Review

Protein kinase inhibitors to treat non-small-cell lung cancer Gabriele Minuti, Armida D’Incecco, Lorenza Landi & Federico Cappuzzo† 1.

Introduction

Istituto Toscano Tumori, Ospedale Civile, Oncologia Medica, Livorno, Italy

2.

EGFR protein kinase inhibitors

3.

Crizotinib in ALK-positive

Introduction: Activating mutations of the EGFR and rearrangement of anaplastic lymphoma kinase (ALK) best illustrate the therapeutic relevance of molecular characterization in NSCLC patients. Areas covered: For this review article, all published data on the most relevant Phase III trials with tyrosine kinase inhibitors (TKIs) for the treatment of NSCLC were collected and analyzed. Expert opinion: Eight Phase III trials clearly established EGFR TKIs as the best therapeutic option for front-line therapy in EGFR-mutated patients. In pretreated NSCLC, EGFR TKIs are considered more effective than standard monotherapy with cytotoxics in presence of classical EGFR mutations, whereas in the EGFR wild-type population, a similar efficacy to docetaxel or pemetrexed in term of survival has been demonstrated. In ALK-translocated NSCLC, a Phase III trial demonstrated the superiority of a multi-target TKI, including ALK, in terms of progression-free survival, response rate and toxicity profile when compared to standard second-line chemotherapy. New agents targeting EGFR or ALK are under evaluation particularly in individuals with acquired resistance to EGFR TKIs or crizotinib.

Expert Opin. Pharmacother. Downloaded from informahealthcare.com by Ondokuz Mayis Univ. on 05/16/14 For personal use only.

NSCLC 4.

Discussion

5.

Expert opinion

Keywords: afatinib, crizotinib, erlotinib, gefitinib, NSCLC Expert Opin. Pharmacother. (2014) 15(9):1203-1213

1.

Introduction

Lung cancer is the leading cause of cancer-related mortality worldwide in men and is secondary only to breast cancer in women [1]. NSCLC accounts for 75% of all lung cancers and includes different subtypes, underlying relevant biological differences. The most part of patients with NSCLC are diagnosed in advanced stage, with a 5-year survival rate of < 5% [2]. In the past years progresses conducted in the field of cancer biology have significantly impacted on NSCLC treatment, mainly in adenocarcinoma histology, and, today, the old ‘one size fit all’ chemotherapeutic approach has been substitute by a novel approach, where treatment selection is based on biological characteristics (Figure 1). Alterations in gene sequence or expression can occur in the cell-signaling and regulatory pathways involved in cell cycle control, apoptosis, proteasome regulation and angiogenesis. As a consequence of genetic alterations, tumor can become highly dependent for proliferation and survival on the function of a single oncogene, defined as ‘driver oncogene’ (Figure 2) [3]. The demonstration of cells with genetic abnormalities that become addicted to oncogenes [4] is a rational basis for the development of targeted therapies. Specifically, the treatment of NSCLC has dramatically changed since the discovery of the biological and therapeutic importance of acquired genetic alterations in two genes that encode pharmacologically targetable tyrosine kinases (TK), EGFR [5-7] and Anaplastic lymphoma kinase (ALK) [8]. Drugs targeting the TK domain of EGFR, such as gefitinib, erlotinib or afatinib, demonstrated superiority over standard chemotherapy in patients harboring activating EGFR mutations (Table 1) [9-16] and crizotinib is now approved by the FDA for patients with ALK translocation [17]. 10.1517/14656566.2014.909412 © 2014 Informa UK, Ltd. ISSN 1465-6566, e-ISSN 1744-7666 All rights reserved: reproduction in whole or in part not permitted

1203

G. Minuti et al.

Article highlights. .

.

Expert Opin. Pharmacother. Downloaded from informahealthcare.com by Ondokuz Mayis Univ. on 05/16/14 For personal use only.

.

.

.

In the past years, progresses conducted in the field of cancer biology have significantly impacted on NSCLC treatment, mainly in adenocarcinoma histology, with the opportunity to identify molecularly defined subgroup of patients. Tailored approach with targeted agents, such as gefitinib, erlotinib or afatinib, demonstrated superiority versus standard chemotherapy in patients harboring activating EGFR mutations, and crizotinib is now approved for patients with anaplastic lymphoma kinase (ALK) translocation. An important option for patients not progressing after standard first-line chemotherapy is represented by maintenance therapy. Several studies investigated the role of erlotinib or gefitinib in this setting. Unfortunately, after an initial response, a consistent proportion of EGFR-mutant and ALK-positive patients inevitably progress. Several approaches are under clinical evaluation to overcome acquired resistance, including the use of novel and more selective tyrosine kinase inhibitors or combined strategies with inhibitors of multiple pathways. The possibility to obtain additional tumor tissue at progression or to explore biomarkers in plasma will represent the backbone of clinical trials evaluating novel targeted agents, aiming to offer newer tailored treatments for the cure of NSCLC patients.

This box summarizes key points contained in the article.

Aim of the present paper is to review the available data from Phase III trials with protein kinase inhibitors in advanced NSCLC. 2.

EGFR protein kinase inhibitors

The first successful example of targeted therapy based on driver mutations in lung cancer involved EGFR that encodes for one of the four receptor of the ErbB family, including EGFR (ErbB-1; HER1), HER2 (c-ErbB-2), HER3 (c-ErbB- 3) and HER4 (c-ErbB-4). EGFR gene mutations were first identified in 2004 and classical activating EGFR mutations are localized in exon 19, mainly consisting of an in-frame deletion (45 -50%), and in exon 21, with a L858R point mutation (40 -45%). Preclinical models demonstrated that EGFR mutations increase sensitivity to TK inhibitors (TKIs), most likely through induction of critical structural modifications of the ATPbinding site in the TK domain [6,7]. Additional mutations have been characterized, including mutations in exon 20, classically associated with EGFR TKIs resistance, and other uncommon mutations with an undefined predictive role [18]. Since their identification, it was clear that EGFR mutations, predominantly discovered in adenocarcinoma histology, in Asian race, in never-smokers and in female [5], outlined a distinct subgroup of NSCLC patients. In the past years, eight Phase III trials [9-16] had clearly established that EGFR-mutated 1204

patients benefit more from first-line treatment with EGFR TKIs than from standard chemotherapy with platinum doublet, at least in terms of response rate (RR), progression-free survival (PFS), toxicity profile and quality of life (QoL), with no differences in overall survival (OS), most likely because of crossover effect (Table 1). Gefitinib (ZD 1839, IressaÒ, AstraZeneca) and erlotinib (OSI 774, TarcevaÒ, Genentech) are orally bioavailable synthetic anilinoquinazoline compounds that selectively bind to the ATP-binding site of EGFR TK intracellular domain, blocking EGFR autophosphorylation. Afatinib (GilotrifÒ, Boehringer Ingelheim) is a highly selective anilinoquinazoline that irreversibly binds directly to the ATP site in the kinase domain of both EGFR and human epidermal growth factor receptor 2 (HER2). In European countries, gefitinib and afatinib are approved for patients with EGFR mutations only, whereas erlotinib is also approved in second or third line for EGFR wild-type patients. Several and large Phase III trials evaluated the efficacy of reversible EGFR TKIs both in unselected and selected NSCLC population as front-line therapy.

