Acquired Resistance to Sotorasib and Adagrasib

Biological mechanisms of acquired resistance to sotorasib and adagrasib have been described in both pre-clinical and clinical experiences. Similar to what is known concerning other oncogenic drivers in NSCLC [186], new-onset KRAS-activating mutations and KRAS amplification can mediate acquired resistance through RAS signaling pathway activation [187]. With the development of KRAS inhibitors, some findings highlighting the diversity of KRAS mutations emerged in response to their use, thereby limiting the potential development of efficient next-generation KRAS c.34G > T (p.G12C) inhibitors [188,189,190]. Some of these mutations display differential sensitivity to either sotorasib or adagrasib, thus providing a theoretical rationale for a sequencing strategy [188,189,190].
In the preclinical and clinical setting, presumed mechanisms of acquired resistance were described in 45% of cases for adagrasib and 60% of cases for sotorasib. These observations provide a rationale for combined therapy to prevent or delay acquired resistance [177,190]. These resistance mechanisms can involve on-target or bypass alterations, which lead to the abnormal activation of downstream or connecting signaling pathways as well as phenotypic transformation mechanisms to either SCLC or SCC [188,189,190].

10.2. Acquired Resistance Mechanisms to Either Sotorasib or Adagrasib

Mutations in KRAS that confer acquired resistance to KRAS c.34G > T (p.G12C) inhibitors have been described in vitro and in the clinical setting [188,189,190] and occur primarily in switch-II binding pocket, thereby altering drug binding [187].
Patients who progressed on adagrasib have shown secondary mutations that alter drug binding [190]. In this subgroup of patients, the secondary mutations were expressed as double mutant alleles in Ba/F3 cell lines in order to shape their mechanistic impacts on KRAS c34G > T (p.G12C) inhibitor sensitivity. Awad et al. [190] described that Switch-II binding pocket mutations, including c.204G > T (p.R68S) and codons H95 and Y96, conferred resistance to Adagrasib in Ba/F3 cells, in contrast with the control KRAS c.34G > T (p.G12C) allele. According to their findings, the Y96C mutation caused cross-resistance to both adagrasib and sotorasib in vitro. In sotorasib-resistant clones, the KRAS c.38G > A (p.G13D) variant was the most frequent secondary mutation (i.e., 23% of KRAS secondary mutations), followed by c.203G > T (p.R68M) and c.175G > T (p.A59S) (21% and 2%, respectively). In adagrasib-resistant clones, KRAS Q99L was the most frequent secondary mutation (i.e., 52.8% of KRAS secondary mutations), followed by codon Y96 and c.204G > T (p.R68S) (i.e., 15, 3% and 13.9%, respectively) [186].
Tanaka et al. described how RM-018 (a novel KRAS c.34G > T (p.G12C) inhibitor that exploits cyclophilin A to bind and inhibit KRAS c.34G > T (p.G12C) in its GDP bound state) could overcome KRAS codon Y96 resistance mutation [189].
Other mutations were involved in in vitro resistance to MRTX1257 (a compound highly related to adagrasib and sotorasib) through the inhibition of GTP hydrolysis or the increase of GDP to GTP nucleotide exchange (like mutations occurring at codons 13, 59, 61, 117 and 146) [188,190,191,192,193,194].
The abovementioned in vitro experiments highlight the different sensitivities to KRAS c.34G > T (p.G12C) inhibitors for different acquired mutations. Some mutations like G13D and A59S remained partially sensitive to adagrasib while they conferred strong resistance to sotorasib [186]. In the same way, codon Q99 and H95 secondary mutations, which conferred resistance to adagrasib, remained sensitive to sotorasib [188,190]. In the plethora of shared mutations between sotorasib and adagrasib, only the ones in codon Y96 proved to confer strong cross-resistance to both KRAS c.34G > T (p.G12C) inhibitors [188]. The trials in clinical setting highlighted the main mechanisms of acquired resistance to sotorasib and adagrasib through secondary mutations within the drug-binding pocket and activating mutations in KRAS. Up to 53% of patients who showed any putative mechanisms of resistance to adagrasib had at least one acquired KRAS mutation or KRAS amplification. Acquired mutations within the switch II binding pocket– Y96 codon, c.204G > T (p.R68S), H95 codon, likewise mutations in KRAS c.35G > A (p.G12D) and c.35G > T (p.G12V) were reported. A single patient showed a high level of KRAS c.34G > T (p.G12C) amplification at relapse.
Regarding other alterations associated with resistance to KRAS c.34G > T (p.G12C) inhibitors, among 32 NSCLC patients enrolled in Codebreak 100 and Codebreak 101, acquired resistance was identified in 78% of cases (including alterations in KRAS, NRAS, BRAF, EGFR, FGFR2, MYC and other genes). Among them, four patients had pG12D, pG12V, pG12F and pV8L changes, while three patients had KRAS amplifications [177]. The c.35G > T (p.G12V) mutation in this report was shown to decrease the antiproliferative effects of both sotorasib and adagrasib.
Sotorasib has been described as improving the therapeutic efficacy of other targeted agents, i.e., with HER kinase inhibitors (i.e afatinib), SHP2 inhibitors (RMC-4550) and MEK inhibitors (trametinib), and in this setting, several studies are ongoing (Table 6 and Table 7). This might emerge as a potential strategy to overcome acquired resistance to these compounds [166]. A synergic activity of adagrasib was found when this drug was associated with other targeted agents (afatanib or RMC 4550) in xenograph models of NSCLC. Other efficient mechanisms that might enhance adagrasib efficacy have been described in vivo, like targeting downstream KRAS effectors such as mTOR and cyclid D family [180].

11. Activation of RTKs and RAS Downstream Signaling Pathways

One of the main mechanisms of acquired resistance is the activation of bypass signaling pathways described in oncogene-driven NSCLC, whether these are vertical and/or parallel pathways [186]. Targeting these pathways in addition to direct KRAS inhibition may be useful for overcoming both ab initio and acquired resistance mechanisms, and may result in more durable responses as well.
Tumoral heterogeneity of resistant tumors is explained partially by the co-occurrence of numerous mechanisms of acquired resistance to adagrasib [189], including mutations in NRAS (i.e., c.182A > T, p.Q61L; c.181C > A, p.Q61K; and c.182A > G, p.Q61R), BRAF c.1799T > A (p.V600E) and MAP2K1 (i.e., c.171G > C, p.K57N and c.167A > C, p.Q56P) genes [189]. In particular, it has been proposed that the polyclonal RAS-MAPK reactivation is a central resistance mechanism, specifying that NRAS c.181C > A (p.Q61K) not only was mechanistically confirmed as a mutation on adagrasib resistance but was also found to confer resistance to sotorasib in vitro as well [177]. Other mutations or amplifications detected in five NSCLC patients with progressive disease on adagrasib were found in RET, PIK3CA and MET as defined as well as acquired bypass mechanisms of resistance. Moreover, other alterations in the clinical setting were described in the RAS–MAPK pathway as acquiring resistance to sotorasib like in NRAS, BRAF, EGFR, FGFR2 and MET, expressed as mutations or amplifications of these genes [177,190].
In order to overcome acquired resistance to KRAS c.34G > T (p.G12C), several promising strategies are being elaborated, like targeting the specific RTK involved in bypassing the signaling pathways either directly or by inhibiting upstream and downstream effectors of KRAS c34G > T(G12C). In this perspective, different strategies are currently being proposed [191,192,193,194,195,196]. The combination of sotorasib with crizotinib has proved to be more efficient in inhibiting ERK, AKT and MET activation in H23 sotorasib-resistant cells that acquired MET amplification, compared to monotherapy. Likewise, this combination induced apoptosis and notably decreased tumor growth in MET-amplified H23 sotorasib-resistant xenograft models. In the same direction, the combination of sotorasib and capmatinib (a specific MET inhibitor) decreased cell viability [195].
Even though these results that support the further targeting of downstream or upstream effectors of KRAS c.G34 > T (pG12C) might seem to suggest an auspicious strategy, it might not be sufficient to bypass resistance. As a consequence, several clinical trials in development stages are evaluating the efficacy of KRAS c.34G > T (p.G12C) inhibitors in combination with chemotherapy [197,198] or with targeted therapies like cetuximab, afatinib, pembrolizumab and SHP2, mTOR and CDK4/6 inhibitors [178,197,199,200] (Table 8).
CDK4/6 are key regulators of the cell cycle and can be affected by KRAS through multiple signaling pathways, such as the RAF-MEK-ERK and PI3K/AKT pathways. Hallin et al. showed that in several MRTX849-refractory models, the combination of MRTX849 and palbociclib led to significant general tumor regression, which may be due to the increased inhibition of the retinoblastoma protein/E2F transcription factor pathway [180]. Trials evaluating the combination of CDK4/6 inhibitors with KRAS c.34G > T (p.G12C) selective inhibitors are ongoing [180] (Table 8).
Another promising strategy for overcoming resistance might be the inhibition of aurora kinase A (AURKA), an enzyme implicated in cell division. ARS-1620 (a potent, orally bioavailable covalent inhibitor of KRAS c.34G > T (p.G12C), when combined with alisertib (AURKA inhibitor), was observed to have a significant synergistic effect in ARS-1620-refractory KRAS c.34G > T (p.G12C) mutant cancer. The mechanistic inhibition of AURKA has been shown not only to impair normal cell division and proliferation but also to affect KRAS–c RAF interaction, leading to a challenging activation of RAF [201].
Lastly, it appears that the PI3K-AKT pathway is less dependent on the RAS pathway and less sensitive to KRAS c.34G > T (p.G12C) inhibitors; nevertheless, enhanced antitumor activity was seen when adagrasib was combined with afatinib, RMC-550, vistusertib (mTOR) and palbociclib in xenograft models [180]. Combined strategies targeting upstream effectors (i.e., SOS1 or SHP2) and the PI3KAKT- mTOR pathway could be relevant in the near future.

12. MEK Inhibition and Therapeutic Strategies to Overcome on-Target Mechanisms of Acquired Resistance to KRAS c.34G > T (p.G12C) Inhibitors

The combination of sotorasib with inhibitors of HER kinase, EGFR, SHP2, PI3K/AKT and MEK was evaluated in vitro, and the results of a CodeBreak 101 study (NCT04185883) favored the combination of MEK inhibitors (trametinib) with sotorasib versus one single agent. The addition of a MEK inhibitor to docetaxel demonstrated better outcomes in patients with KRAS-mutant NSCLC compared with either single-agent treatment, combination with erlotinib or docetaxel alone [202,203]. Different combinatorial strategies with a KRAS c.34G > T (p.G12C) inhibitor and/or MEK with chemotherapy are being evaluated in in vitro and in the clinical setting [204,205]. MEK inhibitors might constitute an alternative strategy in MET-amplified tumors, as preclinical experiences highlighted the importance of the MAPK pathway in this setting [206] (Table 7). The MEK inhibitor selumetinib showed interesting activity in vitro when associated with sotorasib as assessed by the complete inhibition of ERK phosphorylation in H23 MET-amplified sotorasib-resistant cells [195]. In spite of the preclinical efficacy of these therapeutic strategies, expanding on these results in dedicated clinical trials will prove complex due to the small proportion of patients who present these alterations.
In a phase 1b study, the addition of the oral MEK inhibitor binimetinib to the treatment combination of cisplatin and pemetrexed as first-line therapy in advanced KRAS-mutated metastatic NSCLC showed no signal of anti-tumor activity in terms of PFS or OS. (Froesch et al., 2021). Novel strategies focused on KRAS scaffold targets, namely SOS1 and SHP2, are being actively investigated to overcome acquired resistance to KRAS c.34G > T (p.G12C) inhibitors. SHP2 is a tyrosine phosphatase that is required for complete RAS-MAPK activation, and it is essential for KRAS-mutant carcinogenesis [207].
Multiple SHP2 inhibitors are being investigated both as monotherapy and in combination with KRAS c.34G > T (p.G12C) inhibitors like RMC 4630, RMC 4550, RLY 1971, TNO15 [180,201,208,209,210,211], (clinicaltrials.gov NCT 04252339, NCT03114319, NCT04699188). A novel inhibition strategy into KRAS c.34G > T (p.G12C) therapeutic approaches has yielded a new target in the SOS1 catalytic domain that affects the EGFR/MAPK pathway. Through the inhibition of SOS1, the binding of KRAS to GTP and its constitutively active state is reduced. BI-3046 and BI 1,701,963 are orally bioavailable small-molecule SOS1 inhibitors, that bind to the catalytic domain of SOS1, thereby preventing the interaction with KRAS. They showed effectiveness in KRAS-driven cancers when combined with MEK inhibitors (trametinib) [212].
Further on, the identification of BI-3406 in pre-clinical studies (a highly potent selective small-molecule SOS1 inhibitor that binds to the catalytic domain of SOS1 thereby prevents the interaction with KRAS) allowed for broadening the spectrum of activity by demonstrating that the therapeutic efficacy of this molecule was not limited only to KRAS c.34G > T (p.G12C) mutant models but also showed activity in KRAS c.35G > T (p.G12V) (SW620cells), KRAS c.38G > A (p.G13D) (LoVo cells) and KRAS c.34G > A (p.G12S) (A549 cells) as well by inducing tumor growth inhibition in xenograph models [213]. Moreover, BI-3406 was found to prevent ERK activation rebound following sotorasib treatment in vitro [213].
Importantly, BI-3406 demonstrated promising pre-clinical results in overcoming acquired resistance to KRAS c.34G > T (p.G12C) inhibitors when combined with treatments that target downstream KRAS c.34G > T (p.G12C) effectors. BI-3406 attenuates the feedback reactivation induced by MEK inhibitors enhancing as a consequence the sensitivity of KRAS-dependent cancers to MEK inhibition. Combined SOS1 and MEK inhibition represents a novel and effective therapeutic concept for addressing KRAS-driven tumors. Finally, the combination of sotorasib and BI-3406 decreased RAS-GTP levels and Inhibit ERK phosphorylation, while it affected less the AKT activation in MET-amplified sotorasib resistant H23 cells [195].

13. Lineage Plasticity and Acquisition of Features of Epithelial-To-Mesenchymal Transition as a Mechanisms of Acquired Resistance

Despite the genetic and histologic transformation to either SCLC or SCC representing a well-known biological mechanism of acquired resistance to targeted therapies in NSCLC [192,193], the evidence of trans-differentiation to SCLC in the context of acquired resistance to KRAS c.34G > T (p.G12C)-targeted therapies is scarce and limited to clinical case reports [190]. Other co-mutations like STK11 deletion seem to drive lineage transformation to SCC in KRAS mutated lung AC [214].
Another particular and praiseworthy resistance mechanism is represented by the epithelial-to-mesenchymal transition (EMT). The ability of acquisition of EMT features in in vitro and in xenograph cells is interpreted as a potential acquired resistance mechanism to sotorasib in the setting of KRAS c.34G > T (p.G12C) mutant NSCLC [196].
In vitro efficacy was demonstrated by Adachi et al. when IGF1R and FGFR1 were targeted: RTKs that intermediated acquired resistance to KRAS c34 G > T (p.G12C) inhibitors through EMT [196]. The researchers reported as well that EMT might be the cause of both intrinsic and acquired resistance by activating the PI3K pathway in the presence of the KRAS G12C inhibitor.
The triple combination of sotorasib, linsitinib (IGF1R inhibitor) and infigratinib (FGFR1 inhibitor), in contrast with doublet therapies (sotorasib with either linsitinib or infigatinib), showed efficacy by decreasing the cell proliferation and phosphorylation of AKT, ERK and S6 in H358-sotorasib-resistant cells [196]. Furthermore, this triple combination was shown to be the only treatment that reversed acquired resistance to sotorasib in vivo through the successful suppression of PI3K-AKT, MAPK and S6 phosphorylation [196].

