Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 A RAS Renaissance: Emerging Targeted Therapies for KRAS-Mutated Non–Small Cell Lung Cancer Neil Vasan1, Julie Boyer2, and Roy S. Herbst2 1 Department of Internal Medicine, Massachusetts General Hospital, Boston, MA 2 Yale Cancer Center and Smilow Cancer Hospital at Yale-New Haven, New Haven, CT Grant Support Research reported in this publication was supported by the National Institutes of Health under award number MSTP TG 2T32GM07205 (N. Vasan). Corresponding author: Roy S. Herbst, MD, PhD Yale Medical Oncology PO Box 208028 New Haven, CT 06520-8028 ph (203) 785-6879 fax (203) 737-5698 email: [email protected] Running title: A RAS Renaissance Disclosure of Potential Conflicts of Interest Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 2 No potential conflicts of interest were disclosed. Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 3 Abstract Of the numerous oncogenes implicated in human cancer, the most common and perhaps the most elusive to target pharmacologically is RAS. Since the discovery of RAS in the 1960s, numerous studies have elucidated the mechanism of activity, regulation, and intracellular trafficking of the RAS gene products, and of its regulatory pathways. These pathways yielded druggable targets, such as farnesyltransferase during the 1980s-1990s. Unfortunately, early clinical trials investigating farnesyltransferase inhibitors yielded disappointing results, and subsequent interest by pharmaceutical companies in targeting RAS waned. However, recent advances including the identification of novel regulatory enzymes (e.g. Rce1, Icmt, Pdeδ), siRNA-based synthetic lethality screens, and fragment-based small molecule screens have resulted in a "Ras renaissance", signified by new Ras and Ras-pathway targeted therapies that have led to new clinical trials of patients with Ras-driven cancers. This review gives an overview of KRas signaling pathways with an emphasis on novel targets and targeted therapies, using non-small cell lung cancer (NSCLC) as a case example. Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 4 Introduction Three RAS genes encode four proteins: HRas, KRas4a, KRas4b, and NRas (1). These proteins are GTPases, which function as molecular switches: "on" when bound to GTP and "off" when bound to GDP. Ras-GTP can bind to numerous partner proteins, termed "effectors", and these Ras-effector interactions lead to a cascade of downstream signaling events (2). In normal cells, Ras signaling is crucial for proliferation, differentiation, and survival (3). The hydrolysis of GTP to GDP by Ras is a slow process, and therefore Ras cycles between these states with the aid of regulatory proteins. GTPase activating proteins (GAPs) catalyze the hydrolysis of GTP to GDP (“on to off”); while guanine nucleotide exchange factors (GEFs) catalyze the dissociation of GDP, with GTP binding afterwards due to its high concentration in cells (“off to on”) (4) (Figure 1A). However, this pathway is co-opted by oncogenic mutations in Ras. Among the four Ras isoforms, the most common mutations are at amino acid positions G12, G13, and Q61 (5). Crystal structures of Ras proteins have modeled these mutants' mechanisms of activation. Q61 mutants prevent coordination of a water molecule necessary for GTP hydrolysis (6), while G12 and G13 mutants prevent binding of Ras to its GAP and interfere with the orientation of Q61. These mutants result in Ras-GTP in an "on state", driving oncogenesis (7) (Figure 1B). The Ras proteins are important mediators of cell signaling. There is a wide range of Ras effector proteins, notably Raf (MAP kinase pathway), PI3 kinase (Akt/mTOR pathway), and RalGDS (Ral pathway). These effectors (which represent only a subset of downstream Ras signaling nodes) are highly complex with numerous redundancies and interactions between pathways (8). Dysregulated Ras signaling results in increased proliferation, decreased apoptosis, disrupted cellular metabolism, and increased angiogenesis, all seminal hallmarks of cancer (9) Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 5 (Figure 1B). Ras mutations: differences from isoform to amino acid RAS is the most commonly mutated oncogene in cancer (8), with distinct Ras isoforms detected in various cancers (10). KRas is the most commonly mutated isoform. Listed in order of percentage of cases, KRas mutations are most common in cancers of the pancreas, colon, biliary tract, and lung (the majority of which are adenocarcinomas); NRas mutations are most common in cancers of the skin (malignant melanoma) and hematopoietic system [acute myeloid leukemia (AML)]; HRas mutations are most common in cancers of the head and neck (squamous cell carcinoma) and urinary tract (transitional cell carcinoma). Ras mutations are much less common in cancers of the breast, central nervous system, or prostate (5) (Figure 2A). Why certain cancers seem to be driven preferentially by specific isoforms remains an outstanding question. Another unsettled issue in oncogenesis is the differential role, if any, among different Ras activating point mutations. In lung cancer, the most common mutations are KRas G12C, G12V, and G12D (11). Other KRas-driven cancers have different mutation frequencies: in the colon G12D, G12V, and G13D; in the pancreas G12D, G12V, and G12R; and in the biliary tract G12D, G12V, and G12S (Figure 2B). Unlike in HRas and NRas, KRas Q61 oncogenic mutations are very rare (7). In vitro, KRas G12V and G12R have greater transforming ability as shown by soft agar colony formation (12). Unexpectedly, there is no correlation between the GTPase activity of the mutant and its propensity to transform (13). However, once transformed, certain mutations are more aggressive than others. Mice with KRas G12V, G12R, and G12D had higher-stage lung tumors compared to KRas G12C or wildtype (14). In lung cancer patients, KRas G12C resulted Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 6 in increased sensitivity to pemetrexed and taxol compared to G12V and