EGFR TKIs front line in unselected NSCLC The two large randomized Phase III trials (INTACT 1 and INTACT 2), evaluating the role of gefitinib in combination with standard chemotherapy in first-line setting, failed to demonstrate any benefit in terms of survival for patients receiving gefitinib, even when the analysis was restricted to patients harboring EGFR mutation [19,20]. As observed for gefitinib, the combination of erlotinib with platinum-based chemotherapy (carboplatin and paclitaxel in TRIBUTE, cisplatin and gemcitabine in TALENT) did not demonstrate any gain in term of OS over chemotherapy alone [21,22]. Gridelli et al. explored the possibility of giving erlotinib as front-line treatment in unselected NSCLC in a randomized Phase III trial. The TORCH study [23] was conducted in a cohort of 760 NSCLC patients to evaluate whether first-line erlotinib followed at progression by cisplatin plus gemcitabine was not inferior in term of OS to the reverse standard sequence. As expected, median PFS was significantly in favor of standard arm, and, most importantly for clinical practice, survival was statistically longer for patients treated with chemotherapy first. Therefore, the TORCH trial confirmed that upfront chemotherapy is the gold standard for unselected population and that EGFR TKIs can be offered as front-line only in patients harboring activating EGFR mutations. An important clinical question is whether erlotinib could be indicated as front-line treatment in EGFR-unselected NSCLC unfit for chemotherapy. In the TOPICAL study, erlotinib was compared to placebo in patients with a poor performance status or unsuitable for chemotherapy [24]. No difference in OS was observed, confirming that front-line erlotinib is not beneficial in EGFR undefined patients. 2.1

Expert Opin. Pharmacother. (2014) 15(9)

TKIs to treat NSCLC

Expert Opin. Pharmacother. Downloaded from informahealthcare.com by Ondokuz Mayis Univ. on 05/16/14 For personal use only.

Stratification for EGFR, ALK and histolory

EGFR Mut+

ALK+

EGFR TKI

Crizotinib

EGFR WT/ALKnon-squamous

EGFR WT/ALKsquamous

Platinum doublet + bevacizumab OR platinum + pemetrexed +/- bevacizumab

Platinum-based doublet

Figure 1. This figure illustrates an algorithm for first-line treatment in metastatic NSCLC based on biological information. ALK: Anaplastic lymphoma kinase; Mut: Mutated; TKI: Tyrosine kinase inhibitor; WT: Wild-type.

UNKNOWN ≈ 50% KRAS 26.9% EGFR 9.4% ALK 4% MET 4% RET 1.9% ROS 11.7% BRAF 1.6% HER 20.9% PIK3CA 2.6%

Figure 2. This figure illustrates targetable oncogenic drivers in lung adenocarcinoma. Data taken from [3,72,73]. ALK: Anaplastic lymphoma kinase; HER2: Human epidermal growth factor receptor 2; MET: Mesenchymal-epithelial transition; PIK3CA: Phosphatidylinositol 3 kinase.

EGFR TKIs front line in selected NSCLC The IPASS trial was the first Phase III trial conducted in a NSCLC population selected for clinical characteristics predictive for presence of EGFR mutations. In this study, untreated Asiatic patients with advanced lung adenocarcinoma, non- or former/light smokers were randomized to receive gefitinib or carboplatin-paclitaxel [9]. The primary objective was to assess the noninferiority of gefitinib versus carboplatin-paclitaxel in terms of PFS. The study exceeded the primary objective and demonstrated superiority of gefitinib relative to chemotherapy in terms of PFS in the intent-to-treat (ITT) population. Patients assigned to the gefitinib arm had a significant reduction in the risk of progression (hazard ratio [HR] = 0.78, p < 0.001), with no difference in OS probably because of the confounding effect of post-study treatments. Importantly, the PFS 2.2

benefit observed in this trial was confined to patients harboring EGFR mutations, with a clear detrimental effect of gefitinib in individuals with EGFR wild-type. In the First-SIGNAL trial [10], another Asian study conducted in a clinically selected population, gefitinib significantly improved PFS (HR = 0.81, p = 0.044) and offered better QoL when compared to chemotherapy, with a similar OS for the two arms of treatment. Similar to the IPASS study, the benefit in PFS favored the gefitinib arm only in patients with EGFR mutations. Therefore, both the IPASS and the First-SIGNAL trials demonstrated that EGFR TKI can be offered as front-line therapy only in EGFR-mutated patients and clinical characteristics are not sufficient for a proper patient selection. Two randomized Phase III studies have been performed in Japanese patients to compare the efficacy of gefitinib versus chemotherapy in the first-line setting. Both trials, the WJTOG3405 [11] and the NEJ002 [12], confirmed that in NSCLC patients with EGFR mutations, gefitinib produces higher RR and longer PFS than platinum-doublet chemotherapy. In 2011, Zhou et al. published the results of the OPTIMAL trial, a Phase III study comparing erlotinib with carboplatin and gemcitabine in Chinese patients harboring EGFR mutations [13]. A striking HR of 0.16 for PFS (13.1 vs 4.6 months, p < 0.0001) was reported for subjects receiving the experimental treatment with erlotinib. The possibility of translating these findings to Western populations was a major theme of discussion. Rosell et al. duplicated the results of erlotinib over standard first-line chemotherapy in European EGFR-mutated population in a large Phase III trial (EURTAC) [14]. In this study, patients receiving erlotinib had a 63% relative reduction in risk of progression compared to chemotherapy (9.7 vs 5.2 months; HR = 0.37, p < 0.0001), leading to the approval of the drug in firstline setting only in EGFR-mutated patients.