14. Immune Check Point Inhibitors in KRAS-Mutant NSCLC, Current and Future Therapeutic Prospectives

As previously mentioned, KRAS-mutant NSCLC patients are expected to benefit from immune checkpoint inhibitors (ICIs) [136], and this has been demonstrated in particular in KRAS c.34G > T (p.G12C) mutant individuals [137,138].
Patients with KRAS c.34G > T (p.G12C) aberrations were included in frontline immunotherapy clinical trials including KEYNOTE-189, KEYNOTE-042, KEYNOTE-024 and CheckMate-227. An exploratory analysis was performed in KEYNOTE-042 aiming to assess the prevalence of KRAS mutations and their associations with ICI efficacy. Findings from this exploratory analysis showed that pembrolizumab monotherapy was associated with a 58% reduced risk of death (HR = 0.42 [95% CI, 0.22–0.81]) in patients with any KRAS mutation and a 72% (HR = 0.28 [95% CI, 0.09–0.86]) reduced risk in patients with the KRAS c.34G > T (p.G12C) mutation compared with chemotherapy [215].
The benefit from immunotherapy was limited by concomitant mutations like STK11 [174].
Sotorasib and adagrasib have shown enhanced antitumor efficacy when combined with anti-PD-1/PD-L1 therapy. Clinical trials are under development to investigate this interaction [216]. In genetically engineered mouse models with KRAS c.34G > T (p.G12C), the combination of adagrasib with an anti-PD-1 resulted in a complete and prolonged response in vivo and increased PFS compared with single-agent monotherapy [217].
Currently, multiple trials are investigating the potential of KRAS c.34G > T (p.G12C) inhibitors with existing immune checkpoint inhibitors (Table 9).

15. Co-Occurring Mutations Might Alter Clinical Benefit of ICIs

In preclinical studies, KRAS c.34G > T (p.G12C) direct inhibitors were noted to upregulate a pro-inflammatory tumor microenvironment and enhance anti-tumor T cell activity [166].
KRAS/STK11 aberrations, on the other hand, promote a so-called ‘cold tumor’ immune microenvironment, generally with limited PD-L1 expression and reduced infiltrating CD8+ T cells [218]. While TP53 co-mutation seems to be associated with a better response to ICIs, STK11 and KEAP1 co-mutations seem to impair response to ICIs
Indeed, TP53/KRAS co-mutated patients show higher PD-L1 expression, increased count of CD8 + TILs [141] and a higher mutational load compared with either STK11 or KEAP1 co-mutated patients; these findings may explain the better PFS in favor of patients harboring TP53/KRAS mutations treated with anti-PD-1 therapy compared with patients without mutations in TP53 and KRAS genes [141]. An exception is KRAS c.35G > A (p.G12D) mutation combined with TP53 mutations, which predict a lower TMB and as a consequence, less efficacy of immunotherapies in lung AC [219,220].
As aforementioned, STK11 and KEAP1 co-occurring mutations have been associated with ICI resistance in KRAS-mutant patients, independently of PD-L1 expression. Skoulidis et al. showed STK11/LKB1 alterations as a major driver of primary resistance to PD-1 blockade in KRAS-mutant patients, identifying STK11/LKB1 alterations as the most prevalent genomic driver of primary resistance to PD-1/PD-L1 inhibitors in KRAS-mutant NSCLC [87,174,219]
Patients harboring KRAS, STK11 and/or KEAP 1 co-mutations treated with car-boplatin-paclitaxel combined with either atezolizumab, bevacizumab or atezolizumab and bevacizumab had a worse outcome in terms of PFS and OS compared with STK11 and KEAP1 wt patients as showed in the post hoc analysis of the IMpower 150 trial [219].
Recently, Feng et al. reported a positive association between PD-L1 expression (i.e., ≥50%) and levels of stromal SHP2 (p = 0.039) [220]. In the same study, SHP2 was also found to promote a pro-inflammatory microenvironment through a higher expression of CD8+/CD4+ T-cells and CD64+ macrophages found in the stromal compartment of patients with high stromal SHP2 expression. Finally, the authors reported that despite small subgroup sizes, our subgroups analyses showed that patients with high SHP2 expression and PD-L1 ≥ 1% had significantly prolonged PFS and OS [220,221]. Based on the above-described preclinical results, several clinical trials are currently ongoing evaluating the association of anti-PD-1/PD-L1 agents with either sotorasib or adagrasib (Table 9).
The presence of the SWI/SNF related, matrix-associated, actin-dependent regulator of chromatin, subfamily A, member 4 (SMARCA4) are co-mutations in KRAS-mutated NSCLC (nearly 10% of cases) and are associated with poor immunotherapy outcomes [222,223,224,225]. This poor response could be associated with the influence of SMARCA4 alterations that decrease the production of the SMARCA4 protein, which is involved in chromatin remodeling and it is important in regulating the binding of transcription factors to DNA [226]. However, further studies are required in this setting.

16. Conclusions

Although neglected until few years ago, the clinical role of KRAS mutations has now gained a central role in the oncologic management of lung cancer patients. In recent years, it has become evident that the occurrence of KRAS mutations in lung cancer patients may reflect an opportunity for new drug inhibition, essentially with those directly targeting the G12C variant. Sotorasib is currently approved in this context, but other molecules are now under investigation in several clinical trials. Mechanisms of primary and acquired resistance, including mutations in KRAS as well as in other genes not belonging to the same MAP kinase pathway, may lead to new, innovative therapeutic options, with other TKI but including ICI as well, thus significantly prolonging patients’ survival.

Author Contributions

Writing—original draft preparation: E.C., S.E. and G.D.; writing—review and editing: E.C., S.E., G.D., M.C., L.W., M.F. and P.F.; supervision: L.W., M.F. and P.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

P.F. declares advisor activities (participation to advisory boards) for the following companies: Roche, Takeda, Pfizer, Novartis, MSD, BMS, Lilly, Janssen. L.W. declares advisor activities (participation to advisory boards) for the following companies: MSD, BMS, AstraZeneca, Roche, Amgen, Takeda, Merck, Novartis. M.C. declares advisor activities (participation to advisory boards) for the following companies: Servier. These activities do not influence the presentation or interpretation of the data reported in this review. All the other authors declare no conflicts of interest.

