Review
KRAS Mutations in Lung Cancer

https://doi.org/10.1016/j.cllc.2012.09.007Get rights and content

Abstract

Epidermal growth factor receptor (EGFR) gene mutations and increased EGFR copy numbers have been associated with a favorable response to EGFR tyrosine kinase inhibitors (TKI) in patients with non–small-cell lung cancer (NSCLC), and several markers have been identified that predict response to treatment. Lung adenocarcinomas also harbor activating mutations in the downstream GTPase, v-Ki-ras2 Kirsten rat sarcoma viral oncogene (KRAS), and mutations in EGFR and KRAS appear to be mutually exclusive. Even though KRAS mutations were identified in NSCLC tumors more than 20 years ago, we have only just begun to appreciate the clinical value of determining KRAS tumor status. Recent studies indicate that patients with mutant KRAS tumors fail to benefit from adjuvant chemotherapy and do not respond to EGFR inhibitors. There is a clear need for therapies specifically developed for patients with KRAS-mutant NSCLC. In this review, we summarize the clinical and pathologic characteristics of patients with NSCLC and with KRAS mutations, describe work that explores the predictive and prognostic influence of KRAS mutations, and provide an overview of the “synthetic lethal” interactions and current approaches to targeting KRAS-mutant NSCLC.

Introduction

Lung cancer is the leading cause of cancer-related death worldwide, with non–small-cell lung cancer (NSCLC) being the predominant form of the disease. Most lung cancer is caused by the accumulation of genomic alterations and the two most commonly mutated oncogenes encode for the epidermal growth factor receptor (EGFR) and KRAS. The KRAS gene together with the v-Ha-ras Harvey rat sarcoma viral oncogene and the neuroblastoma RAS viral oncogene encode a family of membrane-bound 21 kd guanosine triphosphate (GTP) binding proteins that regulate cell growth, differentiation, and apoptosis by interacting with multiple effectors, including those in the mitogen-activated protein kinase (MAPK), signal transducer and activator of transcription (STAT), and phosphoinositide 3-kinase (PI3K) signaling cascades.1 Mutations in the KRAS gene are frequently found in different human cancers, such as cancer of the lung, large intestine, pancreas, and biliary tract.2 Almost 15% to 25% of patients with NSCLC have KRAS mutations, and more than 97% of KRAS-mutant cases affect exon 2 and 3 (G12, G13, and Q61) (Figure 1).3 These mutations impair the intrinsic GTPase activity of RAS and confer resistance to GTPase activators, thereby causing RAS to accumulate in its active GTP-bound state, which sustains the activation of RAS signaling.4

Several studies have shown that EGFR and KRAS mutations are mutually exclusive, which suggests that they have functionally equivalent roles in lung tumorigenesis.5, 6, 7 Similar to EGFR mutations, KRAS mutations also appear to be associated with distinct clinical and pathologic characteristics, and vary according to tumor histology, ethnicity, and smoking history.8 KRAS mutations occur more frequently in lung adenocarcinomas (approximately 30%) and less frequently in the squamous cell carcinoma subtype (approximately 5%).9 There are ethnic differences that play a role with regard to the occurrence of KRAS mutations, which varies from that of EGFR mutations. Although EGFR mutations are more frequent in Asian (approximately 30%-40%) than white populations (10%-15%), KRAS mutations more commonly affect whites, with a frequency of 25% to 50% vs. 5% to 15%, respectively.8, 9, 10, 11 Unlike EGFR mutations, which occur more frequently in tumors in patients who are never-smokers, a large number of studies have found a significant association between tobacco or smoking habit and the presence of KRAS mutations.1, 10, 11, 12 Moreover, as has been described in other genes, for example, TP53, it appears that tobacco smoke causes certain types of KRAS mutations, thus making it possible to identify a molecular signature for the carcinogenic effects of cigarette smoke.13 In both KRAS and TP53, transversions (substituting a pyrimidine for a purine or purine for a pyrimidine) are more common than transitions (substituting purine for purine or pyrimidine for pyrimidine) in patients with a history of cigarette smoking compared with never-smokers.13 Further investigation is warranted to determine whether this etiologic heterogeneity within KRAS-mutant lung adenocarcinomas is associated with differences in cooperating genetic lesions and overall biologic behavior.

