Review
ALK translocation and crizotinib in non-small cell lung cancer: An evolving paradigm in oncology drug development

https://doi.org/10.1016/j.ejca.2012.02.001Get rights and content

Abstract

Advances in our understanding of tumour biology have encouraged reassessment of tumour classification by the site of origin in favour of molecular characteristics and/or oncogenic drivers amenable to treatment. The identification of EML4-anaplastic lymphoma kinase (ALK) as an oncogenic driver in non-small cell lung cancer (NSCLC) early in the clinical development of crizotinib and the observation of promising clinical responses in patients with NSCLC harbouring ALK translocations accelerated its clinical development in ALK-positive NSCLC. Phase I and II trials of crizotinib in patients with ALK-positive advanced NSCLC reported notably high response rates that tended to be rapid and of prolonged duration. Crizotinib was well tolerated; treatment-related adverse events were typically gastrointestinal (grade 1/2) and visual disorders (almost exclusively grade 1). Crizotinib provided NSCLC symptom relief and maintained quality of life. Based on the phase I and II trial data, the US Food and Drug Administration granted approval of crizotinib in August 2011. The consistency of the crizotinib data to date suggests accurate selection of the target population for crizotinib treatment. The ability to molecularly select patients likely to respond to an investigational agent argues that future clinical development of targeted agents should be re-evaluated. Updated trial designs incorporating molecular testing, early use of enrichment biomarkers and intermediary endpoints may accelerate and optimise clinical evaluation of targeted agents. Such trial designs should allow rapid clinical evaluation, minimise exposure of patients to therapies unlikely to be of benefit and, potentially, allow accelerated drug approval in molecularly specified populations.

Introduction

Crizotinib clinical development has focused primarily on molecularly selected patients with anaplastic lymphoma kinase (ALK) translocations. Following the identification of EML4-ALK as an oncogenic driver in non-small cell lung cancer (NSCLC) early in the clinical development of crizotinib and the observation of promising clinical responses in patients with NSCLC harbouring ALK translocations, ALK-positive NSCLC became a focus for the clinical development of crizotinib.1, 2 Trials with crizotinib have consistently reported notably high response rates, with responses of prolonged duration, often rapidly achieved.1, 2, 3, 4, 5 In addition, crizotinib was well tolerated and provided symptomatic relief whilst maintaining quality of life. Accelerated Food and Drug Administration (FDA) approval of crizotinib has been granted based on the phase I and II trial data.4, 5, 6, 7 Advances in our understanding of tumour biology are overturning the classification of tumours by site of origin in favour of grouping by molecular characteristics and key oncogenic drivers amenable to pharmacologic modulation.8, 9 This progress, together with the realistic expectation of achieving impressive tumour responses, argues that the current approach of evaluating drugs via large empirical trials in unselected patient populations should be re-evaluated for targeted drugs. Updated trial designs incorporating customised testing, use of enrichment biomarkers as early as possible and intermediary endpoints will accelerate and optimise clinical evaluation of targeted agents.10

Matching patients with tumours harbouring ‘drugable’ genetic abnormalities with appropriate molecularly targeted agents can have dramatic results. High response rates were reported with imatinib in interferon-resistant chronic myeloid leukaemia (CML) (target: BCR-ABL; cytogenetic response rate: 54%) and gastrointestinal stromal tumour (GIST) (target: KIT; objective response rate [ORR] 54%), and with dasatinib in imatinib-resistant Philadelphia chromosome-positive leukaemias (target: BCR-ABL; haematological response rate: 92% for patients with chronic-phase CML and 70% for patients with accelerated-phase CML, CML with blast crisis or Philadelphia chromosome-positive acute lymphoblastic leukaemia).11, 12, 13 Treatment of women with breast cancer overexpressing human epidermal growth factor receptor 2 (HER2) with trastuzumab resulted in an obvious improvement in survival and dramatic responses to endothelial growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) were observed in patients with NSCLC harbouring EGFR sensitising mutations (approximately 10% of the unselected Caucasian patients enroled in early trials).14, 15, 16 The IPASS trial, which compared gefitinib with combination chemotherapy in the first-line treatment of NSCLC, was a landmark study that not only redefined standard therapy for patients with EGFR sensitising mutations, but also clearly demonstrated that patient selection for targeted agents must be made on the basis of molecular characteristics.15, 17

