251
|
Dietel M, Bubendorf L, Dingemans AMC, Dooms C, Elmberger G, García RC, Kerr KM, Lim E, López-Ríos F, Thunnissen E, Van Schil PE, von Laffert M. Diagnostic procedures for non-small-cell lung cancer (NSCLC): recommendations of the European Expert Group. Thorax 2015; 71:177-84. [PMID: 26530085 PMCID: PMC4752623 DOI: 10.1136/thoraxjnl-2014-206677] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 07/21/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND There is currently no Europe-wide consensus on the appropriate preanalytical measures and workflow to optimise procedures for tissue-based molecular testing of non-small-cell lung cancer (NSCLC). To address this, a group of lung cancer experts (see list of authors) convened to discuss and propose standard operating procedures (SOPs) for NSCLC. METHODS Based on earlier meetings and scientific expertise on lung cancer, a multidisciplinary group meeting was aligned. The aim was to include all relevant aspects concerning NSCLC diagnosis. After careful consideration, the following topics were selected and each was reviewed by the experts: surgical resection and sampling; biopsy procedures for analysis; preanalytical and other variables affecting quality of tissue; tissue conservation; testing procedures for epidermal growth factor receptor, anaplastic lymphoma kinase and ROS proto-oncogene 1, receptor tyrosine kinase (ROS1) in lung tissue and cytological specimens; as well as standardised reporting and quality control (QC). Finally, an optimal workflow was described. RESULTS Suggested optimal procedures and workflows are discussed in detail. The broad consensus was that the complex workflow presented can only be executed effectively by an interdisciplinary approach using a well-trained team. CONCLUSIONS To optimise diagnosis and treatment of patients with NSCLC, it is essential to establish SOPs that are adaptable to the local situation. In addition, a continuous QC system and a local multidisciplinary tumour-type-oriented board are essential.
Collapse
Affiliation(s)
- Manfred Dietel
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Anne-Marie C Dingemans
- Department of Respiratory Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Christophe Dooms
- Respiratory Division, University Hospitals KU Leuven, Leuven, Belgium
| | - Göran Elmberger
- Department of Laboratory Medicine, Pathology, Örebro University Hospital, Örebro, Sweden
| | - Rosa Calero García
- Department of Radiology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Keith M Kerr
- Aberdeen University Medical School, Aberdeen, UK
| | - Eric Lim
- Academic Division of Thoracic Surgery, The Royal Brompton Hospital and Imperial College, London, UK
| | - Fernando López-Ríos
- Laboratorio de Dianas Terapéuticas, Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul E Van Schil
- Department of Thoracic and Vascular Surgery, Antwerp University Hospital, Antwerp, Belgium
| | | |
Collapse
|
252
|
Sacco PC, Sgambato A, Casaluce F, Maione P, Rossi A, Palazzolo G, Napolitano A, Gridelli C. Agents in the preclinical development stage for non-small cell lung cancer. Expert Rev Anticancer Ther 2015; 15:1361-6. [PMID: 26485341 DOI: 10.1586/14737140.2015.1092875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Despite recent advancements in identifying distinct molecular subtypes with driver oncogenes and advances in developing targeted treatments such as epidermal growth factor receptor and anaplastic lymphoma kinase inhibitors, current therapeutic approaches for tumors with no driver mutation have achieved a plateau of effectiveness. Thus, the overall outlook of lung cancer survival for most patients remains dismal. Moreover, the inevitable acquisition of resistance to targeted therapies has prompted significant efforts to develop second-generation inhibitors. In recent years, several agents for targeted therapy of lung cancers are rapidly migrating from bench to bedside and multiple small molecule inhibitors with activity against distinct receptors, genes or molecular pathways have been developed.
Collapse
Affiliation(s)
| | - Assunta Sgambato
- b ² Department of Clinical and Experimental Medicine, Second University of Naples, Napoli, Italy
| | - Francesca Casaluce
- b ² Department of Clinical and Experimental Medicine, Second University of Naples, Napoli, Italy
| | - Paolo Maione
- a ¹ Division of Medical Oncology, S. G. Moscati Hospital, Avellino, Italy
| | - Antonio Rossi
- a ¹ Division of Medical Oncology, S. G. Moscati Hospital, Avellino, Italy
| | | | - Alba Napolitano
- d 4 Division of Pharmacy, S. G. Moscati Hospital, Avellino, Italy
| | - Cesare Gridelli
- a ¹ Division of Medical Oncology, S. G. Moscati Hospital, Avellino, Italy
| |
Collapse
|
253
|
Lim SM, Choi J, Chang JH, Sohn J, Jacobson K, Policht F, Schulz J, Cho BC, Kim SH. Lack of ROS1 Gene Rearrangement in Glioblastoma Multiforme. PLoS One 2015; 10:e0137678. [PMID: 26366867 PMCID: PMC4569301 DOI: 10.1371/journal.pone.0137678] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 08/19/2015] [Indexed: 01/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of brain tumor, and the prognosis remains poor. Rearrangement of ROS1 gene, which was shown to have an oncogenic potential, was previously discovered in GBM cell lines. In this pilot study, we aimed to identify the incidence of ROS1 rearrangement in GBM patient tissues to explore novel biomarkers for therapeutic strategy. Formalin-fixed and paraffin-embedded (FFPE) tissue sections from 109 patients with GBM were screened for ROS1 rearrangement by anti-ROS immunohistochemistry (IHC) and ROS1 break-apart fluorescent in situ hybridization (FISH) assays. O6-methylguanine-DNA methyltransferase (MGMT) gene promoter methylation and Isocitrate dehydrogenase 1 (IDH1) mutation status were also assessed. All samples were interpreted by two experienced pathologists who were blinded to the clinical data. A total of 109 samples were collected and all samples were examined for ROS1 rearrangement by IHC and FISH assays, and none was found to harbor ROS1 rearrangement. MGMT gene methylation was found in 42 (39.2%) cases, and IDH1 mutation was found in 6 (5.5%) cases. In this study, ROS1 rearrangement was not identified in GBM patients, and thus it is difficult to classify ROS1 rearrangement as a novel molecular subset in GBM patients for now.