Expert Opin. Pharmacother. (2014) 15(9)

1205

G. Minuti et al.

Table 1. First-line Phase III trials comparing standard chemotherapy with platinum doublet versus EGFR TKIs in EGFR-mutated population. Study

EGFR-TKI versus Chemotherapy

Patients (n)

RR (%)

PFS (months)

HR p value

OS (months)

HR p value

IPASS* [9]

Gefitinib versus CBDCA + TXL Gefitinib versus CDDP + GEM Gefitinib versus CDDP + TXT Gefitinib versus CBDCA + TXL Erlotinib versus CBDCA + GEM Erlotinib versus Platinum-doublet Afatinib versus CDDP + PEM Afatinib versus CDDP + GEM

97 versus 111 159 versus 150 88 versus 89 114 versus 114 82 versus 72 77 versus 76 230 versus 115 242 versus 122

71.2 versus 47.3 84.6 versus 37.5 62.1 versus 32.2 73.7 versus 30.7 83.0 versus 36.0 54.5 versus 10.5 56.0 versus 23.0 66.9 versus 23.0

9.5 versus NR 8.0 versus 6.3 9.2 versus 6.3 10.4 versus 5.5 13.1 versus 4.6 NR

0.48

21.6 versus 21.9 27.2 versus 25.6 36.0 versus 39.0 27.7 versus 26.6 NR

1.00

Expert Opin. Pharmacother. Downloaded from informahealthcare.com by Ondokuz Mayis Univ. on 05/16/14 For personal use only.

FIRST SIGNAL* [10] WJTOG 3405 [11] NEJ 002 [12]

OPTIMAL [13]

EURTAC [14]

LUX LUNG 3 [15] LUX LUNG 6 [16]

11.1 versus 6.9 11.0 versus 5.6

< 0.0001 0.54 0.008 0.48 < 0.001 0.36 < 0.001 0.16 < 0.0001 0.34 < 0.0001 0.58 0.0004 0.28 < 0.0001

0.99 1.04 NR 1.18 NR 0.89 0.48 1.06 0.06

22.9 versus 20.8

NR

NR

NR

NR

0.95 0.75

*Shown data are restricted to EGFR-mutant population. CBDCA: Carboplatin; CDDP: Cisplatin; GEM: Gemcitabine; HR: Hazard ratio; NR: Not reported; OS: Overall survival; PEM: Pemetrexed; PFS: Progression-free survival; RR: Response rate; TKI: Tyrosine kinase inhibitor; TXL: Paclitaxel; TXT: Docetaxel.

The role of afatinib in first-line setting has been investigated in two Phase III studies. The LUX-LUNG 3 trial [15] compared afatinib with cisplatin plus pemetrexed in patients with advanced lung adenocarcinoma harboring EGFR mutations. The primary end point, PFS, assessed by independent review has been significantly prolonged (11.1 vs 6.9 months) in the afatinib arm compared to chemotherapy arm (HR = 0.58; p = 0.001) and particularly in patients with classical EGFR mutations (HR = 0.47; p = 0.001). Recently, Wu et al. published the results of the LUX-LUNG 6 trial [16], conducted in Asian patients harboring EGFR mutations and randomized to afatinib or cisplatin plus gemcitabine. The study showed that patients treated with afatinib had a significantly longer PFS than individuals receiving chemotherapy (median PFS = 11.0 vs 5.6 months, HR = 0.28, p < 0.0001), as well as higher RR (66.9 vs 23.0%, p < 0.0001). Therefore, in clinical practice, we have three EGFR TKIs available for EGFR-mutated patients, two reversible (gefitinib and erlotinib) and one irreversible (afatinib). At the present time, no study directly compared these three agents in patients with EGFR mutations. Nevertheless, indirect comparisons suggested that the three agents are equally effective with higher toxicity, particularly skin rash and diarrhea, reported in the afatinib studies. Recently, Pfizer announced that the 1206

ARCHER 1009 study [25], a Phase III, multicenter, double-blinded trial comparing dacomitinib (PF00299804, Pfizer, USA), an irreversible EGFR, HER2 and HER4 inhibitor, with erlotinib in pretreated NSCLC, did not met its primary end point, reinforcing the hypothesis that efficacy differences among currently available EGFR TKIs are minimal, if any. Resistance to EGFR TKIs and new generation EGFR TKIs

2.3

In the vast majority of patients with EGFR-activating mutations, an EGFR TKI treatment induces a dramatic tumor shrinkage with a median PFS ranging from 8 to 13 months. Unfortunately, after an initial response, all patients inevitably progress. Several mechanisms responsible for acquired resistance have been described, including the T790M missense mutation in exon 20 of EGFR [26], additional secondary EGFR mutations [27], MET amplification [28], HER2 amplification [26], small cell transformation [26], BRAF mutations [29] or combined events (Figure 3) [26]. One of the possible strategies to overcome resistance is to intensify EGFR inhibition through the use of compounds with additional activity against other ERBB family receptors. Second-generation EGFR TKIs, such as neratinib, dacomitinib and afatinib, showed promising activity in preclinical models [30-32]. Nevertheless,

Expert Opin. Pharmacother. (2014) 15(9)

TKIs to treat NSCLC

EGFR TKIs in maintenance or as second-line therapy

Expert Opin. Pharmacother. Downloaded from informahealthcare.com by Ondokuz Mayis Univ. on 05/16/14 For personal use only.

2.4

T790M 60% MET amplification ≈ 4% HER2 amplification 8% Small cell transformation 1% Concomitant mecanisms of resistance 10% UNKNOWN ≈ 18%

Figure 3. This figure illustrates the principal events responsible for acquired resistance in EGFR-mutant patient treated with EGFR TKIs. Data taken from [26,28]. HER2: Human epidermal growth factor receptor 2; MET: Mesenchymal-epithelial transition; TKIs: Tyrosine kinase inhibitors.

afatinib and neratinib demonstrated only a modest efficacy in clinical trials [33-35]. This lack of clinical activity in EGFR TKIs refractory patients is probably due to the high concentrations required to inhibit the T790M-mutated form of EGFR and the dose-related toxicity limitations derived from the concurrent inhibition of the wild-type form. Finally, as previously reported, an intensified strategy with irreversible EGFR TKIs, such as dacomitinib, failed to demonstrate a better outcome in pretreated NSCLC when compared to erlotinib [25]. Alternative strategies to overcome EGFR TKIs resistance, currently under clinical evaluation in Phase III trial, include the continuation of EGFR inhibition along with chemotherapy at the time of progression (the IMPRESS trial, NCT01544179) or the ‘re-challenge’ with EGFR TKIs after chemotherapy (the ICARUS trial, NCT01530334). Therefore, the ideal inhibitor should be more active and less toxic than older compounds. Covalent pyrimidine EGFR inhibitors seem to be good candidate to test in clinical, considering their ability to simultaneously inhibit EGFR mutant clone, including T790M, and spare EGFR wild-type. Preliminary results of a Phase I/II study with CO-1686, an oral covalent TKI, suggested a relevant activity in T790M-mutated NSCLC patients previously exposed to first-generation EGFR TKIs with a good tolerability, lacking the typical class adverse events [36]. Similarly, the ongoing Phase I trial with AZD9291, an oral and irreversible third-generation selective inhibitor of both EGFR-activating mutation and T790M, showed tumor shrinkage in a promising percentage of EGFR TKIs refractory patients, especially in those with T790M-positive NSCLC [37].