References

  1. Malumbres, M.; Barbacid, M. RAS oncogenes: The first 30 years. Nat. Cancer 2003, 3, 459–465, Erratum in: Nat. Rev. Cancer 2003, 3, 708. [Google Scholar] [CrossRef] [PubMed]
  2. Chen, K.; Zhang, Y.; Qian, L.; Wang, P. Emerging strategies to target RAS signaling in human cancer therapy. J. Hematol. Oncol. 2021, 14, 116. [Google Scholar] [CrossRef] [PubMed]
  3. Moore, A.R.; Rosenberg, S.C.; McCormick, F.; Malek, S. RAS-targeted therapies: Is the undruggable drugged? Nat. Rev. Drug Discov. 2020, 19, 533–552, Erratum in: Nat. Rev. Drug. Discov. 2020, 19, 902. [Google Scholar] [CrossRef]
  4. Wang, C.; Fakih, M. Targeting KRAS in Colorectal Cancer. Curr. Oncol. Rep. 2021, 23, 28. [Google Scholar] [CrossRef] [PubMed]
  5. Reck, M.; Carbone, D.; Garassino, M.; Barlesi, F. Targeting KRAS in non-small-cell lung cancer: Recent progress and new approaches. Ann. Oncol. 2021, 32, 1101–1110. [Google Scholar] [CrossRef] [PubMed]
  6. Prior, I.A.; Hood, F.E.; Hartley, J.L. The Frequency of Ras Mutations in Cancer. Cancer Res. 2020, 80, 2969–2974. [Google Scholar] [CrossRef]
  7. The AACR Project GENIE Consortium; André, F.; Arnedos, M.; Baras, A.S.; Baselga, J.; Bedard, P.L.; Berger, M.F.; Bierkens, M.; Calvo, F.; Cerami, E.; et al. AACR Project GENIE: Powering Precision Medicine through an International Consortium. Cancer Discov. 2017, 7, 818–831. [Google Scholar] [CrossRef]
  8. Uras, I.Z.; Moll, H.P.; Casanova, E. Targeting KRAS Mutant Non-Small-Cell Lung Cancer: Past, Present and Future. Int. J. Mol. Sci. 2020, 21, 4325. [Google Scholar] [CrossRef]
  9. Pao, W.; Wang, T.Y.; Riely, G.J.; Miller, V.A.; Pan, Q.; Ladanyi, M.; Zakowski, M.F.; Heelan, R.T.; Kris, M.G.; Varmus, H.E. KRAS Mutations and Primary Resistance of Lung Adenocarcinomas to Gefitinib or Erlotinib. PLoS Med. 2005, 2, e17. [Google Scholar] [CrossRef]
  10. Eberhard, D.A.; Johnson, B.E.; Amler, L.C.; Goddard, A.D.; Heldens, S.L.; Herbst, R.S.; Ince, W.L.; Jänne, P.A.; Januario, T.; Johnson, D.H.; et al. Mutations in the Epidermal Growth Factor Receptor and in KRAS Are Predictive and Prognostic Indicators in Patients With Non–Small-Cell Lung Cancer Treated With Chemotherapy Alone and in Combination With Erlotinib. J. Clin. Oncol. 2005, 23, 5900–5909. [Google Scholar] [CrossRef]
  11. Román, M.; Baraibar, I.; López, I.; Nadal, E.; Rolfo, C.; Vicent, S.; Gil-Bazo, I. KRAS oncogene in non-small cell lung cancer: Clinical perspectives on the treatment of an old target. Mol. Cancer 2018, 17, 33. [Google Scholar] [CrossRef] [PubMed]
  12. Yun, J.; Rago, C.; Cheong, I.; Pagliarini, R.; Angenendt, P.; Rajagopalan, H.; Schmidt, K.; Willson, J.K.V.; Markowitz, S.; Zhou, S.; et al. Glucose Deprivation Contributes to the Development of KRAS Pathway Mutations in Tumor Cells. Science 2009, 325, 1555–1559. [Google Scholar] [CrossRef] [PubMed]
  13. Ying, H.; Kimmelman, A.C.; Lyssiotis, C.A.; Hua, S.; Chu, G.C.; Fletcher-Sananikone, E.; Locasale, J.W.; Son, J.; Zhang, H.; Coloff, J.L.; et al. Oncogenic Kras Maintains Pancreatic Tumors through Regulation of Anabolic Glucose Metabolism. Cell 2012, 149, 656–670. [Google Scholar] [CrossRef] [PubMed]
  14. Nicholson, A.G.; Tsao, M.S.; Beasley, M.B.; Borczuk, A.C.; Brambilla, E.; Cooper, W.A.; Dacic, S.; Jain, D.; Kerr, K.M.; Lantuejoul, S.; et al. The 2021 WHO Classification of Lung Tumors: Impact of Advances since 2015. J. Thorac. Oncol. 2022, 17, 362–387. [Google Scholar] [CrossRef]
  15. Edkins, S.; O’Meara, S. Recurrent KRAS codon 146 mutations in human colorectal cancer. Cancer Biol. Ther. 2006, 5, 928–932. [Google Scholar] [CrossRef]
  16. Kris, M.G.; Johnson, B.E.; Kwiatkowski, D.J.; Iafrate, A.J.; Wistuba, I.I.; Aronson, S.L.; Shyr, Y.; Khuri, F.R.; Rudin, C.M.; Garon, E.B.; et al. Identification of driver mutations in tumor specimens from 1,000 patients with lung adenocarcinoma: The NCI’s Lung Cancer Mutation Consortium (LCMC). J. Clin. Oncol. 2011, 29, CRA7506. [Google Scholar] [CrossRef]
  17. The Cancer Genome Atlas (TCGA) Research Network. Comprehensive genomic characterization of squamous cell lung cancers. Nature 2012, 489, 519–525. [Google Scholar] [CrossRef]
  18. Judd, J.; Karim, N.A.; Khan, H.; Naqash, A.R.; Baca, Y.; Xiu, J.; VanderWalde, A.M.; Mamdani, H.; Raez, L.E.; Nagasaka, M.; et al. Characterization of KRAS Mutation Subtypes in Non–small Cell Lung Cancer. Mol. Cancer Ther. 2021, 20, 2577–2584. [Google Scholar] [CrossRef]
  19. Boch, C.; Kollmeier, J.; Roth, A.; Stephan-Falkenau, S.; Misch, D.; Grüning, W.; Bauer, T.T.; Mairinger, T. The frequency of EGFR and KRAS mutations in non-small cell lung cancer (NSCLC): Routine screening data for central Europe from a cohort study. BMJ Open 2013, 3, e002560. [Google Scholar] [CrossRef]
  20. Renaud, S.; Falcoz, P.-E.; Schaëffer, M.; Guenot, D.; Romain, B.; Olland, A.; Reeb, J.; Santelmo, N.; Chenard, M.-P.; Legrain, M.; et al. Prognostic value of the KRAS G12V mutation in 841 surgically resected Caucasian lung adenocarcinoma cases. Br. J. Cancer 2015, 113, 1206–1215. [Google Scholar] [CrossRef] [Green Version]
  21. Sebastian, M.; Eberhardt, W.E.; Hoffknecht, P.; Metzenmacher, M.; Wehler, T.; Kokowski, K.; Alt, J.; Schütte, W.; Büttner, R.; Heukamp, L.C.; et al. KRAS G12C-mutated advanced non-small cell lung cancer: A real-world cohort from the German prospective, observational, nation-wide CRISP Registry (AIO-TRK-0315). Lung Cancer 2021, 154, 51–61. [Google Scholar] [CrossRef] [PubMed]
  22. Zhou, W.; Christiani, D.C. East meets West: Ethnic differences in epidemiology and clinical behaviors of lung cancer between East Asians and Caucasians. Chin. J. Cancer 2011, 30, 287–292. [Google Scholar] [CrossRef] [PubMed]
  23. Dearden, S.; Stevens, J.; Wu, Y.L.; Blowers, D. Mutation incidence and coincidence in non small-cell lung cancer: Meta-analyses by ethnicity and histology (mutMap). Ann. Oncol. 2019, 24, 2371–2376. [Google Scholar] [CrossRef] [PubMed]
  24. Tímár, J. The clinical relevance of KRAS gene mutation in non-small-cell lung cancer. Curr. Opin. Oncol. 2014, 26, 138–144. [Google Scholar] [CrossRef]
  25. Yang, H.; Liang, S.-Q.; Schmid, R.A.; Peng, R.-W. New Horizons in KRAS-Mutant Lung Cancer: Dawn after Darkness. Front. Oncol. 2019, 9, 953. [Google Scholar] [CrossRef]
  26. Chatziandreou, I.; Tsioli, P.; Sakellariou, S.; Mourkioti, I.; Giannopoulou, I.; Levidou, G.; Korkolopoulou, P.; Patsouris, E.; Saetta, A.A. Comprehensive Molecular Analysis of NSCLC; Clinicopathological Associations. PLoS ONE 2015, 10, e0133859. [Google Scholar] [CrossRef]
  27. Barlesi, F.; Mazieres, J.; Merlio, J.-P.; Debieuvre, D.; Mosser, J.; Lena, H.; Ouafik, L.H.; Besse, B.; Rouquette, I.; Westeel, V.; et al. Routine molecular profiling of patients with advanced non-small-cell lung cancer: Results of a 1-year nationwide programme of the French Cooperative Thoracic Intergroup (IFCT). Lancet 2016, 387, 1415–1426. [Google Scholar] [CrossRef]
  28. Gobbini, E.; Galetta, D.; Tiseo, M.; Graziano, P.; Rossi, A.; Bria, E.; Di Maio, M.; Rossi, G.; Gregorc, V.; Riccardi, F.; et al. Molecular profiling in Italian patients with advanced non-small-cell lung cancer: An observational prospective study. Lung Cancer 2017, 111, 30–37. [Google Scholar] [CrossRef]
  29. Martorell, P.M.; Huerta, M.; Quilis, A.C.; Abellán, R.; Seda, E.; Blesa, S.; Chaves, F.J.; Beltrán, D.D.; Keränen, S.R.; Franco, J.; et al. Coexistence of EGFR, KRAS, BRAF, and PIK3CA Mutations and ALK Rearrangement in a Comprehensive Cohort of 326 Consecutive Spanish Nonsquamous NSCLC Patients. Clin. Lung Cancer 2017, 18, e395–e402. [Google Scholar] [CrossRef]
  30. Colombino, M.; Sardinian Lung Cancer (SLC) Study Group; Paliogiannis, P.; Cossu, A.; Santeufemia, D.A.; Sini, M.C.; Casula, M.; Palomba, G.; Manca, A.; Pisano, M.; et al. EGFR, KRAS, BRAF, ALK, and cMET genetic alterations in 1440 Sardinian patients with lung adenocarcinoma. BMC Pulm. Med. 2019, 19, 209. [Google Scholar] [CrossRef] [Green Version]
  31. Elghissassi, I.; Inrhaoun, H.; Boukir, A.; Kettani, F.; Gamra, L.; Mestari, A.; Jabri, L.; Bensouda, Y.; Mrabti, H.; Errihani, H. Frequency and Spectrum of KRAS Mutations in Moroccan Patients with Lung Adenocarcinoma. ISRN Oncol. 2014, 2014, 192493. [Google Scholar] [CrossRef] [PubMed]
  32. Kim, E.Y.; Kim, A.; Kim, S.K.; Kim, H.J.; Chang, J.; Ahn, C.M.; Lee, J.S.; Shim, H.S.; Chang, Y.S. KRAS oncogene substitutions in Korean NSCLC patients: Clinical implication and relationship with pAKT and RalGTPases expression. Lung Cancer 2014, 85, 299–305. [Google Scholar] [CrossRef] [PubMed]
  33. 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 2014, 110, 2812–2820. [Google Scholar] [CrossRef] [PubMed]
  34. Nadal, E.; Chen, G.; Prensner, J.R.; Shiratsuchi, H.; Sam, C.; Zhao, L.; Kalemkerian, G.P.; Brenner, D.; Lin, J.; Reddy, R.M.; et al. KRAS-G12C Mutation Is Associated with Poor Outcome in Surgically Resected Lung Adenocarcinoma. J. Thorac. Oncol. 2014, 9, 1513–1522. [Google Scholar] [CrossRef]
  35. Arrieta, O.; Ramírez-Tirado, L.-A.; Báez-Saldaña, R.; Peña-Curiel, O.; Soca-Chafre, G.; Macedo-Perez, E.-O. Different mutation profiles and clinical characteristics among Hispanic patients with non-small cell lung cancer could explain the “Hispanic paradox”. Lung Cancer 2015, 90, 161–166. [Google Scholar] [CrossRef]
  36. Dhieb, D.; Belguith, I.; Capelli, L.; Chiadini, E.; Canale, M.; Bravaccini, S.; Yangui, I.; Boudawara, O.; Jlidi, R.; Boudawara, T.; et al. Analysis of Genetic Alterations in Tunisian Patients with Lung Adenocarcinoma. Cells 2019, 8, 514. [Google Scholar] [CrossRef]
  37. Izumi, M.; Suzumura, T.; Ogawa, K.; Matsumoto, Y.; Sawa, K.; Yoshimoto, N.; Tani, Y.; Watanabe, T.; Kaneda, H.; Mitsuoka, S.; et al. Differences in molecular epidemiology of lung cancer among ethnicities (Asian vs. Caucasian). J. Thorac. Dis. 2020, 12, 3776–3784. [Google Scholar] [CrossRef]
  38. Masykura, N.; Zaini, J.; Syahruddin, E.; Andarini, S.L.; Hudoyo, A.; Yasril, R.; Ridwanuloh, A.; Hidajat, H.; Nurwidya, F.; Utomo, A. Impact of smoking on frequency and spectrum of K-RAS and EGFR mutations in treatment naive Indonesian lung cancer patients. Lung Cancer Targets Ther. 2019, 10, 57–66. [Google Scholar] [CrossRef]
  39. Matsumoto, S.; Tsuchihara, K.; Yoh, K.; Zenke, Y.; Kohno, T.; Ishii, G.; Tsuta, K.; Umemura, S.; Niho, S.; Ohmatsu, H.; et al. A new nationwide genomic screening system in Japan for the development of targeted therapies against advanced non-small lung cancers with rare driver mutations. J. Clin. Oncol. 2014, 32, 11007. [Google Scholar] [CrossRef]
  40. Mehta, A.; Dobersch, S.; Romero-Olmedo, A.J.; Barreto, G. Epigenetics in lung cancer diagnosis and therapy. Cancer Metastasis Rev. 2015, 34, 229–241. [Google Scholar] [CrossRef]
  41. Perri, F.; Pisconti, S.; Scarpati, G.D.V. P53 mutations and cancer: A tight linkage. Ann. Transl. Med. 2016, 4, 522. [Google Scholar] [CrossRef] [PubMed]
  42. Dang, A.-T.H.; Tran, V.-U.; Tran, T.-T.; Pham, H.-A.T.; Le, D.-T.; Nguyen, L.; Nguyen, N.-V.; Nguyen, T.-H.T.; Nguyen, C.; Le, H.T.; et al. Actionable Mutation Profiles of Non-Small Cell Lung Cancer patients from Vietnamese population. Sci. Rep. 2020, 10, 2707. [Google Scholar] [CrossRef] [PubMed]
  43. Lee, J.; Tan, A.C.; Zhou, S.; Yoon, S.; Liu, S.; Masuda, K.; Hayashi, H.; Batra, U.; Kim, D.-W.; Goto, Y.; et al. Clinical Characteristics and Outcomes in Advanced KRAS-Mutated NSCLC: A Multicenter Collaboration in Asia (ATORG-005). JTO Clin. Res. Rep. 2021, 3, 100261. [Google Scholar] [CrossRef] [PubMed]
  44. McIntyre, A.; Ganti, A.K. Lung cancer-A global perspective. J. Surg. Oncol. 2017, 115, 550–554. [Google Scholar] [CrossRef] [PubMed]
  45. Mezni, F.; Mlika, M.; Boussen, H.; Ghedira, H.; Fenniche, S.; Faten, T.; Loriot, M.-A. About molecular profile of lung cancer in Tunisian patients. J. Immunoass. Immunochem. 2018, 39, 99–107. [Google Scholar] [CrossRef]
  46. Reinersman, J.M.; Johnson, M.L.; Riely, G.J.; Chitale, D.A.; Nicastri, A.D.; Soff, G.A.; Schwartz, A.G.; Sima, C.S.; Ayalew, G.; Lau, C.; et al. Frequency of EGFR and KRAS Mutations in Lung Adenocarcinomas in African Americans. J. Thorac. Oncol. 2011, 6, 28–31. [Google Scholar] [CrossRef]
  47. McQuitty, E.; Zhang, W.; Hendrickson, H.; Tio, F.O.; Jagirdar, J.; Olsen, R.; Cagle, P.T. Lung Adenocarcinoma Biomarker Incidence in Hispanic Versus Non-Hispanic White Patients. Arch. Pathol. Lab. Med. 2014, 138, 390–394. [Google Scholar] [CrossRef]
  48. Arrieta, O.; Cardona, A.F.; Martín, C.; Más-López, L.; Corrales-Rodríguez, L.; Bramuglia, G.; Castillo-Fernandez, O.; Meyerson, M.; Amieva-Rivera, E.; Campos-Parra, A.D.; et al. Updated Frequency of EGFR and KRAS Mutations in NonSmall-Cell Lung Cancer in Latin America: The Latin-American Consortium for the Investigation of Lung Cancer (CLICaP). J. Thorac. Oncol. 2015, 10, 838–843. [Google Scholar] [CrossRef]
