Review Article
Ann Mutagen. 2017; 1(1): 1002.
Therapeutic Approaches of Non-Small Cell Lung Cancer (NSCLC) with KRAS Mutations
Mohamed F. Salama*
Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Egypt
*Corresponding author: Salama MF, Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Egypt
Received: March 20, 2017; Accepted: April 05, 2017; Published: April 13, 2017
Abstract
Mutations in KRAS are among the most commonly observed mutations in Non-Small Cell Lung Cancer (NSCLC) patients. However, different therapeutic approaches targeting mutant KRAS so far were not efficient. Targeting KRAS downstream signaling pathways such as MAPK, ERK is a promising tool to control the disease. In the current review, the different therapeutic strategies are briefly discussed.
Keywords: Non-small cell lung cancer; KRAS mutations; Therapy
Introduction
Lung cancer is associated with the highest cancer-related mortalities all over the world [1]. Several oncogenic mutations have been linked to the development of lung cancer. KRAS mutations are among those mutations that exist in about quarter of Non-Small Cell Lung Cancer (NSCLC) patients [2]. Mutations in Epidermal Growth Factor Receptor (EGFR) have also been observed in NSCLC patients. Mutations in EGFR and KRAS have been shown to be mutually exclusive in patients with NSCLC [3]. However, double mutations have recently been reported in some cases [4]. KRAS mutations can also coexist with other mutations such as p53and STK11 [5,6].
RAS is a GTP kinase that has been discovered almost 60 years ago. In NSCLC, KRAS missense substitutions mutations are mainly observed at codon 12, codon 13, and to a lesser extent at codon 61 [7]. G12C is the main KRAS mutation found in lung cancer patients that accounts for about 40% and is mostly observed in smokers. Other mutations include G12V and G12D that account for 22% and 16% of mutations, respectively [8,9]. The available information regarding the prognostic significance of KRAS mutations in NSCLC patients are scarce and elusive. In an earlier report, NSCLC patients with KRAS mutations has been shown to have a shorter overall survival (OS) compared to patients with wild-type KRAS [10]. In another study conducted on patients treated with first-line platinum-based chemotherapy, KRAS mutations have been shown to mildly affect OS [11]. However, in a recent study, analysis of data from patients treated with EGFR-tyrosine kinase inhibitor failed to demonstrate any difference in survival between wild-type and mutant KRAS tumors [12]. Moreover, KRAS mutation has recently been shown to be associated with poor prognosis in patients with lung adenocarcinoma with bone metastasis [13]. The type of mutated codon could also affect the disease outcome [9]. Codon 12 mutation, G12V, has been shown to be associated with poor prognosis [10,14].
Similar to its prognostic value, the predictive role of KRAS mutations in response to chemotherapy is also contradictory. Several studies did not show any predictive role of KRAS mutations in efficient response to chemotherapy [15-17]. A recent meta-analysis conducted on patients with advanced NSCLC following first line chemotherapy demonstrated that KRAS mutations decreased Overall Response Rate (ORR) and Progression Free Survival (PFS) [18]. An earlier retrospective analysis demonstrated a limited role of KRAS mutation in Asian patients with advanced NSCLC [19].
G12C and G12V mutations activate several downstream signaling cascades including RAL pathway and thus are associated with poor prognosis [20]. On the other hand, G12D mutation induces RAF/ MAPK/PI3K signaling [20]. Collectively, mutations in KRAS result in constitutively active protein independent of upstream signals due to loss of GTPase activity with subsequent activation of several downstream pathways such as MAPK, and AKT/mTOR. Therefore, targeting these signaling pathways is the preferred approach to treat lung cancer patients with KRAS mutations.
RAS Signaling
In normal cells, RAS is usually inactive and bound to GDP until it is triggered by external stimuli that exchange GDP for GTP forming an active molecule. Consequently, GTPase activating proteins inactivate RAS through hydrolysis of GTP. Mutations in KRAS are associated with loss of GTPase activity leading to constitutively active protein [21]. The signaling pathways downstream of KRAS (Figure 1) are in turn switched on including MAPK, ERK, AKT/mTOR leading to increased proliferation, angiogenesis, and resistance to apoptosis that favors tumor growth [21].
Figure 1: KRAS signaling in NSCLC. Mutations in KRAS are associated with a constitutively active RAS protein with the induction of downstream signaling cascades such as MEK/ERK1, 2 and PI3K/AKT/mTOR pathways that leads to proliferation, migration, invasion, and angiogenesis.
