251
|
De Pas T, Pala L, Catania C, Conforti F. Molecular and clinical features of second-generation anaplastic lymphoma kinase inhibitors: ceritinib. Future Oncol 2017; 13:2629-2644. [DOI: 10.2217/fon-2017-0262] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The discovery of ALK rearrangement in non-small-cell lung cancer (NSCLC) triggered rapid clinical development of a family of specific drugs targeting this alteration, called ALK inhibitors. Despite high rate of responses, the vast majority of patients treated with first-generation ALK inhibitor crizotinib will ultimately develop disease progression. The second-generation ALK inhibitor, ceritinib, is an oral, small-molecule that inhibits the ALK kinase activity with a potency 20-fold greater than crizotinib, being able to tackle some of the principal mechanisms of resistance to crizotinib. Evidences from five large prospective clinical trials have so far showed impressive activity of ceritinib in ALK inhibitor pretreated and naive NSCLC patients. This review will focus on the preclinical and clinical data available regarding ceritinib pharmacology, clinical efficacy and safety profile.
Collapse
Affiliation(s)
- Tommaso De Pas
- Medical Oncology of Melanoma & Sarcoma Unit, European Institute of Oncology, Via Ripamonti, 435, 20141 Milan, Italy
| | - Laura Pala
- Medical Oncology of Melanoma & Sarcoma Unit, European Institute of Oncology, Via Ripamonti, 435, 20141 Milan, Italy
| | - Chiara Catania
- Medical Oncology Unit of Respiratory Tract, European Institute of Oncology, Via Ripamonti, 435, 20141 Milan, Italy
| | - Fabio Conforti
- Medical Oncology of Melanoma & Sarcoma Unit, European Institute of Oncology, Via Ripamonti, 435, 20141 Milan, Italy
| |
Collapse
|
252
|
Lovly CM, Iyengar P, Gainor JF. Managing Resistance to EFGR- and ALK-Targeted Therapies. Am Soc Clin Oncol Educ Book 2017; 37:607-618. [PMID: 28561721 PMCID: PMC10183098 DOI: 10.1200/edbk_176251] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Targeted therapies have transformed the management of non-small cell lung cancer (NSCLC) and placed an increased emphasis on stratifying patients on the basis of genetic alterations in oncogenic drivers. To date, the best characterized molecular targets in NSCLC are the epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK). Despite steady advances in targeted therapies within these molecular subsets, however, acquired resistance to therapy is near universal. Recent preclinical models and translational efforts have provided critical insights into the molecular mechanisms of resistance to EGFR and ALK inhibitors. In this review, we present a framework for understanding resistance to targeted therapies. We also provide overviews of the molecular mechanisms of resistance and strategies to overcome resistance among EGFR-mutant and ALK-rearranged lung cancers. To date, these strategies have centered on the development of novel next-generation inhibitors, rationale combinations, and use of local ablative therapies, such as radiotherapy.
Collapse
Affiliation(s)
- Christine M Lovly
- From the Division of Hematology and Oncology, Vanderbilt University School of Medicine, Nashville, TN; Department of Radiation Oncology, Thoracic Disease Oriented Team, Thoracic Radiation Oncology Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX; Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - Puneeth Iyengar
- From the Division of Hematology and Oncology, Vanderbilt University School of Medicine, Nashville, TN; Department of Radiation Oncology, Thoracic Disease Oriented Team, Thoracic Radiation Oncology Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX; Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - Justin F Gainor
- From the Division of Hematology and Oncology, Vanderbilt University School of Medicine, Nashville, TN; Department of Radiation Oncology, Thoracic Disease Oriented Team, Thoracic Radiation Oncology Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX; Harvard Medical School, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
253
|
Rolfo C, Arias D, Gandia JF, Manca P, Santos E, Raez LE. Ceritinib: an orphan drug for ALK positive non-small cell lung cancer with robust clinical evidence. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1406347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Christian Rolfo
- Phase I - Early Clinical Trials Unit, Antwerp University Hospital & Center for Oncological Research (CORE), Edegem, Belgium
| | - David Arias
- Phase I - Early Clinical Trials Unit, Antwerp University Hospital & Center for Oncological Research (CORE), Edegem, Belgium
- Ourense University Hospital Complex, Ourense, Spain
| | - Jose Ferri Gandia
- Phase I - Early Clinical Trials Unit, Antwerp University Hospital & Center for Oncological Research (CORE), Edegem, Belgium
| | - Paolo Manca
- Phase I - Early Clinical Trials Unit, Antwerp University Hospital & Center for Oncological Research (CORE), Edegem, Belgium
- Medical Oncology Department, Campus Bio-Medico University, Rome, Italy
| | - Edgardo Santos
- Oncology Department, Lynn Cancer Institute, Boca Raton, FL, USA
| | - Luis E. Raez
- Thoracic Oncology Program, Memorial Cancer Institute, Memorial Health Care System, Pembroke Pines, FL, USA
| |
Collapse
|
254
|
Itchins M, Chia PL, Hayes SA, Howell VM, Gill AJ, Cooper WA, John T, Mitchell P, Millward M, Clarke SJ, Solomon B, Pavlakis N. Treatment of ALK-rearranged non-small cell lung cancer: A review of the landscape and approach to emerging patterns of treatment resistance in the Australian context. Asia Pac J Clin Oncol 2017; 13 Suppl 3:3-13. [PMID: 28795492 DOI: 10.1111/ajco.12754] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Since the identification of anaplastic lymphoma kinase (ALK) gene rearrangements in non-small cell lung cancer (NSCLC) in 2005, the treatment of ALK-rearranged NSCLC (ALK+ NSCLC) has evolved at a rapid pace. This molecularly distinct subset of NSCLC has uniquely important biology, clinicopathologic features and mechanisms of drug resistance which impact on the choice of treatment for a patient with this disease. There are multiple ALK tyrosine kinase inhibitors now available in clinical practice with efficacy data continuing to emerge and guide the optimal treatment algorithm. A detailed search of medical databases and clinical trial registries was conducted to capture all relevant articles on this topic enabling an updated detailed overview of the landscape of management of ALK-rearranged NSCLC.
Collapse
Affiliation(s)
- M Itchins
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia.,Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Northern Cancer Institute, St Leonards, New South Wales, Australia
| | - P L Chia
- Medical Oncology Unit, Olivia Newton John Cancer and Wellness Centre, Austin Health, Melbourne.,Olivia Newton-John Cancer Research Institute, Melbourne, Australia.,Department of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - S A Hayes
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia.,Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - V M Howell
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia.,Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - A J Gill
- Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - W A Cooper
- Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia.,School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
| | - T John
- Medical Oncology Unit, Olivia Newton John Cancer and Wellness Centre, Austin Health, Melbourne.,Olivia Newton-John Cancer Research Institute, Melbourne, Australia.,Department of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - P Mitchell
- Medical Oncology Unit, Olivia Newton John Cancer and Wellness Centre, Austin Health, Melbourne.,Olivia Newton-John Cancer Research Institute, Melbourne, Australia.,Department of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - M Millward
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia.,Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Linear Clinical Research, Nedlands, Western Australia, Australia
| | - S J Clarke
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia.,Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Northern Cancer Institute, St Leonards, New South Wales, Australia.,Department of Medical Oncology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - B Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - N Pavlakis
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia.,Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Northern Cancer Institute, St Leonards, New South Wales, Australia.,Department of Medical Oncology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
255
|
Reck M. Pembrolizumab as first-line therapy for metastatic non-small-cell lung cancer. Immunotherapy 2017; 10:93-105. [PMID: 29145737 DOI: 10.2217/imt-2017-0121] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
This review describes trials evaluating the monoclonal antibody pembrolizumab (an immunotherapy that blocks the interaction between programmed death-1 and programmed death-ligand 1 and 2 [PD-L1/PD-L2]) as first-line therapy for advanced non-small-cell lung cancer (NSCLC). In the Phase III KEYNOTE-024 study, pembrolizumab monotherapy significantly improved progression-free survival (primary end point) and overall survival, and was associated with fewer adverse events compared with platinum-based chemotherapy in patients with NSCLC with PD-L1 expression on ≥50% of tumor cells. In cohort G of the Phase I/II KEYNOTE-021 study, pembrolizumab plus pemetrexed and carboplatin significantly improved objective response rate (primary end point) and progression-free survival versus pemetrexed and carboplatin alone, and had manageable toxicity in patients with nonsquamous NSCLC. These results have changed first-line management of advanced NSCLC.
Collapse
Affiliation(s)
- Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North (ARCN), German Center of Lung Research (DZL), Grosshansdorf, Germany
| |
Collapse
|
256
|
Di Lorenzo R, Ahluwalia MS. Targeted therapy of brain metastases: latest evidence and clinical implications. Ther Adv Med Oncol 2017; 9:781-796. [PMID: 29449898 PMCID: PMC5808839 DOI: 10.1177/1758834017736252] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 09/18/2017] [Indexed: 12/16/2022] Open
Abstract
Brain metastases (BM) occur in 20-40% of patients with cancer and 60-75% of patients with BM become symptomatic. Due to an aging population and advances in the treatment of primary cancers, patients are living longer and are more likely to experience complications from BM. The diagnosis of BM drastically worsens long-term survival rates, with multiple metastases being a poor prognostic factor. Until recently, the mainstay of treatment consisted of stereotactic radiosurgery (SRS), surgical resection, whole brain radiation therapy (WBRT), or a combination of these modalities. Systemic chemotherapy has been felt largely ineffective in the treatment of BM due to the presence of the blood-brain barrier (BBB), which includes efflux pumps on brain capillaries. Over the past decade however, researchers have identified therapeutic agents that are able to cross the BBB. These findings could make a multimodality treatment approach possible, consisting of surgery, radiation, immunotherapy, and targeted therapy, which could lead to better disease control in this patient population and prolong survival. In this review, we discuss present evidence on available targeted therapies and their role in the treatment of BM from primary tumors with the highest prevalence of central nervous system (CNS) involvement, specifically non-small cell lung cancer (NSCLC), breast cancer melanoma, and renal cell carcinoma.
Collapse
Affiliation(s)
- Rodica Di Lorenzo
- Brain Tumor and Neuro-Oncology Center, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Manmeet S Ahluwalia
- Brain Tumor and Neuro-Oncology Center, Cleveland Clinic Foundation, 9500 Euclid Avenue, CA-51, Cleveland, OH 44195, USA
| |
Collapse
|
257
|
Lin JJ, Shaw AT. Recent Advances in Targeting ROS1 in Lung Cancer. J Thorac Oncol 2017; 12:1611-1625. [PMID: 28818606 PMCID: PMC5659942 DOI: 10.1016/j.jtho.2017.08.002] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 08/05/2017] [Accepted: 08/08/2017] [Indexed: 01/03/2023]
Abstract
ROS1 is a validated therapeutic target in NSCLC. In a phase I study, the multitargeted MET proto-oncogene, receptor tyrosine kinase/anaplastic lymphoma kinase/ROS1 inhibitor crizotinib demonstrated remarkable efficacy in ROS1-rearranged NSCLCs and consequently gained approval by the United States Food and Drug Administration and by the European Medicines Agency in 2016. However, similar to other oncogene-driven lung cancers, ROS1-rearranged lung cancers treated with crizotinib eventually acquire resistance, leading to disease relapse. Novel ROS1 inhibitors and therapeutic strategies are therefore needed. Insights into the mechanisms of resistance to ROS1-directed tyrosine kinase inhibitors are now beginning to emerge and are helping to guide the development of new ROS1 inhibitors. This review discusses the biology and diagnosis of ROS1-rearranged NSCLC, and current and emerging treatment options for this disease. Future challenges in the field are highlighted.