  49. Bacchi, C.E.; Ciol, H.; Queiroga, E.M.; Benine, L.C.; Silva, L.H.; Ojopi, E.B. Epidermal growth factor receptor and KRAS mutations in Brazilian lung cancer patients. Clinics 2012, 67, 419–424. [Google Scholar] [CrossRef]
  50. Zheng, R.; Yin, Z.; Alhatem, A.; Lyle, D.; You, B.; Jiang, A.S.; Liu, D.; Jobbagy, Z.; Wang, Q.; Aisner, S.; et al. Epidemiologic Features of NSCLC Gene Alterations in Hispanic Patients from Puerto Rico. Cancers 2020, 12, 3492. [Google Scholar] [CrossRef]
  51. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  52. Siegel, R.; Naishadham, D.; Jemal, A. Cancer statistics. CA Cancer J. Clin. 2012, 62, 10–29. [Google Scholar] [CrossRef] [PubMed]
  53. Egleston, B.L.; Meireles, S.I.; Flieder, D.B.; Clapper, M.L. Population-Based Trends in Lung Cancer Incidence in Women. Semin. Oncol. 2009, 36, 506–515. [Google Scholar] [CrossRef] [PubMed]
  54. Kligerman, S.; White, C. Epidemiology of Lung Cancer in Women: Risk Factors, Survival, and Screening. Am. J. Roentgenol. 2011, 196, 287–295. [Google Scholar] [CrossRef]
  55. Hitchman, S.C.; Fong, G.T. Gender empowerment and female-to-male smoking prevalence ratios. Bull. World Health Organ. 2011, 89, 195–202. [Google Scholar] [CrossRef]
  56. Rivera, M.P. Lung Cancer in Women: Differences in Epidemiology, Biology, Histology, and Treatment Outcomes. Semin. Respir. Crit. Care Med. 2013, 34, 792–801. [Google Scholar] [CrossRef]
  57. Doll, R.; Hill, A.B. Smoking and Carcinoma of the Lung. Br. Med. J. 1950, 2, 739–748. [Google Scholar] [CrossRef]
  58. Chapman, A.M.; Sun, K.Y.; Ruestow, P.; Cowan, D.M.; Madl, A.K. Lung cancer mutation profile of EGFR, ALK, and KRAS: Meta-analysis and comparison of never and ever smokers. Lung Cancer 2016, 102, 122–134. [Google Scholar] [CrossRef]
  59. Redig, A.J.; Chambers, E.S.; Lydon, C.A.; Dahlberg, S.E.; Alden, R.S.; Janne, P.A. Genomic complexity in KRAS mutant non-small cell lung cancer (NSCLC) from never/light-smokers v smokers [abstract no. 9087]. J. Clin. Oncol. 2016, 34, 9087. [Google Scholar] [CrossRef]
  60. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=STK11 (accessed on 14 July 2022).
  61. Saber, A.; Van Der Wekken, A.J.; Kerner, G.S.M.A.; Berge, M.V.D.; Timens, W.; Schuuring, E.; Ter Elst, A.; Berg, A.V.D.; Hiltermann, T.J.N.; Groen, H.J.M. Chronic Obstructive Pulmonary Disease Is Not Associated with KRAS Mutations in Non-Small Cell Lung Cancer. PLoS ONE 2016, 11, e0152317. [Google Scholar] [CrossRef]
  62. Keohavong, P.; Di, Y.P. Pulmonary Inflammation and KRAS Mutation in Lung Cancer. Adv. Exp. Med. Biol. 2021, 1303, 71–87. [Google Scholar] [CrossRef] [PubMed]
  63. Ahrendt, S.A.; Decker, P.A.; Alawi, E.A.; Zhu, Y.-R.; Sanchez-Cespedes, M.; Yang, S.C.; Haasler, G.B.; Kajdacsy-Balla, A.; Demeure, M.J.; Sidransky, D. Cigarette smoking is strongly associated with mutation of the K-ras gene in patients with primary adenocarcinoma of the lung. Cancer 2001, 92, 1525–1530. [Google Scholar] [CrossRef]
  64. Riely, G.J.; Kris, M.G.; Rosenbaum, D.; Marks, J.; Li, A.; Chitale, D.A.; Nafa, K.; Riedel, E.R.; Hsu, M.; Pao, W.; et al. Frequency and Distinctive Spectrum of KRAS Mutations in Never Smokers with Lung Adenocarcinoma. Clin. Cancer Res. 2008, 14, 5731–5734. [Google Scholar] [CrossRef]
  65. Forbes, S.A.; Bindal, N.; Bamford, S.; Cole, C.; Kok, C.Y.; Beare, D.; Jia, M.; Shepherd, R.; Leung, K.; Menzies, A.; et al. COSMIC: Mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 2011, 39, D945–D950. [Google Scholar] [CrossRef] [PubMed]
  66. Garassino, M.C.; Marabese, M.; Rusconi, P.; Rulli, E.; Martelli, O.; Farina, G.; Scanni, A.; Broggini, M. Different types of K-Ras mutations could affect drug sensitivity and tumour behaviour in non-small-cell lung cancer. Ann. Oncol. 2011, 22, 235–237. [Google Scholar] [CrossRef]
  67. Matakidou, A.; Eisen, T.; Houlston, R. Systematic review of the relationship between family history and lung cancer risk. Br. J. Cancer 2005, 93, 825–833. [Google Scholar] [CrossRef] [PubMed]
  68. Coté, M.L.; Liu, M.; Bonassi, S.; Neri, M.; Schwartz, A.G.; Christiani, D.C.; Spitz, M.R.; Muscat, J.E.; Rennert, G.; Aben, K.K.; et al. Increased risk of lung cancer in individuals with a family history of the disease: A pooled analysis from the International Lung Cancer Consortium. Eur. J. Cancer 2012, 48, 1957–1968. [Google Scholar] [CrossRef]
  69. Santos, E.; Martin-Zanca, D.; Reddy, E.P.; Pierotti, M.A.; Della Porta, G.; Barbacid, M. Malignant activation of a K-ras oncogene in lung carcinoma but not in normal tissue of the same patient. Science 1984, 223, 661–664. [Google Scholar] [CrossRef]
  70. Brenner, D.R.; Boffetta, P.; Duell, E.J.; Bickeböller, H.; Rosenberger, A.; McCormack, V.; Muscat, J.E.; Yang, P.; Wichmann, H.-E.; Brueske-Hohlfeld, I.; et al. Previous Lung Diseases and Lung Cancer Risk: A Pooled Analysis from the International Lung Cancer Consortium. Am. J. Epidemiol. 2012, 176, 573–585. [Google Scholar] [CrossRef]
  71. Serraino, D.; Boschini, A.; Carrieri, P.; Pradier, C.; Dorrucci, M.; Maso, L.D.; Ballarini, P.; Pezzotti, P.; Smacchia, C.; Pesce, A.; et al. Cancer risk among men with, or at risk of, HIV infection in southern Europe. AIDS 2000, 14, 553–559. [Google Scholar] [CrossRef]
  72. Jones, R.N.; Hughes, J.M.; Weill, H. Asbestos exposure, asbestosis, and asbestos-attributable lung cancer. Thorax 1996, 51, S9–S15. [Google Scholar] [CrossRef] [PubMed]
  73. Egilman, D.; Reinert, A. Lung cancer and asbestos exposure: Asbestosis is not necessary. Am. J. Ind. Med. 1996, 30, 398–406. [Google Scholar] [CrossRef]
  74. Narayanan, P.K.; Goodwin, E.H.; Lehnert, B.E. Alpha particles initiate biological production of superoxide anions and hydrogen peroxide in human cells. Cancer Res. 1997, 57, 3963–3971. [Google Scholar]
  75. Consonni, D.; De Matteis, S.; Pesatori, A.C.; Bertazzi, P.A.; Olsson, A.C.; Kromhout, H.; Peters, S.; Vermeulen, R.C.; Pesch, B.; Brüning, T.; et al. Lung cancer risk among brick layers in a pooled analysis of case-control studies. Int. J. Cancer 2015, 136, 360–371. [Google Scholar] [CrossRef] [PubMed]
  76. Putila, J.J.; Guo, N.L. Association of Arsenic Exposure with Lung Cancer Incidence Rates in the United States. PLoS ONE 2011, 6, e25886. [Google Scholar] [CrossRef]
  77. Omenn, G.S.; Goodman, G.E.; Thornquist, M.D.; Balmes, J.; Cullen, M.R.; Glass, A.; Keogh, J.P.; Meyskens, F.L.; Valanis, B.; Williams, J.H.; et al. Effects of a combination of betacarotene and vitamin A on lung cancer and cardiovascular disease. N. Engl. J. Med. 1996, 334, 1150–1155. [Google Scholar] [CrossRef]
  78. Woodson, K.; Tangrea, J.A.; Barrett, M.J.; Virtamo, J.; Taylor, P.R.; Albanes, D. Serum alpha-tocopherol and subsequent risk of lung cancer among male smokers. J. Natl. Cancer Inst. 1999, 91, 1738–1743. [Google Scholar] [CrossRef]
  79. Bagnardi, V.; Rota, M.; Botteri, E.; Tramacere, I.; Islami, F.; Fedirko, V.; Scotti, L.; Jenab, M.; Turati, F.; Pasquali, E.; et al. Alcohol consumption and site-specific cancer risk: A comprehensive dose–response meta-analysis. Br. J. Cancer 2015, 112, 580–593. [Google Scholar] [CrossRef]
  80. Aldington, S.; Harwood, M.; Cox, B.; Weatherall, M.; Beckert, L.; Hansell, A.; Pritchard, A.; Robinson, G.; Beasley, R.; on behalf of the Cannabis and Respiratory Disease Research Group. Cannabis use and risk of lung cancer: A case-control study. Eur. Respir. J. 2008, 31, 280–286. [Google Scholar] [CrossRef]
  81. Simanshu, D.K.; Nissley, D.V.; McCormick, F. RAS Proteins and Their Regulators in Human Disease. Cell 2017, 170, 17–33. [Google Scholar] [CrossRef]
  82. Khan, I.; Rhett, J.M.; O’Bryan, J.P. Therapeutic targeting of RAS: New hope for drugging the “undruggable”. Biochim. Biophys. Acta Mol. Cell Res. 2019, 1867, 118570. [Google Scholar] [CrossRef] [PubMed]
  83. Soh, J.; Okumura, N.; Lockwood, W.W.; Yamamoto, H.; Shigematsu, H.; Zhang, W.; Chari, R.; Shames, D.S.; Tang, X.; MacAulay, C.; et al. Oncogene Mutations, Copy Number Gains and Mutant Allele Specific Imbalance (MASI) Frequently Occur Together in Tumor Cells. PLoS ONE 2009, 4, e7464. [Google Scholar] [CrossRef] [PubMed]
  84. Ihle, N.T.; Byers, L.A.; Kim, E.S.; Saintigny, P.; Lee, J.J.; Blumenschein, G.R.; Tsao, A.; Liu, S.; Larsen, J.E.; Wang, J.; et al. Effect of KRAS Oncogene Substitutions on Protein Behavior: Implications for Signaling and Clinical Outcome. JNCI J. Natl. Cancer Inst. 2012, 104, 228–239. [Google Scholar] [CrossRef] [PubMed]
  85. Slagter, S.N.; Van Vliet-Ostaptchouk, J.V.; Vonk, J.M.; Boezen, H.M.; Dullaart, R.P.F.; Kobold, A.C.M.; Feskens, E.J.; Van Beek, A.P.; Van Der Klauw, M.M.; Wolffenbuttel, B.H. Associations between smoking, components of metabolic syndrome and lipoprotein particle size. BMC Med. 2013, 11, 195. [Google Scholar] [CrossRef] [PubMed]
  86. Yilmaz, A.; Mohamed, N.; Patterson, K.A.; Tang, Y.; Shilo, K.; Villalona-Calero, M.A.; Davis, M.E.; Zhou, X.-P.; Frankel, W.; Otterson, G.A.; et al. Clinical and Metabolic Parameters in Non-Small Cell Lung Carcinoma and Colorectal Cancer Patients with and without KRAS Mutations. Int. J. Environ. Res. Public Health 2014, 11, 8645–8660. [Google Scholar] [CrossRef] [PubMed]
  87. Ferrer, I.; Zugazagoitia, J.; Herbertz, S.; John, W.; Paz-Ares, L.; Schmid-Bindert, G. KRAS-Mutant non-small cell lung cancer: From biology to therapy. Lung Cancer 2018, 124, 53–64. [Google Scholar] [CrossRef]
  88. Aran, V.; Zalis, M.; Montella, T.; de Sousa, C.A.M.; Ferrari, B.L.; Gil Ferreira, C. Evaluation of KRAS Concomitant Mutations in Advanced Lung Adenocarcinoma Patients. Medicina 2021, 57, 1039. [Google Scholar] [CrossRef]
  89. Araujo, L.H.; Souza, B.M.; Leite, L.R.; Parma, S.A.F.; Lopes, N.P.; Malta, F.S.V.; Freire, M.C.M. Molecular profile of KRAS G12C-mutant colorectal and non-small-cell lung cancer. BMC Cancer 2021, 21, 193. [Google Scholar] [CrossRef]
  90. Arbour, K.C.; Jordan, E.; Kim, H.R.; Dienstag, J.; Yu, H.A.; Sanchez-Vega, F.; Lito, P.; Berger, M.; Solit, D.B.; Hellmann, M.; et al. Effects of Co-occurring Genomic Alterations on Outcomes in Patients with KRAS-Mutant Non–Small Cell Lung Cancer. Clin. Cancer Res. 2018, 24, 334–340. [Google Scholar] [CrossRef]
  91. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=KEAP1 (accessed on 14 July 2022).
  92. Skoulidis, F.; Byers, L.A.; Diao, L.; Papadimitrakopoulou, V.A.; Tong, P.; Izzo, J.; Behrens, C.; Kadara, H.; Parra, E.R.; Canales, J.R.; et al. Co-occurring Genomic Alterations Define Major Subsets of KRAS-Mutant Lung Adenocarcinoma with Distinct Biology, Immune Profiles, and Therapeutic Vulnerabilities. Cancer Discov. 2015, 5, 860–877. [Google Scholar] [CrossRef]
  93. Liu, C.; Zheng, S.; Jin, R.; Wang, X.; Wang, F.; Zang, R.; Xu, H.; Lu, Z.; Huang, J.; Lei, Y.; et al. The superior efficacy of anti-PD-1/PD-L1 immunotherapy in KRAS-mutant non-small cell lung cancer that correlates with an inflammatory phenotype and increased immunogenicity. Cancer Lett. 2020, 470, 95–105. [Google Scholar] [CrossRef] [PubMed]
  94. Chen, N.; Fang, W.; Lin, Z.; Peng, P.; Wang, J.; Zhan, J.; Hong, S.; Huang, J.; Liu, L.; Sheng, J.; et al. KRAS mutation-induced upregulation of PD-L1 mediates immune escape in human lung adenocarcinoma. Cancer Immunol. Immunother. 2017, 66, 1175–1187. [Google Scholar] [CrossRef] [PubMed]
  95. Schoenfeld, A.J.; Rizvi, H.; Bandlamudi, C.; Sauter, J.L.; Travis, W.D.; Rekhtman, N.; Plodkowski, A.J.; Perez-Johnston, R.; Sawan, P.; Beras, A.; et al. Clinical and molecular correlates of PD-L1 expression in patients with lung adenocarcinomas. Ann. Oncol. 2020, 31, 599–608. [Google Scholar] [CrossRef] [PubMed]
  96. Villaruz, L.C.; Socinski, M.A.; Cunningham, D.E.; Chiosea, S.I.; Burns, T.F.; Siegfried, J.M.; Dacic, S. The prognostic and predictive value of KRAS oncogene substitutions in lung adenocarcinoma. Cancer 2013, 119, 2268–2274. [Google Scholar] [CrossRef]
  97. Shepherd, F.A.; Domerg, C.; Hainaut, P.; Jänne, P.A.; Pignon, J.-P.; Graziano, S.; Douillard, J.-Y.; 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. 2013, 31, 2173–2181. [Google Scholar] [CrossRef]
  98. Mascaux, C.; Iannino, N.; Martin, B.; Paesmans, M.; Berghmans, T.; Dusart, M.; Haller, A.; Lothaire, P.; Meert, A.P.; Noel, S.; et al. The role of RAS oncogene in survival of patients with lung cancer: A systematic review of the literature with metanalysis. Br. J. Cancer 2005, 92, 131–139. [Google Scholar] [CrossRef]
  99. Gautschi, O.; Huegli, B.; Ziegler, A.; Gugger, M.; Heighway, J.; Ratschiller, D.; Mack, P.; Gumerlock, P.; Kung, H.; Stahel, R.; et al. Origin and prognostic value of circulating KRAS mutations in lung cancer patients. Cancer Lett. 2007, 254, 265–273. [Google Scholar] [CrossRef]