KRAS-Targeted Therapeutic Approaches
Most of therapeutic strategies that were developed to treat NSCLC patients with KRAS mutations are targeting its downstream signaling pathways such as RAF, MEK, ERK, PI3K. However, thorough understanding of these signaling pathways is crucial before developing therapeutic agents. For instance, MEK is activated by RAF, which in turn activates ERK that stimulates several downstream targets including transcription factors and protein kinases responsible for resistance of apoptosis, cell invasion, proliferation, and cell cycle progression [21]. Therefore, it is a real challenge to identify which arm in the signaling pathway is needed to be targeted to inhibit tumor progression. In addition, understanding of the main signaling pathways driven within the context of different KRAS mutations is indispensable for developing effective therapeutic strategies of NSCLC patients [21-23]. Over the past twenty years, several therapeutic agents against KRAS have been tested either by using pharmacologic inhibitors or its downregulation by siRNA. In addition, targeting KRAS downstream signaling pathways has also been examined.
Different strategies have been developed to target activated oncogenic GTP-RAS protein. First, by a competitive inhibitor that interferes with KRAS interaction with GTP [24-27]. The other approach was by targeting membrane binding of RAS [28]. However, inhibiting the membrane localization of RAS by an inhibitor, salirasib, was not efficient in phase II clinical trial [29]. A prospective trial study conducted in 2011 showed a remarkable effect of sorafenib treatment of NSCLC patients with KRAS mutation than those treated with erlotinib [30]. However, due to its toxicity, it is uncertain that sorafenib will be a good therapeutic agent for NSCLC patients with KRAS mutations.
Recently, a small inhibitor molecule that specifically binds to G12C mutant KRAS with no effect on wild-type protein has been developed that renders mutant protein to bind GDP instead of GTP [27]. That pre-clinical finding could pave the road to develop a second generation of the inhibitor to be used for phase I or phase II trials.
Immunotherapy
Immunotherapy is considered as a breakthrough therapeutic approach for cancer. The recent advanced immunotherapeutic strategies are based on the inhibition of protective mechanisms employed by cancer cells against immune cells. This is achieved by blocking certain immune checkpoints, such as Programmed cell Death-1 (PD-1). Recently, targeting PD-1 and its ligand PDL-1in clinical trials has demonstrated exceptional responses in NSCLC patients [31,32]. However, the response to immunotherapy is currently limited to a small number of patients [33]. Interestingly, It has recently been shown that patients with lung adenocarcinoma with KRAS and or/TP53 mutations exhibited more sensitivity to PD-1 targeted immunotherapy [34]. These findings suggest a potential predictive significance of KRAS mutation in immunotherapy.
Alternative Therapeutic Approaches
Since targeting KRAS has been shown to be inefficient, an alternative approach by inhibiting its downstream target such as MAPK could be more effective. Therefore, using sorafenib, a multidrug TKI, in a phase II trials was promising in controlling the disease [30,35,36]. However, in phase III MISSION trial, the analyses of group with KRAS mutations did not show any effectiveness of sorafenib [37].
Another candidate downstream of RAS is MEK1/2. An oral non- ATP competitive MEK1/2 inhibitor, selumetinib was developed [38]. Treatment of advanced cancer patients with that inhibitor achieved a good tumor response in early phase [39]. However, it showed little effect in phase II trials [40]. Combined treatment of selumetinib and docetaxel exhibited a good synergistic tumor regressive effect in vivo [22]. When similar strategy applied in an early phase study, manageable side effects were observed [41]. Therefore, a phase II study was performed in NSCLC patients with KRAS mutations who received combined selumetinib and docetaxel therapy and it showed improvement in PFS [42]. However, results of a subsequent phase III study did not show any effect of combined selumetinib and docetaxel therapy on OS, PFS, or ORR [43]. Therefore, the production of selumetinib was stopped by AstraZeneca.
A second allosteric MEK1/2 inhibitor, trametinib was also tested in a phase I trial in NSCLC patients with mutant KRAS and exhibited a stable disease response in 53% of patients [44]. Consequently, in a phase II trial, trametinib-treated patients had no effect on PFS. However, in three patients, 80% tumor regression was observed [45].
Cyclin Dependent Protein Kinases (CDKs) are the main regulators of cell cycle and therefore are potential therapeutic targets in NSCLC with KRAS mutations. Synthetic lethality was induced after targeting CDK in KRAS mutant NSCLC in in vitro and in vivo preclinical studies, suggesting that CDK plays a key role in tumorigenesis [46]. The first CDK inhibitor, flavopiridol was developed that showed little effect in phase II trials [47]. A new CDK inhibitor palbociclib has been shown to have a good effect in ER+ breast cancer patients [48]. However, its use in clinical trials for treatment of NSCLC patients is ongoing.