The relevance and ethical acceptability of randomised studies for clinical development are therefore highly questionable in poor-prognosis disease where the investigational arm is likely to be markedly more effective than the control arm. Recently, this issue came to the attention of the media when two young male cousins with melanoma enroled in a randomised trial of the investigational agent vemurafenib (PLX4032) versus a marginally active standard chemotherapy. The cousin diagnosed and randomised first received vemurafenib and responded within 2 months, whilst the cousin diagnosed second was randomised to the control arm and progressed quickly. With crossover disallowed, this was obviously very distressing for the patients, their families and the attending physician.18 Conversely, imatinib entered phase II study in GIST on the basis of compelling preclinical data and a single highly encouraging case study.12 Responses in the initial phase II trial were considered ‘remarkable’ and led to FDA approval in 2002.12, 19 The subsequent phase III study tested different doses of imatinib rather than including a control arm.20 For GIST, it was recognised that there simply was no effective treatment option for comparison.12 Timelines for the development of such agents are shortening as our understanding of tumour biology and our ability to select the true patient population increase; whilst 41 years elapsed between the discovery of BCR-ABL and initial trials with imatinib, it was less than 10 years for agents modulating more recently identified targets (KIT: 1998; BRAF: 2002).21

Several potential oncogenic drivers have been identified in NSCLC, including EGFR, BRAF, KRAS, MET, HER2 and ALK.22, 23, 24 The investigation of driver mutations has led to the development of specific molecularly targeted therapies, most notably gefitinib and erlotinib (both EGFR inhibitors, now known to be effective first-line therapy for tumours with EGFR mutations).15, 25, 26, 27 The early development of gefitinib and erlotinib was hampered by the lack of detailed molecular knowledge of lung cancer and its molecular subtypes, and clinical progress was slow as a result. Continued research into EGFR mutations and diagnosis developed our understanding of the molecular basis of NSCLC, and made molecular testing a familiar concept in this disease.

Section snippets

Anaplastic lymphoma kinase (ALK): a specific oncogenic driver

The nucleophosmin (NPM)–anaplastic lymphoma kinase (ALK) fusion protein was originally identified as an oncogenic driver in patients with anaplastic large-cell lymphoma (ALCL) in the early-to-mid 1990s and it quickly became apparent that ALK-positive and ALK-negative ALCLs represent distinct clinical entities.28, 29, 30, 31 Chromosomal translocations fusing ALK with a number of binding partners and resulting in ALK activation have since been described in other human cancers, including

Efficacy

Crizotinib, a potent and selective ATP-competitive inhibitor of c-Met and ALK receptor tyrosine kinases and oncogenic variants, was first studied clinically in the phase I trial.1, 68, 69, 70 In contrast with EGFR TKIs, where identification of the receptor to treatment of patients with a pharmacologic modulator took 26 years, crizotinib entered clinical testing in patients with ALK-translocated NSCLC early, approximately 4 months after ALK-fusion was first identified in that disease. Dramatic

Crizotinib and the future clinical study of targeted agents

ALK-positive NSCLC is a discrete, molecularly defined clinical entity with distinct clinical characteristics. Appropriately case-matched/adjusted retrospective analyses suggest that ALK-positive patients may have a similar-to-worse clinical prognosis compared with ALK-negative patients.65, 66, 67 Clinical data for crizotinib, in the context of historical data for ALK-positive patients who did not receive crizotinib, suggest that the natural history of ALK-positive NSCLC can be fundamentally

Conflict of interest statement

Giorgio Scagliotti has received honoraria from Eli Lilly, AstraZeneca, ARIAD, and Roche. Rolf A. Stahel has served Astellas, AstraZeneca, Bayer, Boeringer Ingelheim, Eli Lilly, Genentech, GSK, Merck Serono, Novartis, Pfizer Oncology, and Roche in a consultant/advisory role, and has received honoraria from AstraZeneca, Eli Lilly, Merck Serono, Novartis, and Roche. Nick Thatcher has served Pfizer Oncology in a consultant/advisory role and has received honoraria and other remuneration from Pfizer

Role of the funding source

The current review resulted from a roundtable meeting in Amsterdam, Holland, on 5 July 2011. This meeting was organised and funded by Pfizer Inc. Pfizer Inc. funded the crizotinib trials reported in this review in addition to being involved in the study design and data collection, analysis and interpretation for these studies.

Acknowledgements

Medical writing support was provided by Christine Arris at ACUMED® (Tytherington, UK) with funding from Pfizer Inc. Editorial support was provided by Roger Wild at ACUMED® (Tytherington, UK) with funding from Pfizer Inc.

References (92)

  • K. Rikova et al.

    Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer

    Cell

    (2007)
  • K. Inamura et al.

    EML4-ALK fusion is linked to histological characteristics in a subset of lung cancers

    J Thorac Oncol

    (2008)
  • S. Perner et al.

    EML4-ALK fusion lung cancer: a rare acquired event

    Neoplasia

    (2008)
  • K. Shinmura et al.

    EML4-ALK fusion transcripts, but no NPM-, TPM3-, CLTC-, ATIC-, or TFG-ALK fusion transcripts, in non-small cell lung carcinomas

    Lung Cancer

    (2008)
  • M.P. Martelli et al.

    EML4-ALK rearrangement in non-small cell lung cancer and non-tumor lung tissues

    Am J Pathol

    (2009)
  • A.T. Shaw et al.

    Effect of crizotinib on overall survival in advanced NSCLC harboring anaplastic lymphoma kinase gene rearrangement: a retrospective analysis

    Lancet Oncol

    (2011)
  • M. Zillhardt et al.

    An orally available small-molecule inhibitor of c-Met, PF-2341066, reduces tumor burden and metastasis in a preclinical model of ovarian cancer metastasis

    Neoplasia

    (2010)
  • D.R. Camidge et al.

    Anaplastic lymphoma kinase gene rearrangements in non-small cell lung cancer are associated with prolonged progression-free survival on pemetrexed

    J Thorac Oncol

    (2011)
  • J.O. Lee et al.

    Anaplastic lymphoma kinase translocation: a predictive biomarker of pemetrexed in patients with non-small cell lung cancer

    J Thorac Oncol

    (2011)
  • E.L. Kwak et al.

    Clinical activity observed in a phase I dose escalation trial of an oral c-met and ALK inhibitor, PF-02341066

    J Clin Oncol

    (2009)
  • E.L. Kwak et al.

    Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer

    N Engl J Med

    (2010)
  • Bang Y, Kway EL, Shaw AT, et al. Clinical activity of the oral ALK inhibitor PF-02341066 in ALK-positive patients with...
  • Camidge DR, Bang Y, Kwak EL, et al. Progression-free survival (PFS) from a phase I study of crizotinib (PF-02341066) in...
  • Crinò L, Kim D-W, Riely G, et al. Initial phase II results with crizotinib in advanced ALK-positive non-small cell lung...
  • Pfizer Inc. Xalkori Prescribing Information. http://www.accessdata.fda.gov/drugsatfda_docs/label/2011/202570s000lbl.pdf...
  • Blackhall FH, Petersen JA, Wilner K, et al. PROFILE 1005: preliminary patient-reported outcomes (PROs) from an ongoing...
  • National Cancer Institute, National Human Genome Research Institute. The Cancer Genome Atlas....
  • I.B. Weinstein et al.

    Oncogene addiction

    Cancer Res

    (2008)
  • T.A. Yap et al.

    Envisioning the future of early anticancer drug development

    Nat Rev Cancer

    (2010)
  • B.J. Druker et al.

    Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome

    N Engl J Med

    (2001)
  • G.D. Demetri et al.

    Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors

    N Engl J Med

    (2002)
  • M. Talpaz et al.

    Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias

    N Engl J Med

    (2006)
  • M.S. Tsao et al.

    Erlotinib in lung cancer – molecular and clinical predictors of outcome

    N Engl J Med

    (2005)
  • T.S. Mok et al.

    Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma

    N Engl J Med

    (2009)
  • F.A. Shepherd

    Molecular selection trumps clinical selection

    J Clin Oncol

    (2011)
  • New York Times. New Drugs Stir Debate on Rules of Clinical Trials....
  • R. Dagher et al.

    Approval summary: imatinib mesylate in the treatment of metastatic and/or unresectable malignant gastrointestinal stromal tumors

    Clin Cancer Res

    (2002)
  • C.D. Blanke et al.

    Phase III randomized, intergroup trial assessing imatinib mesylate at two dose levels in patients with unresectable or metastatic gastrointestinal stromal tumors expressing the kit receptor tyrosine kinase: S0033

    J Clin Oncol

    (2008)
  • K. Garber

    ALK, lung cancer, and personalized therapy: portent of the future?

    J Natl Cancer Inst

    (2010)
  • F. Janku et al.

    Targeted therapy in non-small-cell lung cancer – is it becoming a reality?

    Nat Rev Clin Oncol

    (2010)
  • M. Fukuoka et al.

    Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS)

    J Clin Oncol

    (2011)
  • F.A. Shepherd et al.

    Erlotinib in previously treated non-small-cell lung cancer

    N Engl J Med

    (2005)
  • S.W. Morris et al.

    Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin’s lymphoma

    Science

    (1994)
  • D. Bischof et al.

    Role of the nucleophosmin (NPM) portion of the non-Hodgkin’s lymphoma-associated NPM-anaplastic lymphoma kinase fusion protein in oncogenesis

    Mol Cell Biol

    (1997)
  • M. Shiota et al.

    The clinicopathological features of anaplastic large cell lymphomas expressing p80NPM/ALK

    Leuk Lymphoma

    (1996)
  • E. Lin et al.

    Exon array profiling detects EML4-ALK fusion in breast, colorectal, and non-small cell lung cancers

    Mol Cancer Res

    (2009)
  • Cited by (80)

    • Human genetic diversity in health and disease

      2020, Biodiversity and Biomedicine: Our Future
    View all citing articles on Scopus
    View full text