  100. Sasaki, H.; Okuda, K.; Kawano, O.; Endo, K.; Yukiue, H.; Yokoyama, T.; Yano, M.; Fujii, Y. Nras and Kras mutation in Japanese lung cancer patients: Genotyping analysis using LightCycler. Oncol. Rep. 2007, 18, 623–628. [Google Scholar] [CrossRef]
  101. Aredo, J.V.; Padda, S.K.; Kunder, C.A.; Han, S.S.; Neal, J.W.; Shrager, J.B.; Wakelee, H.A. Impact of KRAS mutation subtype and concurrent pathogenic mutations on non-small cell lung cancer outcomes. Lung Cancer 2019, 133, 144–150. [Google Scholar] [CrossRef]
  102. Park, S.; Kim, J.-Y.; Lee, S.-H.; Suh, B.; Keam, B.; Kim, T.M.; Kim, N.-W.; Heo, D.S. KRAS G12C mutation as a poor prognostic marker of pemetrexed treatment in non-small cell lung cancer. Korean J. Intern. Med. 2017, 32, 514–522. [Google Scholar] [CrossRef]
  103. Izar, B.; Zhou, H.; Heist, R.S.; Azzoli, C.G.; Muzikansky, A.; Scribner, E.E.; Bernardo, L.A.; Dias-Santagata, D.; Iafrate, A.J.; Lanuti, M. The Prognostic Impact of KRAS, Its Codon and Amino Acid Specific Mutations, on Survival in Resected Stage I Lung Adenocarcinoma. J. Thorac. Oncol. 2014, 9, 1363–1369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Yu, H.A.; Sima, C.S.; Shen, R.; Kass, S.; Gainor, J.; Shaw, A.; Hames, M.; Iams, W.; Aston, J.; Lovly, C.; et al. Prognostic Impact of KRAS Mutation Subtypes in 677 Patients with Metastatic Lung Adenocarcinomas. J. Thorac. Oncol. 2015, 10, 431–437. [Google Scholar] [CrossRef] [PubMed]
  105. Tao, L.; Miao, R.; Mekhail, T.; Sun, J.; Meng, L.; Fang, C.; Guan, J.; Jain, A.; Du, Y.; Allen, A.; et al. Prognostic Value of KRAS Mutation Subtypes and PD-L1 Expression in Patients With Lung Adenocarcinoma. Clin. Lung Cancer 2021, 22, e506–e511. [Google Scholar] [CrossRef] [PubMed]
  106. Nusrat, M.; Roszik, J.; Holla, V.; Cai, T.; Hong, M.; Coker, O.; Johnson, B.; Ahnert, J.R.; Janku, F.; Kopetz, S.; et al. Therapeutic vulnerabilities among KRAS G12C mutant (mut) advanced cancers based on co-alteration (co-alt) patterns. J. Clin. Oncol. 2020, 38, 3625. [Google Scholar] [CrossRef]
  107. Scheffler, M.; Ihle, M.A.; Hein, R.; Merkelbach-Bruse, S.; Scheel, A.H.; Siemanowski, J.; Brägelmann, J.; Kron, A.; Abedpour, N.; Ueckeroth, F.; et al. K-ras Mutation Subtypes in NSCLC and Associated Co-occuring Mutations in Other Oncogenic Pathways. J. Thorac. Oncol. 2019, 14, 606–616. [Google Scholar] [CrossRef]
  108. Wang, A.; Wang, H.; Liu, Y.; Zhao, M.; Zhang, H.; Lu, Z.; Fang, Y.; Chen, X.; Liu, G. The prognostic value of PD-L1 expression for non-small cell lung cancer patients: A meta-analysis. Eur. J. Surg. Oncol. 2015, 41, 450–456. [Google Scholar] [CrossRef]
  109. Cefalì, M.; Epistolio, S.; Ramelli, G.; Mangan, D.; Molinari, F.; Martin, V.; Freguia, S.; Mazzucchelli, L.; Froesch, P.; Frattini, M.; et al. Correlation of KRAS G12C Mutation and High PD-L1 Expression with Clinical Outcome in NSCLC Patients Treated with Anti-PD1 Immunotherapy. J. Clin. Med. 2022, 11, 1627. [Google Scholar] [CrossRef]
  110. Rodenhuis, S.; Boerrigter, L.; Top, B.; Slebos, R.J.; Mooi, W.J.; Veer, L.V.; Van Zandwijk, N. Mutational activation of the K-ras oncogene and the effect of chemotherapy in advanced adenocarcinoma of the lung: A prospective study. J. Clin. Oncol. 1997, 15, 285–291. [Google Scholar] [CrossRef]
  111. Schiller, J.H.; Adak, S.; Feins, R.H.; Keller, S.M.; Fry, W.A.; Livingston, R.B.; Hammond, M.E.M.; Wolf, B.; Sabatini, L.; Jett, J.; et al. Lack of Prognostic Significance of p53 and K-ras Mutations in Primary Resected Non–Small-Cell Lung Cancer on E4592: A Laboratory Ancillary Study on an Eastern Cooperative Oncology Group Prospective Randomized Trial of Postoperative Adjuvant Therapy. J. Clin. Oncol. 2001, 19, 448–457. [Google Scholar] [CrossRef]
  112. Sun, J.-M.; Hwang, D.W.; Ahn, J.S.; Ahn, M.-J.; Park, K. Prognostic and Predictive Value of KRAS Mutations in Advanced Non-Small Cell Lung Cancer. PLoS ONE 2013, 8, e64816. [Google Scholar] [CrossRef]
  113. Tsao, M.-S.; Aviel-Ronen, S.; Ding, K.; Lau, D.; Liu, N.; Sakurada, A.; Whitehead, M.; Zhu, C.-Q.; Livingston, R.; Johnson, D.H.; et al. Prognostic and Predictive Importance of p53 and RAS for Adjuvant Chemotherapy in Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2007, 25, 5240–5247. [Google Scholar] [CrossRef] [PubMed]
  114. Chaft, J.E.; Rusch, V.; Ginsberg, M.S.; Paik, P.K.; Finley, D.J.; Kris, M.G.; Price, K.A.; Azzoli, C.G.; Fury, M.G.; Riely, G.J.; et al. Phase II Trial of Neoadjuvant Bevacizumab Plus Chemotherapy and Adjuvant Bevacizumab in Patients with Resectable Nonsquamous Non–Small-Cell Lung Cancers. J. Thorac. Oncol. 2013, 8, 1084–1090. [Google Scholar] [CrossRef] [PubMed]
  115. Ghimessy, Á.K.; Gellert, Á.; Schlegl, E.; Hegedus, B.; Raso, E.; Barbai, T.; Timar, J.; Ostoros, G.; Megyesfalvi, Z.; Gieszer, B.; et al. KRAS Mutations Predict Response and Outcome in Advanced Lung Adenocarcinoma Patients Receiving First-Line Bevacizumab and Platinum-Based Chemotherapy. Cancers 2019, 11, 1514. [Google Scholar] [CrossRef] [PubMed]
  116. D’Incecco, A.; Andreozzi, M.; Ludovini, V.; Rossi, E.; Capodanno, A.; Landi, L.; Tibaldi, C.; Minuti, G.; Salvini, J.; Coppi, E.; et al. PD-1 and PD-L1 expression in molecularly selected non-small-cell lung cancer patients. Br. J. Cancer 2015, 112, 95–102. [Google Scholar] [CrossRef]
  117. Zalcman, G.; Beau-Faller, M.; Creveuil, C.; De Fraipont, F.; Mounawar, M.; Richard, N.; Bergot, E.; Favrot, M.; Morin, F.; Milleron, B. Use of Ras effector RASSF1A promoter gene methylation and chromosome 9p loss of heterozygosity (LOH) to predict progression-free survival (PFS) in perioperative chemotherapy (CT) phase III trial IFCT-0002 in resectable non-small cell lung cancer. J. Clin. Oncol. 2008, 26, 7500. [Google Scholar] [CrossRef]
  118. Winton, T.; Livingston, R.; Johnson, D.; Rigas, J.; Johnston, M.; Butts, C.; Cormier, Y.; Goss, G.; Inculet, R.; Vallieres, E.; et al. Vinorelbine plus cisplatin vs. observation in resected non-small-cell lung cancer. N. Engl. J. Med. 2005, 352, 2589–2597. [Google Scholar] [CrossRef]
  119. Jia, Y.; Jiang, T.; Li, X.; Zhao, C.; Zhang, L.; Zhao, S.; Liu, X.; Qiao, M.; Luo, J.; Shi, J.; et al. Characterization of distinct types of KRAS mutation and its impact on first-line platinum-based chemotherapy in Chinese patients with advanced non-small cell lung cancer. Oncol. Lett. 2017, 14, 6525–6532. [Google Scholar]
  120. Cserepes, M.; Ostoros, G.; Lohinai, Z.; Raso, E.; Barbai, T.; Timar, J.; Rozsas, A.; Moldvay, J.; Kovalszky, I.; Fabian, K.; et al. Subtype-specific KRAS mutations in advanced lung adenocarcinoma: A retrospective study of patients treated with platinum-based chemotherapy. Eur. J. Cancer 2014, 50, 1819–1828. [Google Scholar] [CrossRef]
  121. Kranenburg, O.; Gebbink, M.F.; Voest, E.E. Stimulation of angiogenesis by Ras proteins. Biochim. Biophys. Acta 2004, 1654, 23–37. [Google Scholar] [CrossRef]
  122. Tang, Y.; Kim, M.; Carrasco, D.; Kung, A.L.; Chin, L.; Weissleder, R. In vivo Assessment of RAS-Dependent Maintenance of Tumor Angiogenesis by Real-time Magnetic Resonance Imaging. Cancer Res. 2005, 65, 8324–8330. [Google Scholar] [CrossRef]
  123. Sparmann, A.; Bar-Sagi, D. Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell 2004, 6, 447–458. [Google Scholar] [CrossRef] [PubMed]
  124. Matsuo, Y.; Campbell, P.M.; Brekken, R.A.; Sung, B.; Ouellette, M.M.; Fleming, J.B.; Aggarwal, B.B.; Der, C.J.; Guha, S. K-Ras Promotes Angiogenesis Mediated by Immortalized Human Pancreatic Epithelial Cells through Mitogen-Activated Protein Kinase Signaling Pathways. Mol. Cancer Res. 2009, 7, 799–808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Sunaga, N.; Imai, H.; Shimizu, K.; Shames, D.S.; Kakegawa, S.; Girard, L.; Sato, M.; Kaira, K.; Ishizuka, T.; Gazdar, A.F.; et al. Oncogenic KRAS-induced interleukin-8 overexpression promotes cell growth and migration and contributes to aggressive phenotypes of non-small cell lung cancer. Int. J. Cancer 2012, 130, 1733–1744. [Google Scholar] [CrossRef] [PubMed]
  126. Murillo, M.M.; Zelenay, S.; Nye, E.; Castellano, E.; Lassailly, F.; Stamp, G.; Downward, J. RAS interaction with PI3K p110α is required for tumor-induced angiogenesis. J. Clin. Investig. 2014, 124, 3601–3611. [Google Scholar] [CrossRef] [PubMed]
  127. Chin, L.; Tam, A.; Pomerantz, J.; Wong, M.; Holash, J.; Bardeesy, N.; Shen, Q.; O’Hagan, R.; Pantginis, J.; Zhou, H.; et al. Essential role for oncogenic Ras in tumour maintenance. Nature 1999, 400, 468–472. [Google Scholar] [CrossRef]
  128. Okada, F.; Rak, J.W.; Croix, B.S.; Lieubeau, B.; Kaya, M.; Roncari, L.; Shirasawa, S.; Sasazuki, T.; Kerbel, R.S. Impact of oncogenes in tumor angiogenesis: Mutant K-ras up-regulation of vascular endothelial growth factor/vascular permeability factor is necessary, but not sufficient for tumorigenicity of human colorectal carcinoma cells. Proc. Natl. Acad. Sci. USA 1998, 95, 3609–3614. [Google Scholar] [CrossRef]
  129. Grunstein, J.; Roberts, W.G.; Mathieu-Costello, O.; Hanahan, D.; Johnson, R.S. Tumor-derived expression of vascular endothelial growth factor is a critical factor in tumor expansion and vascular function. Cancer Res. 1999, 59, 1592–1598. [Google Scholar]
  130. Linardou, H.; Dahabreh, I.J.; Kanaloupiti, D.; Siannis, F.; Bafaloukos, D.; Kosmidis, P.; Papadimitriou, C.A.; Murray, S. Assessment of somatic k-RAS mutations as a mechanism associated with resistance to EGFR-targeted agents: A systematic review and meta-analysis of studies in advanced non-small-cell lung cancer and metastatic colorectal cancer. Lancet Oncol. 2008, 9, 962–972. [Google Scholar] [CrossRef]
  131. Mao, C.; Qiu, L.-X.; Liao, R.-Y.; Du, F.-B.; Ding, H.; Yang, W.-C.; Li, J.; Chen, Q. KRAS mutations and resistance to EGFR-TKIs treatment in patients with non-small cell lung cancer: A meta-analysis of 22 studies. Lung Cancer 2010, 69, 272–278. [Google Scholar] [CrossRef]
  132. Massarelli, E.; Varella-Garcia, M.; Tang, X.; Xavier, A.C.; Ozburn, N.C.; Liu, D.D.; Bekele, B.N.; Herbst, R.S.; Wistuba, I.I. KRAS Mutation Is an Important Predictor of Resistance to Therapy with Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Non–Small-Cell Lung Cancer. Clin. Cancer Res. 2007, 13, 2890–2896. [Google Scholar] [CrossRef]
  133. Metro, G.; Chiari, R.; Duranti, S.; Siggillino, A.; Fischer, M.J.; Giannarelli, D.; Ludovini, V.; Bennati, C.; Marcomigni, L.; Baldi, A.; et al. Impact of specific mutant KRAS on clinical outcome of EGFR-TKI-treated advanced non-small cell lung cancer patients with an EGFR wild type genotype. Lung Cancer 2012, 78, 81–86. [Google Scholar] [CrossRef] [PubMed]
  134. Zer, A.; Ding, K.; Lee, S.M.; Goss, G.D.; Seymour, L.; Ellis, P.M.; Hackshaw, A.; Bradbury, P.A.; Han, L.; O’Callaghan, C.J.; et al. Pooled Analysis of the Prognostic and Predictive Value of KRAS Mutation Status and Mutation Subtype in Patients with Non–Small Cell Lung Cancer Treated with Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors. J. Thorac. Oncol. 2016, 11, 312–323. [Google Scholar] [CrossRef]
  135. Fiala, O.; Pesek, M.; Finek, J.; Benesova, L.; Belsanova, B.; Minarik, M. The dominant role of G12C over other KRAS mutation types in the negative prediction of efficacy of epidermal growth factor receptor tyrosine kinase inhibitors in non–small cell lung cancer. Cancer Genet. 2013, 206, 26–31. [Google Scholar] [CrossRef] [PubMed]
  136. Sun, L.; Hsu, M.; Cohen, R.B.; Langer, C.J.; Mamtani, R.; Aggarwal, C. Association between KRAS variant status and outcomes with first-line immune checkpoint inhibitor-based therapy in patients with advanced non-small-cell lung cancer. JAMA Oncol. 2021, 7, 937–939. [Google Scholar] [CrossRef] [PubMed]
  137. Gadgeel, S.; Rodriguez-Abreu, D.; Felip, E.; Esteban, E.; Speranza, G.; Reck, M.; Hui, R.; Boyer, M.; Garon, E.; Horinouchi, H.; et al. 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. 2019, 30, xi64–xi65. [Google Scholar] [CrossRef]
  138. Herbst, R.; Lopes, G.; Kowalski, D.; Kasahara, K.; Wu, Y.-L.; De Castro, G.; Cho, B.; Turna, H.; Cristescu, R.; Aurora-Garg, D.; et al. LBA4 Association of KRAS mutational status with response to pembrolizumab monotherapy given as first-line therapy for PD-L1-positive advanced non-squamous NSCLC in Keynote-042. Ann. Oncol. 2019, 30, xi63–xi64. [Google Scholar] [CrossRef]
  139. Mazieres, J.; Tomasini, P.; Lusque, A.; Boucekine, M.; Gautschi, O.; Cortot, A.; Couraud, S.; Thai, A.; Ng, T.; Greillier, L.; et al. 1279P Impact of KRAS mutations and subtypes on efficacy of immune-checkpoint inhibitors (ICI) in non-small cell lung cancer (NSCLC). Ann. Oncol. 2020, 31, S826–S827. [Google Scholar] [CrossRef]
  140. Mazieres, J.; Drilon, A.; Lusque, A.B.; Mhanna, L.; Cortot, A.; Mezquita, L.; Thai, A.A.; 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. 2019, 30, 1321–1328. [Google Scholar] [CrossRef]