In 1999, Graziano et al14 investigated the prognostic effect of KRAS mutations in stage I and II resected NSCLC and found no statistical differences in overall survival (OS) for patients who had KRAS positive and negative mutations. In addition, Keohavong et al15 found no association between KRAS mutations and survival in 173 patients with adenocarcinoma and adenosquamous NSCLC. In the study by Lu et al,16 in which the prognostic role of a panel of 6 biomarkers, including KRAS mutations, was evaluated in completely resected stage I NSCLC, KRAS mutations were not found to affect OS. By contrast, Slebos et al17 reported that KRAS codon-12 point mutations are a negative prognostic factor for disease-free-survival and OS in a series of 69 patients who were surgically treated for NSCLC. The findings were also consistent after adjustment for factors such as stage, tumor size, and differentiation.17 In another 2 studies, the investigators examined tumor tissue from patients with NSCLC and found that the KRAS mutation–positive group had worse survival than the KRAS mutation-negative group. The same results were observed in another Japanese study in which the OS rate of patients with NSCLC and with wild-type (WT) KRAS was found to be better than the OS of patients whose tumors harbored KRAS mutations.18, 19, 20

In a prospective series of 365 patients with resected early-stage NSCLC treated at Massachusetts General Hospital, KRAS mutations were found only in smokers and were associated with worse survival only in stage I disease but not in the whole population.21 Finally, in 2005, a systematic review and meta-analysis of 28 studies, including a total of 3620 patients, showed that the presence of KRAS mutations confers a significantly worse prognosis, and, in a subgroup analysis according to histology, KRAS mutation resulted in a statistically significant prognostic factor for survival only for adenocarcinoma.22 By reviewing the above data (Table 1), it can be supposed that KRAS mutations could be a negative prognostic factor in NSCLC.

The activated EGFR may phosphorylate a wide array of intracellular signaling cascades, such as the rat sarcoma viral oncogene (RAS)/v-raf murine leukemia viral oncogene homolog (RAF)/murine thymoma viral oncogene homolog (MEK) (mitogen-activated protein kinase)/ERK (extracellular-signal-regulated kinase) and the phosphatidylinositol 3-kinase (PI3K/AKT) pathways, which lead to cancer proliferation, increased metastasis potential, and neoangiogenesis.23 Inhibition of EGFR kinase activities by EGFR tyrosine kinase inhibitors (TKI), such as gefitinib (Iressa; AstraZeneca, Inc.; London, UK) or erlotinib (Tarceva; Hoffman-La Roche, Basel, Switzerland) results in effective treatment of patients with NSCLC and with EGFR mutations, with dramatic clinical and radiologic response. Overall, EGFR mutation carriers have a response rate (RR) of approximately 75%, compared with <10% for patients with WT EGFR, and, furthermore, patients with EGFR mutation have longer progression-free survival (PFS) and OS.24, 25, 26, 27, 28, 29, 30

By contrast, although KRAS mutations were identified in NSCLC tumors more than 20 years ago, we are only now starting to appreciate their clinical value. Colorectal cancer has long been known to harbor KRAS mutations. Clinical assessments of cetuximab and panitumumab in patients with metastatic colorectal cancer, showed responses for these agents of between 10% and 30% (single-agent monoclonal antibody and plus chemotherapy, respectively).31 In view of the fact that KRAS mutations occur in a substantial proportion of metastatic colorectal cancer (30%-50%) and are negative prognostic factors, they were assessed as potential markers for the response of individuals to anti-EGFR agents.31 There has been particular interest in second-line treatment of metastatic colorectal cancer, in which similar data to NSCLC concerning correlations of nonresponsiveness, KRAS mutations, and anti-EGFR monoclonal antibodies have been described.31 Although, in colorectal carcinomas, an important number of studies attest to the value of KRAS mutational status as a predictive value of resistance to anti-EGFR therapies, the role of KRAS mutations in relation to NSCLC is much less clear.9, 32, 33 Progress in this field has been hampered by relatively small studies; different methods of molecular analysis; and by heterogeneity in histologic subtypes, stage, treatment administered, and survival criteria used. However, recent findings among patients treated with adjuvant chemotherapy or EGFR inhibitors highlight that KRAS might yet be an important biomarker for NSCLC and worthy of further research (Table 2).9