In clinical practice, not all EGFR-mutated NSCLC patients receive EGFR TKIs as front-line therapy, which is related to several factors, including the small amount of tumor tissue, facilities available or simply the time necessary for obtaining the results of the test. In a consistent proportion of NSCLC patients, the results of EGFR testing is available once frontline chemotherapy was already started or sometimes at the time of front-line therapy failure. In pretreated patients, Phase III trials [38] showed that erlotinib and gefitinib are active in patients harboring activating EGFR mutations, with a modest but statistically significant efficacy over placebo in EGFR wild-type individuals, demonstrated only for erlotinib [39,40]. In second-line setting, several trials and two meta-analyses [38,41] showed that both gefitinib and erlotinib produced a similar survival than standard second-line chemotherapy irrespective of any clinical or biological characteristics. Five Phase III studies (TITAN, HORGE, TAILOR, DELTA and PROSE trials) compared erlotinib with standard second-line chemotherapy [42-46]. In all trials, no difference in survival was detected between erlotinib and chemotherapy (Table 2), including studies specifically conducted in the EGFR wild-type population and without any confounding effect of the crossover, such as the TAILOR trial [44]. A meta-analysis of four randomized studies comparing gefitinib to docetaxel as second-line therapy demonstrated that both agents had similar efficacy in terms of OS with a more favorable toxicity profile for gefitinib [47]. More recently, a large meta-analysis showed no difference in survival between standard second-line chemotherapy and EGFR TKIs in EGFR wild-type population [38]. Overall, available data indicated that in pretreated NSCLC selection based on EGFR mutation status is less relevant than in front-line setting and that an EGFR TKI represents a reasonable option for second-line therapy even in the EGFR wild-type population. An important option for patients not progressing after standard first-line chemotherapy is represented by maintenance therapy and several studies investigated the role of erlotinib or gefitinib in this specific setting. The SATURN [48] trial was a Phase III study comparing erlotinib to placebo in patients with no evidence of progression following four cycles of first-line platinum-based chemotherapy. The co-primary end points were PFS in all patients and PFS in patients overexpressing EGFR by immunohistochemistry (IHC). PFS was significantly prolonged with erlotinib in all patients (HR = 0.71, p < 0.0001) and in EGFR IHCpositive (HR = 0.69, p < 0.0001). Importantly, PFS subgroup analysis showed a benefit produced by erlotinib in all patients, irrespective of any clinical or biological characteristic, including in EGFR wild-type patients (HR = 0.78, p = 0.0185). As expected, presence of EGFR mutations was associated with a marked improvement in PFS with erlotinib (HR = 0.10,

Expert Opin. Pharmacother. (2014) 15(9)

1207

G. Minuti et al.

Expert Opin. Pharmacother. Downloaded from informahealthcare.com by Ondokuz Mayis Univ. on 05/16/14 For personal use only.

Table 2. Second-line Phase III trials comparing standard chemotherapy with erlotinib. Study

Patients (n)

TITAN [42]

424

HORG [43]

297

TAILOR [44]

229

DELTA [45]

301

PROSE* [46]

285

Treatment arms

Erlotinib versus chemotherapy (docetaxel or pemetrexed) Erlotinib versus pemetrexed Erlotinib versus docetaxel Erlotinib versus docetaxel Erlotinib versus chemotherapy (docetaxel or pemetrexed)

PFS (months)

HR p value

OS (months)

HR p value

1.6 versus 2.2

1.19

5.3 versus 5.5

0.96

3.6 versus 2.9 2.4 versus 2.9 2.0 versus 3.2

NR

8.2 versus 10.1 5.4 versus 8.2 14.8 versus 12.2

NR

0.08

0.13 0.72 0.01 1.22 0.09 1.28

NR

0.73

0.98 0.78 0.10 0.91 0.52 1.05

NR 0.12

0.75

*Shown data are restricted to EGFR wild-type NSCLC population. HR: Hazard ratio; NR: Not reported; OS: Overall survival; PFS: Progression-free survival.

p < 0.0001). The PFS improvement produced by erlotinib translated in a significant survival benefit in the whole population (HR = 0.81, p = 0.0088), in EGFR wild-type (HR = 0.77, p = 0.0243), but not in EGFR-mutated patients, probably because of the confounding effect of crossover. Although the primary end point was PFS, OS was assessed as a secondary regulatory end point and contributed to the drug’s approval by the FDA and the European Medicines Agency. Considering the toxicity profile, maintenance therapy with erlotinib was generally well tolerated with a safety profile generally comparable to that observed in other studies with the same agent. Interestingly, patients benefiting more from maintenance erlotinib were individuals with stable disease as best response from first-line platinum-doublet chemotherapy, with no survival benefit in patients responding to chemotherapy [48]. The ATLAS trial was a Phase III study comparing maintenance erlotinib plus bevacizumab versus bevacizumab alone in NSCLC patients not progressing after four cycles of platinum-based chemotherapy plus bevacizumab [49]. Although the study met the primary end point of PFS, the difference in survival was not statistically significant, reducing the potential impact of this strategy on clinical practice. Two additional studies investigated the role of gefitinib as maintenance therapy. The INFORM trial [50] was a Phase III study comparing gefitinib with placebo in NSCLC patients not progressing after four cycles of platinum-based chemotherapy. Although PFS significantly favored the gefitinib arm, the difference in survival was not statistically significant probably because of the confounding effect of the crossover in a study population entirely represented by Asiatic people. The Phase III WJTOG0203 trial [51] compared platinumdoublet chemotherapy up to six cycles to platinum-based chemotherapy for three cycles followed by gefitinib 1208

maintenance, with the aim to demonstrate a survival improvement for the sequential strategy. The study failed to meet the primary end point of OS (HR = 0.86; p = 0.11), despite a possible survival improvement in patients with adenocarcinoma histology. 3.

Crizotinib in ALK-positive NSCLC

Initially reported in 2007 as a result of an inversion in chromosome 2p, which results in the fusion of the N-terminal portion of the echinoderm microtubule-associated proteinlike 4 (EML4) gene with the kinase domain of ALK, ALK translocation represents one of the newest and appealing biomarker in NSCLC [52]. Similar to other oncogenic drivers, this event has been described in adenocarcinoma especially but not exclusively in never-smoker population. Despite its low incidence, ranging 7% of cases, current guidelines consider ALK rearrangements as a standard biomarker to test at diagnosis in order to identify those patients suitable for anti-ALK strategies [53]. Crizotinib (PF 02341066, XalkoriÒ, Pfizer) a multi-target receptor TKI, was initially synthesized as a MET inhibitor [54,55]. In the Phase I trial, A8081001, an enriched cohort of 25 patients harboring a wide range of MET alterations received crizotinib but only in MET-amplified tumors -- such as NSCLC, gastroesophageal carcinoma and glioblastoma -an impressive tumor shrinkage was observed [56-59]. However, the occurrence of similar dramatic responses in those NSCLC cases that carried an ALK rearrangement moved the enthusiasm of researchers toward clinical development of the drug on this molecularly defined setting. Updated results of this Phase I trial confirmed an impressive activity in terms of RR of > 60% with an interesting PFS exceeding 9 months [60].