  141. Dong, Z.-Y.; Zhong, W.-Z.; Liu, S.-Y.; Xie, Z.; Wu, S.-P.; Wu, Y.L. MA15.10 Potential Predictive Value of TP53 and KRAS Mutation Status for Response to PD-1 Blockade Immunotherapy in Lung Adenocarcinoma. J. Thorac. Oncol. 2017, 12, S432–S433. [Google Scholar] [CrossRef]
  142. Lee, M.H.; Yanagawa, J.; Lee, M.H.; Yanagawa, J.; Li, R.; Walser, T.C.; Krysan, K.; Wang, G.; Goldman, J.W.; Garon, E.B.G.; et al. ERK Activation Mediates Increased PD-L1 Expression in KRAS Mutated Premalignant Human Bronchial Epithelial Cells. JTO 2015, 10, S268. [Google Scholar]
  143. Lindsay, C.; Jamal-Hanjani, M.; Forster, M.; Blackhall, F. KRAS: Reasons for optimism in lung cancer. Eur. J. Cancer 2018, 99, 20–27. [Google Scholar] [CrossRef]
  144. Friedlaender, A.; Drilon, A.; Weiss, G.J.; Banna, G.L.; Addeo, A. KRAS as a druggable target in NSCLC: Rising like a phoenix after decades of development failures. Cancer Treat. Rev. 2020, 85, 101978. [Google Scholar] [CrossRef] [PubMed]
  145. Liu, P.; Wang, Y.; Li, X. Targeting the untargetable KRAS in cancer therapy. Acta Pharm. Sin. B 2019, 9, 871–879. [Google Scholar] [CrossRef] [PubMed]
  146. Bar-Sagi, D.; Knelson, E.H.; Sequist, L.V. A bright future for KRAS inhibitors. Nat. Cancer 2020, 1, 25–27. [Google Scholar] [CrossRef] [Green Version]
  147. Huang, L.; Guo, Z.; Wang, F.; Fu, L. KRAS mutation: From undruggable to druggable in cancer. Signal Transduct. Target. Ther. 2021, 6, 386. [Google Scholar] [CrossRef]
  148. Adjei, A.A.; Mauer, A.; Bruzek, L.; Marks, R.S.; Hillman, S.; Geyer, S.; Hanson, L.J.; Wright, J.J.; Erlichman, C.; Kaufmann, S.H.; et al. Phase II Study of the Farnesyl Transferase Inhibitor R115777 in Patients With Advanced Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2003, 21, 1760–1766. [Google Scholar] [CrossRef]
  149. Papke, B.; Der, C.J. Drugging RAS: Know the enemy. Science 2017, 355, 1158–1163. [Google Scholar] [CrossRef]
  150. Cox, A.D.; Der, C.J.; Philips, M.R. Targeting RAS Membrane Association: Back to the Future for Anti-RAS Drug Discovery? Clin. Cancer Res. 2015, 21, 1819–1827. [Google Scholar] [CrossRef]
  151. Riely, G.J.; Johnson, M.L.; Medina, C.; Rizvi, N.A.; Miller, V.A.; Kris, M.G.; Pietanza, M.C.; Azzoli, C.G.; Krug, L.M.; Pao, W.; et al. A Phase II Trial of Salirasib in Patients with Lung Adenocarcinomas with KRAS Mutations. J. Thorac. Oncol. 2011, 6, 1435–1437. [Google Scholar] [CrossRef]
  152. Hymowitz, S.G.; Malek, S. Targeting the MAPK Pathway in RAS Mutant Cancers. Cold Spring Harb. Perspect. Med. 2018, 8, a031492. [Google Scholar] [CrossRef]
  153. Uprety, D.; Adjei, A.A. KRAS: From undruggable to a druggable Cancer Target. Cancer Treat. Rev. 2020, 89, 102070. [Google Scholar] [CrossRef]
  154. Cox, A.D.; Fesik, S.W.; Kimmelman, A.C.; Luo, J.; Der, C.J. Drugging the undruggable RAS: Mission Possible? Nat. Rev. Drug Discov. 2014, 13, 828–851. [Google Scholar] [CrossRef] [PubMed]
  155. Janne, P.A.; van den Heuvel, M.M.; 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 The SELECT-1 Randomized Clinical Trial. JAMA J. Am. Med. Assoc. 2017, 317, 1844–1853. [Google Scholar] [CrossRef]
  156. Kim, D.; Xue, J.Y.H.; Lito, P. Targeting KRAS(G12C): From Inhibitory Mechanism to Modulation of Antitumor Effects in Patients. Cell 2020, 183, 850–859. [Google Scholar] [CrossRef] [PubMed]
  157. Goldman, J.W.; Mazieres, J.; Barlesi, F.; Dragnev, K.H.; Koczywas, M.; Göskel, T.; Cortot, A.B.; Girard, N.; Wesseler, C.; Bischoff, H.; et al. A Randomized Phase III Study of Abemaciclib Versus Erlotinib in Patients with Stage IV Non-small Cell Lung Cancer With a Detectable KRAS Mutation Who Failed Prior Platinum-Based Therapy: JUNIPER. Front. Oncol. 2020, 10, 578756. [Google Scholar] [CrossRef] [PubMed]
  158. Middleton, G.; Fletcher, P.; Popat, S.; Savage, J.; Summers, Y.; Greystoke, A.; Gilligan, D.; Cave, J.; O’Rourke, N.; Brewster, A.; et al. The National Lung Matrix Trial of personalized therapy in lung cancer. Nature 2020, 583, 807–812. [Google Scholar] [CrossRef] [PubMed]
  159. Xie, M.; Xu, X.; Fan, Y. KRAS-Mutant Non-Small Cell Lung Cancer: An Emerging Promisingly Treatable Subgroup. Front Oncol. 2021, 11, 672612. [Google Scholar] [CrossRef]
  160. Ostrem, J.M.; Peters, U.; Sos, M.L.; Wells, J.A.; Shokat, K.M. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature 2013, 503, 548–551. [Google Scholar] [CrossRef]
  161. Janes, M.R.; Zhang, J.; Li, L.-S.; Hansen, R.; Peters, U.; Guo, X.; Chen, Y.; Babbar, A.; Firdaus, S.J.; Darjania, L.; et al. Targeting KRAS Mutant Cancers with a Covalent G12C-Specific Inhibitor. Cell 2018, 172, 578–589.e17. [Google Scholar] [CrossRef]
  162. Lito, P.; Solomon, M.; Li, L.-S.; Hansen, R.; Rosen, N. Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science 2016, 351, 604–608. [Google Scholar] [CrossRef]
  163. Patricelli, M.P.; Janes, M.R.; Li, L.-S.; Hansen, R.; Peters, U.; Kessler, L.V.; Chen, Y.; Kucharski, J.M.; Feng, J.; Ely, T.; et al. Selective Inhibition of Oncogenic KRAS Output with Small Molecules Targeting the Inactive State. Cancer Discov. 2016, 6, 316–329. [Google Scholar] [CrossRef] [PubMed]
  164. Lanman, B.A.; Allen, J.R.; Allen, J.G.; Amegadzie, A.K.; Ashton, K.S.; Booker, S.K.; Chen, J.J.; Chen, N.; Frohn, M.J.; Goodman, G.; et al. Discovery of a Covalent Inhibitor of KRAS(G12C) (AMG 510) for the Treatment of Solid Tumors. J. Med. Chem. 2020, 63, 52–65. [Google Scholar] [CrossRef] [PubMed]
  165. Fell, J.B.; Fischer, J.P.; Baer, B.R.; Blake, J.F.; Bouhana, K.; Briere, D.M.; Brown, K.D.; Burgess, L.E.; Burns, A.C.; Burkard, M.R.; et al. Identification of the Clinical Development Candidate MRTX849, a Covalent KRASG12C Inhibitor for the Treatment of Cancer. J. Med. Chem. 2020, 63, 6679–6693. [Google Scholar] [CrossRef] [PubMed]
  166. Canon, J.; Rex, K.; Saiki, A.Y.; Mohr, C.; Cooke, K.; Bagal, D.; Gaida, K.; Holt, T.; Knutson, C.G.; Koppada, N.; et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 2019, 575, 217–223. [Google Scholar] [CrossRef]
  167. Pantsar, T. KRAS(G12C)–AMG 510 interaction dynamics revealed by all-atom molecular dynamics simulations. Sci. Rep. 2020, 10, 11992. [Google Scholar] [CrossRef]
  168. Hong, D.S.; Fakih, M.G.; Strickler, J.H.; Desai, J.; Durm, G.A.; Shapiro, G.I.; Falchook, G.S.; Price, T.J.; Sacher, A.; Denlinger, C.S.; et al. KRAS(G12C) Inhibition with Sotorasib in Advanced Solid Tumors. N. Engl. J. Med. 2020, 383, 1207–1217. [Google Scholar] [CrossRef]
  169. Ramalingam, S.; Skoulidis, F.; Govindan, R.; Velcheti, V.; Li, B.; Besse, B.; Dy, G.; Kim, D.; Schuler, M.; Vincent, M.; et al. P52.03 Efficacy of Sotorasib in KRAS p.G12C-Mutated NSCLC with Stable Brain Metastases: A Post-Hoc Analysis of CodeBreaK 100. J. Thorac. Oncol. 2021, 16, S1123. [Google Scholar] [CrossRef]
  170. Begum, P.; Goldin, R.D.; Possamai, L.A.; Popat, S. Severe Immune Checkpoint Inhibitor Hepatitis in KRAS G12C-Mutant NSCLC Potentially Triggered by Sotorasib: Case Report. JTO Clin. Res. Rep. 2021, 2, 100213. [Google Scholar] [CrossRef]
  171. Spira, A.I.; Wilson, F.H.; Shapiro, G.; Dooms, C.; Curioni-Fontecedro, A.; Esaki, T.; Barlesi, F.; Cocks, K.; Trigg, A.; Stevinson, K.; et al. Patient-reported outcomes (PRO) from the phase 2 CodeBreaK 100 trial evaluating sotorasib in KRAS p.G12C mutated non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2021, 39, 9057. [Google Scholar] [CrossRef]
  172. Available online: https://ascopost.com/issues/june-10-2021/updated-information-on-sotorasib-dose-comparison-study/ (accessed on 18 June 2022).
  173. Papillon-Cavanagh, S.; Doshi, P.; Dobrin, R.; Szustakowski, J.; Walsh, A.M. STK11 and KEAP1 mutations as prognostic biomarkers in an observational real-world lung adenocarcinoma cohort. ESMO Open 2020, 5, e000706. [Google Scholar] [CrossRef]
  174. Skoulidis, F.; Goldberg, M.E.; Greenawalt, D.M.; Hellmann, M.D.; Awad, M.M.; Gainor, J.F.; Schrock, A.B.; Hartmaier, R.J.; Trabucco, S.E.; Gay, L.; et al. STK11/LKB1 Mutations and PD-1 Inhibitor Resistance in KRAS-Mutant Lung Adenocarcinoma. Cancer Discov. 2018, 8, 822–835. [Google Scholar] [CrossRef] [PubMed]
  175. Hong, D.S.; Bang, Y.J.; Barlesi, F.; Durm, G.A.; Falchook, G.S.; Govindan, R.; Dy, G.K.; Park, K.; Strickler, J.H.; Burns, T.F.; et al. Durability of clinical benefit and biomarkers in patients (pts) with advanced non-small cell lung cancer (NSCLC) treated with AMG 510 (sotorasib). Ann. Oncol. 2020, 31, S1389–S1390. [Google Scholar] [CrossRef]
  176. Skoulidis, F.; Li, B.T.; Dy, G.K.; Shapiro, G.; Bauml, J.; Schuler, M.H.; Addeo, A.; Kato, T.; Besse, B.; Anderson, A. Overall survival and exploratory subgroup analyses from the phase 2 CodeBreaK 100 trial evaluating sotorasib in pretreated KRAS p.G12C mutated non-small cell lung cancer. J. Clin. Oncol. 2021, 39, 9003. [Google Scholar] [CrossRef]
  177. Zhao, Y.; Murciano-Goroff, Y.R.; Xue, J.Y.; Ang, A.; Lucas, J.; Mai, T.T.; Da Cruz Paula, A.F.; Saiki, A.Y.; Mohn, D.; Achanta, P.; et al. Diverse alterations associated with resistance to KRAS(G12C) inhibition. Nature 2021, 599, 679–683. [Google Scholar] [CrossRef]
  178. Jänne, P.; Rybkin, I.; Spira, A.; Riely, G.; Papadopoulos, K.; Sabari, J.; Johnson, M.; Heist, R.; Bazhenova, L.; Barve, M.; et al. KRYSTAL-1: Activity and Safety of Adagrasib (MRTX849) in Advanced/ Metastatic Non–Small-Cell Lung Cancer (NSCLC) Harboring KRAS G12C Mutation. Eur. J. Cancer 2020, 138, S1–S2. [Google Scholar] [CrossRef]
  179. Abstract 9002, 2022 ASCO Annual Meeting. Volume 40, Issue 16. Available online: https://ascopubs.org/doi/abs/10.1200/JCO.2022.40.16_suppl.9002 (accessed on 16 June 2022).
  180. Hallin, J.; Engstrom, L.D.; Hargis, L.; Calinisan, A.; Aranda, R.; Briere, D.M.; Sudhakar, N.; Bowcut, V.; Baer, B.R.; Ballard, J.A.; et al. The KRAS(G12C) Inhibitor MRTX849 Provides Insight toward Therapeutic Susceptibility of KRAS-Mutant Cancers in Mouse Models and Patients. Cancer Discov. 2020, 10, 54–71. [Google Scholar] [CrossRef]
  181. Riely, G.; Ou, S.-H.; Rybkin, I.; Spira, A.; Papadopoulos, K.; Sabari, J.; Johnson, M.; Heist, R.; Bazhenova, L.; Barve, M.; et al. 99O_PR KRYSTAL-1: Activity and preliminary pharmacodynamic (PD) analysis of adagrasib (MRTX849) in patients (Pts) with advanced non–small cell lung cancer (NSCLC) harboring KRASG12C mutation. J. Thorac. Oncol. 2021, 16, S751–S752. [Google Scholar] [CrossRef]
  182. Gu, S.; Cui, D.; Chen, X.; Xiong, X.; Zhao, Y. PROTACs: An Emerging Targeting Technique for Protein Degradation in Drug Discovery. BioEssays 2018, 40, e1700247. [Google Scholar] [CrossRef]
  183. Burslem, G.M.; Smith, B.E.; Lai, A.C.; Jaime-Figueroa, S.; McQuaid, D.C.; Bondeson, D.P.; Toure, M.; Dong, H.; Qian, Y.; Wang, J.; et al. The Advantages of Targeted Protein Degradation over Inhibition: An RTK Case Study. Cell Chem. Biol. 2017, 25, 67–77.e3. [Google Scholar] [CrossRef]
  184. Petrylak, D.P.; Gao, X.; Vogelzang, N.J.; Garfield, M.H.; Taylor, I.; Moore, M.D.; Peck, R.A.; Burris, H.A. First-in-human phase I study of ARV-110, an androgen receptor (AR) PROTAC degrader in patients (pts) with metastatic castrate-resistant prostate cancer (mCRPC) following enzalutamide (ENZ) and/or abiraterone (ABI). J. Clin. Oncol. 2020, 38, 3500. [Google Scholar] [CrossRef]
  185. Snyder, L.B.; Flanagan, J.J.; Qian, Y.; Gough, S.M.; Andreoli, M.; Bookbinder, M.; Cadelina, G.; Bradley, J.; Rousseau, E.; Chandler, J.; et al. Abstract 44: The discovery of ARV-471, an orally bioavailable estrogen receptor degrading PROTAC for the treatment of patients with breast cancer. Cancer Res. 2021, 81, 44. [Google Scholar] [CrossRef]
  186. Camidge, D.R.; Pao, W.; Sequist, L.V. Acquired resistance to TKIs in solid tumours: Learning from lung cancer. Nat. Rev. Clin. Oncol. 2014, 11, 473–481. [Google Scholar] [CrossRef] [PubMed]
  187. Romero, D. Uncovering adagrasib resistance. Nat. Rev. Clin. Oncol. 2021, 18, 541. [Google Scholar] [CrossRef]
  188. Koga, T.; Suda, K.; Fujino, T.; Ohara, S.; Hamada, A.; Nishino, M.; Chiba, M.; Shimoji, M.; Takemoto, T.; Arita, T.; et al. KRAS Secondary Mutations That Confer Acquired Resistance to KRAS G12C Inhibitors, Sotorasib and Adagrasib, and Overcoming Strategies: Insights From the In Vitro Experiments. J. Thorac. Oncol. 2021, 16, 1321–1332. [Google Scholar] [CrossRef] [PubMed]
  189. Tanaka, N.; Lin, J.J.; Li, C.; Ryan, M.B.; Zhang, J.; Kiedrowski, L.A.; Michel, A.G.; Syed, M.U.; Fella, K.A.; Sakhi, M.; et al. Clinical Acquired Resistance to KRAS(G12C) Inhibition through a Novel KRAS Switch-II Pocket Mutation and Polyclonal Alterations Converging on RAS-MAPK Reactivation. Cancer Discov. 2021, 11, 1913–1922. [Google Scholar] [CrossRef]
  190. Awad, M.M.; Liu, S.; Rybkin, I.I.; Arbour, K.C.; Dilly, J.; Zhu, V.W.; Johnson, M.L.; Heist, R.S.; Patil, T.; Riely, G.J.; et al. Acquired Resistance to KRAS(G12C) Inhibition in Cancer. N. Engl. J. Med. 2021, 384, 2382–2393. [Google Scholar] [CrossRef] [PubMed]