Another CDK inhibitor, Roscovitine was evaluated in NSCLC patients. However, it had no effect on survival [47]. Another cell cycle inhibitor, LY2835219 has been tested in xenografts and showed a good effect. A phase III trial (NCT02152631) is currently ongoing and recruiting patients.
Focal Adhesion Kinase (FAK) is another candidate to be targeted in NSCLC with KRAS mutation. FAK is a tyrosine kinase that is involved in cellular adhesion, invasion in different cancer types. FAK inhibition has been shown to induce tumor regression in KRAS mutant mice [49]. Defactinib is a FAK inhibitor that has recently been tested in Asian phase I study in Japanese patients with advanced solid tumors and showed good tolerability [50]. Defactinib has also been used in phase II trial in KRAS mutant NSCLC patients (NCT01951690). However, negative unpublished results were reported at the 16th World Conference on Lung Cancer.
Conclusion
Although KRAS mutations are frequently observed in NSCLC patients, it is apparent that targeting mutant KRAS is a real challenge. The complexity of its downstream signaling pathways and the absence of oncogenic addiction made it difficult to develop an effective therapeutic approach. However, currently there are promising therapeutic trials that could effectively improve the overall survival of patients.
References
- Siegel RL, Miller KD, Jemal A. Cancer statistics. CA Cancer Journal for Clinicians. 2016; 66: 7-30.
- Karnoub AE, Weinberg RA. Ras oncogenes: split personalities. Nature reviews Molecular cell biology. 2008; 9: 517-531.
- Jang TW, Oak CH, Chang HK, Suo SJ, Jung MH. EGFR and KRAS mutations in patients with adenocarcinoma of the lung. The Korean journal of internal medicine. 2009; 24: 48-54.
- Lee T, Lee B, Choi YL, Han J, Ahn MJ, Um SW. Non-small cell lung Cancer with concomitant EGFR, KRAS, and ALK mutation: clinicopathologic features of 12 cases. Journal of pathology and translational medicine. 2016; 50: 197-203.
- Gibbons DL, Byers LA, Kurie JM. Smoking, p53 mutation, and lung cancer. Molecular Cancer Research. 2014; 12: 3-13.
- Pao W, Girard N. New driver mutations in non-small-cell lung cancer. The lancet oncology. 2011; 12: 175-180.
- Tabin CJ, Bradley SM, Bargmann C I, Weinberg RA. Mechanism of activation of a human oncogene. Nature. 1982; 300: 143-149.
- Forbes SA, Bindal N, Bamford S, Cole C, Kok CY, Beare D, et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Research. 2011; 39: D945-D950.
- Garassino M, 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. Annals of oncology. 2011; 22: 235-237.
- Prior IA, Lewis PD, Mattos C. A comprehensive survey of Ras mutations in cancer. Cancer research. 2012; 72: 2457-2467.
- Marabese M, Ganzinelli M, Garassino MC, Shepherd FA, Piva S, Caiola E, et al. KRAS mutations affect prognosis of non-small-cell lung cancer patients treated with first-line platinum containing chemotherapy. Oncotarget. 2015; 6: 34014-34022.
- Zer A, Ding K, Lee SM, Goss GD, Seymour L, Ellis PM, 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. Journal of thoracic oncology. 2016; 11: 312-323.
- Lohinai Z, Klikovits T, Moldvay J, Ostoros G, Raso E, Timar J, et al. KRAS-mutation incidence and prognostic value are metastatic site-specific in lung adenocarcinoma: poor prognosis in patients with KRAS mutation and bone metastasis. Scientific Reports. 2017; 7.
- Stephen AG, Esposito D, Bagni RK, McCormick F. Dragging ras back in the ring. Cancer cell. 2014; 25: 272-281.
- Dong X, Zhao X, Hao Y, Wei Y, Yin Q, Du J. Response to first-line chemotherapy in patients with non–small-cell lung cancer according to epidermal growth factor receptor and K-RAS mutation status. Clinical lung cancer. 2013; 14: 680-687.
- Mellema WW, Dingemans AMC, Thunnissen E, Snijders PJ, Derks J, Heideman DA, et al. KRAS mutations in advanced nonsquamous non-small-cell lung cancer patients treated with first-line platinum-based chemotherapy have no predictive value. Journal of Thoracic Oncology. 2013; 8: 1190-1195.
- O'Byrne KJ, Gatzemeier U, Bondarenko I, Barrios C, Eschbach C, Martens UM, et al. Molecular biomarkers in non-small-cell lung cancer: a retrospective analysis of data from the phase 3 FLEX study. The lancet oncology. 2011; 12: 795-805.
- Zhang Y, Fang W, Yan Y, Wang M, Kang S, Sheng J, et al. The efficacy of first-line chemotherapy is associated with KRAS mutation status in patients with advanced non-small cell lung cancer: a meta-analysis. Medical Oncology. 2015; 32: 61.
- Sun JM, Hwang DW, Ahn JS, Ahn MJ, Park K. Prognostic and predictive value of KRAS mutations in advanced non-small cell lung cancer. PloS one. 2013; 8: e64816.
- Ihle NT, Byers LA, Kim ES, Saintigny P, Lee JJ, Blumenschein GR, et al. Effect of KRAS oncogene substitutions on protein behavior: implications for signaling and clinical outcome. Journal of the National Cancer Institute. 2012; 104: 228-239.
- Vasan N, Boyer JL, Herbst RS. A RAS renaissance: emerging targeted therapies for KRAS-mutated non–small cell lung cancer. Clinical Cancer Research. 2014; 20: 3921-3930.
- Chen Z, Cheng K, Walton Z, Wang Y, Ebi H, Shimamura T, et al. A murine lung cancer co-clinical trial identifies genetic modifiers of therapeutic response. Nature. 2012; 483: 613-617.
- Shackelford DB, Abt E, Gerken L, Vasquez DS, Seki A, Leblanc M, et al. LKB1 inactivation dictates therapeutic response of non-small cell lung cancer to the metabolism drug phenformin. Cancer cell. 2013; 23: 143-158.
- Maurer T, Garrenton LS, Oh A, Pitts K, Anderson DJ, Skelton NJ, et al. Small-molecule ligands bind to a distinct pocket in Ras and inhibit SOS-mediated nucleotide exchange activity. Proceedings of the National Academy of Sciences. 2012; 109: 5299-5304.
- Sun Q, Burke JP, Phan J, Burns MC, Olejniczak ET, Waterson AG, et al. Discovery of small molecules that bind to K‐Ras and inhibit Sos‐mediated activation. Angewandte Chemie. 2012; 124: 6244-6247.
- Shima F, Yoshikawa Y, Ye M, Araki M, Matsumoto S, Liao J, et al. In silico discovery of small-molecule Ras inhibitors that display antitumor activity by blocking the Ras-effector interaction. Proceedings of the National Academy of Sciences. 2013; 110: 8182-8187.
- Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-Ras (G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013; 503: 548-551.
- Zimmermann G, Papke B, Ismail S, Vartak N, Chandra A, Hoffmann M, et al. Small molecule inhibition of the KRAS-PDE [delta] interaction impairs oncogenic KRAS signalling. Nature. 2013; 497: 638-642.
- Riely GJ, Johnson ML, Medina C, Rizvi NA, Miller VA, Kris MG, et al. A phase II trial of Salirasib in patients with lung adenocarcinomas with KRAS mutations. Journal of Thoracic Oncology. 2011; 6: 1435-1437.
- Kim ES, Herbst RS, Wistuba II, Lee JJ, Blumenschein GR, Tsao A, et al. The BATTLE trial: personalizing therapy for lung cancer. Cancer discovery. 2011; 1: 44-53.
- Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, etn al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. New England Journal of Medicine. 2012; 366: 2455-2465.
- O'Sullivan Coyne G, Madan RA, Gulley JL. Nivolumab: promising survival signal oupled with limited toxicity raises expectations. American Society of Clinical Oncology. 2014; 32: 986-988.
- Carbognin L, Pilotto S, Milella M, Vaccaro V, Brunelli M, Calio A, et al. Differential activity of nivolumab, pembrolizumab and MPDL3280A according to the tumor expression of Programmed Death-Ligand-1 (PD-L1): sensitivity analysis of trials in melanoma, lung and genitourinary cancers. PloS one. 2015; 10: e0130142.
- Dong ZY, Zhong W, Zhang XC, Su J, Xie Z, Liu SY, et al. Potential Predictive Value of TP53 and KRAS Mutation Status for Response to PD-1 Blockade Immunotherapy in Lung Adenocarcinoma. Clinical Cancer Research. 2017; 2554: 2016.
- Smit EF, Dingemans AMC, Thunnissen FB, Hochstenbach MM, van Suylen RJ, Postmus PE. Sorafenib in patients with advanced non-small cell lung cancer that harbor K-ras mutations: a brief report. Journal of Thoracic Oncology. 2010; 5: 719-720.
