701
|
Cha YJ, Kim HR, Shim HS. Clinical outcomes in ALK-rearranged lung adenocarcinomas according to ALK fusion variants. J Transl Med 2016; 14:296. [PMID: 27756333 PMCID: PMC5069800 DOI: 10.1186/s12967-016-1061-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 10/11/2016] [Indexed: 12/02/2022] Open
Abstract
Background Clinical outcomes of anaplastic lymphoma kinase (ALK)-rearranged non-small cell lung cancer according to ALK fusion variants are not clear. We aimed to investigate the prevalence of ALK fusion variants and to compare clinical outcomes according to ALK fusion variants. Methods A retrospective analysis was conducted on patients with advanced ALK-rearranged adenocarcinoma treated with chemotherapy and ALK inhibitors. ALK rearrangement was identified by fluorescence in situ hybridization and confirmed by immunohistochemistry. Peptide nucleic acid-mediated quantitative polymerase chain reaction assays, designed to detect 28 types of echinoderm microtubule-associated protein-like 4 (EML)-ALK rearrangements, were performed. Clinicopathological analysis and treatment outcomes with platinum-based chemotherapy, pemetrexed therapy, and ALK inhibitors—including crizotinib and ceritinib—were evaluated. Results A total of 52 patients with ALK-rearranged lung adenocarcinoma were enrolled. EML4-ALK variant 1 (v1) was the most common variant (38.5 %) followed by the non-EML4 variant (36.5 %), EML4-ALK variant 3a/b (19.2 %), and EML4-ALK variant 2 (5.8 %). No clinicopathological distinction was found between the different ALK fusion variants. Treatment response rates for each therapeutic agent did not differ according to ALK fusion variant. However, EML4 variants, especially v1, showed significantly longer progression-free survival (PFS) on pemetrexed treatment than did non-EML4 variants (median 31.1 months versus 5.7 months, P = 0.003). PFS with platinum-based chemotherapy and ALK inhibitors did not differ according to ALK fusion variant. Multivariate survival analysis using Cox’s regression model revealed v1 as the only predictive factor for prolonged PFS on pemetrexed. Conclusions Among ALK fusion variants, v1 is the most common subtype. It showed superior progression-free survival on pemetrexed than did non-EML4 variants. No survival difference was demonstrated between variants treated with crizotinib or ceritinib. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-1061-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yoon Jin Cha
- Department of Pathology, GangNam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Hye Ryun Kim
- Department of Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyo Sup Shim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
702
|
Putora PM, Szentesi K, Glatzer M, Rodriguez R, Müller J, Baty F, Früh M. SUVmax and Tumour Location in PET-CT Predict Oncogene Status in Lung Cancer. Oncol Res Treat 2016; 39:681-686. [PMID: 27855379 DOI: 10.1159/000450622] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/25/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND In non-small cell lung cancer, anaplastic lymphoma kinase gene rearrangement (ALK+) and epidermal growth factor receptor mutations (EGFR+) are targetable with tyrosine kinase inhibitors. PATIENTS AND METHODS 27 patients with ALK+ tumours, who underwent positron emission tomography-computed tomography (PET-CT) prior to any treatment, were identified. 2 equally sized control groups based on consecutive patients with EGFR+ and EGFR/ALK wild-type (wt) were identified. The maximum standardized uptake value (SUVmax), tumour location (central vs. peripheral), as well as patient- and disease-specific characteristics were collected. RESULTS Mutation status was significantly associated with SUVmax (p < 0.008). The median SUVmax of the primary tumour in the lung for ALK+ patients (SUVmax 13) was significantly higher compared to that of the EGFR+ (SUVmax 9.8, p = 0.010) and the EGFR/ALKwt group (SUVmax 9.6, p = 0.022). No difference was observed between the EGFR+ and the EGFR/ALKwt group (p = 0.961). Mutation status was also associated with primary tumour location (p = 0.001). There was a significantly lower rate of central tumours in the EGFR+ group when compared to ALK+ tumours (15%, p = 0.002). Among EGFR/ALKwt tumours, 41% were central compared to 63% of ALK+ tumours (p = 0.235). CONCLUSION On initial PET-CT, ALK+ primary lung tumours showed a higher SUVmax and were more frequently centrally located while peripheral tumours were more likely to be EGFR+.
Collapse
Affiliation(s)
- Paul M Putora
- Department of Radiation Oncology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | | | | | | | | | | | | |
Collapse
|
703
|
Zhang G, Scarborough H, Kim J, Rozhok AI, Chen YA, Zhang X, Song L, Bai Y, Fang B, Liu RZ, Koomen J, Tan AC, Degregori J, Haura EB. Coupling an EML4-ALK-centric interactome with RNA interference identifies sensitizers to ALK inhibitors. Sci Signal 2016; 9:rs12. [PMID: 27811184 DOI: 10.1126/scisignal.aaf5011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Patients with lung cancers harboring anaplastic lymphoma kinase (ALK) gene fusions benefit from treatment with ALK inhibitors, but acquired resistance inevitably arises. A better understanding of proximal ALK signaling mechanisms may identify sensitizers to ALK inhibitors that disrupt the balance between prosurvival and proapoptotic effector signals. Using affinity purification coupled with mass spectrometry in an ALK fusion lung cancer cell line (H3122), we generated an ALK signaling network and investigated signaling activity using tyrosine phosphoproteomics. We identified a network of 464 proteins composed of subnetworks with differential response to ALK inhibitors. A small hairpin RNA screen targeting 407 proteins in this network revealed 64 and 9 proteins that when knocked down sensitized cells to crizotinib and alectinib, respectively. Among these, knocking down fibroblast growth factor receptor substrate 2 (FRS2) or coiled-coil and C2 domain-containing protein 1A (CC2D1A), both scaffolding proteins, sensitized multiple ALK fusion cell lines to the ALK inhibitors crizotinib and alectinib. Collectively, our data set provides a resource that enhances our understanding of signaling and drug resistance networks consequent to ALK fusions and identifies potential targets to improve the efficacy of ALK inhibitors in patients.
Collapse
Affiliation(s)
- Guolin Zhang
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Hannah Scarborough
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jihye Kim
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Andrii I Rozhok
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yian Ann Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Xiaohui Zhang
- Department of Hematopathology and Laboratory Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Lanxi Song
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Yun Bai
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Bin Fang
- Proteomics Core Facility, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Richard Z Liu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - John Koomen
- Department of Molecular Oncology Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Aik Choon Tan
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - James Degregori
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
704
|
Guo J, Zinner R, Zaorsky NG, Guo W, Lu B. Systemic therapy for echinoderm microtubule-associated protein-like 4 anaplastic lymphoma kinase non-small cell lung cancer brain metastases. J Thorac Dis 2016; 8:E1028-E1031. [PMID: 27747054 DOI: 10.21037/jtd.2016.09.09] [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)
- Jenny Guo
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ralph Zinner
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nicholas G Zaorsky
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Wei Guo
- Department of Thoracic Surgery, Ruijin Hospital at Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Bo Lu
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
705
|
Abstract
Clinical pharmacists are important contributors to the care of patients with cancer; it is therefore critical for oncology clinical pharmacists to stay current with new anticancer therapies. This review summarizes the epidemiology and pathogenesis of non-small cell lung cancer, including the most common genetic alterations, as well as the mechanism of action, clinical development, pharmacodynamics and pharmacokinetics of the anaplastic lymphoma kinase inhibitor ceritinib for the treatment of patients with anaplastic lymphoma kinase-positive non-small cell lung cancer. Targeted therapies based on the presence of specific mutations are an important development in the treatment of non-small cell lung cancer. However, acquired resistance to the first anaplastic lymphoma kinase-inhibitor approved by the U.S. Food and Drug Administration, crizotinib, is observed in almost half of patients treated with it. Ceritinib is an oral anaplastic lymphoma kinase-inhibitor that has demonstrated more potent antitumor activity than crizotinib in preclinical models. It was granted accelerated approval in 2014 to treat anaplastic lymphoma kinase-positive metastatic non-small cell lung cancer patients who have progressed on or are intolerant to crizotinib. Ceritinib represents an important alternative second-line therapy for patients with metastatic non-small cell lung cancer who have traditionally limited treatment options.
Collapse
Affiliation(s)
- Trang H Au
- 1 Department of Pharmacotherapy, College of Pharmacy, University of Utah, Salt Lake City, UT, USA.,2 Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | - David D Stenehjem
- 1 Department of Pharmacotherapy, College of Pharmacy, University of Utah, Salt Lake City, UT, USA.,2 Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
706
|
Caccese M, Ferrara R, Pilotto S, Carbognin L, Grizzi G, Caliò A, Brunelli M, Cuppone F, Petraglia S, Scarpa A, Tortora G, Bria E. Current and developing therapies for the treatment of non-small cell lung cancer with ALK abnormalities: update and perspectives for clinical practice. Expert Opin Pharmacother 2016; 17:2253-2266. [DOI: 10.1080/14656566.2016.1242578] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
707
|
Abstract
With the incorporation of targeted therapies in routine cancer therapy, it is imperative that the array of toxicities associated with these agents be well-recognized and managed, especially since these toxicities are distinct from those seen with conventional cytotoxic agents. This review will focus on these renal toxicities from commonly used targeted agents. This review discusses the mechanisms of these side effects and management strategies. Anti-vascular endothelial growth factor (VEGF) agents including the monoclonal antibody bevacizumab, aflibercept (VEGF trap), and anti-VEGF receptor (VEGFR) tyrosine kinase inhibitors (TKIs) all cause hypertension, whereas some of them result in proteinuria. Monoclonal antibodies against the human epidermal growth factor receptor (HER) family of receptors, such as cetuximab and panitumumab, cause electrolyte imbalances including hypomagnesemia and hypokalemia due to the direct nephrotoxic effect of the drug on renal tubules. Cetuximab may also result in renal tubular acidosis. The TKIs, imatinib and dasatinib, can result in acute or chronic renal failure. Rituximab, an anti-CD20 monoclonal antibody, can cause acute renal failure following initiation of therapy because of the onset of acute tumor lysis syndrome. Everolimus, a mammalian target of rapamycin (mTOR) inhibitor, can result in proteinuria. Discerning the renal adverse effects resulting from these agents is essential for safe treatment strategies, particularly in those with pre-existing renal disease.