Expert Opin. Pharmacother. (2014) 15(9)

Expert Opin. Pharmacother. Downloaded from informahealthcare.com by Ondokuz Mayis Univ. on 05/16/14 For personal use only.

TKIs to treat NSCLC

Similar findings were observed in a second single-arm Phase II study, in which 261 pretreated NSCLC ALK-positive patients receiving crizotinib had a probability of response of 60%; notably, responses were long-lasting and PFS ranged 8 months [61]. Based on these results, in August 2011, crizotinib received the FDA approval for the treatment of ALK-positive NSCLC. The superiority of crizotinib versus standard chemotherapy as second-line treatment has been demonstrated in the recently completed PROFILE 1007 Phase III trial [17]. The study, enrolling 347 patients with advanced ALK-rearranged NSCLC who failed one prior platinum-based regimen to receive crizotinib or chemotherapy with either pemetrexed or docetaxel, met its primary end point of PFS. Patients treated with crizotinib had a 51% relative reduction in risk of progression compared with those receiving standard chemotherapy (7.7 vs 3.0 months, HR = 0.49, p < 0.001). Treatment with crizotinib was also associated with higher RR (65 vs 20%, ITT population, p < 0.001) and better toxicity profile. Notably, the subset analyses confirmed a significant PFS benefit in favor of crizotinib arm independently of age (> 65 vs < 65 years), gender, performance status (EGOG PS 0 -- 1 vs 2), histology (adenocarcinoma vs non-adenocarcinoma) and presence of brain metastases. Consistent with what was observed in all Phase III trials with front-line EGFR TKIs [9-16], the improvement in PFS did not translate in a significant advantage in OS in favor of crizotinib therapy. Also in this case, the vast majority of patients assigned to the chemotherapy arm received crizotinib at progression, with an inevitable confounding effect on survival. Despite this, the unusual silhouette of survival curves seems to suggest an inversion in the natural course of ALK-positive disease. Finally, a large randomized Phase III trial (NCT01154140, www.clinicaltrials.gov) comparing crizotinib with the association of platinum and pemetrexed as front line in ALK-positive NSCLC just completed accrual and its results will be available in 2014. Although ALK-positive NSCLCs dramatically respond to crizotinib, no patient can be cured, and after a median of 10 months, the vast majority of patients inevitably relapses and dies because of disease progression. Several mechanisms have been identified as responsible for acquired resistance, including ALK point mutations in the gatekeeper site, ALK amplification or the emergence of a second driver, such as EGFR or KRAS mutations [62]. Moreover, there are a growing number of patients for whom disease progression occurs only in CNS, supporting the hypothesis of an inadequate CNS crizotinib penetration [63]. Different compounds are currently under investigation in crizotinib-refractory patients. LDK378 (Novartis Pharmaceuticals) is an oral potent second-generation ALK inhibitor that demonstrated efficacy in crizotinib-naı¨ve and crizotinib-pretreated patients [64]. Data from a dose-escalating Phase I study (NCT01283516) showed that LDK378 is highly active and well tolerated, with RR of 70 and 73% in the whole population and in crizotinib-resistant patients, respectively, including individuals harboring an ALK

secondary mutation [64]. Due to these impressive results and its favorable toxicity profile, LDK378 received the FDA breakthrough therapy designation for ALK-positive NSCLC in March 2013, in order to accelerate the development of this new compound. Two confirmatory Phase II studies in crizotinib-resistant patients (NCT01685060) and in crizotinib-naı¨ve patients (NCT01685138) with metastatic NSCLC are currently ongoing. In addition, two large Phase III trials of LDK378 versus chemotherapy are now enrolling patients. The first trial (NCT01828112) compares LDK378 with pemetrexed or docetaxel in ALK-positive NSCLC previously exposed to platinum-doublet chemotherapy and crizotinib, whereas the second trial (NCT01828099) compares LDK378 versus standard cisplatin- or carboplatin-pemetrexed as front-line treatment in ALK-positive NSCLC. Further, the first clinical results from the Phase I/II study with CH5424802, a novel and selective ALK inhibitor, have been recently published. This new target agent produced an exciting RR of 94%. More interestingly, patients with brain metastases also experienced durable disease control rate (DCR > 6 months) [65]. Finally, AP26113, a dual ALK/EGFR inhibitor, and the heat shock protein 90 inhibitor ganetespib [66,67] showed interesting activity and a favorable toxicity in early phases of development. Crizotinib in ROS1-positive NSCLC In addition to ALK-rearranged lung cancers, an encouraging antitumor effect emerged for a small fraction of NSCLC patients with ROS1 translocations treated with crizotinib. From a molecular point of view, ALK and ROS1 share a high degree of homology in their TK domains, thus suggesting that ALK TKIs, such as crizotinib, may also inhibit ROS1 and preclinical models confirmed this hypothesis [68,69]. Studies investigating incidence and clinical features of ROS1translocated patients showed that such molecular event is present in approximately 1% of NSCLC, mainly in never- or light smoker, with a prevalent adenocarcinoma histology [70]. Recently, Ou et al. reported the preliminary results of a Phase I study enrolling 20 ROS1-positive NSCLC patients exposed to crizotinib. RR was 50%, including one complete response and nine partial responses with a DCR of 70% after only 2 months of therapy [71]. 3.1

4.

Discussion

In the past few years, new insights in molecular biology rapidly changed the therapeutic landscape of NSCLC with the opportunity to develop specific therapies for a subset of molecularly defined patients. Currently, we have the possibility to identify a ‘driver genetic alteration’ in up to 60% of lung adenocarcinoma (Figure 2) [72,73]. Although many of these gene alterations are relatively rare, their identification with specific and approved tests is crucial and strongly recommended, considering that a tailored approach with target

Expert Opin. Pharmacother. (2014) 15(9)

1209

G. Minuti et al.

agents can deeply impact on patient outcome and change the natural history of the disease, as demonstrated with TKIs for EGFR-mutated patients and, more recently, for ALK-positive patients.

Expert Opin. Pharmacother. Downloaded from informahealthcare.com by Ondokuz Mayis Univ. on 05/16/14 For personal use only.

5.

Expert opinion

Cancer treatment is currently based on individualized therapy, and biomarker analysis represents the most relevant factors for treatment decision. The era of targeted therapy for NSCLC became a reality since the discovery of EGFR mutations. Available data from randomized Phase III trials clearly demonstrated that EGFR TKIs, such as erlotinib, gefitinib or afatinib, are the best option that we can offer today as front-line therapy only in EGFR-mutant patients; therefore, it should be mandatory to obtain EGFR assessment before starting systemic treatment. Other genomic alterations, such as gene amplification and rearrangements, can be detected and characterized as ‘oncogenic driver’ in NSCLC, underlying a potential sensibility to specific compounds. ALK translocation represents one of the newest and most attractive genomic alterations in NSCLC, and despite its quite low incidence, current guidelines consider ALK rearrangements as a standard biomarker to test at diagnosis, considering the deep impact of crizotinib therapy in ALK-positive population. A dramatic tumor shrinkage and a significant gain in outcome is obtained with TKI treatment in these molecularly selected NSCLC patients, but, unfortunately, none with a metastatic disease is definitively cured and a consistent proportion of EGFR-mutant and ALK-positive patients are no longer sensitive to those compounds. Virtually all individuals, after an initial response, finally relapse and die from their disease. Understanding molecular events responsible for development of TKIs acquired or inner resistance is crucial Bibliography

for improving outcome, and several approaches are under clinical evaluation, including the use of novel and more selective TKIs or combined strategies with inhibitors of multiple pathways. Moreover, testing for different biomarkers represents a cost and often is not feasible to all patients, mainly for the limited amount of available tumor tissue. Today, in the era of targeted therapies, all the efforts should be made in all patients with the aim of characterizing the biological portrait of their tumor, offering the best therapy for each patient. That is the reason why many investigators are exploring the possibility of assessing tumor biomarkers in plasma, and new techniques today allow characterizing circulating DNA and circulating tumor cells. These approaches are currently under investigation and are not available in all centers. Finally, the possibility to obtain additional tumor tissue at progression or to explore biomarkers in plasma should be always considered in order to decipher the complex phenomenon of resistance. In the next future, this information will represent the backbone of clinical trials with novel and targeted agents, aiming to offer newer and tailored treatment options for the cure of our patients.

Declaration of interest This manuscript is supported by the Italian Association for Cancer Research IG 2012 -- 13157, Fondazione Ricerca Traslazionale and Istituto Tumori Toscano Project F13/16. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Papers of special note have been highlighted as either of interest () or of considerable interest () to readers.

4.

Weinstein IB. Cancer. Addiction to oncogenes - the achilles heal of cancer. Science 2002;297:63-4

1.

5.

Pao W, Miller V, Zakowski M, et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci USA 2004;10:13306-11

2.

3.

1210

Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin 2013;63:11-30 Goldstraw P, Crowley J, Chansky K, et al. The IASLC Lung Cancer Staging Project: proposals for the revision of the TNM stage groupings in the forthcoming (seventh) edition of the TNM Classification of malignant tumours. J Thorac Oncol 2007;2:706-14 Levy MA, Lovly CM, Pao W. Translating genomic information into clinical medicine: lung cancer as a paradigm. Genome Res 2012;22:2101-18

6.

Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 2004;350:2129-39

7.

Paez JG, Ja¨nne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004;304:1497-500

Expert Opin. Pharmacother. (2014) 15(9)

8.

..

9.

..

10.

Soda M, Choi YL, Enomoto M, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007;448(7153):561-6 The first evidence in literature of ALK-translocation in NSCLC. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 2009;361:947-57 This is the first Phase III study demonstrating that an EGFR tyrosine kinase inhibitor (TKI) is superior to platinum-based chemotherapy in molecularly selected NSCLC patients. Han JY, Park K, Kim SW, et al. First-signal: first-line single-agent iressa versus gemcitabine and cisplatin trial in

TKIs to treat NSCLC

Expert Opin. Pharmacother. Downloaded from informahealthcare.com by Ondokuz Mayis Univ. on 05/16/14 For personal use only.

11.

Mitsudomi T, Morita S, Yatabe Y, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol 2010;11:121-8

12.

Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib or chemotherapy for non--small-cell lung cancer with mutated EGFR. N Engl J Med 2010;362:2380-8

13.

Zhou C, Wu YL, Chen G, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small- cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 2011;12:735-42

14.

15.

16.

17.

..

18.

Rosell R, Carcereny E, Gervais R, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 2012;13:239-46 Sequist LV, Yang JC, Yamamoto N, et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol 2012;31:3327-34 Wu YL, Zhou C, Hu CP, et al. Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-Lung 6): an open-label, randomised phase 3 trial. Lancet Oncol 2014;15(2):213-22 Shaw AT, Kim DW, Nakagawa K, et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med 2013;368:2385-94 The first trial that clearly demonstrated crizotinib superiority over standard second-line chemotherapy in anaplastic lymphoma kinase-positive NSCLC patients. Tokumo M, Toyooka S, Kiura K, et al. The relationship between epidermal growth factor receptor mutations and clinicopathologic features in non-small

T790M mutations in epidermal growth factor receptor-mutant lung adenocarcinomas with acquired resistance to kinase inhibitors. Clin Cancer Res 2006;12:6494-501

cell lung cancers. Clin Cancer Res 2005;11:1167-73

never-smokers with adenocarcinoma of the lung. J Clin Oncol 2012;30:1122-8 19.

20.

21.

22.

Giaccone G, Herbst RS, Manegold C, et al. Gefitinib in combination with gemcitabine and cisplatin in advanced non-small-cell lung cancer: a phase III trial - INTACT 1. J Clin Oncol 2004;22:777-84 Herbst RS, Giaccone G, Schiller JH, et al. Gefitinib in combination with paclitaxel and carboplatin in advanced non-small-cell lung cancer: a phase III trial - INTACT 2. J Clin Oncol 2004;22:785-94 Herbst RS, Prager D, Hermann R, et al. TRIBUTE: a phase III trial of erlotinib hydrochloride (OSI-774) combined with carboplatin and paclitaxel chemotherapy in advanced non-small-cell lung cancer. J Clin Oncol 2005;23:5892-9 Gatzemeier U, Pluzanska A, Szczesna A, et al. Phase III study of erlotinib in combination with cisplatin and gemcitabine in advanced non-small-cell lung cancer: the Tarceva Lung Cancer Investigation Trial. J Clin Oncol 2007;25:1545-52

23.

Gridelli C, Ciardiello F, Gallo C, et al. First-line erlotinib followed by secondline cisplatin-gemcitabine chemotherapy in advanced non-small-cell lung cancer: the TORCH randomized trial. J Clin Oncol 2012;30:3002-11

24.

Lee SM, Khan I, Upadhyay S, et al. First-line erlotinib in patients with advanced non-small-cell lung cancer unsuitable for chemotherapy (TOPICAL): a double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2012;13:1161-70

25.