  191. Aldea, M.; Andre, F.; Marabelle, A.; Dogan, S.; Barlesi, F.; Soria, J.-C. Overcoming Resistance to Tumor-Targeted and Immune-Targeted Therapies. Cancer Discov. 2021, 11, 874–899. [Google Scholar] [CrossRef] [PubMed]
  192. Passaro, A.; Jänne, P.A.; Mok, T.; Peters, S. Overcoming therapy resistance in EGFR-mutant lung cancer. Nat. Cancer 2021, 2, 377–391. [Google Scholar] [CrossRef]
  193. Marine, J.-C.; Dawson, S.-J.; Dawson, M.A. Non-genetic mechanisms of therapeutic resistance in cancer. Nat. Rev. Cancer 2020, 20, 743–756. [Google Scholar] [CrossRef]
  194. Hunter, J.C.; Manandhar, A.; Carrasco, M.A.; Gurbani, D.; Gondi, S.; Westover, K.D. Biochemical and Structural Analysis of Common Cancer-Associated KRAS Mutations. Mol. Cancer Res. 2015, 13, 1325–1335. [Google Scholar] [CrossRef]
  195. Suzuki, S.; Yonesaka, K.; Teramura, T.; Takehara, T.; Kato, R.; Sakai, H.; Haratani, K.; Tanizaki, J.; Kawakami, H.; Hayashi, H.; et al. KRAS Inhibitor Resistance in MET-Amplified KRAS (G12C) Non-Small Cell Lung Cancer Induced By RAS- and Non-RAS-Mediated Cell Signaling Mechanisms. Clin. Cancer Res. 2021, 27, 5697–5707. [Google Scholar] [CrossRef] [PubMed]
  196. Adachi, Y.; Ito, K.; Hayashi, Y.; Kimura, R.; Tan, T.Z.; Yamaguchi, R.; Ebi, H. Epithelial-to-Mesenchymal Transition is a Cause of Both Intrinsic and Acquired Resistance to KRAS G12C Inhibitor in KRAS G12C–Mutant Non–Small Cell Lung Cancer. Clin. Cancer Res. 2020, 26, 5962–5973. [Google Scholar] [CrossRef] [PubMed]
  197. Reck, M.; Spira, A.; Besse, B.; Wolf, J.; Skoulidis, F.; Borghaei, H.; Goto, K.; Park, K.; Griesinger, F.; Felip, E.; et al. 1416TiP CodeBreak 200: A phase III multicenter study of sotorasib (AMG 510), a KRAS(G12C) inhibitor, versus docetaxel in patients with previously treated advanced non-small cell lung cancer (NSCLC) harboring KRAS p.G12C mutation. Ann. Oncol. 2020, 31, S894–S895. [Google Scholar] [CrossRef]
  198. Mok, T.S.K.; Lawler, W.E.; Shum, M.K.; Dakhil, S.R.; Spira, A.I.; Barlesi, F.; Reck, M.; Garassino, M.C.; Spigel, D.R.; Alvarez, D.; et al. KRYSTAL-12: A randomized phase 3 study of adagrasib (MRTX849) versus docetaxel in patients (pts) with previously treated non-small-cell lung cancer (NSCLC) with KRAS(G12C) mutation. J. Clin. Oncol. 2021, 39, TPS9129. [Google Scholar] [CrossRef]
  199. Hong, D.S.; Strickler, J.H.; Fakih, M.; Falchook, G.S.; Li, B.T.; Durm, G.A.; Burns, T.F.; Ramalingam, S.S.; Goldberg, S.B.; Frank, R.C.; et al. Trial in progress: A phase 1b study of sotorasib, a specific and irreversible KRASG12C inhibitor, as monotherapy in non-small cell lung cancer (NSCLC) with brain metastasis and in combination with other anticancer therapies in advanced solid tumors (CodeBreaK 101). J. Clin. Oncol. 2021, 39, TPS2669. [Google Scholar] [CrossRef]
  200. Sabari, J.K.; Park, H.; Tolcher, A.W.; Ou, S.-H.I.; Garon, E.B.; George, B.; Janne, P.A.; Moody, S.E.; Tan, E.Y.; Sen, S.K.; et al. KRYSTAL-2: A phase I/II trial of adagrasib (MRTX849) in combination with TNO155 in patients with advanced solid tumors with KRAS G12C mutation. J. Clin. Oncol. 2021, 39, TPS146. [Google Scholar] [CrossRef]
  201. Xue, J.Y.; Zhao, Y.; Aronowitz, J.; Mai, T.T.; Vides, A.; Qeriqi, B.; Kim, D.; Li, C.; de Stanchina, E.; Mazutis, L.; et al. Abstract 622: Rapid non-uniform adaptation to conformation-specific KRAS G12Cinhibition. Cancer Res. 2020, 80, 622. [Google Scholar] [CrossRef]
  202. Janne, P.A.; Shaw, A.T.; Pereira, J.R.; Jeannin, G.; Vansteenkiste, J.; Barrios, C.H.; Franke, F.A.; Grinsted, L.; Smith, P.D.; Zazulina, V.; et al. Phase II double-blind, randomized study of selumetinib (SEL) plus docetaxel (DOC) versus DOC plus placebo as second-line treatment for advanced KRAS mutant non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2012, 30, 7503. [Google Scholar] [CrossRef]
  203. Carter, C.A.; Rajan, A.; Keen, C.; Szabo, E.; Khozin, S.; Thomas, A.; Brzezniak, C.; Guha, U.; Doyle, L.A.; Steinberg, S.M.; et al. Selumetinib with and without erlotinib in KRAS mutant and KRAS wild-type advanced nonsmall-cell lung cancer. Ann. Oncol. 2016, 27, 693–699. [Google Scholar] [CrossRef]
  204. Li, S.; Liu, S.W.; Akbay, E.A.; Hai, J.; Ambrogio, C.; Zhang, L.; Zhou, F.; Jenkins, R.W.; Adeegbe, D.O.; Gao, P. Assessing Therapeutic Efficacy of MEK Inhibition in a KRAS(G12C)-Driven Mouse Model of Lung Cancer. Clin. Cancer Res. 2018, 24, 4854–4864. [Google Scholar] [CrossRef]
  205. Molina-Arcas, M.; Moore, C.; Rana, S.; van Maldegem, F.; Mugarza, E.; Romero-Clavijo, P.; Herbert, E.; Horswell, S.; Li, L.-S.; Janes, M.R.; et al. Development of combination therapies to maximize the impact of KRAS-G12C inhibitors in lung cancer. Sci. Transl. Med. 2019, 11, aaw7999. [Google Scholar] [CrossRef] [PubMed]
  206. Chiba, M.; Togashi, Y.; Tomida, S.; Mizuuchi, H.; Nakamura, Y.; Banno, E.; Hayashi, H.; Terashima, M.; De Velasco, M.A.; Sakai, K.; et al. MEK inhibitors against MET-amplified non-small cell lung cancer. Int. J. Oncol. 2016, 49, 2236–2244. [Google Scholar] [CrossRef] [PubMed]
  207. Ruess, D.A.; Heynen, G.J.; Ciecielski, K.J.; Ai, J.; Berninger, A.; Kabacaoglu, D.; Görgülü, K.; Dantes, Z.; Wörmann, S.M.; Diakopoulos, K.N.; et al. Mutant KRAS-driven cancers depend on PTPN11/SHP2 phosphatase. Nat. Med. 2018, 24, 954–960. [Google Scholar] [CrossRef] [PubMed]
  208. Miura, Y.; Sunaga, N.; Kaira, K.; Tsukagoshi, Y.; Osaki, T.; Sakurai, R.; Hisada, T.; Girard, L.; Minna, J.D.; Yamada, M. Abstract 4028: Oncogenic KRAS mutations induce PD-L1 overexpression through MAPK pathway activation in non-small cell lung cancer cells. Cancer Res. 2016, 76, 4028. [Google Scholar] [CrossRef]
  209. Ryan, M.B.; de la Cruz, F.F.; Phat, S.; Myers, D.T.; Wong, E.; Shahzade, H.A.; Hong, C.B.; Corcoran, R.B. Vertical Pathway Inhibition Overcomes Adaptive Feedback Resistance to KRAS(G12C) Inhibition. Clin. Cancer Res. 2020, 26, 1633–1643. [Google Scholar] [CrossRef]
  210. Liu, C.; Lu, H.; Wang, H.; Loo, A.; Zhang, X.; Yang, G.; Kowal, C.; Delach, S.; Wang, Y.; Goldoni, S.; et al. Combinations with Allosteric SHP2 Inhibitor TNO155 to Block Receptor Tyrosine Kinase Signaling. Clin. Cancer Res. 2021, 27, 342–354. [Google Scholar] [CrossRef]
  211. Mainardi, S.; Mulero-Sánchez, A.; Prahallad, A.; Germano, G.; Bosma, A.; Krimpenfort, P.; Lieftink, C.; Steinberg, J.D.; De Wit, N.; Gonçalves-Ribeiro, S.; et al. SHP2 is required for growth of KRAS-mutant non-small-cell lung cancer in vivo. Nat. Med. 2018, 24, 961–967. [Google Scholar] [CrossRef]
  212. Gerlach, D.; Gmachl, M.; Ramharter, J.; Teh, J.; Fu, S.-C.; Trapani, F.; Kessler, D.; Rumpel, K.; Botesteanu, D.-A.; Ettmayer, P.; et al. Abstract 1091: BI-3406 and BI 1701963: Potent and selective SOS1:KRAS inhibitors induce regressions in combination with MEK inhibitors or irinotecan. Cancer Res. 2020, 80, 1091. [Google Scholar] [CrossRef]
  213. Hofmann, M.H.; Gmachl, M.; Ramharter, J.; Savarese, F.; Gerlach, D.; Marszalek, J.R.; Sanderson, M.P.; Kessler, D.; Trapani, F.; Arnhof, H.; et al. BI-3406, a Potent and Selective SOS1–KRAS Interaction Inhibitor, Is Effective in KRAS-Driven Cancers through Combined MEK Inhibition. Cancer Discov. 2021, 11, 142–157. [Google Scholar] [CrossRef]
  214. Zhang, H.; Brainson, C.; Koyama, S.; Redig, A.J.; Chen, T.; Li, S.; Gupta, M.; de Alba, C.G.; Paschini, M.; Herter-Sprie, G.S.; et al. Lkb1 inactivation drives lung cancer lineage switching governed by Polycomb Repressive Complex 2. Nat. Commun. 2017, 8, 14922. [Google Scholar] [CrossRef]
  215. Available online: https://www.merck.com/news/data-from-exploratory-analysis-show-mercks-keytruda-pembrolizumab-improved-overall-survival-as-monotherapy-for-the-first-line-treatment-of-metastatic-non-small-cell-lung-cancer-regard/ (accessed on 18 June 2022).
  216. Available online: https://clinicaltrials.gov/ct2/show/NCT05067283 (accessed on 18 June 2022).
  217. Briere, D.M.; Li, S.; Calinisan, A.; Sudhakar, N.; Aranda, R.; Hargis, L.; Peng, D.H.; Deng, J.; Engstrom, L.D.; Hallin, J.; et al. The KRAS(G12C) Inhibitor MRTX849 Reconditions the Tumor Immune Microenvironment and Sensitizes Tumors to Checkpoint Inhibitor Therapy. Mol. Cancer Ther. 2021, 20, 975–985. [Google Scholar] [CrossRef] [PubMed]
  218. Skoulidis, F.; Heymach, J.V. Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat. Rev. Cancer 2019, 19, 495–509. [Google Scholar] [CrossRef] [PubMed]
  219. West, H.; Cappuzzo, F.; Reck, M.; Mok, T.; Jotte, R.; Nishio, M.; Kim, E.; Morris, S.; Shankar, G.; Zou, W.; et al. IMpower150: A post hoc analysis of efficacy outcomes in patients with KRAS, STK11 and KEAP1 mutations. Ann. Oncol. 2020, 31, S817–S818. [Google Scholar] [CrossRef]
  220. Gao, G.; Liao, W.; Ma, Q.; Zhang, B.; Chen, Y.; Wang, Y. KRAS G12D mutation predicts lower TMB and drives immune suppression in lung adenocarcinoma. Lung Cancer 2020, 149, 41–45. [Google Scholar] [CrossRef]
  221. Feng, H.B.; Chen, Y.; Xie, Z.; Jiang, J.; Zhong, Y.M.; Guo, W.B.; Yan, W.Q.; Lv, Z.Y.; Lu, D.X.; Liang, H.L.; et al. High SHP2 expression determines the efficacy of PD-1/PD-L1 inhibitors in advanced KRAS mutant non-small cell lung cancer. Thorac. Cancer 2021, 12, 2564–2573. [Google Scholar] [CrossRef] [PubMed]
  222. Rodriguez-Nieto, S.; Canada, A.; Pros, E.; Pinto, A.I.; Torres-Lanzas, J.; Lopez-Rios, F.; Sanchez-Verde, L.; Pisano, D.G.; Sanchez-Cespedes, M. Massive Parallel DNA Pyrosequencing Analysis of the Tumor Suppressor BRG1/SMARCA4 in Lung Primary Tumors. Hum. Mutat. 2011, 32, E1999–E2017. [Google Scholar] [CrossRef]
  223. Hodges, H.C.; Stanton, B.Z.; Cermakova, K.; Chang, C.-Y.; Miller, E.L.; Kirkland, J.G.; Ku, W.L.; Veverka, V.; Zhao, K.; Crabtree, G.R. Dominant-negative SMARCA4 mutants alter the accessibility landscape of tissue-unrestricted enhancers. Nat. Struct. Mol. Biol. 2018, 25, 61–72. [Google Scholar] [CrossRef]
  224. Alessi, J.V.; Ricciuti, B.; Spurr, L.F.; Gupta, H.; Li, Y.Y.; Glass, C.; Nishino, M.; Cherniack, A.D.; Lindsay, J.; Sharma, B.; et al. SMARCA4 and Other SWItch/Sucrose NonFermentable Family Genomic Alterations in NSCLC: Clinicopathologic Characteristics and Outcomes to Immune Checkpoint Inhibition. J. Thorac. Oncol. 2021, 16, 1176–1187. [Google Scholar] [CrossRef]
  225. Schoenfeld, A.J.; Bandlamudi, C.; Lavery, J.A.; Montecalvo, J.; Namakydoust, A.; Rizvi, H.; Egger, J.V.; Concepcion, C.P.; Paul, S.; Arcila, M.E.; et al. The Genomic Landscape of SMARCA4 Alterations and Associations with Outcomes in Patients with Lung Cancer. Clin. Cancer Res. 2020, 26, 5701–5708. [Google Scholar] [CrossRef]
  226. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=SMARCA4 (accessed on 14 July 2022).
Table 1. Cancers with the highest KRAS mutations frequencies.
Table 1. Cancers with the highest KRAS mutations frequencies.
CancerKRAS Mutations Percentage
PDAC88%
CRC45–50%
Lung AC30–35%
AC: AdenoCarcinoma; CRC: ColoRectal Cancer; PDAC: Pancreatic Ductal AdenoCarcinoma (AACR Project GENIE Consortium, 2017 [7]; Prior et al., 2020 [6]; Uras et al., 2020 [8]; Reck et al., 2021 [5]).
Table 2. Rates of KRAS mutations in NSCLC and subtypes.
Table 2. Rates of KRAS mutations in NSCLC and subtypes.
CancerMutation Rates
KRASKRAS Exon 2 Mutations RateKRAS Exon 3 Mutations RateKRAS Exon 4 Rate Considering the Total of KRAS Mutations
NSCLC25–30%34%1.89%0.24%
Lung AC30–35%27%7%<1%
SCC3–4%7%<3%<0.5%
AC: AdenoCarcinoma; NSCLC: Non-Small Cell Lung Cancer; SCC: Squamous-Cell Carcinoma (AACR Project GENIE Consortium, 2017 [7]).
Table 3. Rates of KRAS exons 2, 3 and 4 mutations in NSCLC.
Table 3. Rates of KRAS exons 2, 3 and 4 mutations in NSCLC.
KRAS MutationsFrequencies
Exon 2 codon 12
c.34G > T (p.G12C)40%
c.35G > T (p.G12V)19–21%
c.35G > A (p.G12D)15–17%
c.35G > C (p.G12A)6%
c.34G > A (p.G12S)4%
c.34G > C (p.G12R)4%
Exon 2 codon 13
c.37G > T (p.G13C)6.8%
c.38G > A (p.G13D)0.8%
c.37G > C (p.G13R)0.6%
c.37G > A (p.G13S)NR
Exon 3 codon 59
c.176C > A (p.A59E)NR
c.176C > G (p.A59G)NR
c.175G > A (p.A59T)NR
Exon 3 codon 61NR
c.183A > C (p.Q61H)1.2%
c.182A > T (p.Q61L)0.4%
c.182A > G (p.Q61R)0.06%
c.180_181delinsAA (p.Q61K)0.02%
c.181C > G (p.Q61E)NR
Exon 4 codon 146
c.436G > C (p.A146P)NR
c.436G > A (p.A146T)NR
c.437C > T (p.A146V)NR
NR: Not Relevant; NSCLC: Non-Small Cell Lung Cancer (AACR Project GENIE Consortium, 2017 [7]).
Table 4. Rates of KRAS mutations in lung AC subdivided in different ethnicities.
Table 4. Rates of KRAS mutations in lung AC subdivided in different ethnicities.
KRAS Mutation Rates
CaucasiansAsiansAfricansAmericans
KRAS exon2
c.34G > T (p.G12C)34%24.5%NA41.2%
c.35G > T (p.G12V)21%1.5%NA19%
c.35G > A (p.G12D)14%25.5%28.5%15%
c.35G > C (p.G12A)9.5%1.3%NA7%
c.34G > A (p.G12S)1%0.6%NA1%
c.34G > C (p.G12R)1%0.6%NA1%
c.37G > T (p.G13C)4%0.7%NA7%
c.38G > A (p.G13D)1–2%0.7%42.8%1%
c.37G > C (p.G13R)0.5%0.5%NA0.5%
c.37G > A (p.G13S)NRNRNRNR
KRAS exon3
c.183A > C (p.Q61H)1%0.8%NA4%