- Dingemans AMC, Mellema WW, Groen HJ, Van Wijk A, Burgers SA, Kunst PW, et al. A phase II study of sorafenib in patients with platinum-pretreated, advanced (stage IIIb or IV) non-small cell lung cancer with a KRAS mutation. Clinical Cancer Research. 2013; 19: 743-751.
- Mok TSK PAL, Wu YL, Novello S, Juhasz E, Aren O, Sun Y, et al. Association between tumor EGFR and KRAS mutation Status and clinical outcomes in NSCLC patients randomized to sorafenib plus Best Supportive Care (BSC) or BSC alone: subanalysis of the phase iii mission trial. Annals of Oncology. 2012; 23: ixe1-ixe30.
- Yeh TC, Marsh V, Bernat BA, Ballard J, Colwell H, Evans RJ, et al. Biological characterization of ARRY-142886 (AZD6244), a potent, highly selective mitogen-activated protein kinase kinase 1/2 inhibitor. Clinical Cancer Research. 2007; 13: 1576-1583.
- Banerji U, Camidge DR., Verheul HM, Agarwal R, Sarker D, Kaye SB, et al. The first-in-human study of the hydrogen sulfate (Hyd-sulfate) capsule of the MEK1/2 inhibitor AZD6244 (ARRY-142886): a phase I open-label multicenter trial in patients with advanced cancer. Clinical Cancer Research. 2010; 16: 1613-1623.
- Hainsworth JD, Cebotaru CL, Kanarev V, Ciuleanu TE, Damyanov D, Stella P, et al. A phase II, open-label, randomized study to assess the efficacy and safety of AZD6244 (ARRY-142886) versus pemetrexed in patients with non-small cell lung cancer who have failed one or two prior chemotherapeutic regimens. Journal of thoracic oncology: official publication of the International Association for the Study of Lung Cancer. 2010; 5: 1630-1636.
- Kim K, Infante J, Cohen R, Burris H, Emeribe U, Curt G, Clemett D, LoRusso P. Abstract B225: A phase I dose-escalation study of selumetinib in combination with docetaxel in patients with advanced solid tumors. AACR. 2011; 10.
- Janne PA, Shaw AT, Pereira JR, Jeannin G, Vansteenkiste J, Barrios C, et al. Selumetinib plus docetaxel for KRAS-mutant advanced non-small-cell lung cancer: a randomised, multicentre, placebo-controlled, phase 2 study. The lancet oncology. 2013; 14: 38-47.
- Janne PA, van den Heuvel M, Barlesi F, Cobo M, Mazieres J, Crino L, et al. Selumetinib in combination with docetaxel as second-line treatment for patients with KRAS-mutant advanced NSCLC: Results from the phase III SELECT-1 trial. Annals of Oncology. 2016; 27.
- Infante JR, Fecher LA, Falchook GS, Nallapareddy S, Gordon MS, Becerra C, et al. Safety, pharmacokinetic, pharmacodynamic, and efficacy data for the oral MEK inhibitor trametinib: a phase 1 dose-escalation trial. The lancet oncology. 2012; 13: 773-781.
- Blumenschein GR, Smit E, Planchard D, Kim DW, Cadranel J, De Pas T, et al. A randomized phase II study of the MEK1/MEK2 inhibitor trametinib (GSK1120212) compared with docetaxel in KRAS-mutant advanced Non-Small-Cell Lung Cancer (NSCLC). Annals of Oncology. 2015; 26: 894-901.
- Puyol M, Martin A, Dubus P, Mulero F, Pizcueta P, Khan G, 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; 18: 63-73.
- Asghar U, Witkiewicz AK, Turner NC, Knudsen ES. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nature reviews Drug discovery. 2015; 14: 130-146.
- Finn RS, Crown JP, Lang I, Boer K, Bondarenko IM, Kulyk, SO, et al. The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): a randomised phase 2 study. The lancet oncology. 2015; 16: 25-35.
- Konstantinidou G, Ramadori G, Torti F, Kangasniemi K, Ramirez RE, Cai Y, et al. RHOA-FAK is a required signaling axis for the maintenance of KRAS-driven lung adenocarcinomas. Cancer discovery. 2013; 3: 444-457.
- Shimizu T, Fukuoka K, Takeda M, Iwasa T, Yoshida T, Horobin J, et al. A first-in-Asian phase 1 study to evaluate safety, pharmacokinetics and clinical activity of VS-6063, a Focal Adhesion Kinase (FAK) inhibitor in Japanese patients with advanced solid tumors. Cancer chemotherapy and pharmacology. 2016; 77: 997-1003.