Collapse
Affiliation(s)
- Jessica J Lin
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Alice T Shaw
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.
| |
Collapse
|
258
|
Dagogo-Jack I, Shaw AT. Crizotinib resistance: implications for therapeutic strategies. Ann Oncol 2017; 27 Suppl 3:iii42-iii50. [PMID: 27573756 DOI: 10.1093/annonc/mdw305] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In 2007, a chromosomal rearrangement resulting in a gene fusion leading to expression of a constitutively active anaplastic lymphoma kinase (ALK) fusion protein was identified as an oncogenic driver in non-small-cell lung cancer (NSCLC). ALK rearrangements are detected in 3%-7% of patients with NSCLC and are particularly enriched in younger patients with adenocarcinoma and a never or light smoking history. Fortuitously, crizotinib, a small molecule tyrosine kinase inhibitor initially developed to target cMET, was able to be repurposed for ALK-rearranged (ALK+) NSCLC. Despite dramatic and durable initial responses to crizotinib; however, the vast majority of patients will develop resistance within a few years. Diverse molecular mechanisms underlie resistance to crizotinib. This review will describe the clinical activity of crizotinib, review identified mechanisms of crizotinib resistance, and end with a survey of emerging therapeutic strategies aimed at overcoming crizotinib resistance.
Collapse
Affiliation(s)
- I Dagogo-Jack
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, USA
| | - A T Shaw
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, USA
| |
Collapse
|
259
|
Abstract
Ceritinib (Zykadia™) is an oral, selective inhibitor of the anaplastic lymphoma kinase (ALK), a receptor tyrosine kinase which, after genetic rearrangement, acts as an oncogenic driver in a proportion of non-small cell lung cancers (NSCLCs). The drug is approved in several countries worldwide for the treatment of patients with ALK-positive, advanced NSCLC who have previously received the first-generation ALK inhibitor crizotinib (indication details may vary by country). Approval was based on its clinical benefit in this setting in the phase I and II trials known as ASCEND-1 and -2. Across these noncomparative studies, 36-56 % of patients achieved a response with ceritinib (at the recommended dosage of 750 mg once daily) and the responses were durable, lasting up to a median of 10 months. Patients survived free from progression for a median of up to 7 months and had a median overall survival of up to 17 months. Moreover, efficacy outcomes in patients with brain metastases were generally consistent with those of the overall study populations. Ceritinib has an acceptable tolerability profile, with gastrointestinal issues, fatigue and liver test abnormalities being the most common adverse reactions. Thus, ceritinib is a valuable treatment option for patients with ALK-positive advanced NSCLC who have already received crizotinib therapy.
Collapse
|
260
|
GCC2-ALK as a targetable fusion in lung adenocarcinoma and its enduring clinical responses to ALK inhibitors. Lung Cancer 2017; 115:5-11. [PMID: 29290262 DOI: 10.1016/j.lungcan.2017.10.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/15/2017] [Accepted: 10/26/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVES ALK, RET and ROS1 fusions have been identified as treatable targets in 5%-15% of non-small-cell lung cancers, and thanks to the advanced sequencing technologies, their new partner genes have been steadily detected. Here we identified a rare fusion of ALK (GCC2-ALK) in a patient with advanced lung adenocarcinoma and monitored the treatment efficacy of ALK inhibitors on this patient. We further performed in vitro functional studies of this fusion protein for evaluating its oncogenic potential. MATERIALS AND METHODS The GCC2-ALK fusion gene was identified by targeted next generation sequencing (NGS) from the tumor DNA samples, and its fusion product was confirmed by Sanger sequencing the cDNA product. The functional study of GCC2-ALK was performed in Ba/F3 cells with cell proliferation and viability assays. The activation of downstream signaling pathways of ALK and their responses to crizotinib inhibition were studied in HEK-293 and 293T cells with ectopic expression of GCC2-ALK. In parallel, disease progression in the patient was monitored by computed tomography scanning and targeted NGS of either liquid or tissue biopsy samples throughout and after crizotinib treatment. RESULTS Similarly to EML4-ALK, the GCC2-ALK fusion protein promotes IL-3-independent growth of Ba/F3 cells. Ectopic expression of GCC2-ALK leads to hyper-activation of ALK downstream signaling that can be inhibited by crizotinib. Crizotinib treatment of the patient resulted in 18 months of progression free survival without any trace of GCC2-ALK fusion in the liquid biopsies. Re-biopsy of a lung lesion at progression identified the re-occurrence of GCC2-ALK. The patient was then administrated with a second-generation ALK inhibitor, ceritinib, and received partial response until the last follow-up. CONCLUSION We identified and functionally validated GCC2-ALK as a constitutively activated fusion in NSCLC. The patient was benefited from crizotinib treatment initially and then ceritinib after progression, suggesting GCC2-ALK as a novel therapeutic target for ALK inhibitors.
Collapse
|
261
|
Abstract
OPINION STATEMENT Major therapeutic advances have occurred over the last several years in the management of advanced ALK+ NSCLC patients. Crizotinib was the first agent approved for the management of ALK+ NSCLC patients after it demonstrated significantly greater clinical benefit compared to chemotherapy. Several next generation ALK inhibitors have demonstrated clinical benefit in patients with crizotinib refractory NSCLC patients including in the CNS. Based on available data, therapy with a next generation ALK inhibitor can be initiated following therapy with crizotinib without any assessment of the molecular mechanisms of resistance. The appropriate therapy for patients with progressive disease following two ALK inhibitors is not well defined. In patients with an ALK-resistant mutation in their tumor, an ALK inhibitor with activity against the mutation would be the most appropriate therapy. In others, chemotherapy and PD-1 directed agents can be considered. Clinical data suggests that ALK+ patients are less likely to benefit from PD-1 directed agents and therefore chemotherapy should be considered prior to these agents for the management of ALK+ NSCLC patients.
Collapse
Affiliation(s)
- Shirish M Gadgeel
- Department of Oncology, Karmanos Cancer Institute/Wayne State University, 4100 John R, 4HWCRC, Detroit, MI, 48201, USA.
| |
Collapse
|
262
|
Sánchez NS, Mills GB, Mills Shaw KR. Precision oncology: neither a silver bullet nor a dream. Pharmacogenomics 2017; 18:1525-1539. [PMID: 29061079 DOI: 10.2217/pgs-2017-0094] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Precision oncology is not an illusion, nor is it the magic bullet that will eradicate all cancers. Precision oncology is simply another weapon in our growing armament against cancer. Rather than honing in on the failures of a relatively young field, one should advocate for integrating its successes into widespread clinical practice, especially for indications, such as: ABL, ALK, BRAF, BRCA1, BRCA2, EGFR, KIT, KRAS, PDGFRA, PDGFRB, ROS1, BCR-ABL, FLT3 and ROS1, where aberrations have been shown to alter responses to US FDA approved drugs - that is, level 1 data. Moreover, to truly assess the promise of precision oncology, we must first begin by defining our expectations for this field. Importantly, we must recognize that the conception of precision oncology arose as an antithesis of the 'one-size fits all' cancer therapeutics approach. Consequently, tools used for evaluating these conventional, large-scale trials, are not directly transferable for assessing nonconventional, smaller-scale trials needed for evaluating precision oncology. Hence, a thorough vetting of precision oncology as another tool of the trade, must first begin by reassessing our expectations for this field, as well as current clinical trial designs and end point measurements. Importantly, we must recognize that most targeted therapy approaches are in their infancy, with only monotherapy approaches being assessed and combination therapies likely being necessary to fulfill the promise of precision oncology.
Collapse
Affiliation(s)
- Nora S Sánchez
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gordon B Mills
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kenna R Mills Shaw
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
263
|
Ichiki M, Wataya H, Yamada K, Tsuruta N, Takeoka H, Okayama Y, Sasaki J, Hoshino T. Preventive effect of kampo medicine (hangeshashin-to, TJ-14) plus minocycline against afatinib-induced diarrhea and skin rash in patients with non-small cell lung cancer. Onco Targets Ther 2017; 10:5107-5113. [PMID: 29123409 PMCID: PMC5661491 DOI: 10.2147/ott.s145613] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Purpose Diarrhea and oral mucositis induced by afatinib can cause devastating quality of life issues for patients undergoing afatinib treatment. Several studies have shown that hangeshashin-to (TJ-14) might be useful for chemotherapy-induced diarrhea and oral mucositis. In this study, we investigated the prophylactic effects of TJ-14 for afatinib-induced diarrhea and oral mucositis and minocycline for afatinib-induced skin rash. Patients and methods First- and second-generation epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors have become the standard first-line treatment in patients with EGFR-mutated non-small cell lung cancer. The incidence of diarrhea was higher with afatinib than with gefitinib, and we conducted a single-arm Phase II study with afatinib. Patients who had previously undergone treatment with afatinib were ineligible. Both TJ-14 (7.5 g/day) and minocycline (100 mg/day) were administered simultaneously from the start of afatinib administration. The primary end point was the incidence of ≥ grade 3 (G3) diarrhea (increase of ≥7 stools/day over baseline) during the first 4 weeks of treatment. The secondary end points were the incidence of ≥ G3 oral mucositis (severe pain interfering with oral intake) and $ G3 skin toxicity (severe or medically significant but not immediately life-threatening). Results A total of 29 patients (nine men and 20 women; median age, 66 years; performance status, 0/1/2: 18/10/1) were enrolled from four centers. Four patients had undergone prior treatment with chemotherapy, including gefitinib or erlotinib. In all, 20 (68.9%) patients and one (3.4%) patient had diarrhea of any grade and ≥ G3, respectively. One (3.4%) patient had ≥ G3 oral mucositis; no patients had ≥ G3 skin rash. A total of 18 (62%) of the 29 patients achieved a partial response. Conclusion The present study indicated a trend in which TJ-14 reduced the risk of afatinib-induced diarrhea and minocycline reduced the risk of afatinib-induced skin rash.
Collapse
Affiliation(s)
- Masao Ichiki
- Department of Respiratory Medicine, Clinical Research Institute, National Hospital Organization, Kyushu Medical Center
| | - Hiroshi Wataya
- Division of Internal Medicine, Saiseikai Fukuoka General Hospital, Fukuoka City
| | - Kazuhiko Yamada
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University, Kurume City
| | - Nobuko Tsuruta
- Department of Respiratory Medicine, Hamanomachi Hospital, Fukuoka City, Fukuoka, Japan
| | - Hiroaki Takeoka
- Department of Respiratory Medicine, Clinical Research Institute, National Hospital Organization, Kyushu Medical Center
| | - Yusuke Okayama
- Department of Respiratory Medicine, Clinical Research Institute, National Hospital Organization, Kyushu Medical Center
| | - Jun Sasaki
- Department of Respiratory Medicine, Clinical Research Institute, National Hospital Organization, Kyushu Medical Center
| | - Tomoaki Hoshino
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University, Kurume City
| |
Collapse
|
264
|
Jain RK, Chen H. Spotlight on brigatinib and its potential in the treatment of patients with metastatic ALK-positive non-small cell lung cancer who are resistant or intolerant to crizotinib. LUNG CANCER (AUCKLAND, N.Z.) 2017; 8:169-177. [PMID: 29075144 PMCID: PMC5648304 DOI: 10.2147/lctt.s126507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In the last decade, there have been major therapeutic advances in the management of patients with anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer. Crizotinib was the first approved ALK inhibitor with significant benefits over chemotherapy. However, patients inevitably develop disease progression especially in central nervous system and acquire resistance to crizotinib. Several next-generation ALK inhibitors have been developed to overcome these resistance mechanisms and have demonstrated clinical benefits in crizotinib-refractory non-small cell lung cancer including in central nervous system. Brigatinib is a second-generation ALK inhibitor with high level of activity against ALK and several other targets. It is active in vitro against many ALK kinase domain mutations including L1196M, E1210K, and G1202R which may mediate acquired resistance to other ALK inhibitors. In Phase I/II and ALTA clinical studies, brigatinib has demonstrated substantial and durable responses and intracranial responses after progression on crizotinib. It has acceptable safety profile, but early pulmonary toxicity has been observed prompting to pursue daily dosing of 180 mg (with lead-in). Overall, 180 mg (with lead-in) has showed consistently better efficacy than 90 mg. In this review, we will discuss in detail these two pivotal trials that led to the accelerated approval for brigatinib and its future directions.
Collapse
Affiliation(s)
- Rohit K Jain
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Hongbin Chen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
265
|
Overall survival with crizotinib and next-generation ALK inhibitors in ALK-positive non-small-cell lung cancer (IFCT-1302 CLINALK): a French nationwide cohort retrospective study. Oncotarget 2017; 8:21903-21917. [PMID: 28423535 PMCID: PMC5400633 DOI: 10.18632/oncotarget.15746] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/10/2017] [Indexed: 11/25/2022] Open
Abstract
Overall survival (OS) with the anaplastic lymphoma kinase (ALK) inhibitor (ALKi) crizotinib in a large population of unselected patients with ALK-positive non-small-cell lung cancer (NSCLC) is not documented. We sought to assess OS with crizotinib in unselected ALK-positive NSCLC patients and whether post-progression systemic treatments affect survival outcomes.
Collapse
|
266
|
Affiliation(s)
- Fengying Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Sai-Hong Ignatius Ou
- Department of Medicine, Division of Hematology Oncology, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA, USA
| |
Collapse
|
267
|
Thress KS, Jacobs V, Angell HK, Yang JCH, Sequist LV, Blackhall F, Su WC, Schuler M, Wolf J, Gold KA, Cantarini M, Barrett JC, Jänne PA. Modulation of Biomarker Expression by Osimertinib: Results of the Paired Tumor Biopsy Cohorts of the AURA Phase I Trial. J Thorac Oncol 2017; 12:1588-1594. [PMID: 28751247 DOI: 10.1016/j.jtho.2017.07.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/07/2017] [Accepted: 07/12/2017] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Osimertinib is an oral, potent, irreversible EGFR tyrosine kinase inhibitor (TKI) selective for EGFR TKI and T790M resistance mutations. To enhance understanding of osimertinib's mechanism of action, we aimed to evaluate the modulation of key molecular biomarkers after osimertinib treatment in paired clinical samples from the phase I AURA trial. METHODS Paired tumor biopsy samples were collected before the study and after 15 plus or minus 7 days of osimertinib treatment (80 or 160 mg daily). Clinical efficacy outcomes were assessed according to whether viable paired biopsy samples could be collected; safety was also assessed. Immunohistochemical analyses assessed key pathway and tumor/immune-relevant markers (phospho-EGFR, phospho-S6, phospho-AKT, programmed death ligand 1, and CD8), with samples scored by image analysis or a pathologist blinded to treatment allocation. RESULTS Predose tumor biopsy samples were collected from 61 patients with EGFR T790M tumors; 29 patients had no viable postdose biopsy sample because of tumor regression or insufficient tumor sample. Evaluable predose and postdose tumor biopsy samples were collected from 24 patients. Objective response rate (ORR) and median progression-free survival (mPFS) were improved in patients from whom a postdose biopsy sample could not be collected (ORR 62% and mPFS 9.7 months [p = 0.027]) compared with those from whom paired samples were collected (ORR 29% and mPFS 6.6 months). Osimertinib modulated key EGFR signaling pathways and led to increased immune cell infiltration. CONCLUSIONS Collection of paired biopsy samples was challenging because of rapid tumor regression after osimertinib treatment, highlighting the difficulties of performing on-study biopsies in patients treated with highly active drugs.
Collapse
Affiliation(s)
- Kenneth S Thress
- IMED Oncology Translational Sciences, AstraZeneca, Gatehouse Park, Waltham, MA
| | - Vivien Jacobs
- IMED Oncology Translational Sciences, AstraZeneca, Alderley Park, Macclesfield, United Kingdom
| | - Helen K Angell
- IMED Oncology Translational Sciences, AstraZeneca, Cambridge, United Kingdom
| | - James Chih-Hsin Yang
- Department of Oncology, National Taiwan University Hospital, Taipei, Republic of China
| | - Lecia V Sequist
- Department of Hematology, Massachusetts General Hospital, Boston, Massachusetts; Department of Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Fiona Blackhall
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, United Kingdom; Department of Medical Oncology, The Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Wu-Chou Su
- National Cheng Kung University Hospital, Tainan, Republic of China
| | - Martin Schuler
- Clinic for Internal Medicine (Tumor Research), West German Cancer Center, University Duisburg-Essen and German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Jürgen Wolf
- Clinic for Internal Medicine, Uniklinik Köln, Köln, Germany
| | - Kathryn A Gold
- Thoracic Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | | | - J Carl Barrett
- IMED Oncology Translational Sciences, AstraZeneca, Gatehouse Park, Waltham, MA
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
268
|
Mayekar MK, Bivona TG. Current Landscape of Targeted Therapy in Lung Cancer. Clin Pharmacol Ther 2017; 102:757-764. [PMID: 28786099 DOI: 10.1002/cpt.810] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/01/2017] [Accepted: 08/01/2017] [Indexed: 12/14/2022]
Abstract
Lung cancer is the leading cause of cancer mortality worldwide. Comprehensive genomic profiling of lung cancers revealed their genetic heterogeneity and complexity and identified numerous targetable oncogenic driver alterations. These molecular profiling efforts have made it possible to exploit the potential of molecularly targeted therapies. Selection of patients for targeted therapies is becoming biomarker-driven, where the oncogenic drivers in patient tumors are first identified, and subsequently patients bearing drug-sensitizing genetic aberrations are matched to the appropriate targeted therapy. Success of this design of clinical trials and practice was first demonstrated in EGFR inhibitor trials in lung cancer and has since been incorporated into subsequent targeted therapy trials including ALK-, ROS1-, and BRAF V600E-targeted therapies. In this review we discuss the current landscape of clinically approved and other promising molecularly targeted approaches for the treatment of lung cancers, the challenges with these approaches, and the strategies that could be deployed to overcome these challenges.
Collapse
Affiliation(s)
- Manasi K Mayekar
- Department of Medicine, University of California, San Francisco, San Francisco, USA.,Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, USA
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, USA.,Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, USA
| |
Collapse
|
269
|
Cho BC, Kim DW, Bearz A, Laurie SA, McKeage M, Borra G, Park K, Kim SW, Ghosn M, Ardizzoni A, Maiello E, Greystoke A, Yu R, Osborne K, Gu W, Scott JW, Passos VQ, Lau YY, Wrona A. ASCEND-8: A Randomized Phase 1 Study of Ceritinib, 450 mg or 600 mg, Taken with a Low-Fat Meal versus 750 mg in Fasted State in Patients with Anaplastic Lymphoma Kinase (ALK)-Rearranged Metastatic Non–Small Cell Lung Cancer (NSCLC). J Thorac Oncol 2017; 12:1357-1367. [DOI: 10.1016/j.jtho.2017.07.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/11/2017] [Accepted: 07/11/2017] [Indexed: 12/22/2022]
|
270
|
Attarian S, Rahman N, Halmos B. Emerging uses of biomarkers in lung cancer management: molecular mechanisms of resistance. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:377. [PMID: 29057237 DOI: 10.21037/atm.2017.07.18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Management of patients with advanced non-small cell lung cancer (NSCLC) has recently been transformed by molecularly targeted and immunotherapeutic agents. In patients with EGFR/ALK/ROS mutated NSCLC, first line molecular therapy is the standard of care. Moreover, immune checkpoint inhibitors are revolutionary treatment options for advanced NSCLC and are now the standard of care in front-line or later line settings. Both classes of agents have led to improved patient outcomes, however, primary resistance and development of acquired resistance to both targeted and immunotherapeutic agents is commonly observed, limiting the use of these agents in clinical settings. In this review, we will discuss the most recent advances in understanding the mechanisms of primary and acquired resistance, progress in the spectrum of assays detecting causative molecular events and the development of new generations of inhibitors to overcome acquired resistance.
Collapse
Affiliation(s)
- Shirin Attarian
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Numa Rahman
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Balazs Halmos
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
271
|
Kobayashi T, Fujimoto H, D'Alessandro-Gabazza C, Gabazza EC, Hataji O. Recent studies move closer to answering questions about sequential therapy for anaplastic lymphoma kinase-rearranged non-small cell lung cancer. J Thorac Dis 2017; 9:2847-2851. [PMID: 29221254 DOI: 10.21037/jtd.2017.08.114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Tetsu Kobayashi
- Department of Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Tsu, Japan
| | - Hajime Fujimoto
- Department of Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Tsu, Japan
| | | | - Esteban C Gabazza
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Tsu, Japan
| | - Osamu Hataji
- Respiratory Center, Matsusaka Municipal Hospital, Matsusaka, Japan
| |
Collapse
|
272
|
West H. Prioritizing molecular markers to test for in the initial workup of advanced non-small cell lung cancer: wants versus needs. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:371. [PMID: 29057231 DOI: 10.21037/atm.2017.08.29] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The current standard of care for molecular marker testing in patients with advanced non-small cell lung cancer (NSCLC) has been evolving over several years and is a product of the quality of the evidence supporting a targeted therapy for a specific molecular marker, the pre-test probability of that marker in the population, and the magnitude of benefit seen with that treatment. Among the markers that have one or more matched targeted therapies, only a few are in the subset for which they should be considered as most clearly worthy of prioritizing to detect in the first line setting in order to have them supplant other first line alternatives, and in only a subset of patients, as defined currently by NSCLC histology. Specifically, this currently includes testing for an activating epidermal growth factor receptor (EGFR) mutation or an anaplastic lymphoma kinase (ALK) or ROS1 rearrangement. This article reviews the history and data supporting the prioritization of these markers in patients with non-squamous NSCLC, a histologically selected population in whom the probability of these markers combined with the anticipated efficacy of targeted therapies against them is high enough to favor these treatments in the first line setting. In reviewing the evidence supporting this very limited core subset of most valuable molecular markers to detect in the initial workup of such patients, we can also see the criteria by which other actionable markers need to reach in order to be widely recognized as reliably valuable enough to warrant prioritization to detect in the initial workup of advanced NSCLC as well.
Collapse
Affiliation(s)
- Howard West
- Medical Oncology, Swedish Cancer Institute, Seattle, WA, USA
| |
Collapse
|
273
|
Gainor JF, Shaw AT. Fast, Food and Ceritinib in ALK-Positive NSCLC. J Thorac Oncol 2017; 12:1341-1343. [PMID: 28838710 DOI: 10.1016/j.jtho.2017.07.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 07/19/2017] [Indexed: 11/18/2022]
Affiliation(s)
- Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston.
| | - Alice T Shaw
- Department of Medicine, Massachusetts General Hospital, Boston
| |
Collapse
|
274
|
Nayar G, Ejikeme T, Chongsathidkiet P, Elsamadicy AA, Blackwell KL, Clarke JM, Lad SP, Fecci PE. Leptomeningeal disease: current diagnostic and therapeutic strategies. Oncotarget 2017; 8:73312-73328. [PMID: 29069871 PMCID: PMC5641214 DOI: 10.18632/oncotarget.20272] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/20/2017] [Indexed: 12/28/2022] Open
Abstract
Leptomeningeal disease has become increasingly prevalent as novel therapeutic interventions extend the survival of cancer patients. Although a majority of leptomeningeal spread occurs secondary to breast cancer, lung cancer, and melanoma, a wide variety of malignancies have been reported as primary sources. Symptoms on presentation are equally diverse, often involving a combination of neurological deficits with the possibility of obstructive hydrocephalus. Diagnosis is definitively made via cerebrospinal fluid cytology for malignant cells, but neuro-imaging with high quality T1-weighted magnetic resonance imaging can aid diagnosis and localization. While leptomeningeal disease is still a terminal, late-stage complication, a variety of treatment modalities, such as intrathecal chemotherapeutics and radiation therapy, have improved median survival from 4–6 weeks to 3–6 months. Positive prognosticative factors for survival include younger age, high performance scores, and controlled systemic disease. In looking to the future, diagnostics that improve early detection and chemotherapeutics tailored to the primary malignancy will likely be the most significant advances in improving survival.
Collapse
Affiliation(s)
- Gautam Nayar
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA.,Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Tiffany Ejikeme
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA.,Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Pakawat Chongsathidkiet
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA.,Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Aladine A Elsamadicy
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA.,Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Kimberly L Blackwell
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey M Clarke
- Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
| | - Shivanand P Lad
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Peter E Fecci
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA.,Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.,Department of Pathology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
275
|
Abstract
目的 观察微信平台管理对肺癌化疗患者主要照顾者消化系统和照顾负担评分效果分析.
方法 选取80例肺癌化疗患者主要照顾者作为研究对象, 按照随机数字表法分为对照组和观察组各40例, 对照组给予心理护理、饮食护理、健康宣教和减轻思想负担等护理. 观察组在对照组基础上给予微信平台管理护理. 观察2组消化系统症状评分和照顾负担评分变化情况.
结果 观察组腹胀、早饱、嗳气、恶心、食欲评分均比对照组降低, 差异均有统计学意义(P<0.05). 干预前2组主要照顾者负担量表评分比较, 差异无统计学意义(P>0.05). 经过干预后2组评分均下降, 与干预前比较差异有统计学意义(P<0.05); 且观察组干预后得分明显低于对照组, 差异有统计学意义(P<0.05).
结论 微信平台管理能降低肺癌化疗患者主要照顾者消化系统评分和照顾负担评分.
Collapse
|
276
|
Efficacy of alectinib in central nervous system metastases in crizotinib-resistant ALK-positive non-small-cell lung cancer: Comparison of RECIST 1.1 and RANO-HGG criteria. Eur J Cancer 2017. [PMID: 28646771 DOI: 10.1016/j.ejca.2017.05.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND Central nervous system (CNS) progression is common in patients with anaplastic lymphoma kinase-positive (ALK+) non-small-cell lung cancer (NSCLC) receiving crizotinib. Next-generation ALK inhibitors have shown activity against CNS metastases, but accurate assessment of response and progression is vital. Data from two phase II studies in crizotinib-refractory ALK+ NSCLC were pooled to examine the CNS efficacy of alectinib, a CNS-active ALK inhibitor, using Response Evaluation Criteria in Solid Tumours (RECIST version 1.1) and Response Assessment in Neuro-Oncology high-grade glioma (RANO-HGG) criteria. METHODS Both studies enrolled patients aged ≥18 years who had previously received crizotinib. NP28761 was conducted in North America and NP28673 was a global study. All patients received 600 mg oral alectinib twice daily and had baseline CNS imaging. CNS response for those with baseline CNS metastases was determined by an independent review committee. RESULTS Baseline measurable CNS disease was identified in 50 patients by RECIST and 43 by RANO-HGG. CNS objective response rate was 64.0% by RECIST (95% confidence interval [CI]: 49.2-77.1; 11 CNS complete responses [CCRs]) and 53.5% by RANO-HGG (95% CI: 37.7-68.8; eight CCRs). CNS responses were durable, with consistent estimates of median duration of 10.8 months with RECIST and 11.1 months with RANO-HGG. Of the 39 patients with measurable CNS disease by both RECIST and RANO-HGG, only three (8%) had CNS progression according to one criteria but not the other (92% concordance rate). CONCLUSION Alectinib demonstrated promising efficacy in the CNS for ALK+ NSCLC patients pretreated with crizotinib, regardless of the assessment criteria used.
Collapse
|
277
|
Pooled Systemic Efficacy and Safety Data from the Pivotal Phase II Studies (NP28673 and NP28761) of Alectinib in ALK-positive Non-Small Cell Lung Cancer. J Thorac Oncol 2017; 12:1552-1560. [PMID: 28689043 PMCID: PMC6886236 DOI: 10.1016/j.jtho.2017.06.070] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/27/2017] [Accepted: 06/29/2017] [Indexed: 11/23/2022]
Abstract
Introduction: Alectinib demonstrated clinical efficacy and an acceptable safety profile in two phase II studies (NP28761 and NP28673). Here we report the pooled efficacy and safety data after 15 and 18 months more follow-up than in the respective primary analyses. Methods: Enrolled patients had ALK receptor tyrosine kinase gene (ALK)-positive NSCLC and had progressed while taking, or could not tolerate, crizotinib. Patients received oral alectinib, 600 mg twice daily. The primary end point in both studies was objective response rate assessed by an independent review committee (IRC) using the Response Evaluation Criteria in Solid Tumors, version 1.1. Secondary end points included disease control rate, duration of response, progression-free survival, overall survival, and safety. Results: The pooled data set included 225 patients (n = 138 in NP28673 and n = 87 in NP28761). The response-evaluable population included189 patients (84% [n = 122 in NP28673 and n = 67 in NP28761]). In the response-evaluable population, objective response rate as assessed by the IRC was 51.3% (95% confidence interval [CI]: 44.0–58.6 [all PRs]), the disease control rate was 78.8% (95% CI: 72.3–84.4), and the median duration of response was 14.9 months (95% CI: 11.1–20.4) after 58% of events. Median progression-free survival as assessed by the IRC was 8.3 months (95% CI: 7.0–11.3) and median overall survival was 26.0 months (95% CI: 21.4–not estimable). Grade 3 or higher adverse events (AEs) occurred in 40% of patients, 6% of patients had treatment withdrawn on account of AEs, and 33% had AEs leading to dose interruptions/modification. Conclusions: This pooled data analysis confirmed the robust systemic efficacy of alectinib in ALK-positive NSCLC with a durable response rate. Alectinib also had an acceptable safety profile with a longer duration of follow-up.
Collapse
|
278
|
Santarpia M, Daffinà MG, D’Aveni A, Marabello G, Liguori A, Giovannetti E, Karachaliou N, Gonzalez Cao M, Rosell R, Altavilla G. Spotlight on ceritinib in the treatment of ALK+ NSCLC: design, development and place in therapy. Drug Des Devel Ther 2017; 11:2047-2063. [PMID: 28740365 PMCID: PMC5503498 DOI: 10.2147/dddt.s113500] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The identification of echinoderm microtubule-associated protein-like 4 (EML4) and anaplastic lymphoma kinase (ALK) fusion gene in non-small cell lung cancer (NSCLC) has radically changed the treatment of a subset of patients harboring this oncogenic driver. Crizotinib was the first ALK tyrosine kinase inhibitor to receive fast approval and is currently indicated as the first-line therapy for advanced, ALK-positive NSCLC patients. However, despite crizotinib's efficacy, patients almost invariably progress, with the central nervous system being one of the most common sites of relapse. Different mechanisms of acquired resistance have been identified, including secondary ALK mutations, ALK copy number alterations and activation of bypass tracks. Different highly potent and brain-penetrant next-generation ALK inhibitors have been developed and tested in NSCLC patients with ALK rearrangements. Ceritinib, a structurally distinct and selective ALK inhibitor, showed 20 times higher potency than crizotinib in inhibiting ALK and had activity against the most common crizotinib-resistant mutations, including L1196M and G1269A, in preclinical models. In Phase I and II studies, ceritinib demonstrated pronounced activity in both crizotinib-naïve and crizotinib-refractory patients, with responses observed regardless of the presence of ALK resistance mutations. Ceritinib was the first ALK inhibitor to be approved for the treatment of crizotinib-refractory, ALK-rearranged NSCLC, and recent results from a Phase III study have demonstrated superior efficacy compared to standard chemotherapy in the first- and second-line setting. We provide an extensive overview of ceritinib from the design of the compound through preclinical data until efficacy and toxicity results from Phase I-III clinical studies. We review the molecular alterations associated with resistance to ceritinib and highlight the importance of obtaining tumor biopsy at progression to tailor therapy based upon the underlying resistance mechanism. We finally provide an outlook on novel rational therapeutic combinations.
Collapse
Affiliation(s)
- Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Maria Grazia Daffinà
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Alessandro D’Aveni
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Grazia Marabello
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Alessia Liguori
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Nanoscience and Nanotechnologies, CNR-Nano, Institute of Nanoscience and Nanotechnology
- Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Niki Karachaliou
- Institute of Oncology Rosell (IOR), University Hospital Sagrat Cor
| | - Maria Gonzalez Cao
- Oncology Department, Institute of Oncology Rosell (IOR), Quirón-Dexeus University Institute, Barcelona
| | - Rafael Rosell
- Cancer Biology and Precision Medicine Program, Germans Trias i Pujol Research Institute
- Catalan Institute of Oncology, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Giuseppe Altavilla
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| |
Collapse
|
279
|
Bendaly E, Dalal AA, Culver K, Galebach P, Bocharova I, Foster R, Sasane M, Macalalad AR, Guérin A. Monitoring for and Characterizing Crizotinib Progression: A Chart Review of ALK-Positive Non-Small Cell Lung Cancer Patients. Adv Ther 2017; 34:1673-1685. [PMID: 28578501 DOI: 10.1007/s12325-017-0551-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Crizotinib is recommended as first-line therapy for ALK-positive non-small cell lung cancer (NSCLC), but within a year of treatment initiation many patients develop resistance. With the recent approval of second-generation ALK inhibitors, this study assessed how physicians monitor for and diagnose progression and how they alter treatment following progression on crizotinib. METHODS A panel of oncologists from the United States were surveyed regarding their monitoring practices and criteria for diagnosing progression on crizotinib. The physicians also retrospectively provided data (March-June 2016) from the medical charts of their adult patients with locally advanced or metastatic ALK-positive NSCLC who progressed on crizotinib after the approval (April 2014) of the first second-generation ALK inhibitor, ceritinib. RESULTS A total of 28 physicians responded to the survey. Data was abstracted on 74 patients. In the physician survey, most physicians (71%) reported monitoring for radiographic progression every 3-4 months. When new lesions were detected, physician response varied. Following a symptomatic isolated lesion, most physicians (75%) would add local therapy and resume crizotinib. Following multiple symptomatic lesions, 96% and 64% of physicians would switch to a new therapy depending on whether the lesions were extracranial or isolated to the brain, respectively. For the patient cohort, physician-defined progression on crizotinib was diagnosed after a median of 10 months, and within 30 days of diagnosis, 86% of patients discontinued crizotinib. Among all patients who discontinued crizotinib, 77% switched to ceritinib, 14% to chemotherapy, and 1% to alectinib. The remaining 7% did not receive additional systemic antineoplastic therapy. CONCLUSION The findings from this physician survey and retrospective chart review study suggest that physician response to the development of new lesions in crizotinib-treated ALK-positive NSCLC patients varies with location and extent of the lesions. Once patients were considered to have progressed, most of them were immediately switched to ceritinib. FUNDING Novartis Pharmaceuticals Corporation.
Collapse
Affiliation(s)
- Edmond Bendaly
- Marion General Hospital, Medical Oncology, 831 N Theatre Rd, Marion, IN, USA
| | - Anand A Dalal
- Novartis Pharmaceutical Corporation, 1 Health Plaza, East Hanover, NJ, USA
| | - Kenneth Culver
- Novartis Pharmaceutical Corporation, 1 Health Plaza, East Hanover, NJ, USA
| | | | | | - Rebekah Foster
- Analysis Group, Inc., 111 Huntington Ave, Boston, MA, USA
| | - Medha Sasane
- Novartis Pharmaceutical Corporation, 1 Health Plaza, East Hanover, NJ, USA
| | | | - Annie Guérin
- Analysis Group, Inc., 111 Huntington Ave, Boston, MA, USA.
| |
Collapse
|
280
|
Carlson JJ, Canestaro W, Ravelo A, Wong W. The cost-effectiveness of alectinib in anaplastic lymphoma kinase-positive (ALK+) advanced NSCLC previously treated with crizotinib. J Med Econ 2017; 20:671-677. [PMID: 28332433 DOI: 10.1080/13696998.2017.1302453] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Introduction Anaplastic lymphoma kinase (ALK) targeting drugs provide an important option for advanced non-small cell lung cancer patients with this distinct tumor type; however, there is considerable uncertainty as to which drug provides the optimal value after crizotinib treatment. This study estimated the cost-utility of alectinib vs ceritinib from a US payer perspective. Methods A cost-utility model was developed using partition survival methods and three health states: progression-free (PF), post-progression (PP), and death. Survival data were derived from the key clinical trials (alectinib: NP28761 & NP28673, ceritinib: ASCEND I and II). Costs included drugs, adverse events, and supportive care. Utilities were based on trial data and the literature. One-way and probabilistic sensitivity analyses (PSA) were performed to assess parameter uncertainty. Results Treatment with alectinib vs ceritinib resulted in increases of 2.55 months in the PF state, 0.44 quality adjusted life-years (QALYs), and $13,868, yielding a mean cost/QALY of $31,180. In the PSA, alectinib had a 96% probability of being cost-effective at a willingness-to-pay of $100,000/QALY. Drivers of model results were drug costs and utilities in the PF health state. The ICER ranged from $10,600-$65,000 per QALY in scenario analyses, including a sub-group analysis limited to patients with prior chemotherapy and crizotinib treatment. Conclusions Treatment with alectinib in ALK + crizotinib-treated patients increased time progression-free and QALYs vs ceritinib. The marginal cost increase was driven by longer treatment durations with alectinib. This model demonstrates that alectinib may be considered a cost-effective treatment after progression on crizotinib.
Collapse
Affiliation(s)
- J J Carlson
- a University of Washington , Seattle , WA , USA
| | - W Canestaro
- a University of Washington , Seattle , WA , USA
| | - A Ravelo
- b Genentech, Inc. , South San Francisco , CA , USA
| | - W Wong
- b Genentech, Inc. , South San Francisco , CA , USA
| |
Collapse
|
281
|
The between Now and Then of Lung Cancer Chemotherapy and Immunotherapy. Int J Mol Sci 2017; 18:ijms18071374. [PMID: 28653990 PMCID: PMC5535867 DOI: 10.3390/ijms18071374] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 06/23/2017] [Accepted: 06/25/2017] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is the most common cancer worldwide. Disappointingly, despite great effort in encouraging screening or, at least, a close surveillance of high-risk individuals, most of lung cancers are diagnosed when already surgically unresectable because of local advancement or metastasis. In these cases, the treatment of choice is chemotherapy, alone or in combination with radiotherapy. Here, we will briefly review the most successful and recent advances in the identification of novel lung cancer genetic lesions and in the development of new drugs specifically targeting them. However, lung cancer is still the leading cause of cancer-related mortality also because, despite impressive initial responses, the patients often develop resistance to novel target therapies after a few months of treatment. Thus, it is literally vital to continue the search for new therapeutic options. So, here, on the basis of our recent findings on the role of the tumor suppressor CCDC6 protein in lung tumorigenesis, we will also discuss novel therapeutic approaches we envision for lung cancer.
Collapse
|
282
|
Sekine I. What is the optimal first-line treatment for advanced anaplastic lymphoma kinase-rearranged non-small cell lung cancer? J Thorac Dis 2017; 9:E447-E450. [PMID: 28616306 DOI: 10.21037/jtd.2017.04.43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ikuo Sekine
- Department of Medical Oncology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
283
|
Zhu Q, Hu H, Weng DS, Zhang XF, Chen CL, Zhou ZQ, Tang Y, Xia JC. Pooled safety analyses of ALK-TKI inhibitor in ALK-positive NSCLC. BMC Cancer 2017; 17:412. [PMID: 28606126 PMCID: PMC5469041 DOI: 10.1186/s12885-017-3405-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 06/07/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The anaplastic lymphoma kinase tyrosine kinase inhibitors (ALK-TKIs) have been administered to patients with ALK-positive non-small cell lung cancer for a long period of time and show a promising response. However, the differences in the toxicity profiles among these drugs are still unclear. METHODS We performed a comprehensive search of the MEDLINE, EMBASE, WEB OF SCIENCE and COCHRANE databases from the drugs' inception to May 2016 to identify clinical trials. Severe adverse events (AEs) (grade ≥ 3) based on the ALK-TKI type were analysed. RESULTS Seventeen trials published between 2011 and 2016, including a total of 1826 patients, were eligible for analysis. Patients in 10 trials (n = 1000) received crizotinib, patients in 5 trials (n = 601) received ceritinib and patients in 2 trials (n = 225) received alectinib. The overall frequencies of treatment-related death and AEs due to treatment withdrawal were 0.9% (12/1365) and 5.5% (85/1543), respectively. Moreover, the frequency of severe AEs in patients treated with ceritinib was significantly higher than patients treated with crizotinib or alectinib, especially for hepatotoxicity, fatigue and some of gastrointestinal symptoms. Additionally, significant difference in the elevated lipase and amylase levels (grade ≥ 3) were detected between ceritinib and crizotinib/alectinib, whereas neutropenia was less frequent. CONCLUSIONS ALK-TKIs were safe for ALK-positive patients. Moreover, statistically significant differences in some severe AEs among ceritinib, crizotinib and alectinib were detected in present study.
Collapse
Affiliation(s)
- Qian Zhu
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Hao Hu
- Department of Thoracic Surgery, Medical College of Nanchang University, Nanchang, 330006 People’s Republic of China
| | - De-Sheng Weng
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Xiao-Fei Zhang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Chang-Long Chen
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Zi-Qi Zhou
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Yan Tang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Jian-Chuan Xia
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| |
Collapse
|
284
|
Management of ceritinib therapy and adverse events in patients with ALK-rearranged non-small cell lung cancer. Lung Cancer 2017; 111:51-58. [PMID: 28838397 DOI: 10.1016/j.lungcan.2017.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/05/2017] [Accepted: 06/08/2017] [Indexed: 12/18/2022]
Abstract
Anaplastic lymphoma kinase rearrangement (ALK+) occurs in approximately 2-7% of patients with non-small cell lung cancer (NSCLC), contributing to a considerable number of patients with ALK+ NSCLC worldwide. Ceritinib is a next generation ALK inhibitor (ALKi), approved by the European Medicines Agency in 2015. In the first-in-human, phase I study, ceritinib demonstrated rapid and durable responses in ALK patients previously treated with a different ALKi and in those who were ALKi-naive. As ceritinib is starting to be used routinely for the treatment of patients with ALK+ NSCLC, experience is growing with regard to ideal therapy management. In this review we provide a brief background to the development of ceritinib. The optimal treatment management and adverse events associated with ceritinib in clinical trials and in clinical practice are then discussed in detail, and where applicable, an expert consensus on specific recommendations are made. In clinical trials, the most common adverse events related to ceritinib are nausea, vomiting, and diarrhea. However, the majority of these are mild and, in the opinion of the authors, can be effectively managed with dose modifications. Based on clinical data, ceritinib has demonstrated efficacy as a first-line therapy and in patients who have relapsed on crizotinib, including those with brain metastases at baseline. Unfortunately, at some point, all patients experience progressive disease, with the central nervous system being a common site of metastases. Recommendations are made for continuing treatment beyond disease progression as long as a clinical benefit to patients is observed. Here, we review management of ceritinib treatment and adverse events and make recommendations on optimal management of patients.
Collapse
|
285
|
Waqar SN, Morgensztern D, Govindan R. Systemic Treatment of Brain Metastases. Hematol Oncol Clin North Am 2017; 31:157-176. [PMID: 27912831 DOI: 10.1016/j.hoc.2016.08.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Lung cancer continues to be the leading cause of cancer-related mortality in the United States. Brain metastases are a significant problem in patients with lung cancer and have conventionally been treated with whole-brain radiation. This article reviews the data for systemic chemotherapy to treat brain metastasis from lung cancer and examines the activity of small molecule tyrosine kinase inhibitors for the targeted therapy for brain metastases from EGFR-mutant and ALK-rearranged non-small cell lung cancer. Future directions for evaluating the role of immunotherapy in treating brain metastasis are also discussed.
Collapse
Affiliation(s)
- Saiama N Waqar
- Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8056, St Louis, MO 63110, USA.
| | - Daniel Morgensztern
- Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8056, St Louis, MO 63110, USA
| | - Ramaswamy Govindan
- Section of Medical Oncology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8056, St Louis, MO 63110, USA
| |
Collapse
|
286
|
Shaw AT, Kim TM, Crinò L, Gridelli C, Kiura K, Liu G, Novello S, Bearz A, Gautschi O, Mok T, Nishio M, Scagliotti G, Spigel DR, Deudon S, Zheng C, Pantano S, Urban P, Massacesi C, Viraswami-Appanna K, Felip E. Ceritinib versus chemotherapy in patients with ALK-rearranged non-small-cell lung cancer previously given chemotherapy and crizotinib (ASCEND-5): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol 2017; 18:874-886. [PMID: 28602779 DOI: 10.1016/s1470-2045(17)30339-x] [Citation(s) in RCA: 427] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 11/27/2022]
Abstract
BACKGROUND Ceritinib is a next-generation anaplastic lymphoma kinase (ALK) inhibitor, which has shown robust anti-tumour efficacy, along with intracranial activity, in patients with ALK-rearranged non-small-cell lung cancer. In phase 1 and 2 studies, ceritinib has been shown to be highly active in both ALK inhibitor-naive and ALK inhibitor-pretreated patients who had progressed after chemotherapy (mostly multiple lines). In this study, we compared the efficacy and safety of ceritinib versus single-agent chemotherapy in patients with advanced ALK-rearranged non-small-cell lung cancer who had previously progressed following crizotinib and platinum-based doublet chemotherapy. METHODS In this randomised, controlled, open-label, phase 3 trial, we recruited patients aged at least 18 years with ALK-rearranged stage IIIB or IV non-small-cell lung cancer (with at least one measurable lesion) who had received previous chemotherapy (one or two lines, including a platinum doublet) and crizotinib and had subsequent disease progression, from 99 centres across 20 countries. Other inclusion criteria were a WHO performance status of 0-2, adequate organ function and laboratory test results, a life expectancy of at least 12 weeks, and having recovered from previous anticancer treatment-related toxicities. We randomly allocated patients (1:1; with blocking [block size of four]; stratified by WHO performance status [0 vs 1-2] and presence or absence of brain metastases) to oral ceritinib 750 mg per day fasted (in 21 day treatment cycles) or chemotherapy (intravenous pemetrexed 500 mg/m2 or docetaxel 75 mg/m2 [investigator choice], every 21 days). Patients who discontinued chemotherapy because of progressive disease could cross over to the ceritinib group. The primary endpoint was progression-free survival, assessed by a masked independent review committee using Response Evaluation Criteria in Solid Tumors 1.1 in the intention-to-treat population, assessed every 6 weeks until month 18 and every 9 weeks thereafter. This trial is registered with ClinicalTrials.gov, number NCT01828112, and is ongoing but no longer recruiting patients. FINDINGS Between June 28, 2013, and Nov 2, 2015, we randomly allocated 231 patients; 115 (50%) to ceritinib and 116 (50%) to chemotherapy (40 [34%] to pemetrexed, 73 [63%] to docetaxel, and three [3%] discontinued before receiving treatment). Median follow-up was 16·5 months (IQR 11·5-21·4). Ceritinib showed a significant improvement in median progression-free survival compared with chemotherapy (5·4 months [95% CI 4·1-6·9] for ceritinib vs 1·6 months [1·4-2·8] for chemotherapy; hazard ratio 0·49 [0·36-0·67]; p<0·0001). Serious adverse events were reported in 49 (43%) of 115 patients in the ceritinib group and 36 (32%) of 113 in the chemotherapy group. Treatment-related serious adverse events were similar between groups (13 [11%] in the ceritinib group vs 12 [11%] in the chemotherapy group). The most frequent grade 3-4 adverse events in the ceritinib group were increased alanine aminotransferase concentration (24 [21%] of 115 vs two [2%] of 113 in the chemotherapy group), increased γ glutamyltransferase concentration (24 [21%] vs one [1%]), and increased aspartate aminotransferase concentration (16 [14%] vs one [1%] in the chemotherapy group). Six (5%) of 115 patients in the ceritinib group discontinued because of adverse events compared with eight (7%) of 116 in the chemotherapy group. 15 (13%) of 115 patients in the ceritinib group and five (4%) of 113 in the chemotherapy group died during the treatment period (from the day of the first dose of study treatment to 30 days after the final dose). 13 (87%) of the 15 patients who died in the ceritinib group died because of disease progression and two (13%) died because of an adverse event (one [7%] cerebrovascular accident and one [7%] respiratory failure); neither of these deaths were considered by the investigator to be treatment related. The five (4%) deaths in the chemotherapy group were all due to disease progression. INTERPRETATION These findings show that patients derive significant clinical benefit from a more potent ALK inhibitor after failure of crizotinib, and establish ceritinib as a more efficacious treatment option compared with chemotherapy in this patient population. FUNDING Novartis Pharmaceuticals Corporation.
Collapse
Affiliation(s)
- Alice T Shaw
- Massachusetts General Hospital, Boston, MA, USA.
| | - Tae Min Kim
- Seoul National University Hospital, Seoul, Korea
| | - Lucio Crinò
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Cesare Gridelli
- SG Moscati Hospital, Città Ospedaliera, Contrada Amoretta, Avellino, Italy
| | | | - Geoffrey Liu
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | | | - Alessandra Bearz
- Istituto di Ricovero e Cura a Carattere Scientifico-Centro di Riferimento Oncologico, Aviano, Italy
| | | | - Tony Mok
- State Key Laboratory of South China, Chinese University of Hong Kong, Shatin, China
| | - Makoto Nishio
- Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | | | | | | | | | | | | | | | - Enriqueta Felip
- Vall d'Hebron University Hospital, Barcelona, Spain; Vall d'Hebron Institute of Oncology, Barcelona, Spain
| |
Collapse
|
287
|
Lin L, Zhao J, Kong N, He Y, Hu J, Huang F, Han J, Cao X. Meta-analysis of the incidence and risks of interstitial lung disease and QTc prolongation in non-small-cell lung cancer patients treated with ALK inhibitors. Oncotarget 2017; 8:57379-57385. [PMID: 28915678 PMCID: PMC5593649 DOI: 10.18632/oncotarget.18283] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 05/03/2017] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND To conduct a systematic review and meta-analysis to assess the overall incidence and risk of interstitial lung disease (ILD) and QTc prolongation associated with anaplastic lymphoma kinase (ALK)-tyrosine kinase inhibitors (-TKIs) in non-small-cell lung cancer (NSCLC) patients. RESULTS A total of 1,770 patients from 8 clinical trials were included. The incidences of high-grade ILD and QTc prolongation was 2.5% (95% CI 1.7-3.6%), and 2.8% (95% CI 1.8-4.3%), respectively. Meta-analysis demonstrated that the use of ALK-TKIs in NSCLC patients significantly increased the risk of developing high-grade ILD (Peto OR, 3.27, 95%CI: 1.18-9.08, p = 0.023) and QTc prolongation (Peto OR 7.51, 95% CI, 2.16-26.15; p = 0.002) in comparison with chemotherapy alone. MATERIALS AND METHODS A systematic literature search was performed to identify related citations up to January 31, 2017. Data were extracted, and summary incidence rates, Peto odds ratios (Peto ORs), and 95% confidence intervals (CIs) were calculated. CONCLUSIONS The use of ALK-TKIs significantly increases the risk of developing high-grade ILD and QTc prolongation in lung cancer patients. Clinicians should pay attention to the risks of severe ILD and QTc prolongation with the administration of these drugs.
Collapse
Affiliation(s)
- Liping Lin
- Department of Oncology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| | - Juanjuan Zhao
- School of Nursing, Sun Yat-sen University, Guangzhou, 510000, China
| | - Ning Kong
- Department of Ophthalmology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| | - Yan He
- Department of Oncology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| | - Jiazhu Hu
- Department of Oncology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| | - Fuxi Huang
- Department of Oncology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| | - Jianjun Han
- Department of Oncology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| | - Xiaolong Cao
- Department of Oncology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| |
Collapse
|
288
|
Lim SM, Kim HR, Lee JS, Lee KH, Lee YG, Min YJ, Cho EK, Lee SS, Kim BS, Choi MY, Shim HS, Chung JH, La Choi Y, Lee MJ, Kim M, Kim JH, Ali SM, Ahn MJ, Cho BC. Open-Label, Multicenter, Phase II Study of Ceritinib in Patients With Non-Small-Cell Lung Cancer Harboring ROS1 Rearrangement. J Clin Oncol 2017; 35:2613-2618. [PMID: 28520527 DOI: 10.1200/jco.2016.71.3701] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose ROS1 rearrangement is a distinct molecular subset of non-small-cell lung cancer (NSCLC). We investigated the efficacy and safety of ceritinib in patients with ROS1-rearranged NSCLC. Patients and Methods We enrolled 32 patients with advanced NSCLC who tested positive for ROS1 rearrangement by fluorescent in situ hybridization. Ceritinib 750 mg was administered once daily. The primary end point was objective response rate. The secondary end points were disease control rate; duration of response; progression-free survival; overall survival; toxicity; and concordance among fluorescent in situ hybridization, immunohistochemistry, and next-generation sequencing. Results Between June 7, 2013, and February 1, 2016, 404 patients underwent ROS1 prescreening, and 32 patients with ROS1 rearrangement were enrolled. All patients except two were crizotinib-naïve. At the time of data cutoff, the median follow-up was 14.0 months, and 18 patients (56%) had discontinued treatment. Of the 32 patients enrolled, 28 were evaluable for response by independent radiologic review. Objective response rate was 62% (95% CI, 45% to 77%), with one complete response and 19 partial responses; duration of response was 21.0 months (95% CI, 17 to 25 months); and disease control rate was 81% (95% CI, 65% to 91%). The median progression-free survival was 9.3 months (95% CI, 0 to 22 months) for all patients and 19.3 months (95% CI, 1 to 37 months) for crizotinib-naïve patients. The median overall survival was 24 months (95% CI, 5 to 43 months). Of the eight patients with brain metastases, intracranial disease control was reported in five (63%; 95% CI, 31% to 86%). The most common adverse events (majority, grade 1 or 2) for all treated patients were diarrhea (78%), nausea (59%), and anorexia (56%). Conclusion Ceritinib demonstrated potent clinical activity in patients with ROS1-rearranged NSCLC who were heavily treated previously with multiple lines of chemotherapy.
Collapse
Affiliation(s)
- Sun Min Lim
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Hye Ryun Kim
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Jong-Seok Lee
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Ki Hyeong Lee
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Yun-Gyoo Lee
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Young Joo Min
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Eun Kyung Cho
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Sung Sook Lee
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Bong-Seog Kim
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Moon Young Choi
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Hyo Sup Shim
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Jin-Haeng Chung
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Yoon La Choi
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Min Jeong Lee
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Maria Kim
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Joo-Hang Kim
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Siraj M Ali
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Myung-Ju Ahn
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Byoung Chul Cho
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| |
Collapse
|
289
|
Karachaliou N, Santarpia M, Gonzalez Cao M, Teixido C, Sosa AE, Berenguer J, Rodriguez Capote A, Altavilla G, Rosell R. Anaplastic lymphoma kinase inhibitors in phase I and phase II clinical trials for non-small cell lung cancer. Expert Opin Investig Drugs 2017; 26:713-722. [PMID: 28463570 DOI: 10.1080/13543784.2017.1324572] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Crizotinib is a first-in-class ALK tyrosine kinase inhibitor (TKI), which has proven its superiority over standard platinum-based chemotherapy for the first-line therapy of ALK-rearranged non-small cell lung cancer (NSCLC) patients. The development of acquired resistance to crizotinib represents an ongoing challenge with the central nervous system being one of the most common sites of relapse. Ceritinib and alectinib are approved second-generation ALK TKIs. Several novel ALK inhibitors, more potent and with different selectivity compared to crizotinib, are currently in development. Areas covered: This review will focus on new ALK inhibitors, currently in phase 1 or 2 clinical studies. We will also comment on the mechanisms of resistance to ALK inhibition and the strategies to delay or overcome resistance. Expert opinion: The therapeutic management of ALK-rearranged NSCLC has been greatly improved. Next-generation ALK inhibitors have shown differential potency against ALK rearrangements and ALK resistance mutations. The molecular profile of the tumor at the time of disease progression to crizotinib is crucial for the sequencing of novel ALK TKIs. Ongoing clinical studies will address key issues, including the optimal therapeutic algorithm and whether combinational approaches are more effective than single ALK inhibition for the outcome of ALK-rearranged NSCLC patients.
Collapse
Affiliation(s)
- Niki Karachaliou
- a Institute of Oncology Rosell (IOR), University Hospital Sagrat Cor , Barcelona , Spain
| | - Mariacarmela Santarpia
- b Medical Oncology Unit, Department of Human Pathology 'G. Barresi,' University of Messina , Messina , Italy
| | - Maria Gonzalez Cao
- c Institute of Oncology Rosell (IOR) , Quirón-Dexeus University Institute , Barcelona , Spain
| | - Cristina Teixido
- d Pangaea Oncology , Quirón-Dexeus University Institute , Barcelona , Spain
| | - Aaron E Sosa
- a Institute of Oncology Rosell (IOR), University Hospital Sagrat Cor , Barcelona , Spain
| | - Jordi Berenguer
- d Pangaea Oncology , Quirón-Dexeus University Institute , Barcelona , Spain
| | | | - Giuseppe Altavilla
- b Medical Oncology Unit, Department of Human Pathology 'G. Barresi,' University of Messina , Messina , Italy
| | - Rafael Rosell
- f Germans Trias i Pujol Research Institute , Badalona , Spain.,g Catalan Institute of Oncology , Germans Trias i Pujol University Hospital , Badalona , Spain
| |
Collapse
|
290
|
Ceritinib for non-small cell lung cancer. Aust Prescr 2017; 40:82-83. [PMID: 28507404 DOI: 10.18773/austprescr.2017.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
291
|
Wang VE, Young L, Ali S, Miller VA, Urisman A, Wolfe J, Bivona TG, Damato B, Fogh S, Bergsland EK. A Case of Metastatic Atypical Neuroendocrine Tumor with ALK Translocation and Diffuse Brain Metastases. Oncologist 2017; 22:768-773. [PMID: 28507205 PMCID: PMC5507651 DOI: 10.1634/theoncologist.2017-0054] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/08/2017] [Indexed: 01/10/2023] Open
Abstract
A challenge in precision medicine is the identification of actionable driver mutations. Alterations can be identified within the tumor tissue, by small biopsy or fine‐needle aspirates, or by noninvasive methods, such as circulating tumor cells or circulating tumor DNA. This article presents a case of atypical neuroendocrine tumor metastatic to the bone and brain for which circulating tumor DNA analysis found an ALK translocation. A challenge in precision medicine requires identification of actionable driver mutations. Critical to such effort is the deployment of sensitive and well‐validated assays for mutation detection. Although identification of such alterations within the tumor tissue remains the gold standard, many advanced non‐small cell lung cancer cases have only limited tissue samples, derived from small biopsies or fine‐needle aspirates, available for testing. More recently, noninvasive methods using either circulating tumor cells or tumor DNA (ctDNA) have become an alternative method for identifying molecular biomarkers and screening patients eligible for targeted therapies. In this article, we present a case of a 52‐year‐old never‐smoking male who presented with widely metastatic atypical neuroendocrine tumor to the bones and the brain. Molecular genotyping using DNA harvested from a bone metastasis was unsuccessful due to limited material. Subsequent ctDNA analysis revealed an ALK translocation. The clinical significance of the mutation in this particular cancer type and therapeutic strategies are discussed. Key Points. To our knowledge, this index case represents the first reported ALK translocation identified in an atypical carcinoid tumor. Liquid biopsy such as circulating tumor DNA is a feasible alternative platform for identifying sensitizing genomic alterations. Second‐generation ALK inhibitors represent a new paradigm for treating ALK‐positive patients with brain metastases.
Collapse
Affiliation(s)
- Victoria E. Wang
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- Correspondence: Victoria E. Wang, M.D., Ph.D., Department of Medicine, University of California, San Francisco, 1600 Divisadero Street, San Francisco, California 94115, USA
| | - Lauren Young
- Foundation Medicine, Cambridge, Massachusetts, USA
| | - Siraj Ali
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | - Anatoly Urisman
- Department of Medicine Pathology, University of California, San Francisco, San Francisco, California, USA
| | - John Wolfe
- Department of Pathology, Santa Rosa Memorial Hospital, Santa Rosa, California, USA
| | - Trever G. Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Bertil Damato
- Department of Medicine Ophthalmology, University of California, San Francisco, San Francisco, California, USA
| | - Shannon Fogh
- Department of Medicine Radiation Oncology, University of California, San Francisco, San Francisco, California, USA
| | - Emily K. Bergsland
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
292
|
Choi SH, Kim DH, Choi YJ, Kim SY, Lee JE, Sung KJ, Kim WS, Choi CM, Rho JK, Lee JC. Multiple receptor tyrosine kinase activation related to ALK inhibitor resistance in lung cancer cells with ALK rearrangement. Oncotarget 2017; 8:58771-58780. [PMID: 28938595 PMCID: PMC5601691 DOI: 10.18632/oncotarget.17680] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/22/2017] [Indexed: 01/30/2023] Open
Abstract
The activation of alternative receptor tyrosine kinases (RTKs) is known to mediate resistance to ALK inhibitors. However, the role of multiple RTK activation in resistance has yet to be determined. Two crizotinib-resistant (H3122/CR-1 and H3122/CR-2) and one TAE684-resistant (H2228/TR) cell lines were established. Multi-RTK arrays and Western blots were performed to detect the activation of bypass signals. There were no secondary mutations in the sequencing. EGFR and MET were activated in H3122/CR-1 cells whereas EGFR and IGF1R were activated in H3122/CR-2 cells. Concomitant activation of MET did not contribute to resistance as crizotinib completely suppressed both p-MET and p-ALK in H3122/CR-1 cells, whose survival was not affected by crizotinib. However, combined inhibition of EGFR and ALK was effective in controlling this resistant cell line. In H3122/CR-2 cells, the inhibition of both ALK and IGF1R could effectively suppress cell growth, whereas simultaneous inhibition of ALK and EGFR brought about a less-effective suppression, indicating that IGF1R activation is the main resistance mechanism. H2228/TR cells showed activation of the HER family (EGFR, ErbB2, and ErbB3). Afatinib, a pan-HER inhibitor, was more potent in suppressing resistant cells than gefitinib when combined with crizotinib, which suggests that coactivation of ErbB2 and ErbB3 also contributes to resistance. Interestingly, all three resistant cell lines responded well to AUY922, which can inhibit ALK, EGFR, and IGF1R activity. Activation of multiple RTKs can occur during acquired resistance to ALK inhibitors, in which case the dominant or significant bypass signal should be identified to provide a more appropriate combination therapy.
Collapse
Affiliation(s)
- Se Hoon Choi
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Dong Ha Kim
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Yun Jung Choi
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Seon Ye Kim
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Jung-Eun Lee
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Ki Jung Sung
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Woo Sung Kim
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Chang-Min Choi
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Department of Oncology, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Jin Kyung Rho
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Jae Cheol Lee
- Department of Oncology, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| |
Collapse
|
293
|
Kim DW, Tiseo M, Ahn MJ, Reckamp KL, Hansen KH, Kim SW, Huber RM, West HL, Groen HJM, Hochmair MJ, Leighl NB, Gettinger SN, Langer CJ, Paz-Ares Rodríguez LG, Smit EF, Kim ES, Reichmann W, Haluska FG, Kerstein D, Camidge DR. Brigatinib in Patients With Crizotinib-Refractory Anaplastic Lymphoma Kinase-Positive Non-Small-Cell Lung Cancer: A Randomized, Multicenter Phase II Trial. J Clin Oncol 2017; 35:2490-2498. [PMID: 28475456 DOI: 10.1200/jco.2016.71.5904] [Citation(s) in RCA: 448] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose Most crizotinib-treated patients with anaplastic lymphoma kinase gene ( ALK)-rearranged non-small-cell lung cancer (ALK-positive NSCLC) eventually experience disease progression. We evaluated two regimens of brigatinib, an investigational next-generation ALK inhibitor, in crizotinib-refractory ALK-positive NSCLC. Patients and Methods Patients were stratified by brain metastases and best response to crizotinib. They were randomly assigned (1:1) to oral brigatinib 90 mg once daily (arm A) or 180 mg once daily with a 7-day lead-in at 90 mg (180 mg once daily [with lead-in]; arm B). Investigator-assessed confirmed objective response rate (ORR) was the primary end point. Results Of 222 patients enrolled (arm A: n = 112, 109 treated; arm B: n = 110, 110 treated), 154 (69%) had baseline brain metastases and 164 of 222 (74%) had received prior chemotherapy. With 8.0-month median follow-up, investigator-assessed confirmed ORR was 45% (97.5% CI, 34% to 56%) in arm A and 54% (97.5% CI, 43% to 65%) in arm B. Investigator-assessed median progression-free survival was 9.2 months (95% CI, 7.4 to 15.6) and 12.9 months (95% CI, 11.1 to not reached) in arms A and B, respectively. Independent review committee-assessed intracranial ORR in patients with measurable brain metastases at baseline was 42% (11 of 26 patients) in arm A and 67% (12 of 18 patients) in arm B. Common treatment-emergent adverse events were nausea (arm A/B, 33%/40%), diarrhea (arm A/B, 19%/38%), headache (arm A/B, 28%/27%), and cough (arm A/B, 18%/34%), and were mainly grades 1 to 2. A subset of pulmonary adverse events with early onset (median onset: day 2) occurred in 14 of 219 treated patients (all grades, 6%; grade ≥ 3, 3%); none occurred after escalation to 180 mg in arm B. Seven of 14 patients were successfully retreated with brigatinib. Conclusion Brigatinib yielded substantial whole-body and intracranial responses as well as robust progression-free survival; 180 mg (with lead-in) showed consistently better efficacy than 90 mg, with acceptable safety.
Collapse
Affiliation(s)
- Dong-Wan Kim
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Marcello Tiseo
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Myung-Ju Ahn
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Karen L Reckamp
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Karin Holmskov Hansen
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Sang-We Kim
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Rudolf M Huber
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Howard L West
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Harry J M Groen
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Maximilian J Hochmair
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Natasha B Leighl
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Scott N Gettinger
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Corey J Langer
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Luis G Paz-Ares Rodríguez
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Egbert F Smit
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Edward S Kim
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - William Reichmann
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Frank G Haluska
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - David Kerstein
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - D Ross Camidge
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| |
Collapse
|
294
|
Tuzi A, Bolzacchini E, Suter MB, Giaquinto A, Passaro A, Gobba S, Vallini I, Pinotti G. Biopsy and re-biopsy in lung cancer: the oncologist requests and the role of endobronchial ultrasounds transbronchial needle aspiration. J Thorac Dis 2017; 9:S405-S409. [PMID: 28603652 PMCID: PMC5459872 DOI: 10.21037/jtd.2017.04.09] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/28/2017] [Indexed: 12/21/2022]
Abstract
As the leading cause of death worldwide, lung cancer has proven itself incurable in the advanced stages. For early stages, endobronchial ultrasounds transbronchial needle aspiration (EBUS-TBNA) is now considered the standard to assess mediastinal lymph node, to define the multimodality therapeutic approach. In recent years, EBUS-TBNA has extended its use also in the metastatic and locally recurrent disease. New molecules, with specific mutations that give resistance to current target therapies, have made re-biopsy at disease progression an important assessment, with therapeutic and clinical implication. Here we present the oncologist's point of view on EBUS-TBNA in the staging process, at recurrence and progression.
Collapse
Affiliation(s)
| | | | | | | | - Antonio Passaro
- Medical Oncology, Istituto Europeo di Oncologia (IEO), Milan, Italy
| | | | | | | |
Collapse
|
295
|
Treatment Patterns and Early Outcomes of ALK-Positive Non-Small Cell Lung Cancer Patients Receiving Ceritinib: A Chart Review Study. Adv Ther 2017; 34:1145-1156. [PMID: 28405961 DOI: 10.1007/s12325-017-0527-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Indexed: 01/19/2023]
Abstract
INTRODUCTION This study aimed to provide the first real-world description of the characteristics, treatments, dosing patterns, and early outcomes of patients with ALK-positive non-small cell lung cancer (NSCLC) who received ceritinib in US clinical practice. METHODS US oncologists provided data from medical charts of adult patients diagnosed with locally advanced or metastatic ALK-positive NSCLC who received ceritinib following crizotinib. Patient characteristics, treatment patterns, ceritinib dosing, early outcomes, and occurrence of gastrointestinal adverse events (AEs) by dose and instructions on food intake were assessed, and Kaplan-Meier analysis was used to describe clinician-defined progression-free survival (PFS) on ceritinib. RESULTS Medical charts of 58 ALK-positive NSCLC patients treated with ceritinib were reviewed (median age 63 years; 41% male; 21% with prior chemotherapy experience). At ceritinib initiation, 44 patients had multiple distant metastases, most commonly in the liver (60%), bone (53%), and brain (38%). Initial ceritinib dose varied: 71% received 750 mg, 19% 600 mg, and 10% 450 mg. Although median follow-up after ceritinib initiation was short (3.8 months), most patients achieved either a complete or partial response (69%) on ceritinib, regardless of metastatic sites present at initiation or initial dose. Median PFS on ceritinib was 12.9 months. 17% of patients had a gastrointestinal AE reported during follow-up. The majority of events occurred in patients instructed to fast; no patients instructed to take a lower dose of ceritinib with food reported gastrointestinal AEs. CONCLUSION These early findings of ceritinib use in clinical practice suggest that ceritinib is effective at treating crizotinib-experienced ALK-positive NSCLC patients, regardless of metastatic sites or initial dose, and dosing ceritinib with food may lead to fewer gastrointestinal AEs. Future studies with larger sample size and longer follow-up are warranted, including an ongoing randomized trial to assess the gastrointestinal tolerability of ceritinib 450 and 600 mg with low-fat meals. FUNDING Novartis Pharmaceutical Corporation.
Collapse
|
296
|
Chen G, Chen X, Zhang Y, Yan F, Fang W, Yang Y, Hong S, Miao S, Wu M, Huang X, Luo Y, Zhou C, Gong R, Huang Y, Zhou N, Zhao H, Zhang L. A large, single-center, real-world study of clinicopathological characteristics and treatment in advanced ALK-positive non-small-cell lung cancer. Cancer Med 2017; 6:953-961. [PMID: 28374971 PMCID: PMC5430086 DOI: 10.1002/cam4.1059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/19/2017] [Accepted: 02/22/2017] [Indexed: 11/10/2022] Open
Abstract
Crizotinib has achieved astonishing success in advanced non-small-cell lung cancer (NSCLC) patients harboring anaplastic lymphoma kinase (ALK) rearrangement. However, no real-world studies described the clinicopathological characteristics and treatment of such patients in China. Patients were consecutively collected from Sun Yat-sen University Cancer Center. Chi-square test was applied to explore the relationship between ALK fusion status and metastasis sites. Kaplan-Meier methods and multivariable analyses were used to estimate progression-free survival (PFS). A total of 291 advanced NSCLC patients (ALK (+), N = 97; both ALK & epidermal growth factor receptor (EGFR) (-), N = 194) were enrolled. The occurrence of brain metastasis in ALK-positive patients was significantly higher than double-negative ones both at baseline (26.5% vs. 16.5%, P = 0.038) and during treatment (25.8% vs. 11.9%, P = 0.003), but opposite for pleural effusion (6.2% vs. 26.9%, P < 0.001 at baseline; 3.1% vs. 10.3%, P = 0.031 during treatment). ALK-positive patients of 53.6% used crizotinib, whereas others only received chemotherapy (37.1%) or supportive care (9.3%). Usage of crizotinib prolonged PFS compared with chemotherapy in ALK-positive patients (median PFS 17.6 m vs. 4.8 m, P < 0.001). ALK-positive NSCLC had more brain metastasis and less pleural effusion than double-negative ones. Crizotinib showed better PFS than chemotherapy in advanced ALK-positive NSCLC at any line. However, half advanced ALK-positive patients never received crizotinib, which was grim and need improving.
Collapse
|
297
|
Martin P, Leighl NB. Review of the use of pretest probability for molecular testing in non-small cell lung cancer and overview of new mutations that may affect clinical practice. Ther Adv Med Oncol 2017; 9:405-414. [PMID: 28607579 PMCID: PMC5455881 DOI: 10.1177/1758834017704329] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 03/14/2017] [Indexed: 11/17/2022] Open
Abstract
This article considers the use of pretest probability in non-small cell lung cancer (NSCLC) and how its use in EGFR testing has helped establish clinical guidelines on selecting patients for EGFR testing. With an ever-increasing number of molecular abnormalities being identified and often limited tissue available for testing, the use of pretest probability will need to be increasingly considered in the future for selecting investigations and treatments in patients. In addition we review new mutations that have the potential to affect clinical practice.
Collapse
Affiliation(s)
- Petra Martin
- Division of Medical Oncology, Princess Margaret Cancer Centre, 5-105 610 University Avenue, Toronto, ON, M5G 2M9, Canada
| | - Natasha B Leighl
- Division of Medical Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
298
|
Zhang G, Wang H, Ma Z. [Recent Advances and Prospect of Advanced Non-small Cell Lung Cancer Targeted
Therapy: Focus on Small Molecular Tyrosine Kinase Inhibitors]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2017; 20:278-286. [PMID: 28442018 PMCID: PMC5999678 DOI: 10.3779/j.issn.1009-3419.2017.04.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 02/10/2017] [Accepted: 03/07/2017] [Indexed: 11/22/2022]
Abstract
At present the treatment of advanced non-small cell lung cancer enters a targeted era and develops rapidly. New drugs appear constantly. Small molecular tyrosine kinase inhibitors have occupied the biggest piece of the territory, which commonly have a clear biomarker as predictor, and show remarkable effect in specific molecular classification of patients. The epidermal growth factor tyrosine kinase inhibitors such as gefitinib, erlotinib, icotinib and anaplastic lymphoma kinase tyrosine kinase inhibitors crizotinib have brought a milestone advance. In recent years new generations of tyrosine kinase inhibitors have achieved a great success in patients with acquired resistance to the above two kinds of drugs. At the same time new therapeutic targets are constantly emerging. So in this paper, we reviewed and summarized the important drugs and clinical trails on this topic, and made a prospect of the future development.
Collapse
Affiliation(s)
- Guowei Zhang
- Department of Internal Medicine, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital,
Zhengzhou 450003, China
| | - Huijuan Wang
- Department of Internal Medicine, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital,
Zhengzhou 450003, China
| | - Zhiyong Ma
- Department of Internal Medicine, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital,
Zhengzhou 450003, China
| |
Collapse
|
299
|
Lau YY, Gu W, Lin T, Viraswami-Appanna K, Cai C, Scott JW, Shi M. Assessment of drug-drug interaction potential between ceritinib and proton pump inhibitors in healthy subjects and in patients with ALK-positive non-small cell lung cancer. Cancer Chemother Pharmacol 2017; 79:1119-1128. [PMID: 28424965 DOI: 10.1007/s00280-017-3308-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 04/11/2017] [Indexed: 12/31/2022]
Abstract
PURPOSE The impact of proton pump inhibitors (PPIs) on the pharmacokinetics (PK) and efficacy of ceritinib was evaluated. METHODS A healthy subject drug-drug interaction (DDI) study was conducted to assess the effect of esomeprazole on the PK of a single 750 mg dose of ceritinib. To further investigate the impact of PPIs on the PK and efficacy of ceritinib in ALK-positive cancer patients, two subgroup analyses were performed. Analysis 1 evaluated ceritinib steady-state trough concentration (Ctrough,ss) and overall response rate (ORR) by concomitant use of PPIs in patients from the ASCEND-1, -2, and -3 studies; analysis 2 evaluated ceritinib single-dose and steady-state AUC0-24h and C max by concomitant PPI use in patients from ASCEND-1 using a definition of PPI usage similar to that used in the healthy subject study. RESULTS In the healthy subject study, co-administration of a single 750 mg dose of ceritinib with esomeprazole 40 mg for 6 days decreased ceritinib AUC0-∞ by 76% and C max by 79%. However, based on subgroup analysis 1, patients had similar C trough,ss and ORR regardless of concomitant PPI usage. Based on analysis 2, co-administration of a single 750 mg ceritinib dose with PPIs for 6 days in patients suggested less effect on ceritinib exposure than that observed in healthy subjects as AUC0-24h decreased by 30% and C max decreased by 25%. No clinically meaningful effect on steady-state exposure was observed after daily dosing. CONCLUSIONS Long-term administration of ceritinib with PPIs does not adversely affect the PK and efficacy of ceritinib in ALK-positive cancer patients.
Collapse
Affiliation(s)
- Yvonne Y Lau
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA.
| | - Wen Gu
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA
| | - Tiffany Lin
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA
| | | | - Can Cai
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA
| | - Jeffrey W Scott
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA
| | - Michael Shi
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA
| |
Collapse
|
300
|
Tan AC, Heimberger AB, Menzies AM, Pavlakis N, Khasraw M. Immune Checkpoint Inhibitors for Brain Metastases. Curr Oncol Rep 2017; 19:38. [DOI: 10.1007/s11912-017-0596-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|