c.182A > T (p.Q61L)0.5%0.4%NA1%
c.182A > G (p.Q61R)0.06%0.05%NA0.05
c.180_181delinsAA (p.Q61K)0.02%NRNA0.02
c.181C > G (p.Q61E)NRNRNANR
c.176C > A (p.A59E)NRNRNANR
c.176C > G (p.A59G)NRNRNANR
c.175G > A (p.A59T)NRNRNANR
KRAS exon4NRNRNANR
c.436G > C (p.A146P)NRNRNANR
c.436G > A (p.A146T)NRNRNANR
c.437C > T (p.A146V)NRNRNANR
AC, AdenoCarcinoma; NA: Not Available; NR: Not Relevant (Boch et al., 2013 [19]; Elghissassi et al., 2014 [31]; Kim et al., 2014 [32]; Li et al., 2014 [33]; Nadal et al., 2014 [34]; Arrieta and Ramírez-Tirado et al., 2015 [35]; Dhieb et al., 2019 [36]; Judd et al., 2021 [18]).
Table 5. Direct KRAS inhibition, ongoing studies.
Table 5. Direct KRAS inhibition, ongoing studies.
TitleStatusConditionInterventionNCT Number
A Study of Sotorasib (AMG 510) in Participants With Stage IV NSCLC Whose Tumors Harbor a KRAS p.G12C Mutation in Need of First-line Treatment (CodeBreaK201)Recruiting
-
NSCLC
-
Sotorasib
NCT04933695
FIH Study of JAB-21822 in Adult Patients With Advanced Solid Tumors Harboring KRAS G12C Mutation in ChinaRecruiting
-
NSCLC
-
Colorectal cancer
-
Solid Tumor
-
JAB-21822
NCT05009329
Testing the Use of Targeted Treatment (AMG 510) for KRAS G12C Mutated Advanced Non-squamous Non-small Cell Lung Cancer (A Lung-MAP Treatment Trial)Recruiting
-
Recurrent and metastatic Non-Squamous NSCLC
-
Sotorasib
NCT04625647
Sotorasib in Advanced KRASG12C-mutated Non-small Cell Lung Cancer Patients With Comorbidities (SOLUCOM)Recruiting
-
Recurrent or metastatic NSCLC
-
Mutation cancer
-
Sotorasib
NCT05311709
Study to Evaluate D-1553 in Subjects With Solid TumorsRecruiting
-
Solid Tumor,
-
NSCLC
-
CRC
-
D-1553
NCT04585035
CRC: ColoRectal Cancer; NSCLC: Non-Small Cell Lung Cancer.
Table 6. KRAS scaffold inhibition (SHP2/SOS1), ongoing studies.
Table 6. KRAS scaffold inhibition (SHP2/SOS1), ongoing studies.
TitleStatusConditionInterventionNCT Number
SHP2 Inhibitor BBP-398 in Combination With Nivolumab in Patients With Advanced Non-Small Cell Lung Cancer With a KRAS MutationRecruiting
-
NSCLC
-
Solid Tumor
-
BBP-398 with nivolumab
NCT05375084
Combination Therapy of RMC-4630 and LY3214996 in Metastatic KRAS-mutant Cancers (SHERPA)Recruiting
-
Pancreatic Cancer
-
CRC
-
NSCLC
-
KRAS Mutation-Related Tumors
-
RMC-4630
-
LY3214996
NCT04916236
Combination Study of RMC-4630 and Sotorasib for NSCLC Subjects With KRASG12C Mutation After Failure of Prior Standard TherapiesRecruiting
-
NSCLC
-
RMC-4630
-
Sotorasib
NCT05054725
Dose-Escalation/Expansion of RMC-4630 and Cobimetinib in Relapsed/Refractory Solid Tumors and RMC-4630 and Osimertinib in EGFR Positive Locally Advanced/Metastatic NSCLCActive, not recruiting
-
Solid tumor
-
RMC-4630
-
Cobimetinib
-
Osimertinib
NCT03989115
A Phase 1 Study of the SHP2 Inhibitor BBP-398 in Combination With Nivolumab in Patients With Advanced Non-Small Cell Lung Cancer With a KRAS MutationRecruiting
-
NSCLC
-
Solid Tumor
-
BBP-398 with Nivolumab
NCT05375084
A Dose Escalation Study of SHP2 Inhibitor in Patients With Solid Tumors Harboring KRAS of EGFR MutationsRecruiting
-
NSCLC
-
CRC
-
Pancreatic Cancer
-
Solid Tumor
-
HBI-2376
NCT05163028
Adagrasib in Combination With TNO155 in Patients With Cancer (KRYSTAL 2)Active, not recruiting
-
Advanced Cancer
-
Metastatic Cancer
-
Malignant Neoplastic Disease
-
MRTX849
-
TNO155
NCT04330664
Study of JDQ443 in Patients With Advanced Solid Tumors Harboring the KRAS G12C Mutation (KontRASt-01)Recruiting
-
KRAS G12C Mutant Solid Tumors
-
NSCLC
-
CRC
-
JDQ443
-
TNO155
-
Tislelizumab
NCT04699188
AC: AdenoCarcinoma; CRC: ColoRectal Cancer; NSCLC: Non-Small Cell Lung Cancer.
Table 7. Combination strategies of KRAS inhibition and upstream/downstream pathways (EGFR/RAS/RAF/MEK/HER2), ongoing studies.
Table 7. Combination strategies of KRAS inhibition and upstream/downstream pathways (EGFR/RAS/RAF/MEK/HER2), ongoing studies.
TitleStatusConditionInterventionNCT Number
Assess Efficacy & Safety of Selumetinib in Combination With Docetaxel in Patients Receiving 2nd Line Treatment for v-Ki-ras2 Kirsten Rat Sarcoma Viral Oncogene Homolog (KRAS) Positive NSCLC (SELECT-1)Active, not recruiting
-
Locally Advanced or Metastatic
-
NSCLC
-
Selumetinib
-
Docetaxel
-
Placebo
-
Pegylated G-CSF
NCT01933932
Trametinib and Docetaxel in Treating Patients With Recurrent or Stage IV KRAS Mutation Positive Non-small Cell Lung CancerActive, not recruiting
-
KRAS Gene Mutation
-
Recurrent ore metastatic NSCLC
-
Docetaxel
-
Laboratory Biomarker Analysis
-
Trametinib
NCT02642042
Rigosertib Plus Nivolumab for KRAS+ NSCLC Patients WhoProgressed on First-Line TreatmentRecruiting
-
Metastatic NSCLC
-
AC
-
Rigosertib
-
Nivolumab
NCT04263090
Binimetinib and Hydroxychloroquine in Patients With Advanced KRAS-mutant Non-Small Cell Lung CancerRecruiting
-
NSCLC
-
KRAS Mutation-Related Tumors
-
Binimetinib
-
Hydroxychloroquine
NCT04735068
MEK Inhibitor Combined With Anlotinib in the Treatment of KRAS-mutated Advanced Non-small Cell Lung CancerRecruiting
-
NSCLC
-
Trametinib
-
Anlotinib
NCT04967079
Regorafenib and Methotrexate in Treating Participants With Recurrent or Metastatic KRAS Mutated Non-Small Cell Lung CancerActive, not recruiting
-
KRAS Gene Mutation
-
Metastatic Malignant Neoplasm in the Brain
-
Recurrent or metastatic NSCLC
-
Methotrexate
-
Pharmacokinetic Study
-
Regorafenib
NCT03520842
A Phase I/IB Trial of MEK162 in Combination With Erlotinib in NSCLC Harboring KRAS or EGFR MutationActive, not recruiting
-
Lung cancer
-
NSCLC
-
MEK162
-
Erlotinib
NCT01859026
Trametinib and Pembrolizumab in Treating Patients With recurrent NSCLC that is metastatic, unresectable or locally advanced.Active, not recruiting
-
Unreseactable or Metastatic NSCLC
-
Pembrolizumab
-
Trametinib
-
Pharmacokinetic study
NCT03225664
Dose-Escalation/Expansion of RMC-4630 and Cobimetinib in Relapsed/Refractory Solid Tumors and RMC-4630 and Osimertinib in EGFR Positive Locally Advanced/Metastatic NSCLCActive, not recruiting
-
Solid tumor
-
RMC-4630
-
Cobimetinib
-
Osimertinib
NCT03989115
JAB-21822 Activity in Adult Patients With Advanced Solid Tumors Harboring KRAS G12C MutationRecruiting
-
Advanced Solid Tumor
-
NSCLC
-
CRC
-
JAB-21822 (KRAS G12C inhibitor)
-
Cetuximab (EGFR inhibitor)
NCT05002270
Pembrolizumab and Trametinib in Treating Patients With Stage IV Non-Small Cell Lung Cancer and KRAS Gene Mutations.Active, not recruiting
-
KRAS Gene Mutation
-
Recurrent or Metastatic Non-Squamous NSCLC.
-
Pembrolizumab
-
Trametinib
NCT03299088
Tarlox and Sotorasib in Patients With KRAS G12C MutationsRecruiting
-
NSCLC
-
Sotorasib and Tarloxotinib
NCT05313009
Phase 1/2 Study of VS-6766 + Sotorasib in G12C NSCLC Patients (RAMP203)Recruiting
-
NSCLC
-
KRAS Activating Mutation
-
VS-6766 and sotorasib
NCT05074810
A Study of VS-6766 v. VS-6766 + Defactinib in Recurrent G12V, Other KRAS and BRAF Non-Small Cell Lung Cancer (RAMP202)Recruting
-
NSCLC
-
KRAS Activating Mutation
-
VS-6766 •Drug: VS-6766 and Defactinib
NCT04620330
A Study to Evaluate the Safety, Pharmacokinetics, and Activity of GDC-6036 Alone or in Combination in Participants With Advanced or Metastatic Solid Tumors With a KRAS G12C MutationRecruiting
-
NSCLC
-
CRC
-
Advanced Solid Tumors
-
GDC-6036
-
Atezolizumab
-
Cetuximab
-
Bevacizumab
-
Erlotinib
-
GDC-1971
-
Inavolisib
NCT04449874
Phase 1b/2 Study of Futibatinib in Combination With Binimetinib in Patients With Advanced KRAS-mutant CancerRecruiting
-
Advanced or Metastatic Solid Tumors Irrespective of Gene Alterations
-
NSCLC
-
KRAS Gene Mutation
-
Futibatinib and Binimetinib
NCT04965818
Phase 1/2 Study of MRTX849 in Patients With Cancer Having a KRAS G12C Mutation KRYSTAL-1Recruiting
-
Advanced Cancer
-
Metastatic Cancer
-
Malignant Neoplastic Disease
-
MRTX849
-
Pembrolizumab
-
Cetuximab
-
Afatinib
NCT03785249
AC: AdenoCarcinoma; CRC: ColoRectal Cancer; NSCLC: Non-Small Cell Lung Cancer.
Table 8. KRAS inhibition and downstream pathways inhibition(PI3K/CD4/CD6), ongoing studies.
Table 8. KRAS inhibition and downstream pathways inhibition(PI3K/CD4/CD6), ongoing studies.
TitleStatusConditionInterventionNCT Number
A Study of Abemaciclib (LY2835219) in Participants with Previously Treated KRAS Mutated Lung Cancer (JUNIPER)Active, not recruiting recruiting
-
NSCLC
-
AC
-
Stage IV
-
Abemaciclib
-
Erlotinib
NCT02152631
PALBOCICLIB + PD-0325901 for NSCLC & Solid TumorsActive, not recruiting
-
KRAS-mutant NSCLC
-
Solid Tumors
-
Palbociclib
-
PD-0325901
NCT02022982
A Study to Evaluate the Safety, Pharmacokinetics, and Activity of GDC-6036 Alone or in Combination in Participants With Advanced or Metastatic Solid Tumors With a KRAS G12C MutationRecruiting
-
NSCLC
-
CRC
-
Advanced Solid Tumors
-
GDC-6036
-
Atezolizumab
-
Cetuximab
-
Bevacizumab
-
Erlotinib
-
GDC-1971
-
Inavolisib
NCT04449874
AC: AdenoCarcinoma; CRC: ColoRectal Cancer; NSCLC: Non-Small Cell Lung Cancer.
Table 9. Immunotherapy and KRAS inhibition, ongoing studies.
Table 9. Immunotherapy and KRAS inhibition, ongoing studies.
TitleStatusConditionInterventionNCT Number
Rigosertib Plus Nivolumab for KRAS+ NSCLC Patients WhoProgressed on First-Line TreatmentRecruiting
-
NSCLC
-
AC
-
Stage IV
-
Rigosertib
-
Nivolumab
NCT04263090
A Phase 1/2, Study Evaluating the Safety, Tolerability, PK, and efficacy of Sotorasib in subjects with solid tumors with a specific KRAS mutation (codeBreak 100)Recruiting
-
KRAS p.G12C mut
-
Advanced Solid tumor
-
Sotorasib
-
Anti PD-1/L1
-
Midazolam
NCT03600883
Trametinib and Pembrolizumab in Treating Patients With recurrent NSCLC that is metastatic, unresectable or locally advanced.Active, not recruiting
-
Metastatic NSCLC
-
Stage IIIA-C lung cancer AJCC v8
-
Stage IVA-B lung cancer AJCC v8
-
Unresectable NSCLC
-
Pembrolizumab
-
Trametinib
-
Pharmacokinetic study
NCT03225664
Phase 2 Trial of MRTX849 Monotherapy and in Combination With Pembrolizumab for NSCLC With KRAS G12C Mutation KRYSTAL-7.Recruiting
-
Advanced NSCLC
-
Metastatic NSCLC
-
MRTX849 Monotherapy
-
MRTX849 in Combination with Pembrolizumab
NCT04613596
Pembrolizumab and Trametinib in Treating Patients With stage IV NSCLC and KRAS gene mutaitons.Active, not recruiting
-
KRAS Gene Mutation
-
Metastatic or recurrent NSCLC
-
Stage IV NSCLC AJCC v7
-
Pembrolizumab
-
Trametinib
NCT03299088
PDR001 in Patients With Non-small Cell Lung Cancer harboring KRAS/NRAS mutation or no actionable genetic abnormalities.Recruiting
-
Cancer
-
Metastatic Lung Cancer.
-
PDR001
NCT03693326
A Study to Evaluate the Safety, Pharmacokinetics, and Activity of GDC-6036 Alone or in Combination in Participants With Advanced or Metastatic Solid Tumors With a KRAS G12C MutationRecruiting
-
NSCLC
-
CRC
-
Advanced Solid Tumors
-
GDC-6036
-
Atezolizumab
-
Cetuximab
-
Bevacizumab
-
Erlotinib
-
GDC-1971
-
Inavolisib
NCT04449874
Phase 1/2 Study of MRTX849 in Patients With Cancer Having a KRAS G12C Mutation KRYSTAL-1Recruiting
-
Advanced Cancer
-
Metastatic Cancer
-
Malignant Neoplastic Disease
-
MRTX849
-
Pembrolizumab
-
Cetuximab
-
Afatinib
NCT03785249
Phase 1/2 Study of MRTX849 in Patients With Cancer Having a KRAS G12C Mutation KRYSTAL-1Recruiting
-
Advanced Cancer
-
Metastatic Cancer
-
Malignant Neoplastic Disease
-
MRTX849
-
Pembrolizumab
-
Cetuximab
-
Afatinib
NCT03785249
A Phase 1 Study of the SHP2 Inhibitor BBP-398 in Combination With Nivolumab in Patients With Advanced Non-Small Cell Lung Cancer With a KRAS MutationRecruiting
-
NSCLC
-
Solid Tumor
-
BBP-398 with Nivolumab
NCT05375084
Study of JDQ443 in patients with advanced solid tumors harboring the KRAS G12 C mutationRecruiting
-
KRAS G12C Mutant Solid Tumors
-
NSCLC
-
CRC
-
JDQ443
-
TNO155
-
Tislelizumab
NCT04699188
A Study of MK-1084 as Monotherapy and in Combination With Pembrolizumab (MK-3475) in Participants With KRASG12C Mutant Advanced Solid Tumors (MK-1084-001)Recruting
-
Advanced Solid Tumors
-
MK-1084
-
Pembrolizumab
NCT05067283
AC: AdenoCarcinoma; CRC: ColoRectal Cancer; NSCLC: Non-Small Cell Lung Cancer.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cekani, E.; Epistolio, S.; Dazio, G.; Cefalì, M.; Wannesson, L.; Frattini, M.; Froesch, P. Molecular Biology and Therapeutic Perspectives for K-Ras Mutant Non-Small Cell Lung Cancers. Cancers 2022, 14, 4103. https://doi.org/10.3390/cancers14174103

AMA Style

Cekani E, Epistolio S, Dazio G, Cefalì M, Wannesson L, Frattini M, Froesch P. Molecular Biology and Therapeutic Perspectives for K-Ras Mutant Non-Small Cell Lung Cancers. Cancers. 2022; 14(17):4103. https://doi.org/10.3390/cancers14174103

Chicago/Turabian Style

Cekani, Elona, Samantha Epistolio, Giulia Dazio, Marco Cefalì, Luciano Wannesson, Milo Frattini, and Patrizia Froesch. 2022. "Molecular Biology and Therapeutic Perspectives for K-Ras Mutant Non-Small Cell Lung Cancers" Cancers 14, no. 17: 4103. https://doi.org/10.3390/cancers14174103

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop