577738

research-article2015

TAR0010.1177/1753465815577738Therapeutic Advances in Respiratory DiseaseA Pender and S Popat

Therapeutic Advances in Respiratory Disease

Review

The efficacy of crizotinib in patients with ALK-positive nonsmall cell lung cancer

Ther Adv Respir Dis 1­–8 DOI: 10.1177/ 1753465815577738 © The Author(s), 2015. Reprints and permissions: http://www.sagepub.co.uk/ journalsPermissions.nav

Alexandra Pender and Sanjay Popat

Abstract:  Molecular profiling of nonsmall cell lung cancer (NSCLC) contributes to better understanding the different molecular subtypes of this heterogeneous group of diseases. The discovery of oncogenic ALK rearrangements in NSCLC and the subsequent success in their therapeutic targeting with crizotinib reinforces the benefits of a precision approach to systemic anticancer therapy. In addition, the rapid development of crizotinib from first discovery thorough accelerated US Food and Drug Administration approval, and late stage confirmatory clinical trials, exemplifies the success of the drug development strategy of close collaboration between clinicians, industry and regulatory authorities. In this review we describe the identification of ALK rearranged NSCLC, clinical characteristics of such patients, and clinical outcomes when treated with crizotinib. Introduction The discovery of oncogenic driver mutations in nonsmall cell lung cancer (NSCLC) and recognition of different molecular subtypes of disease have revolutionized the treatment and prognosis of this disease. The identification of somatic epidermal growth factor receptor (EGFR) mutations sensitive to EGFR tyrosine kinase inhibitors (TKI) and the superiority of EGFR TKIs over chemotherapy in this patient population has changed the management of advanced NSCLC [Mok et al. 2009; Popat et al. 2014]. Identification of a further mutually exclusive molecular subtype of lung adenocarcinoma with ALK rearrangements that is remarkably sensitive to the TKI, crizotinib [Soda et  al. 2007], has again changed clinical practice and emphasizes the importance of somatic molecular analysis alongside histopathological diagnosis [Travis et al. 2013]. Discovery of ALK rearrangements in NSCLC Following the discovery of chromosomal translocations involving the ALK locus in anaplastic large cell lymphoma and inflammatory myofibroblastic tumours, analysis of the transcriptome of a lung adenocarcinoma specimen first described a fusion product of the ALK and EML4 loci [Soda et  al. 2007]. The product of the 2p chromosome translocation is transforming secondary to anaplastic lymphoma kinase (ALK) activity, which is redistributed to the cytoplasm from the cell membrane.

Subsequently, several fusion variants have been identified involving the ALK locus and EML4. ALK fusions with the TFG [Rikova et al. 2007] and KIF5B loci [Takeuchi et al. 2009] have also been identified (Figure 1). Functional ALK rearrangements are estimated to occur in approximately 4–8% of NSCLC and are in the vast majority mutually exclusive of other oncogenic driver mutations such as EGFR and KRAS [Gainor et  al. 2013; Kris et  al. 2014]. Screening of specimens from a cohort of 754 NSCLC patients identified EML4-ALK fusions in 4.24% (32 patients). The E13:A20 (variant 1, 19 cases) and the E6a/b:A20 (variant 3a/b, 10 cases) fusions were the most frequently identified. Other variants, such as E18:A20 and E20:A20, were less commonly identified. The case series did not detect any TGF-ALK or KIF5B-ALK fusions, suggesting the rarity of these gene fusions [Soda et  al. 2012]. Patients with ALK-positive tumours tend to be younger than average for lung cancer and have a history of light or no tobacco exposure [Kwak et  al. 2010].

Correspondence to: Sanjay Popat, BSc MB(Hons) FRCP PhD Department of Medicine, Royal Marsden Hospital, Fulham Road, London, SW3 6JJ. [email protected] Alexandra Pender, MA MBBChir MRCP Department of Medicine, Royal Marsden Hospital, London, UK and Division of Molecular Pathology, Institute of Cancer Research, London, UK

Therapeutic targeting of ALK rearrangements Crizotinib, a competitive inhibitor of both ALK and c-Met kinase, has shown remarkable efficacy in ALK-positive NSCLC since early phase development. A high-throughput kinase inhibitor

http://tar.sagepub.com 1

Downloaded from tar.sagepub.com at UNIV NEBRASKA LIBRARIES on April 11, 2015

Therapeutic Advances in Respiratory Disease  Intron 13 EML4

Intron 19 ALK

E13:A20

Variant 1

Intron 20 EML4

Intron 19 ALK

E20:A20

Variant 2

Exon 6 EML4

Intron 19 ALK

E6:A20

Variant 3a

Intron 6 EML4

Intron 19 ALK

E6:A20

Variant 3b

Exon 14 EML4

Exon 20 ALK

E14:A20

Variant 4

Exon 2 EML4

Exon 20 ALK

E2:A20

Variant 5a

Exon 2 EML4

Intron 19 ALK

E2:A20

Variant 5b

Exon 18 EML4

Exon 20 ALK

E18:A20

Exon 21 EML4

Exon 20 ALK

E21:A20

Intron 5 TFG

Intron 19 ALK

Intron 24 KIF5B

Intron 19 ALK

Figure 1.  Variants of EML4-ALK, TFG-ALK and KIF5BALK fusions identified in NSCLC. Fusions can occur at several locations in EML4 (white), but thus far, intron 19 or exon 20 are the identified fusion points in ALK (black) [Takeuchi et al. 2008, 2009; Choi et al. 2008; Soda et al. 2012]. Nomenclature and variant descriptions are noted in the right panel.