Collapse
Affiliation(s)
- Anum Abbas
- Department of Internal Medicine, School of Medicine, Creighton University, Omaha, NE, USA
| | - Mohsin M Mirza
- Department of Internal Medicine, School of Medicine, Creighton University, Omaha, NE, USA
| | - Apar Kishor Ganti
- Division of Oncology and Hematology, Department of Internal Medicine, VA-Nebraska Western Iowa Health Care System and University of Nebraska Medical Center, Omaha, NE, USA
| | - Ketki Tendulkar
- Division of Nephrology, Department of Internal Medicine, University of Nebraska Medical Center, 983040 Nebraska Medical Center, Omaha, NE, 68198-3040, USA.
| |
Collapse
|
708
|
Shi Y, Sun Y, Yu J, Ding C, Wang Z, Wang C, Wang D, Wang C, Wang Z, Wang M, Zhi X, Lu Y, Feng J, Liu Y, Liu X, Liu W, Wu G, Li X, Li K, Li E, Li W, Chen G, Chen Z, Yu P, Wu N, Wu M, Xiao W, Zhang L, Zhang Y, Zhang S, Yang S, Song X, Lin D, Luo R, Shan L, Zhou C, Zhou Z, Zhao Q, Hu C, Hu Y, Guo Q, Chang J, Huang C, Zeng X, Han B, Han X, Jia B, Han Y, Huang Y. [China Experts Consensus on the Diagnosis and Treatment of Advanced Stage Primary Lung Cancer (2016 Version)]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2016; 19:1-15. [PMID: 26805732 PMCID: PMC5999802 DOI: 10.3779/j.issn.1009-3419.2016.01.01] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Yuankai Shi
- Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing 100021, China
| | - Yan Sun
- Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing 100021, China
| | - Jinming Yu
- Shandong Province Cancer Hospital, Ji'nan 250117, China
| | - Cuimin Ding
- The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Ziping Wang
- Beijing Cancer Hospital, Beijing 100142, China
| | - Changli Wang
- Tianjin Medical University Cancer Institute & Hospital, Tianjin 300070, China
| | - Dong Wang
- Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Cunde Wang
- Yunnan Province Cancer Hospital, Kunming 650118, China
| | | | - Mengzhao Wang
- Peking Union Medical College Hospital, Beijing 100730, China
| | - Xiuyi Zhi
- Beijing Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - You Lu
- West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jifeng Feng
- Jiangsu Cancer Hospital, Nanjing 210009, China
| | - Yunpeng Liu
- The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaoqing Liu
- The 307th Hospital of Chinese People's Liberation Army, Beijing 100071, China
| | - Wei Liu
- Beijing Cancer Hospital, Beijing 100142, China
| | - Gang Wu
- Huazhong University of Science and Technology Union Hospital, Wuhan 430022, China
| | - Xiaomei Li
- Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Kai Li
- Tianjin Medical University Cancer Institute & Hospital, Tianjin 300070, China
| | - Enxiao Li
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Wei Li
- The First Hospital of Jilin University, Changchun 130021, China
| | - Gongyan Chen
- Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Zhengtang Chen
- Xinqiao Hospital of The Third Military Medical University, Chongqing 400037, China
| | - Ping Yu
- Sichuan Cancer Hospital, Chengdu 610047, China
| | - Ning Wu
- Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Milu Wu
- Qinghai University Affiliated Hospital, Xining 810000, China
| | - Wenhua Xiao
- The First Affiliated Hospital of Chinese People's Liberation Army General Hospital, Beijing 100048, China
| | - Li Zhang
- Peking Union Medical College Hospital, Beijing 100730, China
| | - Yiping Zhang
- Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Shucai Zhang
- Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Shujun Yang
- Henan Province Cancer Hospital, Zhengzhou 450008, China
| | - Xia Song
- Shanxi Province Cancer Hospital, Taiyuan 030013, China
| | - Dongmei Lin
- Beijing Cancer Hospital, Beijing 100142, China
| | - Rongcheng Luo
- Nanfang Hospital, Nanfang Medical University, Guangzhou 510515, China
| | - Li Shan
- Cancer Hospital of Xinjiang Medical University, Urumqi 830000, China
| | - Caicun Zhou
- Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Zongmei Zhou
- Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Qiong Zhao
- The First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China
| | - Chengping Hu
- Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yi Hu
- Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Qisen Guo
- Shandong Province Cancer Hospital, Ji'nan 250117, China
| | - Jianhua Chang
- Cancer Hospital, Fudan Universitay, Shanghai 200032, China
| | - Cheng Huang
- Fujian Cancer Hospital, Fuzhou 350014, China
| | - Xuan Zeng
- Peking Union Medical College Hospital, Beijing 100730, China
| | - Baohui Han
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Xiaohong Han
- Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing 100021, China
| | - Bo Jia
- Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing 100021, China
| | - Ying Han
- Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing 100021, China
| | - Yu Huang
- Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing 100021, China
| |
Collapse
|
709
|
Miyawaki M, Yasuda H, Tani T, Hamamoto J, Arai D, Ishioka K, Ohgino K, Nukaga S, Hirano T, Kawada I, Naoki K, Hayashi Y, Betsuyaku T, Soejima K. Overcoming EGFR Bypass Signal-Induced Acquired Resistance to ALK Tyrosine Kinase Inhibitors in ALK-Translocated Lung Cancer. Mol Cancer Res 2016; 15:106-114. [PMID: 27707887 DOI: 10.1158/1541-7786.mcr-16-0211] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/25/2016] [Accepted: 09/22/2016] [Indexed: 11/16/2022]
Abstract
Activation of the EGFR pathway is one of the mechanisms inducing acquired resistance to anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors (TKI) such as crizotinib and alectinib. Ceritinib is a highly selective ALK inhibitor and shows promising efficacy in non-small cell lung cancers (NSCLC) harboring the ALK gene rearrangement. However, the precise mechanism underlying acquired resistance to ceritinib is not well-defined. This study set out to clarify the mechanism in ALK-translocated lung cancer and to find the preclinical rationale overcoming EGFR pathway-induced acquired resistance to ALK-TKIs. To this end, ceritinib-resistant cells (H3122-CER) were established from the H3122 NSCLC cell line harboring the ALK gene rearrangement via long-term exposure to ceritinib. H3122-CER cells acquired resistance to ceritinib through EGFR bypass pathway activation. Furthermore, H3122 cells that became resistant to ceritinib or alectinib through EGFR pathway activation showed cross-resistance to other ALK-TKIs. Ceritinib and afatinib combination treatment partially restored the sensitivity to ceritinib. IMPLICATIONS This study proposes a preclinical rationale to use ALK-TKIs and afatinib combination therapy for ALK-translocated lung cancers that have acquired resistance to ALK-TKIs through EGFR pathway activation. Mol Cancer Res; 15(1); 106-14. ©2016 AACR.
Collapse
Affiliation(s)
- Masayoshi Miyawaki
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hiroyuki Yasuda
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Shinjuku-ku, Tokyo, Japan.
| | - Tetsuo Tani
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Junko Hamamoto
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Daisuke Arai
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kota Ishioka
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Keiko Ohgino
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Shigenari Nukaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Toshiyuki Hirano
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Ichiro Kawada
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Katsuhiko Naoki
- Keio Cancer Center, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Yuichiro Hayashi
- Department of Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Tomoko Betsuyaku
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kenzo Soejima
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Shinjuku-ku, Tokyo, Japan.
| |
Collapse
|
710
|
Calabuig-Fariñas S, Jantus-Lewintre E, Herreros-Pomares A, Camps C. Circulating tumor cells versus circulating tumor DNA in lung cancer-which one will win? Transl Lung Cancer Res 2016; 5:466-482. [PMID: 27826528 DOI: 10.21037/tlcr.2016.10.02] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Liquid biopsies appear to be a reliable alternative to conventional biopsies that can provide both precise molecular data useful for improving the clinical management of lung cancer patients as well as a less invasive way of monitoring tumor behavior. These advances are supported by important biotechnological developments in the fields of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). Analysis of CTCs and ctDNA may be useful in treatment selection, for response monitoring, and in studying resistance mechanisms. This review focuses on the most recent technological achievements and the most relevant clinical applications for lung cancer patients in the CTC and ctDNA fields, highlighting those that are already (or are close to) being implemented in daily clinical practice.