The clinical relevance of KRAS mutational status in patients with NSCLC has been evaluated in a meta-analysis of 1335 patients, both whites and Asians, who were included in 22 studies and were treated with gefitinib or erlotinib.11 Despite the sample's heterogeneity, the pooled results suggest that KRAS mutations act as a negative predictive marker for tumor response in patients with NSCLC treated with anti-EGFR therapies. KRAS mutations were detected in 16% of patients and were more common in smokers and in patients with adenocarcinomas. A subanalysis also indicated that the predictive value of KRAS mutations in response to EGFR TKI was higher in the Asian population than in the white population, which may be partially explained by the difference of EGFR-pathway activation in Asian patients with NSCLC compared with white patients with NSCLC. The data of overall RR indicated that patients with KRAS mutations were less sensitive to EGFR TKI than those with WT KRAS, with 3% of RR vs. 26% of RR, respectively.11 With this in mind, it is important to mention the results of the study by Hamilton et al,34 in which it was confirmed that the pharmacokinetics of erlotinib are different in current smokers and nonsmokers due to the increased metabolic clearance of erlotinib in current smokers. For this reason, smoking may contribute as a confounding variable in the prognosis of KRAS-mutated lung cancer. A potential benefit in survival to erlotinib in KRAS-mutated NSCLC was reported in the Sequential Tarceva in Unresectable NSCLC (SATURN) trial, a large phase III trial that randomized patients not progressing after first-line chemotherapy to receive erlotinib or placebo as maintenance treatment.35 Four hundred ninety-three (55.4%) tumor samples were analyzed for KRAS mutations. Patients treated with erlotinib experienced longer PFS irrespective of KRAS mutational status, with a marginal, although not significant, improvement in survival in the KRAS-mutant population, whereas the presence of KRAS mutations in the placebo arm was a significant negative prognostic factor for PFS.35

The Avastin and Tarceva in Lung With NSCLC (ATLAS) trial is another study that evaluated maintenance treatment with bevacizumab plus placebo or erlotinib in patients with metastatic NSCLC not progressing after 4 cycles of platinum-based chemotherapy. Analysis of KRAS mutations highlighted longer PFS for patients with WT KRAS treated with bevacizumab plus erlotinib but no difference for patients with KRAS-mutant p between the 2 arms.36 Finally, in the IRESSA NSCLC Trial Evaluating Response and Survival against Taxotere (INTEREST) study, a large phase III trial that compared gefitinib and docetaxel as second-line therapy in metastatic NSCLC, no differences in PFS and OS were detected in both treatment arms according to KRAS status.37 Therefore, although patients who harbor a KRAS mutation do not respond to EGFR TKIs, a minimal survival effect cannot be discounted. Therefore, at the present time, KRAS testing is not recommended as criteria for precluding any patient with NSCLC from therapy with EGFR TKIs.

Analysis of recent data suggests that KRAS mutations may affect the outcome of patients with NSCLC who received chemotherapy. In the adjuvant setting, data from the JBR10 trial suggested no benefit from adjuvant chemotherapy in patients with KRAS mutations.38 In the Lung Adjuvant Cisplatin Evaluation (LACE)-biomarkers (Bio) pooled analysis, the prognostic and predictive role of KRAS mutations was investigated in 1532 patients treated with adjuvant chemotherapy. Among evaluable patients, 300 harbored KRAS mutations (275 codon 12, 24 codon 13, 1 codon 14) and those with KRAS codon 13 mutations had significantly poorer outcomes with adjuvant chemotherapy.39 Several studies investigated the influence of KRAS mutations on sensitivity to chemotherapy in advanced NSCLC. No significant difference in PFS or OS was detected between patients with WT KRAS and those with KRAS mutation, which indicates that KRAS mutations have no role in prediction of response to standard chemotherapy in NSCLC.40, 41 Even more interesting are the results obtained from the recently completed prospective phase II BATTLE (Biomarker-integrated Approaches of Targeted Therapy for Lung Cancer Elimination) trial in patients with refractory NSCLC who received 1 of 4 testing arms: erlotinib, vandetanib, bexarotene and erlotinib, or sorafenib. The first analysis did not show any correlation between KRAS mutational status and survival in any of the treatment groups. However, patients whose tumors had either mutant KRAS-Gly12Cys or mutant KRAS-Gly12Val had worse PFS compared with patients whose tumors had other mutant KRAS proteins or WT KRAS.42 In the same trial, a 46% 8-week disease control rate (primary endpoint) for patients with mutant KRAS who were treated with sorafenib was found.43

The exciting hypothesis that not all mutant KRAS proteins affect patient survival or downstream signaling in a similar way has previously been described in in vitro assays that used KRAS-overexpressing clones with the amino acid substitutions G12C, G12V, and G12D from the human NSCLC cell line NCI-H1299.44 The researchers reported that expression of a specific KRAS substitution induced a different sensitivity pattern: the expression of G12C was associated with a reduced response to cisplatin and increased sensitivity to Taxol (Bristol-Myers Squibb, New York, NY) and pemetrexed, whereas expression of G12D was only associated with resistance to Taxol treatment and sensitivity to sorafenib. Furthermore, the G12V mutant showed strong sensitivity to cisplatin compared with the WT clones and was slightly more resistant to pemetrexed. However, the researchers did not find differences in the cellular response to EGFR inhibitors erlotinib and to gembcitabine.44 Taken together, these findings have important implications from the clinical point of view; first, a change in the means of defining KRAS-mutated lung tumors (a simple definition may not be sufficient to identify patients with differing capacities of responding to therapy), and second, the urgent need for new strategies for treating KRAS-mutated NSCLC tumors.

Although several strategies to inhibit KRAS have been explored, the quest for therapeutic inhibitors of RAS has fallen short of expectations. An obstacle to the development of specific RAS inhibitors is that mutated RAS proteins have lost their normal enzymatic function and such loss-of-function mutated enzymes are much more difficult to inhibit. At the same time, the relative failure of KRAS inhibitors derives from specificity issues and from differences in how mutant KRAS controls its downstream effectors in different individual tumors. Observation of diverse subsets of KRAS-mutant NSCLCs with distinct molecular profiles and signaling pathway activation supports the idea that the most-effective treatment strategy for KRAS-mutant NSCLC is combined targeting of oncogenic KRAS in addition to other therapeutic agents specific to the molecular profile of the tumor. However, realizing this vision will require a priori knowledge of these abnormalities and of tumor cell's compensatory mechanisms. Numerous investigations have been carried out into the expression of other genes by using semiquantitative reverse transcriptase–polymerase chain reaction, or other techniques, in an attempt to address these issues. Having established that oncogenic KRAS is not NSCLC's Achilles heel, what follows is an attempt to summarize other tumor molecular abnormalities that result in oncogene-specific ‘“synthetic lethal”' interactions that, in turn, provide additional targeted therapy opportunities for KRAS-mutated NSCLC.

One potentially promising approach has been to target pathways acting downstream of RAS (Figure 2). Inhibitors of the PI3K/mTOR (mammalian target of rapamycin) pathway may be active in cancers with PI3K mutations and, when combined with MEK inhibitors, may effectively treat KRAS-mutated lung cancers.45 It has become evident that cancers respond dramatically to therapies that target receptor tyrosine kinases (RTK) when inhibition of the RTK leads to loss of both PI3K and ERK signaling.46, 47, 48, 49 Engelman et al45 treated mice with KRAS-mutant lung tumors with a combination of a PI3K and an MEK inhibitor and found that the combination led to marked synergistic tumor regression. In a very recent work by Janne et al,50 a MEK1/2 inhibitor was prospectively evaluated in advanced KRAS-mutant NSCLC. In this study, patients treated with the combination of selumetinib plus docetaxel had a clinical benefit compared with those who received docetaxel plus placebo.

Serine threonine kinase 11 (STK11) also known as liver kinase B1 (LKB1) is a multitasking tumor suppressor kinase involved in Peutz-Jeghers hereditary cancer syndrome.51 LKB1/serine threonine kinase 11 is somatically inactivated in approximately 30% of NSCLC, and homozygous loss of this gene, in combination with KRAS, results in an aggressive tumor phenotype far more than those of KRAS alone.52, 53 It has been reported that LKB1 inactivation and KRAS activation in NSCLC denotes a functionally distinct set of lung cancer, which displays sensitivity to single-agent treatment with the MEK inhibitor CI-1040 or rapamycin.51

Neurofibromatosis type 1 (NF1) has high gene homology with Ras GTPase activating protein and is able to regulate cell motility and invasion. Loss of NF1 results in hyperactive RAS signaling that occurs in some types of cancer, including NSCLC (Figure 2). It has been reported that additional mutational inactivation of the NF1 Ras GTPase activators, usually mutually exclusive to KRAS mutation, increases the occurrence of persistent RAS activation in NSCLC to approximately 40%.9 Johannessen et al54 reported that the mTOR inhibitor rapamycin potently suppresses the growth of aggressive NF1-associated malignancies in a genetically engineered murine model, which demonstrated them to be a possible effective targeted therapy for this largely untreatable malignancy.

Moreover, the Wilms tumor gene WT1 is required in mutant KRAS cells via regulation of cellular senescence.55 Vicent et al55 identified a novel role for WT1 in cells expressing oncogenic KRAS. The synthetic effect of WT1 loss in these cells was tested and confirmed in primary cells, in a genetically engineered mouse model, and in human cell lines. In both mouse and human cells, the loss of WT1 activated a senescence program in cells expressing oncogenic KRAS but not in cells expressing WT KRAS, which demonstrated a surprising and unexpected role for WT1 as a critical regulator of the cellular mechanisms.55 Further elucidation of this study may provide avenues for therapeutic intervention in a wide range of human cancers, including NSCLC.

The overexpression of oncogenic forms of Ras has been reported to result in nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation, whereas WT p53 has been shown to antagonize NF-κB activity.56, 57, 58, 59, 60, 61 Meylan et al62 established a critical role of the NF-κB pathway in lung cancer cells that contain KRAS and p53 mutations. Interestingly, the proteasome has been shown in several earlier studies to be dependent on KRAS.63 Barbie et al,64 identified the noncanonical IκB kinase, TBK1, as a synthetic lethal partner of oncogenic KRAS (Figure 2). TBK1 and, more generally, NF-κB signaling may represent an alternative method of targeting oncogenic KRAS-driven cancers. Suppression of TBK1 induced apoptosis specifically in human cancer cell lines that depend on oncogenic KRAS expression. In these cells, TBK1 activated NF-κB anti-apoptotic signals that involves cREL (reticuloendotheliosis viral oncogene homolog) and Bcl-xL (B-cell lymphoma 2) that were essential for survival.64

These observations identify TBK1 and NF-κB signaling as essential in KRAS-mutant tumors and provide support for the development of NF-κB inhibitory drugs as targeted therapies for the treatment of patients with defined mutations in KRAS. Furthermore, gene expression analysis indicates that reduced expression of genes such as the mitotic polo-like kinase 1 (PLK1), the anaphase promoting complex/cyclosome and the proteasome results in prometaphase accumulation and the subsequent death of Ras mutant cells.65 These results demonstrate a pharmacologically tractable pathway for potential treatment of cancers harboring Ras mutations.63

Enhancer of zeste homolog 2 (EZH2) is a gene that encodes the histone methyltransferase component of the polycomb-repressive complex-2 and regulates epigenetic gene silencing (Figure 3).66 It has been reported that EZH2 activates Ras and p65/RelA (reticuloendotheliosis viral oncogene homolog A), a measure of NF-κB activity.67 High EZH2 expression has been correlated with poor prognosis in several tumors, including NSCLC.67, 68 A causal role for EZH2 in driving metastasis has been established, and disabled homolog 2–interacting protein has been identified as a key target in this process.69 Recently, it was found that EZH2 is upregulated after cerulein-induced pancreatic injury and is required for tissue repair by promoting the regenerative proliferation of progenitor cells. Loss of EZH2 results in impaired pancreatic regeneration and accelerates KRAS (G12D) driven neoplasia.70 EZH2 has been proposed by Simon and Lange,66 to be a potential therapeutic target and their data underscore the utility of developing EZH2 inhibitors because such agents might suppress both Ras and NF-κB.

The seven in absentia homolog (Siah) family of RING-domain proteins are components of ubiquitin ligase complexes, targeting proteins for proteasomal degradation and have upregulated expression in all major types of lung cancer.71, 72 Siah 2 is a member of the Siah family that has been reported to function in Ras interacting with the negative regulator Spry2 (Sprouty homolog 2 [Drosophila]), which results in its degradation.71 Schmidt et al73 showed that dominant-negative and small hairpin RNA (shRNA) inhibition of Siah potently impaired the growth of xenografts of human pancreatic tumor cells in mice that are characterized by very frequent KRAS mutations and, therefore, provide an ideal model to test a requirement for Siah 2 in Ras-driven cancer. ERK phosphorylation was reduced, which suggests that Siah 2 may be acting through the RAS/MEK/ERK pathway in these cells (Figure 2). In a separate study, Siah2 caused targeted degradation of Spry2 and provided a potential mechanism by which the loss of Siah 2 could impair Ras function.74 The use of a dominant negative Siah 2 protein has been found to reduce lung cancer cell growth, whereas knockdown of Siah2 reduces ERK signaling and cell proliferation, increases apoptosis, reduces colony formation in soft agar, and reduces tumorigenesis of A549 human lung cancer cells when injected into athymic nude mice.72 Therefore, given the involvement of Siah proteins in Ras signaling, blocking the function of Siah2 proteins could be an attractive approach to reducing tumor growth in KRAS-mutated NSCLC.

Kumar et al65 showed that lung cancers with mutations in KRAS and other oncogenes on the RTK/RAS pathway depend upon the erythroid transcription factor, also known as GATA-binding factor 2 (GATA2), for survival, via concurrent regulation of 3 pathways: proteasome machinery; interleukin (IL) 1/NF-κB signaling pathway; and Rho-signaling cascade. GATA2 has no known oncogenic function in its own right and is not controlled by signals emanating from the RTK/RAS pathway.65 Thus, dependency on the GATA2 transcriptional network might evolve as a stress-response mechanism in parallel to oncogene signaling during tumorigenesis and that selection for the network in RAS-pathway mutant cells eventually becomes dependency on continued GATA2 function.65 These investigations revealed a marked attenuation of autochthonous KRAS-mutant NSCLC growth in response to systemic GATA2 loss.65 Dramatic effects have been observed with combined inhibition of the proteasome and Rho-associated serino-threonine protein kinase (Rho/ROCK) signaling with the clinical compounds bortezomib and fasudil, respectively.65

It has been reported that the loss of RNA-binding motif 5 (RBM5) is the most frequent and earliest event in NSCLC.75 RBM5 plays an important role in the induction of cell-cycle arrest and apoptosis through pre–messenger RNA (mRNA) splicing of multiple target genes and inhibits tumor transformation and the progression of several malignancies, including NSCLC.76, 77, 78 In previous studies, it was shown that RBM5 is downregulated by the constitutively activated RAS mutant protein, RAS (G12V), in rat embryonic fibroblast cells, which indicates a correlation between the EGFR and RAS pathways and RBM5 expression.79 Liang et al80 found that RBM5 expression was reduced in NSCLC compared with normal tissues, and this reduction was associated with tobacco smoke, tumor stages, and lymph node metastasis. Moreover, expression of RBM5 mRNA and protein was negatively associated with expression of EGFR and KRAS mRNA and protein in NSCLC tissues, which suggests the presence of a complex regulatory network that involves tumor suppression and oncogenic expression.80 Further evaluation of RBM5 expression and KRAS gene function and interrelationships may lead to the development both of biomarkers to identify high-risk individuals and novel therapeutics.

IL-8 (CXCL8) is a cytokine of the CXC chemokine family that has been implicated in a wide variety of processes, including angiogenesis and metastasis in lung cancer. Lung adenocarcinoma and mucoepidermoid carcinoma cells produce substantial amounts of IL-8 and express both CXCR1 and CXCR2 IL-8 receptors.81 IL-8 is highly expressed in NSCLC in men, smokers, elderly patients, and NSCLCs with pleural involvement, whereas it has been found to play a role in cell growth and migration in oncogenic KRAS-driven NSCLC.82 By using microarray analysis Sunaga et al82 identified, that IL-8 is the most downregulated gene by shRNA-mediated KRAS knockdown in NCI-H1792 NSCLC cells, in which IL-8 is overexpressed. These results indicate that activating mutations of KRAS or EGFR upregulate IL-8 expression in NSCLC. Luppi et al81 blocked the ability of IL-8 to increase cell proliferation by an inhibitor of EGFR tyrosine kinase, by a specific anti-EGFR blocking antibody, and by a panmetalloproteinase inhibitor, whereas similar results were obtained by using the G-protein-coupled receptors (GPCR) inhibitor pertussis toxin.81 Inhibition of the MAPK p42/44 (ERK1/2) also blocked the mitogenic effect of IL-8, which made IL-8 a potential target for oncogenic KRAS-driven NSCLC.81

Another key pathway required during the process of tumorigenesis for KRAS-mutant cancers is oncogene-induced senescence (OIS), a fail-safe program that prevents normal cells from progressing toward malignancy after introduction of a mutant form of an oncogene such as KRAS G12D.83 OIS is an irreversible cell-cycle arrest that is thought to be triggered early during tumorigenesis to inhibit aberrant cell-cycle progression, and prevents premalignant tumors from progressing to malignancy.83 Recently, Twist-related protein 1 (TWIST1), a basic helix-loop-helix transcription factor central to embryogenesis, has been shown to suppress OIS associated with KRASG12D and EGFR2 oncogenes, and favors metastatic dissemination of cancer cells through its ability to induce an epithelial-mesenchymal transition (EMT).84 Tran et al85 found that suppression of TWIST1 may be an effective prosenescence therapy for human lung cancer by providing the first in vivo demonstration that TWIST1 plays an important role in both acceleration and maintenance of KRASG12D-induced autochthonous lung tumorigenesis.

It is clear that RAS pathway mutant cells depend on many transcription factors, the study of which extends the basis for a combinatorial design of targeted therapies in lung cancer. There are many other studies that have attempted to characterize gene expression profiles in KRAS-positive adenocarcinoma. The induction of apoptosis by myelocytomatosis viral oncogene homolog (Myc) inhibition in early-stage KRAS-induced lung adenomas suggests that, in addition to proliferation, endogenous Myc is required to maintain survival of KRAS-induced lung tumors, which suggests that pharmacologic inhibition of Myc offers both specificity and efficacy in the treatment of neoplastic disease.86 Serine/threonine kinase 33 (STK33) promotes cancer cell viability in a kinase activity–dependent manner by selectively regulating suppression of mitochondrial apoptosis mediated through ribosomal protein S6 kinase-1 induced inactivation of the death agonist BAD (Bcl-2-associated death promoter protein) in mutant KRAS-dependent cells.87 Consequently, STK33 is required for survival of mutant KRAS-dependent cell lines, which suggests that small molecule kinase inhibitors of STK33 may be useful to treat KRAS-dependent tumors.88 It has been shown that cells dependent on mutant KRAS exhibit selective sensitivity to suppression of STK33.87 Finally, Puyol et al89 investigated the role of the interphase cyclin-dependent kinases (Cdk) in the development of NSCLC driven by an endogenous KRAS oncogene, with the ultimate goal of identifying potential synthetic lethal interactions between oncogenic KRAS signaling and the loss of Cdk activity. Ablation of Cdk4, but not Cdk2 or Cdk6, induces an immediate senescence response only in lung cells expressing an endogenous KRAS oncogene89 (Figure 4). No such response occurs in lungs expressing a single Cdk4 allele or in other KRAS-expressing tissues.89 More importantly, targeting Cdk4 alleles detectable by computed tomography scanning in advanced tumors also induces senescence and prevents tumor progression.89 These observations suggest that robust and selective pharmacologic inhibition of Cdk4 may provide therapeutic benefit for patients with NSCLC that carries KRAS oncogenes.

Section snippets

Conclusion

Somatic mutations that lead to gain-of-function and constitutive signaling of KRAS pathways represent a possible predictive biomarker for nonresponsiveness to TKI-based strategies. EGFR and KRAS mutations are mutually exclusive, and it could be that the absence of EGFR mutation is a negative predictor of EGFR TKI rather than the presence of KRAS mutation. The incidence of EGFR mutations in tumors responsive to EGFR kinase inhibitors varies from 71% to 100%. Thus, at this point, patients whose

Disclosure

The authors have stated that they have no conflicts of interest.

References (89)

  • Q.B. She et al.

    The BAD protein integrates survival signaling by EGFR/MAPK and PI3K/Akt kinase pathways in PTEN-deficient tumor cells

    Cancer Cell

    (2005)
  • C.M. Johannessen et al.

    TORC1 is essential for NF1-associated malignancies

    Curr Biol

    (2008)
  • T.S. Finco et al.

    Oncogenic Ha-Ras-induced signaling activates NF-kappaB transcriptional activity, which is required for cellular transformation

    J Biol Chem

    (1997)
  • W.C. Huang et al.

    Phosphorylation of CBP by IKKalpha promotes cell growth by switching the binding preference of CBP from p53 to NF-kappaB

    Mol Cell

    (2007)
  • J. Luo et al.

    A genome-wide RNAi screen identifies multiple synthetic lethal interactions with the Ras oncogene

    Cell

    (2009)
  • M.S. Kumar et al.

    The GATA2 transcriptional network is requisite for RAS oncogene-driven non-small cell lung cancer

    Cell

    (2012)
  • J.A. Simon et al.

    Roles of the EZH2 histone methyltransferase in cancer epigenetics

    Mutat Res

    (2008)
  • L.C. Sutherland et al.

    RBM5 as a putative tumor suppressor gene for lung cancer

    J Thorac Oncol

    (2010)
  • F. Luppi et al.

    Interleukin-8 stimulates cell proliferation in non-small cell lung cancer through epidermal growth factor receptor transactivation

    Lung Cancer

    (2007)
  • S. Ansieau et al.

    Induction of EMT by twist proteins as a collateral effect of tumor-promoting inactivation of premature senescence

    Cancer Cell

    (2008)
  • C. Scholl et al.

    Synthetic lethal interaction between oncogenic KRAS dependency and STK33 suppression in human cancer cells

    Cell

    (2009)
  • M. Puyol et al.

    A synthetic lethal interaction between K-Ras oncogenes and Cdk4 unveils a therapeutic strategy for non-small cell lung carcinoma

    Cancer Cell

    (2010)
  • G.J. Riely et al.

    KRAS mutations in non-small cell lung cancer

    Proc Am Thorac Soc

    (2009)
  • J.L. Bos

    Ras oncogenes in human cancer: a review

    Cancer Res

    (1989)
  • M.S. Brose et al.

    BRAF and RAS mutations in human lung cancer and melanoma

    Cancer Res

    (2002)
  • M. Trahey et al.

    A cytoplasmic protein stimulates normal N-ras p21 GTPase, but does not affect oncogenic mutants

    Science

    (1987)
  • H. Shigematsu et al.

    Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers

    J Natl Cancer Inst

    (2005)
  • T. Kosaka et al.

    Mutations of the epidermal growth factor receptor gene in lung cancer: biological and clinical implications

    Cancer Res

    (2004)
  • M. Taron et al.

    Activating mutations in the tyrosine kinase domain of the epidermal growth factor receptor are associated with improved survival in gefitinib-treated chemorefractory lung adenocarcinomas

    Clin Cancer Res

    (2005)
  • K. Okudela et al.

    KRAS gene mutations in lung cancer: particulars established and issues unresolved

    Pathol Int

    (2010)
  • P.J. Roberts et al.

    Personalized medicine in non-small-cell lung cancer: is KRAS a useful marker in selecting patients for epidermal growth factor receptor-targeted therapy?

    J Clin Oncol

    (2010)
  • K.L. Thu et al.

    Lung adenocarcinoma of never smokers and smokers harbor differential regions of genetic alteration and exhibit different levels of genomic instability

    PLoS ONE

    (2012)
  • F. Le Calvez et al.

    TP53 and KRAS mutation load and types in lung cancers in relation to tobacco smoke: distinct patterns in never, former, and current smokers

    Cancer Res

    (2005)
  • G.J. Riely et al.

    Frequency and distinctive spectrum of KRAS mutations in never smokers with lung adenocarcinoma

    Clin Cancer Res

    (2008)
  • S.L. Graziano et al.

    Prognostic significance of K-ras codon 12 mutations in patients with resected stage I and II non-small-cell lung cancer

    J Clin Oncol

    (1999)
  • P. Keohavong et al.

    Detection of K-ras mutations in lung carcinomas: relationship to prognosis

    Clin Cancer Res

    (1996)
  • C. Lu et al.

    Prognostic factors in resected stage I non-small-cell lung cancer: a multivariate analysis of six molecular markers

    J Clin Oncol

    (2004)
  • R.J. Slebos et al.

    K-ras oncogene activation as a prognostic marker in adenocarcinoma of the lung

    N Engl J Med

    (1990)
  • Y. Fukuyama et al.

    K-ras and p53 mutations are an independent unfavourable prognostic indicator in patients with non-small-cell lung cancer

    Br J Cancer

    (1997)
  • C.L. Huang et al.

    Mutations of p53 and K-ras genes as prognostic factors for non-small cell lung cancer

    Int J Oncol

    (1998)
  • M. Miyake et al.

    A novel molecular staging protocol for non-small cell lung cancer

    Oncogene

    (1999)
  • H.H. Nelson et al.

    Implications and prognostic value of K-ras mutation for early-stage lung cancer in women

    J Natl Cancer Inst

    (1999)
  • C. Mascaux et al.

    The role of RAS oncogene in survival of patients with lung cancer: a systematic review of the literature with meta-analysis

    Br J Cancer

    (2005)
  • V.D. Cataldo et al.

    Treatment of non-small-cell lung cancer with erlotinib or gefitinib

    N Engl J Med

    (2011)
  • Cited by (173)

    View all citing articles on Scopus
    View full text