screen against 602 cancer cell lines identified that crizotinib reduced the viability of the NSCLC cell line NCI-H3122, in which an EML4-ALK variant 1 fusion mRNA is detectable, by suppression of Akt and Erk signalling [McDermott et  al. 2008]. In xenograft models of anaplastic large cell lymphoma, crizotinib administration caused dose-dependent tumour responses and tumour cell apoptosis [Christensen et al. 2007]. The dose escalation phase of an open-label phase I study of crizotinib in molecularly unselected advanced NSCLC (PROFILE 1001) initially demonstrated impressive radiological responses in two patients who were subsequently shown to harbour ALK rearrangements [Kwak et  al. 2010]. Subsequently, approximately 1500 samples were prospectively screened using fluorescence in situ hybridization (FISH) for ALK rearrangements to recruit an expanded cohort of 82 ALK-positive patients. Although no subtypes of NSCLC were excluded from molecular screening, the vast majority (96%) of ALK rearrangements were identified in adenocarcinomas; 48% of ALK-positive adenocarcinoma cases had solid histological pattern with mucin and 38% were mixedpattern type adenocarcinoma. All cases within this cohort were negative for EGFR mutation or MET amplification. These patients received a dose of 250 mg crizotinib twice daily (BID).

The overall radiological response rate to crizotinib was 57% [95% confidence interval (CI) 46–68] and 87% of patients had controlled disease (stable or responding to therapy) at 8 weeks, notable in a patient cohort in which 94% had previously treated NSCLC. Later updated data from this trial of 143 patients with ALK-positive advanced NSCLC concluded a 60.8% (95% CI 52.3–68.9) objective response rate, with a median duration of response of 49.1 weeks (95% CI 37.3–75.4); 84% of subjects had received at least one previous systemic treatment prior to crizotinib [Camidge et  al. 2012]. Initial findings from the PROFILE 1005 phase II single arm study of crizotinib in pretreated ALKpositive NSCLC were presented at the American Society of Clinical Oncology meeting in June 2011 [Crino et al. 2011] and were updated at the World Lung Conference in July 2011 [Kim et al. 2011]. In this study, 93% of patients treated with crizotinib had received at least two prior lines of treatment. Of the 76 patients with an evaluable response, 83% had target lesion shrinkage on crizotinib and 41 of 76 patients had a response according to Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Following the interim analysis of this phase II study and the PROFILE 1001 results, the US Food and Drug Administration (FDA) granted accelerated approval for crizotinib in August 2011 [Jefferson, 2011]. Evaluation in January 2012 of the first 261 patients enrolled in PROFILE 1005 showed a progression-free survival (PFS) of 8.5 months (95% CI 6.2–9.9) [Kim et al. 2012], way in excess of the 2.7 months previously observed for docetaxel monotherapy [Shepherd et al. 2000]; 77% of evaluable patients remained on treatment with a median duration of treatment of 25 weeks and median duration of response of 43 weeks (95% CI 36–50). This study also reported a clinically relevant improvement in quality of life on crizotinib (p < 0.05) [Blackhall et al. 2012]. Crizotinib phase III study The PROFILE 1007 randomized phase III trial was opened to objectively demonstrate crizotinib efficacy versus standard-of-care chemotherapy in the second-line advanced NSCLC setting. All participants had ALK-positive advanced NSCLC centrally verified by FISH and had