Boyer M, Janne PA, Mok T, et al. ARCHER: dacomitinib (D; PF00299804) versus erlotinib (E) for advanced (adv) non-small cell lung cancer (NSCLC)----A randomized double-blind phase III study. J Clin Oncol 2012;30(Suppl):TPS7615a

26.

Yu HA, Arcila ME, Rekhtman N, et al. Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers. Clin Cancer Res 2013;19:2240-7 The largest series reporting mechanisms of acquired resistance to EGFR TKIs therapy.

.

27.

Balak MN, Gong Y, Riely GJ, et al. Novel D761Y and common secondary Expert Opin. Pharmacother. (2014) 15(9)

28.

Engelman JA, Zejnullahu K, Mitsudomi T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007;316:1039-43

29.

Ohashi K, Sequist LV, Arcila ME, et al. Lung cancers with acquired resistance to EGFR inhibitors occasionally harbor BRAF mutations but lack mutations in KRAS, NRAS, or MEK1. Proc Natl Acad Sci USA 2012;109:E2127-33

30.

Rabindran SK, Discafani CM, Rosfjord EC, et al. Antitumor activity of HKI-272, an orally active, irreversible inhibitor of the HER-2 tyrosine kinase. Cancer Res 2004;64:3958-65

31.

Engelman JA, Zejnullahu K, Gale CM, et al. PF00299804, an irreversible pan-ERBB inhibitor, is effective in lung cancer models with EGFR and ERBB2 mutations that are resistant to gefitinib. Cancer Res 2007;67:11924-32

32.

Li D, Ambrogio L, Shimamura T, et al. BIBW2992, an irreversible EGFR/ HER2 inhibitor highly effective in preclinical lung cancer models. Oncogene 2008;27:4702-11

33.

Sequist LV, Besse B, Lynch TJ, et al. Neratinib, an irreversible pan-ErbB receptor tyrosine kinase inhibitor: results of a phase II trial in patients with advanced non-small-cell lung cancer. J Clin Oncol 2010;28:3076-83

34.

Miller VA, Hirsh V, Cadranel J, et al. Afatinib versus placebo for patients with advanced, metastatic non-small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): a phase 2b/3 randomised trial. Lancet Oncol 2012;13:528-38

35.

Jackman D, Pao W, Riely GJ, et al. Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. J Clin Oncol 2010;28:357-60

36.

Sequist LV, Soria JC, Gadgeel SM, et al. First-in-human evaluation of CO-1686, an irreversible, selective, and potent tyrosine kinase inhibitor of EGFR T790M. J Clin Oncol 2013;31(Suppl):abstract 2524

1211

G. Minuti et al.

Expert Opin. Pharmacother. Downloaded from informahealthcare.com by Ondokuz Mayis Univ. on 05/16/14 For personal use only.

37.

Ranson M, Pao W, Kim DW, et al. Preliminary results from a Phase I study with AZD9291: an irreversible inhibitor of epidermal growth factor receptor (EGFR) activating and resistance mutations in non-small-cell lung cancer (NSCLC) [abstract 33]. ESMO -- ECCO congress 2013, session: Lung Cancer -- Metastatic; 2013

38.

Lee CK, Brown C, Gralla RJ, et al. Impact of EGFR inhibitor in non-small cell lung cancer on progression-free and overall survival: a meta-analysis. J Natl Cancer Inst 2013;105(9):595-605

39.

Shepherd FA, Rodrigues Pereira J, Ciuleanu T, et al. National Cancer Institute of Canada Clinical Trials Group. Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med 2005;353:123-32

40.

41.

42.

Thatcher N, Chang A, Parikh P, et al. Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebocontrolled, multicentre study (Iressa Survival Evaluation in Lung Cancer). Lancet 2005;366:1527-37 Qi WX, Shen Z, Lin F, et al. Comparison of the efficacy and safety of EFGR tyrosine kinase inhibitor monotherapy with standard second-line chemotherapy in previously treated advanced non-small-cell lung cancer: a systematic review and meta-analysis. Asian Pac J Cancer Prev 2012;13(10):5177-82 Ciuleanu T, Stelmakh L, Cicenas S, et al. Efficacy and safety of erlotinib versus chemotherapy in second-line treatment of patients with advanced, non-small-cell lung cancer with poor prognosis (TITAN): a randomized multicentre, open-label, phase 3 study. Lancet 2012;13(3):300-8

43.

Karampeazis A, Voutsina A, Souglakos J, et al. Pemetrexed versus erlotinib in pretreated patients with advanced non-small cell lung cancer: a Hellenic Oncology Research Group (HORG) randomized phase 3 study. Cancer 2013;119(15):2754-64

44.

Garassino MC, Martelli O, Broggini M, et al. Erlotinib versus docetaxel as second-line treatment of patients with advanced non-small-cell lung cancer and wild-type EGFR tumours (TAILOR):

1212

a randomised controlled trial. Lancet Oncol 2013;14(10):981-8 45.

46.

47.

48.

.

49.

50.

Okano Y, Ando M, Asami K, et al. Randomized phase III trial of erlotinib (E) versus docetaxel (D) as second- or third-line therapy in patients with advanced non-small cell lung cancer (NSCLC) who have wild-type or mutant epidermal growth factor receptor (EGFR): Docetaxel and Erlotinib Lung Cancer Trial (DELTA). 2013 ASCO Annual Meeting, Oral Abstract Session, Lung Cancer - Non-small Cell Metastatic. J Clin Oncol 2013;31(Suppl):abstract 8006 Lazzari C, Novello S, Barni S, et al. Randomized proteomic stratified phase III study of second-line erlotinib (E) versus chemotherapy (CT) in patients with inoperable non-small cell lung cancer (PROSE). 2013 ASCO Annual Meeting, Oral Abstract Session, Lung Cancer - Non-small Cell Metastatic. J Clin Oncol 2013;31(Suppl): abstract LBA8005 Shepherd FA, Douillard J, Fukuoka M, et al. Comparison of gefitinib and docetaxel in patients with pretreated advanced non-small cell lung cancer (NSCLC): meta-analysis from four clinical trials. J Clin Oncol 2009;27(Suppl):S15 Cappuzzo F, Ciuleanu T, Stelmakh L, et al. SATURN investigators. Erlotinib as maintenance treatment in advanced non-small-cell lung cancer: a multicentre, randomised, placebo-controlled phase 3 study. Lancet Oncol 2010;11:521-9 The first study that demonstrated a survival gain with maintenance erlotinib in NSCLC patients, irrespective of any clinical or biological characteristics. Johnson BE, Kabbinavar F, Fehrenbacher L, et al. ATLAS: randomized, double-blind, placebocontrolled, phase IIIB trial comparing bevacizumab therapy with or without erlotinib, after completion of chemotherapy, with bevacizumab for first-line treatment of advanced non--small-cell lung cancer. J Clin Oncol 2013;31:3926-34 Zhang L, Ma S, Song X, et al. Gefitinib versus placebo as maintenance therapy in patients with locally advanced or metastaticnon-small-cell lung cancer

Expert Opin. Pharmacother. (2014) 15(9)

(INFORM; C-TONG 0804): a multicentre, double-blind randomised phase 3 trial. Lancet Oncol 2012;13(5):466-75 51.

Takeda K, Hida T, Sato T, et al. Randomized phase III trial of platinum-doublet chemotherapy followed by gefitinib compared with continued platinum-doublet chemotherapy in Japanese patients with advanced non-small-cell lung cancer: results of a west Japan thoracic oncology group trial (WJTOG0203). J Clin Oncol 2010;28(5):753-60

52.

Koivunen JP, Mermel C, Zejnullahu K, et al. EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clin Cancer Res 2008;14:4275-83

53.

Camidge DR, Doebele RC. Targeting ALK-positive lung cancer --early success and future challanges. Nat Rev Clin Oncol 2012;9:268-77

54.

Cui JJ, Tran-Dube´ M, Shen H, et al. Structure based drug design of crizotinib (PF-02341066), a potent and selective dual inhibitor of mesenchymal-epithelial transition factor (c-MET) kinase and anaplastic lymphoma kinase (ALK). J Med Chem 2011;54:6342-63

55.

Tanizaki J, Okamoto I, Okamoto K, et al. MET tyrosine kinase inhibitor crizotinib (PF-02341066) shows differential antitumor effects in non-small cell lung cancer according to MET alterations. J Thorac Oncol 2011;6:1624-31

56.

Ou SH, Kwak EL, Siwak-Tapp C, et al. Activity of crizotinib (PF02341066), a dual mesenchymal-epithelial transition (MET) and anaplastic lymphoma kinase (ALK) inhibitor, in a non-small cell lung cancer patient with de novo MET amplification. J Thorac Oncol 2011;6:942-6

57.

Lennerz JK, Kwak EL, Ackerman A, et al. MET amplification identifies a small and aggressive subgroup of esophagogastric adenocarcinoma with evidence of responsiveness to crizotinib. J Clin Oncol 2011;29:4803-10

58.

Chi AS, Batchelor TT, Kwak EL, et al. Radiographic and clinical improvement after treatment of a MET-amplified recurrent glioblastoma with a mesenchymal-epithelial transition inhibitor. J Clin Oncol 2012;30:e30-3

TKIs to treat NSCLC

Expert Opin. Pharmacother. Downloaded from informahealthcare.com by Ondokuz Mayis Univ. on 05/16/14 For personal use only.

59.

Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med 2010;363:1693-703

60.

Camidge DR, Bang YJ, Kwak EL, et al. Activity and safety of crizotinib in patients with ALK-positive non-small-cell lung cancer: updated results from a phase 1 study. Lancet Oncol 2012;13:1011-19

61.

Kim DW, Ahn MJ, Yang PC, et al. Updated results of a global phase II study with crizotinib in advanced ALK-positive non-small cell lung cancer (NSCLC) [abstract 2252]. ESMO congress 2012, session: NSCLC, metastatic; 2012

62.

63.

64.

Doebele RC, Aisner DL, Le AT, et al. Analysis of resistance mechanisms to ALK kinase inhibitors in ALK+ NSCLC patients. ASCO meeting 2012. J Clin Oncol 2012;30(Suppl):Abstract 7504 Chu SG, Choe KS, Iyengar P, et al. Isolated central nervous system progression on Crizotinib: an Achilles heel of non-small cell lung cancer with EML4-ALK translocation? Cancer Biol Ther 2012;13(14):1376-83 Shaw AT, Mehra R, Kim DW, et al. Clinical activity of the ALK inhibitor LDK378 in advanced, ALK-positive NSCLC. ASCO meeting 2013.

J Clin Oncol 2013;31(Suppl):Abstract 8010 65.

66.

Seto T, Kiura K, Nishio M, et al. CH5424802 (RO5424802) for patients with ALK-rearranged advanced non-small-cell lung cancer (AF-001JP study): a single-arm, open-label, phase 1--2 study. Lancet Oncol 2013;14:590-8 Camidge DR, Bazhenova L, Salgia R, et al. First-in-human dose-finding study of the ALK/EGFR inhibitor AP26113 in patients with advanced malignancies: updated results. ASCO meeting 2013. J Clin Oncol 2013;31(Suppl):Abstract 8031

67.

Sang J, Acquaviva J, Friedland JC, et al. Targeted inhibition of the molecular chaperone Hsp90 overcomes ALK inhibitor resistance in non-small cell lung cancer. Cancer Discov 2013;3:430-43

68.

Chin LP, Soo RA, Soong R, et al. Targeting ROS1 with anaplastic lymphoma kinase inhibitors: a promising therapeutic strategy for a newly defined molecular subset of non-small-cell lung cancer. J Thorac Oncol 2012;7:1625-30

69.

McDermott U, Iafrate AJ, Gray NS, et al. Genomic alterations of anaplastic lymphoma kinase may sensitize tumors to anaplastic lymphoma kinase inhibitors. Cancer Res 2008;68:3389-95

Expert Opin. Pharmacother. (2014) 15(9)

70.

Bergethon K, Shaw AT, Ignatius Ou SH, et al. ROS1 rearrangements define a unique molecular class of lung cancers. J Clin Oncol 2012;30:863-70

71.

Ou S, Camidge R, Engelman J, et al. Clinical activity of crizotinib in patients with advanced non- small cell lung cancer (NSCLC) harboring ROS1 gene rearrangement. Ann Oncol 2012;23(Suppl):2091a

72.

Ding L, Getz G, Wheeler DA, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature 2008;455:1069-75

73.

Barlesi F, Blons H, Beau-Faller M, et al. Biomarkers (BM) France: results of routine EGFR, HER2, KRAS, BRAF, PI3KCA mutations detection and EML4-ALK gene fusion assessment on the first 10,000 non-small cell lung cancer (NSCLC) patients (pts). ASCO meeting 2013. J Clin Oncol 2013;31(Suppl):Abstract 8000

Affiliation Gabriele Minuti MD, Armida D’Incecco MD, Lorenza Landi MD & Federico Cappuzzo† MD † Author for correspondence Istituto Toscano Tumori, Ospedale Civile, Oncologia Medica, Viale Alfieri 36, 57100-Livorno, Italy Tel: +39 0586223189; Fax: +39 0586223457; E-mail: [email protected]

1213

Protein kinase inhibitors to treat non-small-cell lung cancer.

Activating mutations of the EGFR and rearrangement of anaplastic lymphoma kinase (ALK) best illustrate the therapeutic relevance of molecular characte...
450KB Sizes 0 Downloads 3 Views