Collapse
Affiliation(s)
- Silvia Calabuig-Fariñas
- Molecular Oncology Laboratory, General University Hospital Research Foundation, University General Hospital of Valencia, Valencia, Spain;; Department of Pathology, Universitat de València, Valencia, Spain
| | - Eloísa Jantus-Lewintre
- Molecular Oncology Laboratory, General University Hospital Research Foundation, University General Hospital of Valencia, Valencia, Spain;; Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain
| | - Alejandro Herreros-Pomares
- Molecular Oncology Laboratory, General University Hospital Research Foundation, University General Hospital of Valencia, Valencia, Spain;; Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain
| | - Carlos Camps
- Molecular Oncology Laboratory, General University Hospital Research Foundation, University General Hospital of Valencia, Valencia, Spain;; Department of Medicine, Universitat de València, Valencia, Spain;; Department of Medical Oncology, University General Hospital of Valencia, Valencia, Spain
| |
Collapse
|
711
|
Sullivan I, Planchard D. Editorial on the article entitled "brigatinib efficacy and safety in patients with anaplastic lymphoma kinase ( ALK)-positive non-small cell lung cancer in a phase I/II trial". J Thorac Dis 2016; 8:E1287-E1292. [PMID: 27867609 DOI: 10.21037/jtd.2016.10.57] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ivana Sullivan
- Medical Oncology Department, Gustave Roussy, 114 rue Édouard Vaillant, 94800 Villejuif, France
| | - David Planchard
- Medical Oncology Department, Gustave Roussy, 114 rue Édouard Vaillant, 94800 Villejuif, France
| |
Collapse
|
712
|
Morcos PN, Cleary Y, Guerini E, Dall G, Bogman K, De Petris L, Viteri S, Bordogna W, Yu L, Martin-Facklam M, Phipps A. Clinical Drug-Drug Interactions Through Cytochrome P450 3A (CYP3A) for the Selective ALK Inhibitor Alectinib. Clin Pharmacol Drug Dev 2016; 6:280-291. [PMID: 27545757 DOI: 10.1002/cpdd.298] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 08/17/2016] [Indexed: 11/10/2022]
Abstract
The efficacy and safety of alectinib, a central nervous system-active and selective anaplastic lymphoma kinase (ALK) inhibitor, has been demonstrated in patients with ALK-positive (ALK+) non-small cell lung cancer (NSCLC) progressing on crizotinib. Alectinib is mainly metabolized by cytochrome P450 3A (CYP3A) to a major similarly active metabolite, M4. Alectinib and M4 show evidence of weak time-dependent inhibition and small induction of CYP3A in vitro. We present results from 3 fixed-sequence studies evaluating drug-drug interactions for alectinib through CYP3A. Studies NP28990 and NP29042 enrolled 17 and 24 healthy subjects, respectively, and investigated potent CYP3A inhibition with posaconazole and potent CYP3A induction through rifampin, respectively, on the single oral dose pharmacokinetics (PK) of alectinib. A substudy of the global phase 2 NP28673 study enrolled 15 patients with ALK+ NSCLC to determine the effect of multiple doses of alectinib on the single oral dose PK of midazolam, a sensitive substrate of CYP3A. Potent CYP3A inhibition or induction resulted in only minor effects on the combined exposure of alectinib and M4. Multiple doses of alectinib did not influence midazolam exposure. These results suggest that dose adjustments may not be needed when alectinib is coadministered with CYP3A inhibitors or inducers or for coadministered CYP3A substrates.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Li Yu
- Roche Innovation Center, New York, NY, USA
| | | | | |
Collapse
|
713
|
Emergence of resistance to tyrosine kinase inhibitors in non-small-cell lung cancer can be delayed by an upfront combination with the HSP90 inhibitor onalespib. Br J Cancer 2016; 115:1069-1077. [PMID: 27673365 PMCID: PMC5117788 DOI: 10.1038/bjc.2016.294] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/29/2016] [Accepted: 08/24/2016] [Indexed: 12/21/2022] Open
Abstract
Background: Tyrosine kinase inhibitors, such as crizotinib and erlotinib, are widely used to treat non-small-cell lung cancer, but after initial response, relapse is common because of the emergence of resistance through multiple mechanisms. Here, we investigated whether a frontline combination with an HSP90 inhibitor could delay the emergence of resistance to these inhibitors in preclinical lung cancer models. Methods: The HSP90 inhibitor, onalespib, was combined with either crizotinib or erlotinib in ALK- or EGFR-activated xenograft models respectively (H2228, HCC827). Results: In both models, after initial response to the monotherapy kinase inhibitors, tumour relapse was observed. In contrast, tumour growth remained inhibited when treated with an onalespib/kinase inhibitor combination. Analysis of H2228 tumours, which had relapsed on crizotinib monotherapy, identified a number of clinically relevant crizotinib resistance mechanisms, suggesting that HSP90 inhibitor treatment was capable of suppressing multiple mechanisms of resistance. Resistant cell lines, derived from these tumours, retained sensitivity to onalespib (proliferation and signalling pathways were inhibited), indicating that, despite their resistance to crizotinib, they were still sensitive to HSP90 inhibition. Conclusions: Together, these preclinical data suggest that frontline combination with an HSP90 inhibitor may be a method for delaying the emergence of resistance to targeted therapies.
Collapse
|
714
|
Toyokawa G, Seto T, Takenoyama M, Ichinose Y. Insights into brain metastasis in patients with ALK+ lung cancer: is the brain truly a sanctuary? Cancer Metastasis Rev 2016; 34:797-805. [PMID: 26342831 PMCID: PMC4661196 DOI: 10.1007/s10555-015-9592-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Anaplastic lymphoma kinase (ALK) has been identified to exert a potent transforming activity through its rearrangement in non-small cell lung cancer (NSCLC), and patients (pts) with ALK rearrangement can be treated more successfully with ALK inhibitors, such as crizotinib, alectinib, and ceritinib, than with chemotherapy. Despite the excellent efficacy of ALK inhibitors, resistance to these drugs is inevitably encountered in most ALK-rearranged pts. Cases of resistance are subtyped into three groups, i.e., systemic, oligo, and central nervous system (CNS) types, with the CNS being used to be considered a sanctuary. With regard to the management of CNS lesions in pts with ALK+ NSCLC, a growing body of evidence has gradually demonstrated the intracranial (IC) efficacy of ALK inhibitor (ALKi) in ALK+ NSCLC pts with brain metastases (BMs). Although the efficacy of crizotinib for the CNS lesions remains controversial, a recent retrospective investigation of ALK+ pts with BM enrolled in PROFILE 1005 and PROFILE 1007 demonstrated that crizotinib is associated with a high disease control rate for BM. However, BM comprises the most common site of progressive disease in pts with or without baseline BMs, which is a serious problem for crizotinib. Furthermore, alectinib can be used to achieve strong and long-lasting inhibitory effects on BM. In addition to alectinib, the IC efficacy of other next-generation ALK inhibitors, such as ceritinib, AP26113 and PF-06463922, has been demonstrated. In this article, we review the latest evidence regarding the BM and IC efficacy of ALK inhibitors in pts with ALK+ NSCLC.
Collapse
Affiliation(s)
- Gouji Toyokawa
- Department of Thoracic Oncology, National Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka, 811-1395, Japan.
| | - Takashi Seto
- Department of Thoracic Oncology, National Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka, 811-1395, Japan
| | - Mitsuhiro Takenoyama
- Department of Thoracic Oncology, National Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka, 811-1395, Japan
| | - Yukito Ichinose
- Department of Thoracic Oncology, National Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka, 811-1395, Japan
| |
Collapse
|
715
|
Buetti-Dinh A, O’Hare T, Friedman R. Sensitivity Analysis of the NPM-ALK Signalling Network Reveals Important Pathways for Anaplastic Large Cell Lymphoma Combination Therapy. PLoS One 2016; 11:e0163011. [PMID: 27669408 PMCID: PMC5036789 DOI: 10.1371/journal.pone.0163011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 08/25/2016] [Indexed: 01/01/2023] Open
Abstract
A large subset of anaplastic large cell lymphoma (ALCL) patients harbour a somatic aberration in which anaplastic lymphoma kinase (ALK) is fused to nucleophosmin (NPM) resulting in a constitutively active signalling fusion protein, NPM-ALK. We computationally simulated the signalling network which mediates pathological cell survival and proliferation through NPM-ALK to identify therapeutically targetable nodes through which it may be possible to regain control of the tumourigenic process. The simulations reveal the predominant role of the VAV1-CDC42 (cell division control protein 42) pathway in NPM-ALK-driven cellular proliferation and of the Ras / mitogen-activated ERK kinase (MEK) / extracellular signal-regulated kinase (ERK) cascade in controlling cell survival. Our results also highlight the importance of a group of interleukins together with the Janus kinase 3 (JAK3) / signal transducer and activator of transcription 3 (STAT3) signalling in the development of NPM-ALK derived ALCL. Depending on the activity of JAK3 and STAT3, the system may also be sensitive to activation of protein tyrosine phosphatase-1 (SHP1), which has an inhibitory effect on cell survival and proliferation. The identification of signalling pathways active in tumourigenic processes is of fundamental importance for effective therapies. The prediction of alternative pathways that circumvent classical therapeutic targets opens the way to preventive approaches for countering the emergence of cancer resistance.
Collapse
Affiliation(s)
- Antoine Buetti-Dinh
- Department of Chemistry and Biomedical Sciences, Linnæus University, Kalmar, Sweden
- Linnæus University Centre for Biomaterials Chemistry, Linnæus University, Kalmar, Sweden
- Institute of Computational Science, Faculty of Informatics, Università della Svizzera Italiana, Lugano, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- * E-mail: (ABD); (RF)
| | - Thomas O’Hare
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, United States of America
- Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, United States of America
| | - Ran Friedman
- Department of Chemistry and Biomedical Sciences, Linnæus University, Kalmar, Sweden
- Linnæus University Centre for Biomaterials Chemistry, Linnæus University, Kalmar, Sweden
- * E-mail: (ABD); (RF)
| |
Collapse
|
716
|
Santabarbara G, Maione P, Rossi A, Palazzolo G, Gridelli C. Novel immunotherapy in the treatment of advanced non-small cell lung cancer. Expert Rev Clin Pharmacol 2016; 9:1571-1581. [DOI: 10.1080/17512433.2016.1236681] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
717
|
Goldstein DA, Stemmer SM, Gordon N. The cost and value of cancer drugs - are new innovations outpacing our ability to pay? Isr J Health Policy Res 2016; 5:40. [PMID: 27688873 PMCID: PMC5032240 DOI: 10.1186/s13584-016-0097-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 08/10/2016] [Indexed: 01/05/2023] Open
Abstract
Cancer drug expenditures have been increasing significantly in countries around the world. A recent paper in the IJHPR provides new knowledge and insights into this global phenomenon by analyzing how it is playing out in an Israeli health plan with over two million members, whose state-of-the-art information systems provide an opportunity to explore these changes in a comprehensive, detailed and reliable manner. There is a wide variation in both the cost-effectiveness and the budget impact of individual drugs. These issues also vary when analyzing drugs in other countries due to differential pricing mechanisms. In addition to drug expenditure, the overall cost of cancer care is increasing, partly due to expenditures on non-pharmacologic treatments and diagnostic testing. With the arrival of new therapies, the future of cancer care is exciting. However, there will be many challenges ahead with regard to the ability to pay for such innovations. In this commentary we discuss the current problems and anticipate the future challenges.
Collapse
Affiliation(s)
- Daniel A Goldstein
- Davidoff Cancer Center, Rabin Medical Center, Petach Tikvah, Israel ; Winship Cancer Institute, Emory University, Atlanta, GA USA
| | - Salomon M Stemmer
- Davidoff Cancer Center, Rabin Medical Center, Petach Tikvah, Israel ; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Noa Gordon
- Davidoff Cancer Center, Rabin Medical Center, Petach Tikvah, Israel ; Ben Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
718
|
Pelosi G, Scarpa A, Forest F, Sonzogni A. The impact of immunohistochemistry on the classification of lung tumors. Expert Rev Respir Med 2016; 10:1105-21. [PMID: 27617475 DOI: 10.1080/17476348.2017.1235975] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION To highlight the role of immunohistochemistry to lung cancer classification on the basis of existing guidelines and future perspectives. AREAS COVERED Four orienting key-issues were structured according to an extensive review on the English literature: a) cancer subtyping; b) best biomarkers and rules to follow; c) negative and positive profiling; d) suggestions towards an evidence-based proposal for lung cancer subtyping. A sparing material approach based on a limited number of specific markers is highly desirable. It includes p40 for squamous cell carcinoma ('no p40, no squamous'), TTF1 for adenocarcinoma, synaptophysin for neuroendocrine tumors and vimentin for sarcomatoid carcinoma. A close relationship between genotype and phenotype also supports a diagnostic role for negative profiles. Expert commentary: Highly specific and sensitive IHC markers according to positive and negative diagnostic algorithms seem appropriate for individual patients' lung cancer subtyping.
Collapse
Affiliation(s)
- Giuseppe Pelosi
- a Department of Oncology and Hemato-Oncology , Università degli Studi di Milano , Milan , Italy
| | - Aldo Scarpa
- b Department of Pathology and Diagnostics , University and Hospital Trust of Verona , Verona , Italy.,c ARC-Net Research Centre , University and Hospital Trust of Verona , Verona , Italy
| | - Fabien Forest
- d Department of Pathology , University Hospital Center (CHU), North Hospital , Saint Etienne , France
| | - Angelica Sonzogni
- e Department of Pathology and Laboratory Medicine , Fondazione IRCCS Istituto Nazionale Tumori , Milan , Italy
| |
Collapse
|
719
|
Abstract
The advent of precision medicine in non-small cell lung cancer has remarkably altered the direction of research and improved clinical outcomes. The identification of molecular subsets with differential response to targeted therapies began with the identification of epidermal growth factor receptor mutated tumors in subsets of non-small cell lung cancer (NSCLC). Emboldened by unprecedented response rates to kinase inhibitors seen in that subset, the oncologic community searched for other molecular subsets featuring oncogene addiction. An early result of this search was the discovery of NSCLC driven by activating rearrangements of the anaplastic lymphoma kinase (ALK) gene. In an astoundingly brief period following the recognition of ALK-positive NSCLC, details of the biology, clinicopathologic features, development of targeted inhibitors, mechanisms of therapeutic resistance, and new generations of treatment were elucidated. This review summarizes the current understanding of the pathologic features, diagnostic approach, treatment options, resistance mechanisms, and future research areas for ALK-positive NSCLC.
Collapse
|
720
|
Li J, Fuster MM. Advances in lung cancer with a focus on ATS 2016 updates. J Thorac Dis 2016; 8:S566-8. [PMID: 27606096 DOI: 10.21037/jtd.2016.07.44] [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)
- Jinghong Li
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, La Jolla, CA 92037, USA
| | - Mark M Fuster
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, La Jolla, CA 92037, USA; ; VA San Diego Healthcare System, Medical and Research Sections, La Jolla, CA 92093-9111, USA
| |
Collapse
|
721
|
Skoulidis F, Papadimitrakopoulou VA. Personalized Medicine Tackles Clinical Resistance: Alectinib in ALK-Positive Non-Small Cell Lung Cancer Progressing on First-Generation ALK Inhibitor. Clin Cancer Res 2016; 22:5177-5182. [PMID: 27609840 DOI: 10.1158/1078-0432.ccr-16-1415] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/08/2016] [Indexed: 11/16/2022]
Abstract
Over the last 2 years, our therapeutic armamentarium against genomically defined subgroups of non-small cell lung cancer (NSCLC) has extended to patients with acquired resistance to front-line targeted therapy. Alectinib (Alecensa; Roche/Genentech), a second-generation, orally active, potent, and highly selective inhibitor of anaplastic lymphoma kinase (ALK), is indicated for patients with metastatic, ALK rearrangement-positive NSCLC whose disease has worsened after treatment with crizotinib or who became intolerant to the drug. Alectinib received orphan drug designation, breakthrough therapy designation, priority review status, and accelerated approval by the FDA. Clin Cancer Res; 22(21); 5177-82. ©2016 AACR.
Collapse
Affiliation(s)
- Ferdinandos Skoulidis
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
722
|
Ernani V, Steuer CE, Jahanzeb M. The End of Nihilism: Systemic Therapy of Advanced Non-Small Cell Lung Cancer. Annu Rev Med 2016; 68:153-168. [PMID: 27618751 DOI: 10.1146/annurev-med-042915-102442] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Lung cancer is the leading cause of cancer death in the United States and many other parts of the world. Non-small cell lung cancer (NSCLC) comprises 85-90% of lung cancers. Historically, the expected survival of patients with advanced disease has been estimated in months. In recent years, however, lung cancer has come to be seen as a treatable disease with multiple therapeutic options. Enormous advances in the understanding of its pathways and mechanisms have enabled personalized therapy in NSCLC. The evolving approach to therapy focuses on genomic profiling of the tumors to find molecular targets and develop specific agents for individualized therapy. In addition, maintenance therapy has emerged as a valid approach, and the choice of chemotherapy now varies by histology. Most recently, immunotherapy with checkpoint inhibitors has shown promising results, with impressive durations of response and a tolerable toxicity profile. Together, these discoveries have improved overall survival substantially in patient populations that have access to these advancements. We review the clinical data surrounding these impressive improvements.
Collapse
Affiliation(s)
- Vinicius Ernani
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Conor E Steuer
- Winship Cancer Institute, Emory University, Atlanta, Georgia 30322
| | - Mohammad Jahanzeb
- Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, Deerfield Beach, Florida 33442;
| |
Collapse
|
723
|
Ono A, Murakami H, Serizawa M, Wakuda K, Kenmotsu H, Naito T, Taira T, Koh Y, Ohde Y, Nakajima T, Endo M, Takahashi T. Drastic initial response and subsequent response to two ALK inhibitors in a patient with a highly aggressive ALK-rearranged inflammatory myofibroblastic tumor arising in the pleural cavity. Lung Cancer 2016; 99:151-4. [DOI: 10.1016/j.lungcan.2016.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/24/2016] [Accepted: 07/03/2016] [Indexed: 01/12/2023]
|
724
|
Cassel JB, Del Fabbro E, Arkenau T, Higginson IJ, Hurst S, Jansen LA, Poklepovic A, Rid A, Rodón J, Strasser F, Miller FG. Phase I Cancer Trials and Palliative Care: Antagonism, Irrelevance, or Synergy? J Pain Symptom Manage 2016; 52:437-45. [PMID: 27233136 DOI: 10.1016/j.jpainsymman.2016.02.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 02/06/2016] [Accepted: 02/26/2016] [Indexed: 12/19/2022]
Abstract
This article synthesizes the presentations and conclusions of an international symposium on Phase 1 oncology trials, palliative care, and ethics held in 2014. The purpose of the symposium was to discuss the intersection of three independent trends that unfolded in the past decade. First, large-scale reviews of hundreds of Phase I trials have indicated there is a relatively low risk of serious harm and some prospect of clinical benefit that can be meaningful to patients. Second, changes in the design and analysis of Phase I trials, the introduction of "targeted" investigational agents that are generally less toxic, and an increase in Phase I trials that combine two or more agents in a novel way have changed the conduct of these trials and decreased fears and apprehensions about participation. Third, the field of palliative care in cancer has expanded greatly, offering symptom management to late-stage cancer patients, and demonstrated that it is not mutually exclusive with disease-targeted therapies or clinical research. Opportunities for collaboration and further research at the intersection of Phase 1 oncology trials and palliative care are highlighted.
Collapse
Affiliation(s)
- J Brian Cassel
- Virginia Commonwealth University, Richmond, Virginia, USA.
| | | | - Tobias Arkenau
- Sarah Cannon Research Institute and University College London, London, United Kingdom
| | - Irene J Higginson
- Cicely Saunders Institute, King's College London, London, United Kingdom
| | - Samia Hurst
- Institut d'éthique biomedicale, Centre médical universitaire, Geneva, Switzerland
| | - Lynn A Jansen
- Oregon Health and Science University, Portland, Oregon, USA
| | | | - Annette Rid
- King's College London, London, United Kingdom
| | - Jordi Rodón
- Vall d'Hebron Institut d'Oncologia, Barcelona, Spain
| | | | | |
Collapse
|
725
|
Blackhall F, Cappuzzo F. Crizotinib: from discovery to accelerated development to front-line treatment. Ann Oncol 2016; 27 Suppl 3:iii35-iii41. [PMID: 27573754 DOI: 10.1093/annonc/mdw304] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) is associated with a poor prognosis and low survival rates, providing a strong rationale for the development of new treatment options. The discovery of ALK gene rearrangements in a subset of NSCLC specimens and the identification and development of the first-in-class ALK inhibitor crizotinib provided a personalised treatment option for patients with advanced ALK-positive NSCLC. Crizotinib demonstrated rapid and durable responses in advanced ALK-positive NSCLC patients in phase I and II studies, leading to accelerated FDA approval. Subsequent evaluation in phase III studies showed that crizotinib improved progression-free survival compared with platinum-based doublet chemotherapy in previously untreated patients and compared with pemetrexed or docetaxel in previously treated patients. Crizotinib was shown to have an acceptable safety profile and also to improve quality of life and symptom scores. Overall, crizotinib has been shown to provide a valuable first- and second-line treatment option and is now the first-line standard of care for patients with advanced ALK-positive NSCLC.
Collapse
Affiliation(s)
- F Blackhall
- Christie NHS Foundation Trust, Institute of Cancer Sciences, University of Manchester, Manchester, UK
| | - F Cappuzzo
- Department of Oncology, AUSL della Romagna, Ravenna, Italy
| |
Collapse
|
726
|
Rajan A, Kim C, Heery CR, Guha U, Gulley JL. Nivolumab, anti-programmed death-1 (PD-1) monoclonal antibody immunotherapy: Role in advanced cancers. Hum Vaccin Immunother 2016; 12:2219-31. [PMID: 27135835 PMCID: PMC5027703 DOI: 10.1080/21645515.2016.1175694] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 03/22/2016] [Accepted: 04/02/2016] [Indexed: 12/31/2022] Open
Abstract
The development of immune checkpoint inhibitors has altered the landscape of treatment of advanced cancers. These drugs are well tolerated and have shown clinical activity against a wide variety of solid tumors and hematological malignancies. The durability of response is particularly impressive when compared to other forms of systemic therapy. Nivolumab (Opdivo) is an IgG4 antibody that causes immune checkpoint blockade by diminishing inhibitory signaling through the programmed death receptor-1 pathway. It is approved for treatment of recurrent non-small cell lung cancer, melanoma, and renal cell carcinoma. Efforts to identify biomarkers of response to nivolumab are ongoing. Clinical trials are also being conducted to determine the benefits of combining nivolumab with other forms of treatment including chemotherapy, molecular-targeted therapy, radiation therapy, and other forms of immune therapy. This review outlines the clinical trials that have led to the emergence of nivolumab as a treatment option for patients with advanced cancers.
Collapse
Affiliation(s)
- Arun Rajan
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chul Kim
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christopher R. Heery
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Udayan Guha
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James L. Gulley
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
727
|
Tan DSW, Araújo A, Zhang J, Signorovitch J, Zhou ZY, Cai X, Liu G. Comparative Efficacy of Ceritinib and Crizotinib as Initial ALK –Targeted Therapies in Previously Treated Advanced NSCLC: An Adjusted Comparison with External Controls. J Thorac Oncol 2016; 11:1550-7. [DOI: 10.1016/j.jtho.2016.05.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/11/2016] [Accepted: 05/13/2016] [Indexed: 11/15/2022]
|
728
|
|
729
|
Heydt C, Kostenko A, Merkelbach-Bruse S, Wolf J, Büttner R. ALK evaluation in the world of multiplex testing: Network Genomic Medicine (NGM): the Cologne model for implementing personalised oncology. Ann Oncol 2016; 27 Suppl 3:iii25-iii34. [DOI: 10.1093/annonc/mdw303] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
730
|
Gillis NK, McLeod HL. The pharmacogenomics of drug resistance to protein kinase inhibitors. Drug Resist Updat 2016; 28:28-42. [PMID: 27620953 PMCID: PMC5022787 DOI: 10.1016/j.drup.2016.06.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/17/2016] [Accepted: 06/29/2016] [Indexed: 01/05/2023]
Abstract
Dysregulation of growth factor cell signaling is a major driver of most human cancers. This has led to development of numerous drugs targeting protein kinases, with demonstrated efficacy in the treatment of a wide spectrum of cancers. Despite their high initial response rates and survival benefits, the majority of patients eventually develop resistance to these targeted therapies. This review article discusses examples of established mechanisms of drug resistance to anticancer therapies, including drug target mutations or gene amplifications, emergence of alternate signaling pathways, and pharmacokinetic variation. This reveals a role for pharmacogenomic analysis to identify and monitor for resistance, with possible therapeutic strategies to combat chemoresistance.
Collapse
Affiliation(s)
- Nancy K Gillis
- Eshelman School of Pharmacy, Center for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, NC, United States; H. Lee Moffitt Cancer Center and Research Institute, DeBartolo Family Personalized Medicine Institute, Tampa, FL, United States
| | - Howard L McLeod
- H. Lee Moffitt Cancer Center and Research Institute, DeBartolo Family Personalized Medicine Institute, Tampa, FL, United States; Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
731
|
Insight into drug resistance mechanisms and discovery of potential inhibitors against wild-type and L1196M mutant ALK from FDA-approved drugs. J Mol Model 2016; 22:231. [PMID: 27585676 DOI: 10.1007/s00894-016-3099-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/08/2016] [Indexed: 01/10/2023]
Abstract
Anaplastic lymphoma kinase (ALK) plays a crucial role in multiple malignant cancers. It is known as a well-established target for the treatment of ALK-dependent cancers. Even though substantial efforts have been made to develop ALK inhibitors, only crizotinib, ceritinib, and alectinib had been approved by the U.S. Food and Drug Administration for patients with ALK-positive non-small cell lung cancer (NSCLC). The secondary mutations with drug-resistance bring up difficulties to develop effective drugs for ALK-positive cancers. To give a comprehensive understanding of molecular mechanism underlying inhibitor response to ALK tyrosine kinase mutations, we established an accurate assessment for the extensive profile of drug against ALK mutations by means of computational approaches. The molecular mechanics-generalized Born surface area (MM-GBSA) method based on molecular dynamics (MD) simulation was carried out to calculate relative binding free energies for receptor-drug systems. In addition, the structure-based virtual screening was utilized to screen effective inhibitors targeting wild-type ALK and the gatekeeper mutation L1196M from 3180 approved drugs. Finally, the mechanism of drug resistance was discussed, several novel potential wild-type and L1196M mutant ALK inhibitors were successfully identified.
Collapse
|
732
|
Johnson AC, Dô P, Richard N, Dubos C, Michels JJ, Bonneau J, Gervais R. Identification of I1171N resistance mutation in ALK-positive non-small-cell lung cancer tumor sample and circulating tumor DNA. Lung Cancer 2016; 99:38-40. [DOI: 10.1016/j.lungcan.2016.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/29/2016] [Accepted: 06/11/2016] [Indexed: 10/21/2022]
|
733
|
Improving Selection Criteria for ALK Inhibitor Therapy in Non-Small Cell Lung Cancer: A Pooled-Data Analysis on Diagnostic Operating Characteristics of Immunohistochemistry. Am J Surg Pathol 2016; 40:697-703. [PMID: 26825369 DOI: 10.1097/pas.0000000000000604] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Lung cancer is often diagnosed by molecular markers for prediction and treatment. To date, the golden standard for detection of anaplastic lymphoma kinase (ALK) rearrangements is fluorescence in situ hybridization (FISH). We performed a pooled-data analysis on the diagnostic operating characteristics of immunohistochemistry (IHC) assay on non-small cell lung cancer (NSCLC). We searched Embase, Pubmed, and Springer databases. The results of IHC were evaluated using a modified H-score. We used a 2-level bivariate meta-analysis following a random effect model to summarize sensitivity and specificity and fit hierarchical summary receiver-operating characteristic curves. We also performed sensitivity analysis using different antibodies to investigate potential heterogeneity. Twelve studies consisting of a total of 3754 NSCLC specimens were analyzed. When we defined 1+/2+/3+, 2+/3+, and 3+ as ALK positive, we found the sensitivities to be 99% (95% confidence interval [CI], 97%-100%), 86% (95% CI, 73%-93%), and 56% (95% CI, 36%-74%) and the specificities to be 98% (95% CI, 95%-99%), 99% (95% CI, 99%-100%), and 100% (95% CI, 100%-100%), respectively. We demonstrated that when defining 3+ as positive and 0 as negative the sensitivity was 99% and specificity was 100%. In our sensitivity analysis, we found the sensitivity of D5F3 and 5A4 antibodies to be much higher than that of ALK1. We concluded that IHC scores 0 and 3+ were nearly 100% concordant with FISH-negative and FISH-positive status, respectively. However, IHC scores 1+ and 2+ might require further confirmatory testing by FISH assay. IHC assay using D5F3 and 5A4 antibodies reliably detected NSCLC with ALK rearrangement and may be useful as a screening method to identify these tumors.
Collapse
|
734
|
Wiesweg M, Eberhardt WEE, Reis H, Ting S, Savvidou N, Skiba C, Herold T, Christoph DC, Meiler J, Worm K, Kasper S, Theegarten D, Hense J, Hager T, Darwiche K, Oezkan F, Aigner C, Welter S, Kühl H, Stuschke M, Schmid KW, Schuler M. High Prevalence of Concomitant Oncogene Mutations in Prospectively Identified Patients with ROS1-Positive Metastatic Lung Cancer. J Thorac Oncol 2016; 12:54-64. [PMID: 27575422 DOI: 10.1016/j.jtho.2016.08.137] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/17/2016] [Accepted: 08/18/2016] [Indexed: 02/01/2023]
Abstract
OBJECTIVES Chromosomal rearrangements involving ROS1 define a rare entity of lung adenocarcinomas with exquisite sensitivity to molecularly targeted therapy. We report clinical outcomes and genomic findings of patients with ROS1-positive lung cancer who were prospectively identified within a multiplex biomarker profiling program at the West German Cancer Center. METHODS Standardized immunohistochemical (IHC) analysis, fluorescence in situ hybridization (FISH), and hotspot mutation analyses were performed in 1345 patients with advanced cancer, including 805 patients with metastatic lung adenocarcinoma. Clinical and epidemiological data were retrieved from the institutional database. RESULTS ROS1 positivity by IHC analysis was detected in 25 patients with lung cancer (4.8% of lung adenocarcinomas), including 13 patients (2.5%) with ROS1 FISH positivity with a cutoff of at least 15% of events. Of the ROS1 IHC analysis-positive cases, 36% presented with concomitant oncogenic driver mutations involving EGFR (six cases, five of which were clinically validated by response to EGFR-targeting agents), KRAS (two cases), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha gene (PIK3CA), and BRAF. Three cases initially classified as ROS1 FISH-negative passed the threshold of 15% positive events when repeat biopsies were analyzed at progression. The median overall survival of the ROS1-positive patients (104 months) was significantly superior to that of the 261 patients with EGFR/anaplastic lymphoma kinase/ROS1-negative lung adenocarcinoma (24.4 months, p = 0.044). Interestingly, the overall survival of the 13 ROS1-positive patients with lung cancer from initiation of pemetrexed-based chemotherapy was significantly prolonged when compared with that of 169 pemetrexed-treated patients with EGFR/anaplastic lymphoma kinase/ROS1-negative adenocarcinoma (p = 0.01). CONCLUSIONS ROS1-positive metastatic lung adenocarcinomas frequently harbor concomitant oncogenic driver mutations. Levels of ROS1 FISH-positive events are variable over time. This heterogeneity provides additional therapeutic options if discovered by multiplex biomarker testing and repeat biopsies.
Collapse
Affiliation(s)
- Marcel Wiesweg
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Wilfried E E Eberhardt
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany; Division of Thoracic Oncology, West German Lung Center, Ruhrlandklinik, University Hospital Essen, Essen, Germany
| | - Henning Reis
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Saskia Ting
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nikoleta Savvidou
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Charlotte Skiba
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Thomas Herold
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Daniel C Christoph
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Johannes Meiler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Karl Worm
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stefan Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Dirk Theegarten
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Jörg Hense
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Thomas Hager
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kaid Darwiche
- Division of Interventional Pneumology, West German Lung Center, Ruhrlandklinik, University Hospital Essen, Essen, Germany
| | - Filiz Oezkan
- Division of Interventional Pneumology, West German Lung Center, Ruhrlandklinik, University Hospital Essen, Essen, Germany
| | - Clemens Aigner
- Division of Thoracic Surgery, West German Lung Center, Ruhrlandklinik, University Hospital Essen, Essen, Germany
| | - Stefan Welter
- Division of Thoracic Surgery, West German Lung Center, Ruhrlandklinik, University Hospital Essen, Essen, Germany
| | - Hilmar Kühl
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Martin Stuschke
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany; Department of Radiotherapy, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kurt W Schmid
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Martin Schuler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany; Division of Thoracic Oncology, West German Lung Center, Ruhrlandklinik, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.
| |
Collapse
|
735
|
Abstract
The complexity of cancer chemotherapy requires pharmacists be familiar with the complicated regimens and highly toxic agents used. This column reviews various issues related to preparation, dispensing, and administration of antineoplastic therapy, and the agents, both commercially available and investigational, used to treat malignant diseases. Questions or suggestions for topics should be addressed to Dominic A. Solimando, Jr, President, Oncology Pharmacy Services, Inc., 4201 Wilson Blvd #110-545, Arlington, VA 22203, e-mail: OncRxSvc@comcast.net; or J. Aubrey Waddell, Professor, University of Tennessee College of Pharmacy; Oncology Pharmacist, Pharmacy Department, Blount Memorial Hospital, 907 E. Lamar Alexander Parkway, Maryville, TN 37804, e-mail: waddfour@charter.net.
Collapse
|
736
|
Sundberg TB, Liang Y, Wu H, Choi HG, Kim ND, Sim T, Johannessen L, Petrone A, Khor B, Graham DB, Latorre IJ, Phillips AJ, Schreiber SL, Perez J, Shamji AF, Gray NS, Xavier RJ. Development of Chemical Probes for Investigation of Salt-Inducible Kinase Function in Vivo. ACS Chem Biol 2016; 11:2105-11. [PMID: 27224444 DOI: 10.1021/acschembio.6b00217] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Salt-inducible kinases (SIKs) are promising therapeutic targets for modulating cytokine responses during innate immune activation. The study of SIK inhibition in animal models of disease has been limited by the lack of selective small-molecule probes suitable for modulating SIK function in vivo. We used the pan-SIK inhibitor HG-9-91-01 as a starting point to develop improved analogs, yielding a novel probe 5 (YKL-05-099) that displays increased selectivity for SIKs versus other kinases and enhanced pharmacokinetic properties. Well-tolerated doses of YKL-05-099 achieve free serum concentrations above its IC50 for SIK2 inhibition for >16 h and reduce phosphorylation of a known SIK substrate in vivo. While in vivo active doses of YKL-05-099 recapitulate the effects of SIK inhibition on inflammatory cytokine responses, they did not induce metabolic abnormalities observed in Sik2 knockout mice. These results identify YKL-05-099 as a useful probe to investigate SIK function in vivo and further support the development of SIK inhibitors for treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Thomas B. Sundberg
- Center
for the Development of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Yanke Liang
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Cancer Biology, Dana−Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Huixian Wu
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Hwan Geun Choi
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Cancer Biology, Dana−Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Nam Doo Kim
- Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, Korea
| | - Taebo Sim
- Chemical
Kinomics Research Center, Korea Institute of Science and Technology, Seoul, Korea, 136-791
| | - Liv Johannessen
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Cancer Biology, Dana−Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Adam Petrone
- Center
for the Development of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Bernard Khor
- Center
for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Daniel B. Graham
- Program
in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts 02142, United States
- Department
of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Isabel J. Latorre
- Program
in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Andrew J. Phillips
- Center
for the Development of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Stuart L. Schreiber
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
- Department
of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Howard Hughes Medical Institute, Cambridge, Massachusetts 02142, United States
| | - Jose Perez
- Center
for the Development of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Alykhan F. Shamji
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Nathanael S. Gray
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Cancer Biology, Dana−Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Ramnik J. Xavier
- Center
for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Program
in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts 02142, United States
- Gastrointestinal
Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| |
Collapse
|
737
|
Maddalo D, Ventura A. Somatic Engineering of Oncogenic Chromosomal Rearrangements: A Perspective. Cancer Res 2016; 76:4918-23. [PMID: 27520450 DOI: 10.1158/0008-5472.can-16-0726] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/05/2016] [Indexed: 11/16/2022]
Abstract
The ability to engineer specific mutations in mice has proven essential to advancing our understanding of the molecular basis of cancer. Chromosomal rearrangements, a common and clinically relevant class of cancer-causing mutations, have however remained difficult to faithfully recapitulate in vivo The development of genetic tools for in vivo somatic genome editing has recently overcome this limitation and led to the generation of more sophisticated and accurate preclinical models of human cancers. Here, we review the potential applications of these new technologies to the study of tumor biology and discuss their advantages over more conventional strategies, their limitations, and the remaining challenges. Cancer Res; 76(17); 4918-23. ©2016 AACR.
Collapse
Affiliation(s)
- Danilo Maddalo
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrea Ventura
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
738
|
Affiliation(s)
- Timothy P. Heffron
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
739
|
Dorantes-Heredia R, Ruiz-Morales JM, Cano-García F. Histopathological transformation to small-cell lung carcinoma in non-small cell lung carcinoma tumors. Transl Lung Cancer Res 2016; 5:401-12. [PMID: 27652204 PMCID: PMC5009079 DOI: 10.21037/tlcr.2016.07.10] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lung cancer is the principal cause of cancer-related death worldwide. The use of targeted therapies, especially tyrosine kinase inhibitors (TKIs), in specific groups of patients has dramatically improved the prognosis of this disease, although inevitably some patients will develop resistance to these drugs during active treatment. The most common cancer-associated acquired mutation is the epidermal growth factor receptor (EGFR) Thr790Met (T790M) mutation. During active treatment with targeted therapies, histopathological transformation to small-cell lung carcinoma (SCLC) can occur in 3-15% of patients with non-small-cell lung carcinoma (NSCLC) tumors. By definition, SCLC is a high-grade tumor with specific histological and genetic characteristics. In the majority of cases, a good-quality hematoxylin and eosin (H&E) stain is enough to establish a diagnosis. Immunohistochemistry (IHC) is used to confirm the diagnosis and exclude other neoplasia such as sarcomatoid carcinomas, large-cell carcinoma, basaloid squamous-cell carcinoma, chronic inflammation, malignant melanoma, metastatic carcinoma, sarcoma, and lymphoma. A loss of the tumor-suppressor protein retinoblastoma 1 (RB1) is found in 100% of human SCLC tumors; therefore, it has an essential role in tumorigenesis and tumor development. Other genetic pathways probably involved in the histopathological transformation include neurogenic locus notch homolog (NOTCH) and achaete-scute homolog 1 (ASCL1). Histological transformation to SCLC can be suspected in NSCLC patients who clinically deteriorate during active treatment. Biopsy of any new lesion in this clinical setting is highly recommended to rule out a SCLC transformation. New studies are trying to assess this histological transformation by noninvasive measures such as measuring the concentration of serum neuron-specific enolase.
Collapse
|
740
|
Cadranel J, Park K, Arrieta O, Pless M, Bendaly E, Patel D, Sasane M, Nosal A, Swallow E, Galebach P, Kageleiry A, Stein K, Degun R, Zhang J. Characteristics, treatment patterns, and survival among ALK+ non-small cell lung cancer (NSCLC) patients treated with crizotinib: A chart review study. Lung Cancer 2016; 98:9-14. [DOI: 10.1016/j.lungcan.2016.05.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/12/2016] [Accepted: 05/07/2016] [Indexed: 11/29/2022]
|
741
|
Kinases inhibitors in lung cancer: From benchside to bedside. Biochim Biophys Acta Rev Cancer 2016; 1866:128-40. [DOI: 10.1016/j.bbcan.2016.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 07/01/2016] [Accepted: 07/02/2016] [Indexed: 12/31/2022]
|
742
|
Zago G, Muller M, van den Heuvel M, Baas P. New targeted treatments for non-small-cell lung cancer - role of nivolumab. Biologics 2016; 10:103-17. [PMID: 27536062 PMCID: PMC4975160 DOI: 10.2147/btt.s87878] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Non-small-cell lung cancer (NSCLC) is often diagnosed at an advanced stage of disease, where it is no longer amenable to curative treatment. During the last decades, the survival has only improved significantly for lung cancer patients who have tumors harboring a driver mutation. Therefore, there is a clear unmet need for effective therapies for patients with no mutation. Immunotherapy has emerged as an effective treatment for different cancer types. Nivolumab, a monoclonal inhibitory antibody against PD-1 receptor, can prolong survival of NSCLC patients, with a manageable toxicity profile. In two Phase III trials, nivolumab was compared to docetaxel in patients with, respectively, squamous (CheckMate 017) and non-squamous NSCLC (CheckMate 057). In both trials, nivolumab significantly reduced the risk of death compared to docetaxel (41% and 27% lower risk of death for squamous and non-squamous NSCLC, respectively). Therefore, nivolumab has been approved in the US and in Europe as second-line treatment for advanced NSCLC. Unfortunately, accurate predictive factors for patient selection are lacking, making it difficult to decide who will benefit and who will not. Currently, there are many ongoing trials that evaluate the efficacy of nivolumab in different settings and in combination with other agents. This paper reviews the present literature about the role of nivolumab in the treatment of NSCLC. Particular attention has been given to efficacy studies, toxicity profile, and current and emerging predictive factors.
Collapse
Affiliation(s)
- Giulia Zago
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek (NKI-AvL), Amsterdam, the Netherlands
- Medical Oncology 2, Istituto Oncologico Veneto (IOV), Padova, Italy
| | - Mirte Muller
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek (NKI-AvL), Amsterdam, the Netherlands
| | - Michel van den Heuvel
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek (NKI-AvL), Amsterdam, the Netherlands
| | - Paul Baas
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek (NKI-AvL), Amsterdam, the Netherlands
| |
Collapse
|
743
|
Tyrosine Kinase Receptor Landscape in Lung Cancer: Therapeutical Implications. DISEASE MARKERS 2016; 2016:9214056. [PMID: 27528792 PMCID: PMC4977389 DOI: 10.1155/2016/9214056] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 12/24/2022]
Abstract
Lung cancer is a heterogeneous disease responsible for the most cases of cancer-related deaths. The majority of patients are clinically diagnosed at advanced stages, with a poor survival rate. For this reason, the identification of oncodrivers and novel biomarkers is decisive for the future clinical management of this pathology. The rise of high throughput technologies popularly referred to as “omics” has accelerated the discovery of new biomarkers and drivers for this pathology. Within them, tyrosine kinase receptors (TKRs) have proven to be of importance as diagnostic, prognostic, and predictive tools and, due to their molecular nature, as therapeutic targets. Along this review, the role of TKRs in the different lung cancer histologies, research on improvement of anti-TKR therapy, and the current approaches to manage anti-TKR resistance will be discussed.
Collapse
|
744
|
Cameron L, Solomon B. New Treatment Options for ALK-Rearranged Non-Small Cell Lung Cancer. Curr Treat Options Oncol 2016; 16:49. [PMID: 26318457 DOI: 10.1007/s11864-015-0367-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OPINION STATEMENT ALK rearrangements are present in 3-5% of patients with non-small cell lung cancer (NSCLC) and after epidermal growth factor receptor (EGFR) mutations represent the second molecular target in NSCLC to be validated through phase III clinical trials. The PROFILE 1014 international multicentre phase III trial demonstrated the superiority of crizotinib over standard chemotherapy, establishing crizotinib as standard first-line therapy for patients with advanced ALK-positive NSCLC and indicating the requirement for ALK testing to guide selection of optimal first-line therapy for non-squamous NSCLC. Despite impressive and durable responses, progression on treatment reflecting the development of acquired resistance is inevitable. There are several mechanisms of resistance including ALK kinase mutation or copy number gain, activation of bypass pathways and potentially pharmacokinetic failure of therapy (most commonly in CNS). A broad array of newer generation ALK inhibitors are in development that appear effective in the crizotinib-resistant setting including in patients with intracranial progression. These agents, including ceritinib and alectinib, have a higher potency against ALK kinase than crizotinib, activity against mutations that confer resistance to crizotinib and potentially improved CNS penetration. While in selected patients, continued therapy with crizotinib after local ablative treatments of oligo-progressive systemic or CNS disease may be an option, for many patients use of a newer generation compound will be effective. First-line treatment with newer generation ALK inhibitors may have potential advantages over sequential treatment after crizotinib; however, the optimal sequence of therapy with ALK inhibitors has not been determined and is being explored in ongoing phase III studies.
Collapse
Affiliation(s)
- Laird Cameron
- Department of Medical Oncology, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC, 3002, Australia
| | | |
Collapse
|
745
|
Wang S, Tsui ST, Liu C, Song Y, Liu D. EGFR C797S mutation mediates resistance to third-generation inhibitors in T790M-positive non-small cell lung cancer. J Hematol Oncol 2016; 9:59. [PMID: 27448564 PMCID: PMC4957905 DOI: 10.1186/s13045-016-0290-1] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/16/2016] [Indexed: 01/09/2023] Open
Abstract
T790M mutation is the most common mechanism for resistance to first- and second-generation tyrosine kinase inhibitors (TKI) for epidermal growth factor receptor (EGFR). Several third-generation EGFR mutant selective TKIs are being explored to conquer this resistance. AZD9291 (osimertinib, tagrisso) has been approved for treatment of the metastatic EGFR T790M mutation-positive non-small cell lung cancer. Resistance to AZD9291 has been described. C797S mutation was reported to be a major mechanism for resistance to T790M-targeting EGFR inhibitors. This review summarizes the latest development in identifying the C797S mutation and EAI045, the novel selective inhibitor overcoming the C797S mutant.
Collapse
Affiliation(s)
- Shuhang Wang
- The Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China
| | - Stella T Tsui
- SUNY Stony Brook University, Stony Brook, NY, 11794, USA
| | - Christina Liu
- Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, 60208, USA
| | - Yongping Song
- Henan Cancer Hospital and the affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Delong Liu
- Henan Cancer Hospital and the affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
746
|
Pugliese SB, Neal JW, Kwong BY. Management of Dermatologic Complications of Lung Cancer Therapies. Curr Treat Options Oncol 2016; 16:50. [PMID: 26338208 DOI: 10.1007/s11864-015-0368-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OPINION STATEMENT In recent years, oncogene-directed targeted agents and immunotherapies have expanded the treatment armamentarium for advanced lung cancer and, in particular, non-small cell lung cancer (NSCLC). Along with extended survival, these agents are accompanied by a host of cutaneous complications that affect the skin, hair, and nails. These skin complications range from the well-characterized papulopustular (acneiform) eruption of the epidermal growth factor receptor (EGFR) inhibitors to the emerging characterization of lichenoid skin eruptions seen during treatment with antibodies targeting the programmed cell death protein 1 (PD-1) and programmed cell death protein 1 ligand (PD-L1). When promptly recognized and accurately diagnosed, most cutaneous adverse events can be managed with supportive treatments, avoiding the need to interrupt antitumor therapy. Furthermore, preemptive management of skin problems can lead to significantly decreased severity of many cutaneous complications of these therapies. We encourage close collaboration between dermatologists and oncologists to better characterize cutaneous toxicity, select appropriate management, and avoid unnecessary dose reduction or discontinuation while simultaneously improving patient quality of life.
Collapse
|
747
|
Ceccon M, Merlo MEB, Mologni L, Poggio T, Varesio LM, Menotti M, Bombelli S, Rigolio R, Manazza AD, Di Giacomo F, Ambrogio C, Giudici G, Casati C, Mastini C, Compagno M, Turner SD, Gambacorti-Passerini C, Chiarle R, Voena C. Excess of NPM-ALK oncogenic signaling promotes cellular apoptosis and drug dependency. Oncogene 2016; 35:3854-3865. [PMID: 26657151 PMCID: PMC4907875 DOI: 10.1038/onc.2015.456] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/15/2015] [Indexed: 12/12/2022]
Abstract
Most of the anaplastic large-cell lymphoma (ALCL) cases carry the t(2;5; p23;q35) that produces the fusion protein NPM-ALK (nucleophosmin-anaplastic lymphoma kinase). NPM-ALK-deregulated kinase activity drives several pathways that support malignant transformation of lymphoma cells. We found that in ALK-rearranged ALCL cell lines, NPM-ALK was distributed in equal amounts between the cytoplasm and the nucleus. Only the cytoplasmic portion was catalytically active in both cell lines and primary ALCL, whereas the nuclear portion was inactive because of heterodimerization with NPM1. Thus, about 50% of the NPM-ALK is not active and sequestered as NPM-ALK/NPM1 heterodimers in the nucleus. Overexpression or relocalization of NPM-ALK to the cytoplasm by NPM genetic knockout or knockdown caused ERK1/2 (extracellular signal-regulated protein kinases 1 and 2) increased phosphorylation and cell death through the engagement of an ATM/Chk2- and γH2AX (phosphorylated H2A histone family member X)-mediated DNA-damage response. Remarkably, human NPM-ALK-amplified cell lines resistant to ALK tyrosine kinase inhibitors (TKIs) underwent apoptosis upon drug withdrawal as a consequence of ERK1/2 hyperactivation. Altogether, these findings indicate that an excess of NPM-ALK activation and signaling induces apoptosis via oncogenic stress responses. A 'drug holiday' where the ALK TKI treatment is suspended could represent a therapeutic option in cells that become resistant by NPM-ALK amplification.