2 http://tar.sagepub.com

Downloaded from tar.sagepub.com at UNIV NEBRASKA LIBRARIES on April 11, 2015

A Pender and S Popat received standard-of-care platinum doublet chemotherapy. Participants were randomized 1:1 to crizotinib or either single agent docetaxel or pemetrexed chemotherapy, receiving docetaxel if tumour had squamous histology or in case of prior pemetrexed therapy [Shaw et  al. 2013] and, on progression, patients randomized to chemotherapy crossed over and became eligible for PROFILE 1005. At the time of data analysis, both patient cohorts had been on study for just over 12 months. The primary endpoint of PROFILE 1007 was of PFS, and this was 7.7 months for crizotinib and 3.3 months for chemotherapy [hazard ratio (HR) =0.49, 95% CI 0.37–0.64, p < 0.0001]. The high response rate seen in earlier trials was reproducible with 65% response rate (95% CI 58–72) on crizotinib compared with 9% (95% CI 2–16) with docetaxel and 29% (95% CI 21–39) with pemetrexed, demonstrating a differential improved efficacy with pemetrexed over docetaxel for ALK-positive NSCLC. At time of primary analysis, 49 deaths (28%) had occurred on crizotinib and 47 (27%) on chemotherapy. Only 40% of the number of deaths required for analysis of overall survival (OS) had occurred and there was no significant difference between the two arms (HR =1.02, 95% CI 0.58–1.54, p = 0.54) with median survival of 20.3 months (for crizotinib) versus 22.8 months for chemotherapy. Of note, 64% of chemotherapy patients went on to receive crizotinib on progression. Quality of life was a secondary endpoint and analysis confirmed improvements in quality of life compared with chemotherapy. The time to deterioration of cough, chest pain or dyspnea was significantly longer on crizotinib than chemotherapy (5.6 months versus 1.4 months; HR = 0.54, p < 0.0001). A significant improvement in global quality of life (p < 0.0001) was also observed on crizotinib, in contrast to no improvement on chemotherapy. This study validated the previous accelerated FDA approval of crizotinib based on PROFILE 1001 and 1005 in ALK-positive advanced NSCLC [Kazandjian et  al. 2014] and led to European Medicines Agency (EMA) approval in ALKpositive advanced NSCLC previously treated with one line of platinum-based therapy [Committee for Medicinal Products for Human Use].

Crizotinib in treatment-naïve advanced ALKpositive nonsquamous NSCLC PROFILE 1005 allowed recruitment of patients without previous chemotherapy and the activity of crizotinib reported led to accelerated FDA approval for crizotinib to include patients that had not failed platinum-doublet chemotherapy. However, the randomized evidence that crizotinib is superior to platinum doublet chemotherapy, came from the PROFILE 1014 trial [Mok et  al. 2014]. Here, patients with locally advanced, recurrent or metastatic ALK-positive nonsquamous NSCLC were randomized 1:1 to crizotinib 250 mg BID or up to six cycles of platinum-pemetrexed chemotherapy. Similar proportions of patients had stable treated brain metastases in both study arms (26% versus 28%). The median time from diagnosis in both arms was 1.2 months. Never smokers made up 62% of the crizotinib cohort and 65% of the chemotherapy cohort. The vast majority of patients enrolled had adenocarcinoma (92% crizotinib arm, 93% chemotherapy arm). The 343 patient study showed superior median PFS of 10.9 months on crizotinib versus 7.0 months on chemotherapy (HR 0.454, 95% CI 0.35–0.61, p< 0.0001) and a higher overall response rate of 74% (95% CI 67–81) versus 45% (95% CI 37–53, p< 0.0001). Median time to response was quicker with crizotinib (6.1 weeks, range 2.7-41.4) than with chemotherapy (12.1 weeks, range 5.1–36.7) and median duration of response was prolonged with crizotinib [49 weeks, 95% CI 35.1–60 versus 22.9 weeks (95% CI 18.0-25.1)] with chemotherapy). At the point of data analysis, 120 (71%) chemotherapy patients had crossed over to crizotinib following progressive disease on chemotherapy and 68% of all participants were still in follow up. No significant improvement in median OS survival with crizotinib has been shown as yet (HR 0.821, 95% CI 0.54–1.26, p= 0.1804), likely due to crossover. Toxicities with crizotinib were as in previous trials with this compound. Even though survival data from this study are yet to mature and will be biased by crizotinib crossover, given the large PFS and response rate advantage coupled with a favourable safety profile, crizotinib is likely to become a standard first choice therapy for patients with ALK-positive advanced NSCLC.

http://tar.sagepub.com 3

Downloaded from tar.sagepub.com at UNIV NEBRASKA LIBRARIES on April 11, 2015

Therapeutic Advances in Respiratory Disease  Table 1.  Summary of key clinical trials with crizotinib in NSCLC. Trial

Trial population

n (crizotinib arm)

Design

Crizotinib response rate

Median duration of response

Progression-free survival

PROFILE 1001 [Kwak et al. 2010]

Advanced ALKpositive NSCLC, 84% patients at least 1 prior systemic treatment Advanced ALKpositive non-small cell lung cancer, 47.2% patients at least 2 prior systemic treatments

143

Open-label phase I study

60.8% (95% CI 37.3– 75.4).

49.1 weeks (95% CI 37.3–75.4)

NE

261

Phase II single arm study

59.8% (53.6–65.9)

43 weeks (95% CI 36–50).

8.5 months (95% CI 6.2–9.9)

Advanced ALKpositive NSCLC, one prior systemic treatment

173

65% (95% CI 58–72)

32.1 weeks crizotinib (range 2.1–72.9) versus 24.4 weeks chemotherapy (range 3.0–43.6)

Advanced ALKpositive nonsquamous NSCLC, 1 prior systemic treatment

343

Phase III study: crizotinib versus docetaxel or pemetrexed chemotherapy Phase III study: crizotinib versus platinumpemetrexed chemotherapy

74% (95% CI 67–81)

49 weeks (95% CI 35.1–60) versus 22.9 weeks (95% CI 18.0–25.1) chemotherapy

7.7 months crizotinib versus 3.3 months chemotherapy (HR 0.49, 95% CI 0.37–0.64, p < 0.0001). 10.9 months crizotinib versus 7.0 months chemotherapy (HR 0.454, 95% CI 0.35–0.61, p < 0.0001)

PROFILE 1005 (January 2012 evaluation) [Kim et al. 2012] PROFILE 1007 [Shaw et al. 2013]

PROFILE 1014 [Mok et al. 2014]

CI, confidence interval; HR, hazard ratio; NE, not evaluated; NSCLC, nonsmall cell lung cancer.

Survival benefit with crizotinib Formal evidence for an OS advantage for crizotinib has been difficult to prove given the accelerated nature of the drug development programme, rendering it unethical to withhold drug from ALK-positive patients. Therefore, OS results of PROFILE 1007 have been biased by marked crossover. In order to try to quantify the OS benefit of crizotinib, retrospective analysis of patients screened for the (1001) phase I study was performed comparing survival outcomes for ALKpositive patients that received crizotinib with those that did not [Shaw et al. 2011]. Here, crizotinib-treated ALK-positive patients had not yet reached median OS at the time of analysis, with a 1-year survival of 71% (95% CI 58–91) and a 2-year survival of 57% (95% CI 40–71), and was comparable with OS of a separate control cohort of ALK-negative EGFR mutant NSCLC treated with EGFR-TKI [1 year OS 74% (95% CI 61– 83), 2 year OS 52% (95% CI 38–65)], compared with crizotinib-naïve ALK-positive untreated patients, with a median survival of 20 months (95% CI 13–16).

Brain as a site of progressive disease on crizotinib Crizotinib penetrates the blood–brain barrier poorly [Costa et  al. 2011] and the brain is the commonest (45%) reported site of new disease in ALK-positive patients treated with crizotinib in the PROFILE 1001 and 1005 clinical trials [Otterson et al. 2012]. A retrospective exploratory analysis of patients recruited to PROFILE 1005 and 1007 has shown that 20% of patients with progressive disease developed new brain metastases [Costa et  al. 2015]. Brain as the site of progression in PROFILE 1014 was approximately 15% and crizotinib was numerically superior to platinum-doublet chemotherapy for intracranial time to progression (HR 0.6, 95% CI 0.34–1.05 in all patients; HR 0.45, 95% CI 0.19–1.07 in patients with baseline brain metastases p = not significant) [Solomon et  al. 2014]. Intracranial oligoprogressive disease can be treated with palliative radiation or ablative therapy followed by continuation of crizotinib [Takeda et  al. 2013], systemic chemotherapy or newer small molecule ALK inhibitors [Shaw et al. 2014]. In an intriguing

4 http://tar.sagepub.com

Downloaded from tar.sagepub.com at UNIV NEBRASKA LIBRARIES on April 11, 2015

A Pender and S Popat single case-report, escalation of crizotinib dose to 1000 mg daily has been reported to control oligoprogressive cranial disease in a single patient [Kim et al. 2013]. Isolated central nervous system (CNS) failure can be successfully controlled with local therapy and crizotinib continued beyond progression with a further period of disease stability of at least 4 months [Weickhardt et  al. 2012b; Takeda et  al. 2013]. Retrospective analysis suggests that continuation of crizotinib beyond progression confers a median survival benefit of 11 months (HR 0.38, 95% CI 0.22–0.66; p = 0.0005) compared with other subsequent systemic therapy. Furthermore, limited early phase trials data suggest that new more potent ALK inhibitors seem to have CNS activity in crizotinib refractory patients [Gadgeel et al. 2014]. The optimum treatment of oligoprogressive cranial disease may therefore evolve over the coming years, as these agents become a more routine part of clinical practice. Toxicity profile of crizotinib In the PROFILE 1007 trial, whilst more adverse events were most commonly reported in the crizotinib-treated group after controlling for disease progression (1768 versus 1190 events) [Shaw et  al. 2013], the incidence of grade 3–4 adverse events and serious adverse events was similar in both groups, and many crizotinib-associated adverse events were grade 1–2. Adverse events that occurred with over 5% greater incidence in the crizotinib-treated group were elevated liver aminotransferases, oedema, vision disorders, constipation, nausea, vomiting, dizziness, upper respiratory tract infections and taste changes. Liver dysfunction and hepatitis Grade 3–4 elevated aminotransferases were observed in 16% of patients on crizotinib in PROFILE 1007, with one grade 5 hepatic toxicity, and in general is easily managed by drug hold and dose reduction. However, only 1% of patients required crizotinib discontinuation due to hepatotoxicity and the EMA gives clear guidelines on the management of this toxicity [Committee for Medicinal Products for Human Use, 2014b]. Hypogonadism secondary to crizotinib Low serum testosterone levels have been observed in male NSCLC patients receiving crizotinib that

were not identified from PROFILE 1001 or 1005 [Weickhardt et  al. 2012a]. In a small case series, this has been shown to be independent of age, prior cytotoxic therapy, presence of brain metastases, prior whole brain irradiation, concurrent medications and nutritional status. A marked fall in testosterone is generally noted within days of starting crizotinib and serum testosterone levels change according to dose interruptions and recommencement of therapy in some patients. Falling luteinizing hormone (LH) and follicle-stimulating hormone (FSH) accompany the fall in serum testosterone in some patients, suggesting crizotinib has a central affect, but in others LH and FSH remained elevated or in the normal range, suggesting crizotinib also has an additional direct gonadal effect. Low testosterone levels can contribute to reduced quality of life [Fleishman et al. 2010] and testosterone replacement therapy should be considered in patients with symptomatic, proven hypogonadism on crizotinib therapy. Bradycardia Another effect that has been described in clinical practice since the early development of crizotinib is a decrease in heart rate. A retrospective analysis of all patients enrolled in PROFILE 1005 and the crizotinib arm of PROFILE 1007 study (1053 participants) concluded that the mean decrease in heart rate was 25 beats/minute [standard deviation (SD) 15.8] and that 41.9% of patients had a least one reading of sinus bradycardia (40 mmHg did not accompany sinus bradycardia in the vast majority of cases (99.1%), although this was slightly more common when there was concomitant use of beta-blockers (1.7% had a >40 mmHg decrease in systolic blood pressure, 2% had a >30 mmHg decrease in diastolic blood pressure). Non-Asian patients and older patients (⩾65 years-old) were more likely to develop sinus bradycardia (p = 0.003 and p = 0.0039, respectively). Analysis of a small patient cohort suggests that sinus bradycardia may correlate with maximum tumour shrinkage (p = 0.0205) and overall response rate (p = 0.0195) [Ou et  al. 2013]. Whilst this may reflect drug exposure, this study also found that bradycardic patients remained on crizotinib significantly longer than those with normocardia (52.9 weeks versus 24.6 weeks, p = 0.005), as a potential covariate.

http://tar.sagepub.com 5

Downloaded from tar.sagepub.com at UNIV NEBRASKA LIBRARIES on April 11, 2015

Therapeutic Advances in Respiratory Disease  Molecular mechanisms of crizotinib resistance Several different molecular mechanisms have been observed thus far in crizotinib-resistant NSCLC. As with EGFR TKIs in advanced NSCLC [Kobayashi et  al. 2005], secondary mutations in the ALK kinase domain have been discovered in patients who become resistant to crizotinib treatment. The ALK L1196M mutation corresponds to the position of the EGFR T790M mutation and the resulting amino acid change may similarly sterically inhibit inhibitor binding. Other ALK kinase mutations detected in crizotinib-resistant tumours include L1152R, C1156Y and G1269A – another mutation that may interfere with crizotinib binding [Doebele et al. 2012; Choi et al. 2010; Sasaki et al. 2011]. Other molecular resistance mechanisms of crizotinib resistance, which are again analogous to those seen with EGFR TKIs [Sequist et al. 2011], include ALK amplification and addition of other oncogenic mutations such as in EGFR and KRAS. Loss of the ALK fusion gene, in the absence of any additional mutations, has also been reported in crizotinib-resistant disease [Doebele et  al. 2012]. In addition, in vitro data suggest that EGFR pathway activation, in the absence of EGFR amplification or mutation, can contribute to crizotinib resistance and that this effect can be overcome with dual EGFR and ALK inhibition [Yamaguchi et  al. 2014; Sasaki et al. 2011]. Conclusion Crizotinib is an effective treatment in ALKpositive advanced nonsquamous NSCLC with a tolerable side effect profile and a remarkable response rate. In addition, the median duration of response exceeds that of platinum-pemetrexed chemotherapy. The superiority of crizotinib over standard-of-care chemotherapy in untreated patients has been demonstrated. Intracranial oligoprogressive disease on crizotinib can be treated using various strategies and does not necessarily mandate a change from crizotinib. Particular side effects of crizotinib such as bradycardia and low testosterone may require monitoring but are well tolerated. The success of targeting ALK rearrangements with crizotinib emphasizes the importance of molecular profiling of advanced NSCLC at diagnosis to allow selection of the most appropriate therapeutic agent for disease palliation.

Funding This research received no specific grant from any funding agency in the public, commercial, or notfor-profit sectors. The authors acknowledge NHS funding to the Royal Marsden NHS Foundation Trust/The Institute of Cancer Research NIHR Biomedical Research Centre. Conflict of interest statement S.P. is noncompensated consultant to Pfizer. A.P. declares no conflicts of interest in preparing this article. References Blackhall, F.H., Evans, T. L., Han, J., Salgia, R., Moro-Sibilot, D., Gettinger, S. et al. (2012) Impact of crizotinib treatment on patient-reported symptoms and quality of life (QOL) in advanced ALK-positive non-small cell lung cancer (NSCLC). Ann Oncol 23(Suppl. 9): 400–446. Choi, Y.L., Soda, M., Yamashita, Y., Ueno, T., Takashima, J., Nakajima, T. et al. (2010) EML4ALK mutations in lung cancer that confer resistance to ALK inhibitors. N Engl J Med 363(18): 1734–1739. doi:10.1056/NEJMoa1007478. Choi, Y.L., Takeuchi, K., Soda, M., Inamura, K., Togashi, Y., Hatano, S. et al. (2008) Identification of novel isoforms of the EML4-ALK transforming gene in non-small cell lung cancer. Cancer Res 68(13): 4971–4976. doi:10.1158/0008-5472. CAN-07-6158. Christensen, J.G., Zou, H.Y., Arango, M.E., Li, Q., Lee, J.H., McDonnell, S.R. et al. (2007) Cytoreductive antitumor activity of PF-2341066, a novel inhibitor of anaplastic lymphoma kinase and c-Met, in experimental models of anaplastic largecell lymphoma. Mol Cancer Therapeut 6(12 Pt 1): 3314–3322. doi:10.1158/1535-7163.MCT-07-0365. Commitee for Medicinal Products for Human Use (n.d.) Xalkori European Public Assessment Report. European Medicines Agency. Available at: http://www.ema.europa.euemainindex. jspcurlpagesmedicineshumanmedicineshumanmed. jspmidWcbacd Committee for Medicinal Products for Human Use (2014, April 14) Xalkori, Summary of Product Characteristics. European Medicines Agency. Available at: http://www.ema.europa.euemainindex. jspcurlpagesmedicineshumanmedicineshumanmed. jspmidWcbacd Costa, D.B., Kobayashi, S., Pandya, S.S., Yeo, W.-L., Shen, Z., Tan, W. et al. (2011) CSF concentration of the anaplastic lymphoma kinase inhibitor crizotinib. J Clin Oncol 29(15): e443–e445. doi:10.1200/ JCO.2010.34.1313.

6 http://tar.sagepub.com

Downloaded from tar.sagepub.com at UNIV NEBRASKA LIBRARIES on April 11, 2015

A Pender and S Popat Costa, D.B., Shaw, A.T., Ou, S.-H.I., Solomon, B.J., Riely, G.J., Ahn, M.-J. et al. (2015) Clinical experience with crizotinib in patients with advanced ALK-rearranged non-small-cell lung cancer and brain metastases. J Clin Oncol. Epub ahead of print 26 January 2015. doi:10.1200/JCO.2014.59.0539. Crino, L., Kim, D., Riely, G.J., Janne, P.A., Blackhall, F.H., Camidge, D.R. et al. (2011) Initial phase II results with crizotinib in advanced ALKpositive non-small cell lung cancer (NSCLC): PROFILE 1005. J Clin Oncol 29(15s). Doebele, R.C., Pilling, A.B., Aisner, D.L., Kutateladze, T.G., Le, A.T., Weickhardt, A.J. et al. (2012) Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer. Clin Cancer Res 18(5): 1472–1482. doi:10.1158/1078-0432.CCR-11-2906. Fleishman, S.B., Khan, H., Homel, P., Suhail, M.F., Strebel-Amrhein, R., Mohammad, F. et al. (2010) Testosterone levels and quality of life in diverse male patients with cancers unrelated to androgens. J Clin Oncol 28(34): 5054–5060. doi:10.1200/ JCO.2010.30.3818. Gadgeel, S.M., Gandhi, L., Riely, G.J., Chiappori, A.A., West, H.L., Azada, M.C. et al. (2014) Safety and activity of alectinib against systemic disease and brain metastases in patients with crizotinibresistant ALK-rearranged non-small-cell lung cancer (AF-002JG): results from the dose-finding portion of a phase 1/2 study. Lancet Oncol 15(10): 1119–1128. doi:10.1016/S1470-2045(14)70362-6.

data for crizotinib (PF-02341066) in ALK-positive advanced non-small cell lung cancer (NSCLC): PROFILE 2005. J Thorac Oncol 6(6s). Kim, Y.H., Ozasa, H., Nagai, H., Sakamori, Y., Yoshida, H., Yagi, Y. et al. (2013) High-dose crizotinib for brain metastases refractory to standarddose crizotinib. J Thorac Oncol 8(9): e85–e86. Kobayashi, S., Boggon, T.J., Dayaram, T., Jänne, P.A., Kocher, O., Meyerson, M. et al. (2005) EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med 352(8): 786–792. doi:10.1056/NEJMoa044238. Kris, M.G., Johnson, B.E., Berry, L.D., Kwiatkowski, D.J., Iafrate, A.J., Wistuba, I.I. et al. (2014) Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. J Am Med Assoc 311(19): 1998–2006. doi:10.1001/ jama.2014.3741. Kwak, E.L., Bang, Y.-J., Camidge, D. R., Shaw, A. T., Solomon, B., Maki, R.G. et al. (2010) Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med 363(18): 1693–1703. doi:10.1056/NEJMoa1006448. McDermott, U., Iafrate, A.J., Gray, N.S., Shioda, T., Classon, M., Maheswaran, S. et al. (2008) Genomic alterations of anaplastic lymphoma kinase may sensitize tumors to anaplastic lymphoma kinase inhibitors. Cancer Res 68(9): 3389–3395. doi:10.1158/0008-5472. CAN-07-6186.

Gainor, J.F., Varghese, A.M., Ou, S.H.I., Kabraji, S., Awad, M.M., Katayama, R. et al. (2013) ALK rearrangements are mutually exclusive with mutations in EGFR or KRAS: an analysis of 1,683 patients with non-small cell lung cancer. Clin Cancer Res 19(15): 4273–4281. doi:10.1158/1078-0432.CCR-13-0318.

Mok, T., Kim, D.W., Solomon, B.J., Nakagawa, K., Mekhail, T.M., Felip, E. et al. (2014) First-line crizotinib versus pemetrexed-cisplatin or pemetrexedcarboplatin in patients (pts) with advanced ALKpositive non-squamous non-small cell lung cancer (NSCLC): results of a phase III study (PROFILE 1014). J Clin Oncol 32(5s) [abstr 8002].

Jefferson, E. (2011) FDA approves Xalkori with companion diagnostic for a type of late-stage lung cancer. FDA NEWS RELEASE. Available at: http://www.Fda.Gov/NewsEvents/Newsroom/ PressAnnouncements.Ucm.Htm

Mok, T.S., Wu, Y.-L., Thongprasert, S., Yang, C.-H., Chu, D.-T., Saijo, N. et al. (2009) Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 361(10): 947–957. doi:10.1056/ NEJMoa0810699.

Kazandjian, D., Blumenthal, G.M., Chen, H.-Y., He, K., Patel, M., Justice, R. et al. (2014) FDA approval summary: crizotinib for the treatment of metastatic non-small cell lung cancer with anaplastic lymphoma kinase rearrangements. Oncologist 19(10): e5–e11. doi:10.1634/theoncologist.2014-0241.

Otterson, G.A., Riely, G.J., Shaw, A.T., Crino, L., Kim, D.W., Martins, R. et al. (n.d.) Clinical characteristics of ALK+ NSCLC patients (pt) treated with crizotinib beyond disease progression (PD): potential implications for management. J Clin Oncol 30(15s).

Kim, D., Ahn, M., Yang, P., Liu, X., De Pas, T. and Crinò, L. (2012) Updated results of a global phase II study with crizotinib in advanced ALK-positive nonsmall cell lung cancer (NSCLC). Proc Am Soc Clin Oncol (30s) [abstr 8033].

Ou, S.H.I., Tang, Y., Polli, A., Wilner, K. D. and Schnell, P. (n.d.) Characterization of heart rate (HR) changes during crizotinib treatment: A retrospective analysis of 1,053 ALK+ NSCLC patients. J Clin Oncol 32(5s) [abstr 8106].

Kim, D., Crino, L., Janne, P.A., Blackhall, F., Camidge, D.R., Hirsch, V. et al. (2011) Phase 2

Ou, S.-H.I., Tong, W.P., Azada, M., Siwak-Tapp, C., Dy, J. and Stiber, J.A. (2013) Heart rate decrease

http://tar.sagepub.com 7

Downloaded from tar.sagepub.com at UNIV NEBRASKA LIBRARIES on April 11, 2015

Therapeutic Advances in Respiratory Disease  during crizotinib treatment and potential correlation to clinical response. Cancer 119(11): 1969–1975. doi:10.1002/cncr.28040. Popat, S., Mok, T., Yang, J.C.-H., Wu, Y.-L., Lungershausen, J., Stammberger, U. et al (2014) Afatinib in the treatment of EGFR mutation-positive NSCLC–a network meta-analysis. Lung Cancer 85(2): 230–238. doi:10.1016/j.lungcan.2014.05.007. Rikova, K., Guo, A., Zeng, Q., Possemato, A., Yu, J., Haack, H. et al. (2007) Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 131(6): 1190–1203. doi:10.1016/ j.cell.2007.11.025.

Solomon, B., Felip, E., Blackhall, F.H., Mok, T., Kim, D., Wu, J. et al. (2014) Overall and intracranial (IC) efficacy results and time to symptom deterioration in PROFILE 1014: 1st-line crizotinib vs pemetrexed − platinum chemotherapy (PPC) in patients (pts) with advanced ALK-positive nonsquamous non-small cell lung cancer (NSCLC). Ann Oncol 25(Suppl. 4): iv264–iv270.

Ross, Camidge D.D., Bang, Y.-J., Kwak, E.L., Iafrate, A.J., Varella-Garcia, M., Fox, S.B. et al. (2012) Activity and safety of crizotinib in patients with. Lancet Oncol 13(10): 1011–1019. doi:10.1016/ S1470-2045(12)70344-3.

Takeda, M., Okamoto, I. and Nakagawa, K. (2013) Clinical impact of continued crizotinib administration after isolated central nervous system progression in patients with lung cancer positive for ALK rearrangement. J Thorac Oncol 8(5): 654–657.