G12D, although G12D patients were more likely to respond to sorafenib (15). A recent retrospective analysis of the BATTLE clinical trial (16) (discussed below) found worse progression-free survival for the group of patients with either KRas G12C or G12V, compared to other KRas mutants, or wildtype (1.84 months vs. 3.35 months [P=0.046], vs. 1.95 months) (17). KRas G12C and G12V had increased signaling through Ral and decreased signaling through Akt. This study suggests that targeted treatments and clinical trials in nonsmall cell lung cancer (NSCLC) may need to consider the specific KRas point mutation. Together the in vitro, in vivo, and patient data suggest a greater oncogenic potential for KRas G12V (present in ~20% of KRas-mutated lung cancers) compared to other mutations (18). Inhibiting Ras membrane association A series of enzymes, (Figure 3) beginning with farnesyltransferase (FTase) acts posttranslationally on the C-terminal C-A-A-X motif of Ras resulting in the attachment to membranes through cysteine prenylation (19). Next, Ras trafficks to the endoplasmic reticulum, where its last three amino acids are proteolyzed by Ras converting enzyme (RCE1), and then its C-terminus is methylated by isoprenylcysteine carboxyl methyltransferase (ICMT) (20). In the Golgi, HRas, NRas, and KRas4A are palmitoylated, and the fully processed Ras then trafficks to its final plasma membrane location (21). KRas4b is not palmitoylated but rather associates electrostatically with the membrane through a polybasic stretch in its C-terminus (22). Thus, two modes of membrane association poise Ras isoforms for activation and signaling. FTase Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 7 Initial attempts to inhibit Ras focused on FTase (23). These FTase inhibitors (e.g. lonafarnib, tipifarnib) (24) were oral medications, well tolerated, specific for FTase, and were effective against HRas-transformed cells and HRas-driven murine tumors (25). However, these drugs did not increase survival in clinical trials of patients with KRas-mutated pancreatic cancer (26). Later studies found that with inhibition of FTase, KRas could be alternatively prenylated by geranylgeranyltransferase I (GGTase I) (25). Moreover, dual inhibition of FTase and GGTase I did not decrease levels of prenylated KRas (27). Notably, tipifarnib has shown antitumor activity against AML, chronic myelogenous leukemia (CML), and myeloproliferative disorders (28) (often driven by NRas) and in breast cancer, (29) which warrants further study. Another class of FTase inhibitors has been developed (e.g. salirasib) (30) containing farnesylcysteine, thought to compete for membrane-docking proteins that bind farnesyl moieties. (Figure 3) In vitro, salirasib inhibits all Ras isoforms (31). However, it failed to induce radiographic response or increase survival in a phase II NSCLC trial (32). Together these studies revealed several inherent problems in targeting Ras prenylation. First, there is alternative prenylation with dissimilarities among isoforms—KRas and NRas are prenylated by GGTase I (33)—previously thought to be identical to that of HRas. While HRas mutations are infrequent compared to KRas or NRas, FTase inhibitors would target HRas because HRas does not undergo alternative prenylation by GGTase I (33). Such findings could be exploited in a clinical trial of FTase inhibitors for patients with HRas-driven urothelial cancers. Additionally, any observed decreases in tumor size due to FTase inhibition do not correlate with KRas-mutation status or other mutations in the Ras signaling pathway (34). This suggests alternative mechanisms of resistance such as Ras gene amplification, which has been Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 8 observed as a resistance mechanism to MET tyrosine kinase inhibitors (35) or off-target effects, which could be mediated by the large number (>55) of prenylated substrates (e.g. other small GTPases) (36). This may explain why there is not an adequate single biomarker, such as decreased Ras prenylation, for monitoring FTase inhibitor effects. Nevertheless, there are many current clinical trials investigating combination therapies of FTase inhibitors with other cytotoxic and targeted therapies (37). The failure of FT inhibition as a general strategy for targeting KRas has spurred preclinical studies of the other enzymes in the Ras processing pathway: Rce1, Icmt, and Pdeδ. RCE1, ICMT, PDEδ Disruption of the RCE1 gene slows mouse fibroblast cell growth (38); conversely, in another study, deficiency of RCE1 in a genetically engineered mouse model (GEMM) of KRas-driven myeloproliferative disease actually increased disease progression (39). These paradoxical results may reflect cell specific differences in KRas signaling. Several inhibitors against yeast and human Rce1 have been developed (40), however, these bind with only low micromolar affinity so future optimization must occur before these drugs enter clinical trials. Inactivation of ICMT in the presence of activated KRas in fibroblasts leads to decreased cell growth and xenograft tumor formation (41). Concordantly, inactivation of ICMT in a GEMM of KRas-driven myeloprolifative disease also decreased lung tumor formation (42). Several small molecule inhibitors of Icmt have been developed including cysmethynil, which reduces anchorage-independent cell growth in colon cancer cells (43). Of note, inhibition of Icmt has been shown to be an off-target effect of methotrexate, possibly through an increase in Sadenosylhomocysteine, a methyltransferase inhibitor (44). Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 9 Another recently discovered intracellular trafficking target is Pdeδ (phosphodiesterase delta), a subunit of the cyclic GMP phosphodiesterase complex. Pdeδ also functions as a chaperone protein that binds to farnesylated Ras and enhances the presence of fully processed KRas4b at the plasma membrane, and the presence of depalmitoylated HRas, NRas, and KRas4a at the Golgi (45) (Figure 3). Importantly, downmodulation of Pde6δ decreases oncogenic KRas signaling. A recent paper reports benzimidazole small molecule compounds that inhibit the mammalian Pdeδ-KRas interaction with nanomolar affinity, suppresses oncogenic signaling in vitro in KRAS-mutated pancreatic ductal adenocarcinoma cells and most tantalizingly in vivo in a pancreatic cancer mouse xenograft model (46). These findings likely will herald phase I trials of these novel inhibitors. Direct inhibition of the Ras protein The affinity of GTP for Ras is extremely potent, in the picomolar range (47); thus attempts to inhibit Ras competitively would be difficult. There are several previous reports of Ras small molecule inhibition—nucleotide exchange inhibitors (48) and Ras-Raf inhibitors (49) —but in the absence of clear structural data, it is difficult to know if these effects are direct or indirect. This has led to a search for allosteric inhibitors of Ras. Based on the crystal structure of SOS (a Ras GEF) bound to Ras, Patgiri and colleagues designed a cell-permeable peptide to inhibit Ras activation (50). As shown by NMR spectroscopy, the peptide binds Ras with micromolar affinity at the same location as the analogous region of SOS, but does so about twice as avidly. This results in inhibition of downstream MAP kinase signaling. Two recent groups (51)(52) have used NMR fragment-based lead discovery and Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 10 structure-based drug design to find novel small molecule allosteric inhibitors of KRas. Both groups' inhibitors bind at a hydrophobic pocket between the switch 2 and core beta sheet region of the protein, with micromolar affinity. The binding site is distinct from but partially overlapping with the SOS binding site such that SOS is unable to activate KRas. Several FDA-approved tyrosine kinase inhibitors (e.g. afatinib, ibrutinib) take advantage of irreversible binding to a cysteine amino acid residue close to the active site. This approach inspired the development of irreversible inhibitors of KRas G12C, a specific mutant where the glycine at the 12th position of the KRas protein is mutated to cysteine (53). With sub-micromolar affinity, these inhibitors block SOS-mediated nucleotide exchange, favoring the binding of GDP instead of GTP, and rendering the KRas protein in an “off state.” When bound to KRas G12C, the compounds create a new binding surface mostly involving the switch 2 region. Importantly, they decreased viability and activated apoptosis in a KRas G12C specific lung cancer cell line. While these approaches are tantalizing, more potent drugs that bind with nanomolar affinity would be needed for a viable drug. Nevertheless, these studies provide novel lead compounds for future optimization. Inhibiting downstream Ras signaling Many classes of inhibitors exist against components of the canonical Ras signaling pathway, including Raf, mTOR, PI3K, PI3K/mTOR, Akt, and MEK. Clinical trials with these agents are ongoing, but to date, these drugs have not been shown to be effective against Ras-driven cancers as single agents. This has led to the identification of new targets, combination approaches of multiple targeted therapies, and combination approaches of targeted therapies with conventional cytotoxic chemotherapy. Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 11 Identification of synthetic lethal targets Synthetic lethality is a phenomenon where perturbation in either one of two different genes does not cause cell death, but where perturbation of both leads to cell death (54). In cancer, this initial perturbation could be a mutated oncogene or tumor suppressor, making this an attractive strategy for targeting cancer cells since normal cells would not have the mutation of interest and would theoretically be unaffected by a targeted treatment. This is also a strategy to develop ways to target oncogenes that are not readily “druggable” (e.g. Ras, transcription factors) or tumor suppressors where protein target levels are decreased. The most successful example of this approach in oncology is the use of mTOR and VEGF inhibitors that indirectly target HIF1α in VHL-mutated clear cell renal cell carcinoma (55). While the Ras signaling pathway is complex, it is thought that there may be critical nodes in the pathway that could be exploited. In KRas-mutated NSCLC, several small molecule synthetic lethality screens have yielded lead compounds including lanperisone, which induces oxidative stress in KRas-mutated cells (56), and oncrasin, which is synthetically lethal between KRas and PKCι (protein kinase C iota) and functions through inhibition of RNA polymerase II (57). Moreover, the rheumatoid arthritis drug aurothiomalate, which inhibits PKCι signaling, is currently in phase I clinical trials in NSCLC (58). More recently, RNA silencing technologies have facilitated the identification of new targets that, when deleted, are synthetically lethal with KRas mutations; this can be exploited clinically if the newly identified target has a known inhibitor. For example, in KRas-driven colon cancer, previous RNAi studies have shown the importance of TAK1 (TGF-β activated kinase-1), a MAP kinase kinase kinase and PLK1 (Pololike kinase 1), which functions at the mitotic spindle (59). Here we focus on novel targets in Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 12 preclinical development elucidated through synthetic lethality studies of NSCLC (Table 1). TBK1 Using an shRNA screen targeting genes encoding druggable proteins--kinases, phosphatases, and oncogenes--Barbie et al. identified TANK-binding kinase 1 (TBK1) as essential in KRas-driven cancers (60). These authors validated initially-identified candidate genes through a higherstringency secondary screen in lung adenocarcinoma cells, with TBK1 knockdown leading to the greatest amount of cell death, after KRas itself. TBK1 is linked to KRas through RalB, a small GTPase downstream of KRas that is part of the Ral signaling pathway, and is activated by RalB and Sec5, a component of the exocyst, an intracellular tethering complex (61). Activated TBK1 leads to increased antiapoptotic NF-κB signaling through BCL-XL and the c-Rel protooncogene. TBK1 inhibitors with nanomolar affinity have been developed (62), confirming pharmacological tractability. WT1 Targeting genes from a previously identified KRas transcriptional signature (63), as well as transcriptional regulators, Vicent et al. identified the Wilms tumor 1 transcription factor (WT1) as critical for oncogenic KRas signaling (64). Mechanistically, loss of WT1 decreases proliferation and increases cell senescence in KRas-driven cancers and was confirmed in mouse cell lines, GEMMs, and in human cell lines. Moreover, the authors were able to correlate WT1 expression levels with prognosis in KRas-driven lung cancer patients, strengthening this connection. WT1 is a well-known tumor suppressor in Wilms tumor but recent data have implicated it as a possible oncogene with overexpression in lung cancer (65). While WT1 is Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 13 currently not druggable, further examination of WT1’s role in cell senescence may yield future therapies. CDK4 By querying the role of individual cyclin dependent kinases (CDKs) in KRas driven NSCLC, Puyol et al. found a synthetic lethal interaction by disrupting CDK4, but not other related CDKs, causing cellular senescence (66). This effect was recapitulated in mouse embryonic fibroblasts and in a KRas-driven GEMM. Interestingly, the necessity of CDK4 in KRas-driven cancer was observed only in the lung, and not in the colon or pancreas, pointing to possible tissue specific dependencies in the Ras pathway. Additionally, these authors showed that small molecule inhibition of CDK4 (using a CDK4/CDK6 dual antagonist) resulted in tumor regression, with decreased CDK4-mediated phosphorylation; however, this inhibition did not cause the previously observed senescence, pointing to the need for more potent and specific CDK4 inhibitors as well as using them in combination therapies. GATA-2 Screening KRas-driven human NSCLC cell lines, Kumar et al. discovered the transcription factor GATA-2 as necessary for cell viability, in vitro and in vivo in a lung cancer xenograft model (67). Gene expression analysis revealed multiple upregulated pathways: the proteasome, IL-1 signaling, and Rho/ROCK signaling. The authors showed that GATA-2 normally upregulates the proteasome through Nrf1, IL-1 signaling through TRAF6, and Rho signaling through STAT5. Together these pathways converge onto NF-kB signaling which contributes to KRas-driven oncogenesis. Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 14 As transcriptional factors are notoriously difficult to target, the authors relied on approved drugs that target the newly delineated components of the GATA-2 pathway: bortezomib, which inhibits the proteasome and NF-kB indirectly, and fasudil, which inhibits Rho/ROCK signaling. Together, this combination therapy resulted in tumor regression in a KRas-driven lung cancer GEMM. This non-oncogene addiction to GATA-2 was also confirmed independently by a separate group (68). BCL-XL With the goal of finding genes whose inhibition may cooperate with the MEK inhibitor selumetinib in KRas-mutated cells, Corcoran et al. designed a pooled shRNA drug screen of "druggable" genes and identified BCL-XL, an antiapoptotic gene (69). Using navitoclax, a BCLXL small molecule inhibitor, in combination with selumetinib, they showed increased apoptosis in Kras-driven lung adenocarcinoma cell lines. Alone, selumetinib increases the amount of BIM protein, a proapoptotic factor, but also increases the amount of BIM bound to BCL-XL; BIM must be unbound to induce apoptosis, so its levels induced by selumetinib are insufficient for this effect. However, the combination of selumetinib with navitoclax increases the total amount of BIM, and decreases the amount of BIM/BCL-XL complex. Some of this free BIM forms a complex with MCL-1, and this complex is able to drive apoptosis. Thus the authors proposed that the combination therapy "frees up" more BIM to sufficient levels to drive apoptosis. Demonstrating the robustness and generalizability of this combination therapy, they show increased apoptosis in a large percentage of KRas-driven colorectal, lung, and pancreatic cancer cell lines; in KRas-mutant xenografts; and in a KRas-driven lung cancer GEMM. Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 15 Clinical trials targeting KRas-driven NSCLC Patients with KRas-mutated NSCLC derive less benefit from clinical trials compared to their KRas-wildtype patient counterparts (70) (71). Several targeted therapy clinical trials specifically address patients with KRas-mutated NSCLC. These trials represent the translational extension of the many decades of basic science research on the Ras pathway (Table 2). BATTLE and BATTLE-2 As we have seen, biomarkers (e.g. mutations, overexpression) do not always correlate with effects of a targeted therapy. Moreover, as patients receive multiple treatment modalities, these markers may change even though the therapy is often dictated by the pretreatment tumor genotype. The phase 2 Biomarker-integrated Approaches of Targeted Therapy for Lung Cancer Elimination (BATTLE) trial addressed this issue in a novel manner (16). Ninety-seven patients with chemorefractory NSCLC were randomized into 4 treatment groups based on biomarker analysis of prospectively biopsied tumors: KRas/BRaf mutation, treated with sorafenib; EGFR mutation/copy number, treated with erlotinib; VEGF/VEGFR2 expression, treated with vandetanib; and RXRs/Cyclin D1 expression and CCND1 copy number, treated with bexarotene and erlotinib. Based on the cumulative real-time results of the initial 97 patients over 8 weeks of treatment, 158 additional patients were adaptively randomized to receive the most effective therapy for their particular biomarker profile. In other words, if a patient from the first group with a KRas mutation had an adequate clinical response to sorafenib, there would be a greater Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 16 than 25% chance that another patient with a KRas mutation would be placed into that treatment arm. In total, 20% of the 255 patients randomized had KRas mutations. The overall 8-week disease control rate (DCR) was 46%; patients in the sorafenib arm had the highest DCR at 58%. Post-hoc biomarker analysis showed that relative to the other treatments, sorafenib had a significantly higher DCR in EGFR wildtype patients (64% vs. 33%, P<0.001), and higher but non-statistically significant DCR in KRas mutant patients (61% vs. 32%, P=0.11). Of note, in the KRas/BRaf group, sorafenib gave a higher DCR than erlotinib (79% vs. 14%, P=0.016), with the G12C/G12V group associated with decreased progression free survival (PFS) compared to other KRas mutants or KRas wildtype (1.84 months vs. 3.55 months vs. 2.83 months, P=0.026). These findings are similar to a post-hoc biomarker analysis of the MISSION Phase III trial, where EGFR mutation, but not KRas mutation, predicted increased overall survival with sorafenib as third or fourth line monotherapy (72). Together, this trial shows the feasibility of real-time biomarker analysis and adaptive randomization, and provides a rationalization for further targeted therapy clinical trials, especially combination regimens, for patients with KRas mutations. Of note, the BATTLE-2 trial, currently ongoing, is testing four treatment arms—erlotinib, erlotinib + MK2206 (an Akt inhibitor), selumetinib + MK2206, and sorafenib—with multiple biomarkers including KRas (73). Selumetinib/docetaxel combination therapy A recent phase 2 clinical trial tested a combination of MEK inhibition with cytotoxic chemotherapy (74). Jänne, et al. randomized 87 patients with pretreated KRas mutant NSCLC to Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 17 receive either docetaxel alone or in combination with selumetinib (74); selumetinib alone did not increase overall survival in NSCLC (75). Combination therapy increased overall survival, however, without statistical significance (9.4 months vs. 5.2 months, HR 0.80, 80% CI 0.56– 1.14; one-sided P=0.21). Median PFS was significantly increased (5.3 months vs. 2.1 months, HR 0·58, 80% CI 0.42–0.79; one-sided P=0.014). Of note, 37% (n=16) of patients given the combination therapy had an objective response, as measured by a decrease in tumor burden; however, 82% (n=36) of the combination group had adverse events of grade 3 or 4, mostly neutropenia, febrile neutropenia, and asthenia. Waterfall plots (graphs that depict the continuum of tumor growth, positive to negative for all patients in a study) of response to this therapy were widespread, with five patients in the combination group having a >20% increase in tumor size (74). It would be interesting to know if these patients also had mutations in the Lkb1 tumor suppressor, which has been shown to potentiate resistance to selumetinib/docetaxel combination therapy in a KRas-mutant GEMM (76). It will also be important to understand how MEK signaling cooperates with the microtubule depolymerizing activity of docetaxel and if KRas is necessary for this functional interaction. The results from these two clinical trials represent an important step forward in targeting KRas. They show that patients with KRas mutations have a small response to targeted therapies. However, KRas mutation status is likely not the only marker involved since therapeutic responses in KRas-mutant patients were not sufficiently robust, and since KRas wildtype patients also had a response. It also remains to be seen the effects of other gene mutations, such as those induced by smoking, on KRas, and how this may influence therapy. Other ongoing clinical trials aimed at targeting KRas-mutant NSCLC include: a phase 2 trial of bortezomib (77), a phase 1b/2 trial of retaspimycin (an Hsp90 inhibitor) in combination Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 18 with everolimus (78), and a phase 2 trial of selumetinib + erlotinib vs. selumetinib alone (79) (Table 2). New directions The difficulties of targeting KRas-mutated NSCLC remain daunting, but a renaissance of discoveries—novel modes of inhibition, Ras regulatory proteins, and Ras-dependent targets—are defining a new battery of drugs. In turn, these drugs are informing a new wave of clinical trials. While this process may seem slow and unforgiving, we should be reminded that paradigm changes take time—the Renaissance lasted almost three centuries—and that continued studies of the Ras pathway will likely reveal novel therapies for this subset of patients, providing personalized medicine against this most common oncogene. Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 19 References 1. Barbacid M. ras genes. Annu Rev Biochem 1987;56:779–827. 2. Mitin N, Rossman KL, Der CJ. Signaling interplay in Ras superfamily function. Curr Biol CB 2005;15:R563–574. 3. Bourne HR, Sanders DA, McCormick F. The GTPase superfamily: a conserved switch for diverse cell functions. Nature 1990;348:125–32. 4. Bos JL, Rehmann H, Wittinghofer A. GEFs and GAPs: critical elements in the control of small G proteins. Cell 2007;129:865–77. 5. 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 Res 2011;39:D945–D950. 6. Scheidig AJ, Burmester C, Goody RS. The pre-hydrolysis state of p21(ras) in complex with GTP: new insights into the role of water molecules in the GTP hydrolysis reaction of raslike proteins. Structure 1999;7:1311–24. 7. Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D. RAS oncogenes: weaving a tumorigenic web. Nat Rev Cancer 2011;11:761–74. 8. Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer 2003;3:11–22. 9. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646– 74. 10. Karnoub AE, Weinberg RA. Ras oncogenes: split personalities. Nat Rev Mol Cell Biol 2008;9:517–31. 11. Riely GJ, Marks J, Pao W. KRAS Mutations in Non–Small Cell Lung Cancer. Proc Am Thorac Soc 2009;6:201–5. 12. Seeburg PH, Colby WW, Capon DJ, Goeddel DV, Levinson AD. Biological properties of human c-Ha-ras1 genes mutated at codon 12. Nature 1984;312:71–5. 13. Der CJ, Finkel T, Cooper GM. Biological and biochemical properties of human rasH genes mutated at codon 61. Cell 1986;44:167–76. 14. Leone-Kabler S, Wessner LL, McEntee MF, D’Agostino RB Jr, Miller MS. Ki-ras mutations are an early event and correlate with tumor stage in transplacentally-induced murine lung tumors. Carcinogenesis 1997;18:1163–8. 15. Garassino MC, Marabese M, Rusconi P, Rulli E, Martelli O, Farina G, et al. Different types of K-Ras mutations could affect drug sensitivity and tumour behaviour in non-small-cell lung cancer. Ann Oncol 2011;22:235–7. Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 20 16. Kim ES, Herbst RS, Wistuba II, Lee JJ, Blumenschein GR Jr, Tsao A, et al. The BATTLE trial: personalizing therapy for lung cancer. Cancer Discov 2011;1:44–53. 17. 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. J Natl Cancer Inst. 2012;104:228–39. 18. Miller MS, Miller LD. RAS Mutations and Oncogenesis: Not all RAS Mutations are Created Equally. Front Genet 2011;2:100. 19. Reiss Y, Goldstein JL, Seabra MC, Casey PJ, Brown MS. Inhibition of purified p21ras farnesyl:protein transferase by Cys-AAX tetrapeptides. Cell 1990;62:81–8. 20. Ashby MN. CaaX converting enzymes. Curr Opin Lipidol 1998;9:99–102. 21. Mor A, Philips MR. Compartmentalized Ras/MAPK signaling. Annu Rev Immunol 2006;24:771–800. 22. Hancock JF, Paterson H, Marshall CJ. A polybasic domain or palmitoylation is required in addition to the CAAX motif to localize p21ras to the plasma membrane. Cell 1990;63:133– 9. 23. Rowinsky EK. Lately, it occurs to me what a long, strange trip it’s been for the farnesyltransferase inhibitors. J Clin Oncol 2006;24:2981–4. 24. Basso AD, Kirschmeier P, Bishop WR. Lipid posttranslational modifications. Farnesyl transferase inhibitors. J Lipid Res 2006;47:15–31. 25. Kohl NE, Omer CA, Conner MW, Anthony NJ, Davide JP, Desolms SJ, et al. Inhibition of farnesyltransferase induces regression of mammary and salivary carcinomas in ras transgenic mice. Nat Med 1995;1:792–7. 26. Van Cutsem E, van de Velde H, Karasek P, Oettle H, Vervenne WL, Szawlowski A, et al. Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol 2004;22:1430–8. 27. Lobell RB, Liu D, Buser CA, Davide JP, DePuy E, Hamilton K, et al. Preclinical and clinical pharmacodynamic assessment of L-778,123, a dual inhibitor of farnesyl:protein transferase and geranylgeranyl:protein transferase type-I. Mol Cancer Ther 2002;1:747–58. 28. Gotlib J. Farnesyltransferase inhibitor therapy in acute myelogenous leukemia. Curr Hematol Rep 2005;4:77–84. 29. Johnston SRD, Hickish T, Ellis P, Houston S, Kelland L, Dowsett M, et al. Phase II study of the efficacy and tolerability of two dosing regimens of the farnesyl transferase inhibitor, R115777, in advanced breast cancer. J Clin Oncol 2003;21:2492–9. Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 21 30. Blum R, Cox AD, Kloog Y. Inhibitors of chronically active ras: potential for treatment of human malignancies. Recent Patents Anticancer Drug Discov 2008;3:31–47. 31. Weisz B, Giehl K, Gana-Weisz M, Egozi Y, Ben-Baruch G, Marciano D, et al. A new functional Ras antagonist inhibits human pancreatic tumor growth in nude mice. Oncogene 1999;18:2579–88. 32. 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. J Thorac Oncol 2011;6:1435–7. 33. Whyte DB, Kirschmeier P, Hockenberry TN, Nunez-Oliva I, James L, Catino JJ, et al. Kand N-Ras are geranylgeranylated in cells treated with farnesyl protein transferase inhibitors. J Biol Chem 1997;272:14459–64. 34. Tsimberidou AM, Chandhasin C, Kurzrock R. Farnesyltransferase inhibitors: where are we now? Expert Opin Investig Drugs 2010;19:1569–80. 35. Cepero V, Sierra JR, Corso S, Ghiso E, Casorzo L, Perera T, et al. MET and KRAS gene amplification mediates acquired resistance to MET tyrosine kinase inhibitors. Cancer Res 2010;70:7580–90. 36. Sebti SM, Der CJ. Opinion: Searching for the elusive targets of farnesyltransferase inhibitors. Nat Rev Cancer 2003;3:945–51. 37. www.clinicaltrials.gov [Internet]. Available from: http://www.clinicaltrials.gov/ 38. Bergo MO, Ambroziak P, Gregory C, George A, Otto JC, Kim E, et al. Absence of the CAAX endoprotease Rce1: effects on cell growth and transformation. Mol Cell Biol 2002;22:171–81. 39. Wahlstrom AM, Cutts BA, Karlsson C, Andersson KME, Liu M, Sjogren A-KM, et al. Rce1 deficiency accelerates the development of K-RAS-induced myeloproliferative disease. Blood 2007;109:763–8. 40. Manandhar SP, Hildebrandt ER, Schmidt WK. Small-molecule inhibitors of the Rce1p CaaX protease. J Biomol Screen 2007;12:983–93. 41. Bergo MO, Gavino BJ, Hong C, Beigneux AP, McMahon M, Casey PJ, et al. Inactivation of Icmt inhibits transformation by oncogenic K-Ras and B-Raf. J Clin Invest 2004;113:539–50. 42. Wahlstrom AM, Cutts BA, Liu M, Lindskog A, Karlsson C, Sjogren A-KM, et al. Inactivating Icmt ameliorates K-RAS-induced myeloproliferative disease. Blood 2008;112:1357–65. Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 22 43. Winter-Vann AM, Baron RA, Wong W, dela Cruz J, York JD, Gooden DM, et al. A smallmolecule inhibitor of isoprenylcysteine carboxyl methyltransferase with antitumor activity in cancer cells. Proc Natl Acad Sci U S A 2005;102:4336–41. 44. Winter-Vann AM, Kamen BA, Bergo MO, Young SG, Melnyk S, James SJ, et al. Targeting Ras signaling through inhibition of carboxyl methylation: an unexpected property of methotrexate. Proc Natl Acad Sci U S A 2003;100:6529–34. 45. Chandra A, Grecco HE, Pisupati V, Perera D, Cassidy L, Skoulidis F, et al. The GDI-like solubilizing factor PDEδ sustains the spatial organization and signalling of Ras family proteins. Nat Cell Biol 2012;14:148–58. 46. Zimmermann G, Papke B, Ismail S, Vartak N, Chandra A, Hoffmann M, et al. Small molecule inhibition of the KRAS-PDEδ interaction impairs oncogenic KRAS signalling. Nature 2013;497:638–42. 47. Gysin S, Salt M, Young A, McCormick F. Therapeutic Strategies for Targeting Ras Proteins. Genes Cancer 2011;2:359–72. 48. Palmioli A, Sacco E, Abraham S, Thomas CJ, Di Domizio A, De Gioia L, et al. First experimental identification of Ras-inhibitor binding interface using a water-soluble Ras ligand. Bioorg Med Chem Lett 2009;19:4217–22. 49. Rosnizeck IC, Graf T, Spoerner M, Tränkle J, Filchtinski D, Herrmann C, et al. Stabilizing a weak binding state for effectors in the human ras protein by cyclen complexes. Angew Chem Int Ed Engl 2010;49:3830–3. 50. Patgiri A, Yadav KK, Arora PS, Bar-Sagi D. An orthosteric inhibitor of the Ras-Sos interaction. Nat Chem Biol 2011;7:585–7. 51. 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. Proc Natl Acad Sci U S A 2012;109:5299–304. 52. 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. Angew Chem Int Ed Engl 2012;51:6140–3. 53. Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature 2013;503:548–51. 54. Brough R, Frankum JR, Costa-Cabral S, Lord CJ, Ashworth A. Searching for synthetic lethality in cancer. Curr Opin Genet Dev 2011;21:34–41. 55. Patel PH, Chadalavada RSV, Chaganti RSK, Motzer RJ. Targeting von Hippel-Lindau Pathway in Renal Cell Carcinoma. Clin Cancer Res 2006;12:7215–20. Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 23 56. Shaw AT, Winslow MM, Magendantz M, Ouyang C, Dowdle J, Subramanian A, et al. Selective killing of K-ras mutant cancer cells by small molecule inducers of oxidative stress. Proc Natl Acad Sci U S A 2011;108:8773–8. 57. Guo W, Wu S, Wang L, Wei X, Liu X, Wang J, et al. Antitumor Activity of a Novel Oncrasin Analogue Is Mediated by JNK Activation and STAT3 Inhibition. PLoS ONE [Internet]. 2011 [cited 2014 Jan 27];6. Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3236185/ 58. Mansfield AS, Fields AP, Jatoi A, Qi Y, Adjei AA, Erlichman C, et al. Phase I dose escalation study of the protein kinase C iota inhibitor aurothiomalate for advanced nonsmall cell lung cancer, ovarian cancer, and pancreatic cancer. J Clin Oncol [Internet]. 2013 [cited 2014 Jan 27];31. Available from: http://meetinglibrary.asco.org/content/110586-132 59. Luo J, Emanuele MJ, Li D, Creighton CJ, Schlabach MR, Westbrook TF, et al. A genomewide RNAi screen identifies multiple synthetic lethal interactions with the Ras oncogene. Cell 2009;137:835–48. 60. Barbie DA, Tamayo P, Boehm JS, Kim SY, Moody SE, Dunn IF, et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature 2009;462:108–12. 61. Chien Y, Kim S, Bumeister R, Loo Y-M, Kwon SW, Johnson CL, et al. RalB GTPasemediated activation of the IkappaB family kinase TBK1 couples innate immune signaling to tumor cell survival. Cell 2006;127:157–70. 62. Ou Y-H, Torres M, Ram R, Formstecher E, Roland C, Cheng T, et al. TBK1 directly engages Akt/PKB survival signaling to support oncogenic transformation. Mol Cell 2011;41:458–70. 63. Sweet-Cordero A, Mukherjee S, Subramanian A, You H, Roix JJ, Ladd-Acosta C, et al. An oncogenic KRAS2 expression signature identified by cross-species gene-expression analysis. Nat Genet 2005;37:48–55. 64. Vicent S, Chen R, Sayles LC, Lin C, Walker RG, Gillespie AK, et al. Wilms tumor 1 (WT1) regulates KRAS-driven oncogenesis and senescence in mouse and human models. J Clin Invest 2010;120:3940–52. 65. Oji Y, Miyoshi S, Maeda H, Hayashi S, Tamaki H, Nakatsuka S-I, et al. Overexpression of the Wilms’ tumor gene WT1 in de novo lung cancers. Int J Cancer J Int Cancer 2002;100:297–303. 66. Puyol M, Martín 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. Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 24 67. Kumar MS, Hancock DC, Molina-Arcas M, Steckel M, East P, Diefenbacher M, et al. The GATA2 transcriptional network is requisite for RAS oncogene-driven non-small cell lung cancer. Cell 2012;149:642–55. 68. Steckel M, Molina-Arcas M, Weigelt B, Marani M, Warne PH, Kuznetsov H, et al. Determination of synthetic lethal interactions in KRAS oncogene-dependent cancer cells reveals novel therapeutic targeting strategies. Cell Res 2012;22:1227–45. 69. Corcoran RB, Cheng KA, Hata AN, Faber AC, Ebi H, Coffee EM, et al. Synthetic lethal interaction of combined BCL-XL and MEK inhibition promotes tumor regressions in KRAS mutant cancer models. Cancer Cell 2013;23:121–8. 70. Winton T, Livingston R, Johnson D, Rigas J, Johnston M, Butts C, et al. Vinorelbine plus cisplatin vs. observation in resected non-small-cell lung cancer. N Engl J Med 2005;352:2589–97. 71. Eberhard DA, Johnson BE, Amler LC, Goddard AD, Heldens SL, Herbst RS, 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;23:5900–9. 72. Mok TSK, Paz-Ares L, 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. 2012. 73. BATTLE-2 Program: A Biomarker-Integrated Targeted Therapy Study - ClinicalTrials.gov [Internet]. Available from: http://www.clinicaltrials.gov/ct2/show/NCT01248247 74. Jänne 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. Lancet Oncol 2013;14:38–47. 75. 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. J Thorac Oncol Off Publ Int Assoc Study Lung Cancer 2010;5:1630–6. 76. 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– 7. 77. Bortezomib in KRAS-Mutant Non-Small Cell Lung Cancer in Never Smokers or Those With KRAS G12D - ClinicalTrials.gov [Internet]. Available from: http://www.clinicaltrials.gov/ct2/show/NCT01833143 Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 25 78. Phase 1b/2 Study of Retaspimycin HCl (IPI-504) in Combination With Everolimus in KRAS Mutant Non-small Cell Lung Cancer - ClinicalTrials.gov [Internet]. Available from: http://www.clinicaltrials.gov/ct2/show/NCT01427946 79. Randomized Phase II Study of AZD6244 (MEK Inhibitor) With Erlotinib in KRAS Wild Type Advanced NSCLC and a Randomized Phase II Study of AZD6244 With Erlotinib in Mutant KRAS Advanced NSCLC - ClinicalTrials.gov [Internet]. Available from: http://www.clinicaltrials.gov/ct2/show/NCT01229150 Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 26 Table 1: List of genes that are synthetically lethal with KRas in KRas-mutated lung adenocarcinoma cells, their functions, and inhibitor or inhibitor combinations. Gene Tank-binding kinase 1 (TBK1) Wilms tumor 1 (WT1) Cyclin-dependent kinase 4 (CDK4) GATA-2 B-cell lymphoma-extra large (BCL-XL) Function Kinase Transcription factor, tumor suppressor, oncogene (?) Kinase Transcription factor Anti-apoptotic factor Inhibitors 6aminopyrazolopyrimidine derivatives Referenc e 60, 61, 62 None 64, 65 PD-0332991 Bortezomib + fasudil Selumetinib + navitoclax (ABT-263) 66 67, 68 69 Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 27 Table 2: List of past and present clinical trials targeting KRas-mutated non-small cell lung cancer. Drug combinations, targets, patient responses, and references are tabulated. Drug combination Target Sorafenib Selumetinib + docetaxel Raf, VEGFR MEK 1/2, microtubules Selumetinib + MK2206 MEK 1/2, Akt KRas-mutant patient response (vs. KRas-wildtype patients) Increased DCR (61% vs. 32%, P=0.11) Increased PFS (5.3 mo vs. 2.1 mo, 80% CI 0.42-0.79; one-sided P=0.014) Increased OS (9.4 mo vs. 5.2 mo, 80% CI 0.56-1.14; one-sided P=0.21) Ongoing Reference NCT0040996 8 NCT0089082 5 NCT0124824 7 NCT0183314 3 NCT0142794 6 Bortezomib Proteasome Ongoing Retaspimycin + Hsp90, everolimus mTOR Ongoing Selumetinib + erlotinib vs. MEK 1/2, NCT0122915 selumetinib alone EGFR Ongoing 0 DCR (disease control rate) = CR (complete response) + PR (partial response) + SD (stabilized disease) Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 28 Figure 1A: Normal Ras signaling. Figure 1B: Oncogenic Ras signaling. When Ras is mutated, it is constitutively bound to guanosine triphosphate (GTP) such that its GTPase activating protein (GAP) cannot bind. The activated Ras signals through a multitude of effectors and downstream signaling pathways, a subset of which is shown here. [GEF: Guanine nucleotide exchange factor] Figure 2A: Graph showing the percentage of cancers with Ras mutations in different organ types, arranged in descending frequency. KRas, NRas, and HRas-driven cancers are denoted by color. The frequencies of the predominant histology of that organ-specific KRas-mutated cancer are listed below. For example, 53% of all lung cancers have KRas mutations; of these KRasmutated lung cancers, 53% are adenocarcinomas. These frequencies are likely underestimates as many samples deposited onto COSMIC database are listed as a “nonspecific” histology. Data were accessed on May 15, 2013. Figure 2B: Pie charts showing frequencies of different Kras mutations in Kras-mutated lung, colorectal, pancreatic, and biliary tract cancers. In all cancers, G12D and G12V mutations are common; however each cancer displays a different “KRas profile.” In lung cancer, G12C is the most common mutation, followed by G12V and G12D; while in colorectal, pancreatic, and biliary tract cancers, G13D, G12R, and G12S are the third most common mutations, respectively, after G12D and G12V. Figure 3. Ras processing, trafficking, membrane association, and inhibition. Ras is first farnesylated by farnesyltransferase (FTase). In the endoplasmic reticulum, it is proteolyzed by Ras converting enzyme (RCE1) and methylated by isoprenylcysteine carboxylmethyltransferase (ICMT). HRas, NRas, and KRas4a traffic to the Golgi where they are palmitoylated. Then they traffic to and associate with the plasma membrane. After depalmitoylation, these isoforms are chaperoned by PDE6δ and reaccumulate at the Golgi for another round of palmitoylation. KRas4b is chaperoned by PDE6δ and associates with the plasma membrane through its farnesyl Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 29 group and electrostatically through a positively-charged patch. Membrane association of Ras isoforms is aided by farnesyl-binding membrane docking proteins. Relevant potential inhibitors are indicated in the figure. Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. CCR Reviews RAS-GDP RAS-GDP RAS*-GTP GTP RAS-GTP GTP GDP Pi GDP GAP GEF GAP GEF Pi Effector RAF PI3K RALGDS Normal signaling MEK AKT RAL ERK mTOR RLIP Oncogenic signaling © 2014 American Association for Cancer Research Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. B A Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Figure 1: Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Figure 2: Percentage of cancers with RAS mutations A 60 KRAS 50 NRAS 40 HRAS 30 20 10 0 Pancreas Colorectal Frequency of 78% predominant histology 91% Biliary tract Lung 72% 53% Adenocarcinoma Skin Hematologic Urinary tract 66% 42% Melanoma AML 39% Head and neck 69% Transitional Squamous cell carcinoma B G12V Lung Colorectal Pancreatic Biliary tract G12D G12C G12R G12S G12A G13D © 2014 American Association for Cancer Research CCR Reviews Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Figure 3: Tipifarnib ER SH S CAAX FTASE RAS S CAAX GGTASE S CAAX RAS RAS RCE1 CAAX ICMT Inhibitors (ref. 40) RAS Cysmethynil, methotrexate Palmitoyl transferases PDE6B KRAS4b C +++ Benzimidazole compounds (CO)OMe 6B E PD RAS n tio yla ito S RAS C (CO)OMe lm pa De C (CO)OMe S Golgi S Salirasib Plasma membrane RAS All RAS isoforms +++ Positively charged patch Palmitoyl moiety KRAS4b KRAS4b RAS Non-KRAS4b isoforms (KRAS4a, HRAS, NRAS) Farnesyl moiety Farnesyl-binding membrane docking protein © 2014 American Association Assoc for Cancer Research CCR Reviews Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 3, 2014; DOI: 10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. A RAS Renaissance: Emerging Targeted Therapies for KRAS-Mutated Non-Small Cell Lung Cancer Neil Vasan, Julie Boyer and Roy S. Herbst Clin Cancer Res Published OnlineFirst June 3, 2014. Updated version Author Manuscript E-mail alerts Reprints and Subscriptions Permissions Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-13-1762 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Sign up to receive free email-alerts related to this article or journal. To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at [email protected]. To request permission to re-use all or part of this article, contact the AACR Publications Department at [email protected]. Downloaded from clincancerres.aacrjournals.org on August 9, 2017. © 2014 American Association for Cancer Research.