Collapse
MESH Headings
- Animals
- Apoptosis
- Blotting, Western
- Cell Line, Tumor
- Cell Survival/drug effects
- Cell Survival/genetics
- Cells, Cultured
- Crizotinib
- Dose-Response Relationship, Drug
- Drug Synergism
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Histones/metabolism
- Humans
- Hydrazines/pharmacology
- Lymphoma, Large-Cell, Anaplastic/genetics
- Lymphoma, Large-Cell, Anaplastic/metabolism
- Lymphoma, Large-Cell, Anaplastic/pathology
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Microscopy, Confocal
- Nucleophosmin
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Protein Kinase Inhibitors/pharmacology
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- Pyrazoles/pharmacology
- Pyridines/pharmacology
- RNA Interference
- Signal Transduction
- Transplantation, Heterologous
- Triazoles/pharmacology
Collapse
Affiliation(s)
- Monica Ceccon
- Department of Health Science, University of Milano-Bicocca, Monza, Italy
| | - Maria Elena Boggio Merlo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Luca Mologni
- Department of Health Science, University of Milano-Bicocca, Monza, Italy
| | - Teresa Poggio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Lydia M. Varesio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Matteo Menotti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Silvia Bombelli
- Department of Health Science, University of Milano-Bicocca, Monza, Italy
| | - Roberta Rigolio
- Surgery and Translational Medicine department, University of Milano-Bicocca, Monza, Italy
| | - Andrea D. Manazza
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Filomena Di Giacomo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Chiara Ambrogio
- Molecular Oncology Program, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Giovanni Giudici
- Tettamanti Research Centre, Pediatric Clinic, University of Milano-Bicocca, Monza, Italy
| | | | - Cristina Mastini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Mara Compagno
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
- Department of Pathology, Children’s Hospital and Harvard Medical School, Boston, USA
| | - Suzanne D. Turner
- Division of Molecular Histopathology, Addenbrooke's Hospital Cambridge, Cambridge, UK
| | - Carlo Gambacorti-Passerini
- Department of Health Science, University of Milano-Bicocca, Monza, Italy
- Section of Haematology, San Gerardo Hospital, Monza, Italy
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
- Department of Pathology, Children’s Hospital and Harvard Medical School, Boston, USA
| | - Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| |
Collapse
|
748
|
Crinò L, Ahn MJ, De Marinis F, Groen HJM, Wakelee H, Hida T, Mok T, Spigel D, Felip E, Nishio M, Scagliotti G, Branle F, Emeremni C, Quadrigli M, Zhang J, Shaw AT. Multicenter Phase II Study of Whole-Body and Intracranial Activity With Ceritinib in Patients With ALK-Rearranged Non-Small-Cell Lung Cancer Previously Treated With Chemotherapy and Crizotinib: Results From ASCEND-2. J Clin Oncol 2016; 34:2866-73. [PMID: 27432917 DOI: 10.1200/jco.2015.65.5936] [Citation(s) in RCA: 279] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Phase I data (ASCEND-1) showed ceritinib efficacy in patients with ALK-rearranged non-small-cell lung cancer (NSCLC), regardless of brain metastases status and with or without prior therapy with an inhibitor of the ALK protein. Data are presented from a phase II trial (ASCEND-2) in which ceritinib efficacy and safety were evaluated in patients who had ALK-rearranged NSCLC previously treated with at least one platinum-based chemotherapy and who had experienced progression during crizotinib treatment as their last prior therapy. PATIENTS AND METHODS Patients with advanced ALK-rearranged NSCLC, including those with asymptomatic or neurologically stable baseline brain metastases, received oral ceritinib 750 mg/d. Whole-body and intracranial responses were investigator assessed (according to RECIST version 1.1). Patient-reported outcomes were evaluated with the Lung Cancer Symptom Scale and European Organisation for Research and Treatment of Cancer surveys (the core-30 and the 13-item lung cancer-specific quality-of-life questionnaires). RESULTS All 140 patients enrolled had received two or more previous treatment regimens, and all patients had received crizotinib. The median duration of exposure and the follow-up time with ceritinib were 8.8 months (range, 0.1 to 19.4 months) and 11.3 months (range, 0.1 to 18.9 months), respectively. Investigator-assessed overall response rate was 38.6% (95% CI, 30.5% to 47.2%). Secondary end points, all investigator assessed, included disease control rate (77.1%; 95% CI, 69.3% to 83.8%), time to response (median, 1.8 months; range, 1.6 to 5.6 months), duration of response (median, 9.7 months; 95% CI, 7.1 to 11.1 months), and progression-free survival (median, 5.7 months; 95% CI, 5.4 to 7.6 months). Of 100 patients with baseline brain metastases, 20 had active target lesions at baseline; investigator-assessed intracranial overall response rate was 45.0% (95% CI, 23.1% to 68.5%). The most common adverse events (majority, grade 1 or 2) for all treated patients were nausea (81.4%), diarrhea (80.0%), and vomiting (62.9%). Patient-reported outcomes showed a trend toward improved symptom burden. The global quality-of-life score was maintained during treatment. CONCLUSION Consistent with its activity in ASCEND-1, ceritinib treatment provided clinically meaningful and durable responses with manageable tolerability in chemotherapy- and crizotinib-pretreated patients, including those with brain metastases.
Collapse
Affiliation(s)
- Lucio Crinò
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA.
| | - Myung-Ju Ahn
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Filippo De Marinis
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Harry J M Groen
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Heather Wakelee
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Toyoaki Hida
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Tony Mok
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - David Spigel
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Enriqueta Felip
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Makoto Nishio
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Giorgio Scagliotti
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Fabrice Branle
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Chetachi Emeremni
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Massimiliano Quadrigli
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Jie Zhang
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Alice T Shaw
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| |
Collapse
|
749
|
Gainor JF, Dardaei L, Yoda S, Friboulet L, Leshchiner I, Katayama R, Dagogo-Jack I, Gadgeel S, Schultz K, Singh M, Chin E, Parks M, Lee D, DiCecca RH, Lockerman E, Huynh T, Logan J, Ritterhouse LL, Le LP, Muniappan A, Digumarthy S, Channick C, Keyes C, Getz G, Dias-Santagata D, Heist RS, Lennerz J, Sequist LV, Benes CH, Iafrate AJ, Mino-Kenudson M, Engelman JA, Shaw AT. Molecular Mechanisms of Resistance to First- and Second-Generation ALK Inhibitors in ALK-Rearranged Lung Cancer. Cancer Discov 2016; 6:1118-1133. [PMID: 27432227 DOI: 10.1158/2159-8290.cd-16-0596] [Citation(s) in RCA: 872] [Impact Index Per Article: 96.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/14/2016] [Indexed: 11/16/2022]
Abstract
Advanced, anaplastic lymphoma kinase (ALK)-positive lung cancer is currently treated with the first-generation ALK inhibitor crizotinib followed by more potent, second-generation ALK inhibitors (e.g., ceritinib and alectinib) upon progression. Second-generation inhibitors are generally effective even in the absence of crizotinib-resistant ALK mutations, likely reflecting incomplete inhibition of ALK by crizotinib in many cases. Herein, we analyzed 103 repeat biopsies from ALK-positive patients progressing on various ALK inhibitors. We find that each ALK inhibitor is associated with a distinct spectrum of ALK resistance mutations and that the frequency of one mutation, ALKG1202R, increases significantly after treatment with second-generation agents. To investigate strategies to overcome resistance to second-generation ALK inhibitors, we examine the activity of the third-generation ALK inhibitor lorlatinib in a series of ceritinib-resistant, patient-derived cell lines, and observe that the presence of ALK resistance mutations is highly predictive for sensitivity to lorlatinib, whereas those cell lines without ALK mutations are resistant. SIGNIFICANCE Secondary ALK mutations are a common resistance mechanism to second-generation ALK inhibitors and predict for sensitivity to the third-generation ALK inhibitor lorlatinib. These findings highlight the importance of repeat biopsies and genotyping following disease progression on targeted therapies, particularly second-generation ALK inhibitors. Cancer Discov; 6(10); 1118-33. ©2016 AACRSee related commentary by Qiao and Lovly, p. 1084This article is highlighted in the In This Issue feature, p. 1069.
Collapse
Affiliation(s)
- Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Leila Dardaei
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Satoshi Yoda
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Luc Friboulet
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts. Gustave Roussy Cancer Campus, Université Paris Saclay, INSERM U981, Paris, France
| | - Ignaty Leshchiner
- Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Ryohei Katayama
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts. Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ibiayi Dagogo-Jack
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Shirish Gadgeel
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan
| | - Katherine Schultz
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Manrose Singh
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Emily Chin
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Melissa Parks
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Dana Lee
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Richard H DiCecca
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Elizabeth Lockerman
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Tiffany Huynh
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jennifer Logan
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Long P Le
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Ashok Muniappan
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Subba Digumarthy
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Colleen Channick
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Colleen Keyes
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Gad Getz
- Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Dora Dias-Santagata
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Rebecca S Heist
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jochen Lennerz
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Lecia V Sequist
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Cyril H Benes
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jeffrey A Engelman
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| | - Alice T Shaw
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
750
|
Gainor JF. Alectinib-a new chapter in the management of ALK-positive lung cancer. Transl Lung Cancer Res 2016; 5:343-6. [PMID: 27411885 DOI: 10.21037/tlcr.2016.03.05] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|