Sasaki, T., Koivunen, J., Ogino, A., Yanagita, M., Nikiforow, S., Zheng, W. et al. (2011) A novel ALK secondary mutation and EGFR signaling cause resistance to ALK kinase inhibitors. Cancer Res 71(18): 6051–6060. doi:10.1158/0008-5472.CAN11-1340.

Takeuchi, K., Choi, Y.L., Soda, M., Inamura, K., Togashi, Y., Hatano, S. et al. (2008) Multiplex reverse transcription-PCR screening for EML4-ALK fusion transcripts. Clin Cancer Res 14(20): 6618–6624. doi:10.1158/1078-0432.CCR-08-1018.

Sequist, L.V., Waltman, B.A., Dias-Santagata, D., Digumarthy, S., Turke, A.B., Fidias, P. et al. (2011) Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Trans Med 3(75): 75ra26. Shaw, A.T., Kim, D.-W., Mehra, R., Tan, D.S.W., Felip, E., Chow, L.Q.M. et al. (2014) Ceritinib in ALK-rearranged non–small-cell lung cancer. N Engl J Med 370(13): 1189–1197. doi:10.1056/ nejmoa1311107. Shaw, A.T., Kim, D.-W., Nakagawa, K., Seto, T., Crino, L., Ahn, M.-J. et al. (2013) Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med 368(25): 2385–2394. doi:10.1056/ NEJMoa1214886. Shaw, A.T., Yeap, B.Y., Solomon, B.J., Riely, G.J. and Gainor, J. (2011) Effect of crizotinib on overall survival in patients with advanced non-small-cell lung cancer harbouring ALK gene rearrangement: a retrospective analysis. Lancet Oncol 12(11): 1004–1012. doi:10.1016/S1470-2045(11)70232-7. Shepherd, F.A., Dancey, J., Ramlau, R., Mattson, K., Gralla, R., O’Rourke, M. et al. (2000) Prospective randomized trial of docetaxel versus best supportive care in patients with non-small-cell lung cancer previously treated with platinum-based chemotherapy. J Clin Oncol 18(10): 2095–2103.

Visit SAGE journals online http://tar.sagepub.com

SAGE journals

Soda, M., Isobe, K., Inoue, A., Maemondo, M., Oizumi, S., Fujita, Y. et al. (2012) A prospective PCR-based screening for the EML4-ALK oncogene in non-small cell lung cancer. Clin Cancer Res 18(20): 5682–5689. doi:10.1158/1078-0432.CCR-11-2947.

Soda, M., Choi, Y.L., Enomoto, M., Takada, S., Yamashita, Y., Ishikawa, S. et al. (2007) Identification of the transforming EML4-ALK fusion gene in nonsmall-cell lung cancer. Nature 448(7153): 561–566. doi:10.1038/nature05945.

Takeuchi, K., Choi, Y.L., Togashi, Y., Soda, M., Hatano, S., Inamura, K. et al. (2009) KIF5BALK, a novel fusion oncokinase identified by an immunohistochemistry-based diagnostic system for ALK-positive lung cancer. Clin Cancer Res 15(9): 3143–3149. doi:10.1158/1078-0432.CCR-08-3248. Travis, W.D., Brambilla, E., Noguchi, M., Nicholson, A.G., Geisinger, K., Yatabe, Y. et al. (2013) Diagnosis of lung cancer in small biopsies and cytology: implications of the 2011 International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society Classification. Arch Pathol Lab Med 137(5): 668–684. doi:10.5858/arpa.2012-0263-RA. Weickhardt, A.J., Rothman, M.S., Salian-Mehta, S., Kiseljak-Vassiliades, K., Oton, A.B., Doebele, R.C. et al. (2012a) Rapid-onset hypogonadism secondary to crizotinib use in men with metastatic nonsmall cell lung cancer. Cancer 118(21): 5302–5309. doi:10.1002/cncr.27450. Weickhardt, A.J., Scheier, B., Burke, J.M., Gan, G., Lu, X., Bunn, P.A.J. et al. (2012b) Local ablative therapy of oligoprogressive disease prolongs disease control by tyrosine kinase inhibitors in oncogeneaddicted non-small-cell lung cancer. J Thorac Oncol 7(12): 1807–1814. Yamaguchi, N., Lucena-Araujo, A.R., Nakayama, S., de Figueiredo-Pontes, L.L., Gonzalez, D.A., Yasuda, H. et al. (2014) Dual ALK and EGFR inhibition targets a mechanism of acquired resistance to the tyrosine kinase inhibitor crizotinib in ALK rearranged lung cancer. Lung Cancer 83(1): 37–43. doi:10.1016/ j.lungcan.2013.09.019.

8 http://tar.sagepub.com

Downloaded from tar.sagepub.com at UNIV NEBRASKA LIBRARIES on April 11, 2015

The efficacy of crizotinib in patients with ALK-positive nonsmall cell lung cancer.

Molecular profiling of nonsmall cell lung cancer (NSCLC) contributes to better understanding the different molecular subtypes of this heterogeneous gr...
784KB Sizes 0 Downloads 12 Views