Collapse
Affiliation(s)
- Sun Min Lim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Junjeong Choi
- Department of Pharmacy, College of Pharmacy, Yonsei University, Seoul, Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Jinyoung Sohn
- JE-UK Institute for Cancer Research, JEUK Co., Ltd., Gumi-City, Kyungbuk, Korea
| | - Kristine Jacobson
- Abbott Molecular Diagnostics, Des Plaines, Illinois, United States of America
| | - Frank Policht
- Abbott Molecular Diagnostics, Des Plaines, Illinois, United States of America
| | - John Schulz
- Abbott Molecular Diagnostics, Des Plaines, Illinois, United States of America
| | - Byoung Chul Cho
- Department of Internal Medicine, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (BCC); (SHK)
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (BCC); (SHK)
| |
Collapse
|
254
|
Passiglia F, Bronte G, Castiglia M, Listì A, Calò V, Toia F, Cicero G, Fanale D, Rizzo S, Bazan V, Russo A. Prognostic and predictive biomarkers for targeted therapy in NSCLC: for whom the bell tolls? Expert Opin Biol Ther 2015; 15:1553-66. [DOI: 10.1517/14712598.2015.1071348] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
255
|
|
256
|
Boolell V, Alamgeer M, Watkins DN, Ganju V. The Evolution of Therapies in Non-Small Cell Lung Cancer. Cancers (Basel) 2015; 7:1815-46. [PMID: 26371045 PMCID: PMC4586797 DOI: 10.3390/cancers7030864] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/03/2015] [Accepted: 09/07/2015] [Indexed: 11/16/2022] Open
Abstract
The landscape of advanced non-small lung cancer (NSCLC) therapies has rapidly been evolving beyond chemotherapy over the last few years. The discovery of oncogenic driver mutations has led to new ways in classifying NSCLC as well as offered novel therapeutic targets for anticancer therapy. Targets such as epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) gene rearrangements have successfully been targeted with appropriate tyrosine kinase inhibitors (TKIs). Other driver mutations such as ROS, MET, RET, BRAF have also been investigated with targeted agents with some success in the early phase clinical setting. Novel strategies in the field of immune-oncology have also led to the development of inhibitors of cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed death-1 receptor (PD-1), which are important pathways in allowing cancer cells to escape detection by the immune system. These inhibitors have been successfully tried in NSCLC and also now bring the exciting possibility of long term responses in advanced NSCLC. In this review recent data on novel targets and therapeutic strategies and their future prospects are discussed.
Collapse
Affiliation(s)
- Vishal Boolell
- Department of Medical Oncology, Monash Medical Centre, 823-865 Centre Road, East Bentleigh VIC 3165, Australia.
- Hudson Institute of Medical Research, Monash University, 27-31 Wright Street, Clayton VIC 3168, Australia.
| | - Muhammad Alamgeer
- Department of Medical Oncology, Monash Medical Centre, 823-865 Centre Road, East Bentleigh VIC 3165, Australia.
- Hudson Institute of Medical Research, Monash University, 27-31 Wright Street, Clayton VIC 3168, Australia.
| | - David N Watkins
- Hudson Institute of Medical Research, Monash University, 27-31 Wright Street, Clayton VIC 3168, Australia.
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney NSW 2010, Australia.
- UNSW Faculty of Medicine, St Vincent's Clinical School, 390 Victoria Street, Darlinghurst, Sydney NSW 2010, Australia.
- Department of Thoracic Medicine, St Vincent's Hospital, 390 Victoria Street, Darlinghurst, Sydney NSW 2010, Australia:.
| | - Vinod Ganju
- Department of Medical Oncology, Monash Medical Centre, 823-865 Centre Road, East Bentleigh VIC 3165, Australia.
- Hudson Institute of Medical Research, Monash University, 27-31 Wright Street, Clayton VIC 3168, Australia.
| |
Collapse
|
257
|
The Evolution of Therapies in Non-Small Cell Lung Cancer. Cancers (Basel) 2015. [PMID: 26371045 DOI: 10.3390/cancers7030864+cancers7030864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The landscape of advanced non-small lung cancer (NSCLC) therapies has rapidly been evolving beyond chemotherapy over the last few years. The discovery of oncogenic driver mutations has led to new ways in classifying NSCLC as well as offered novel therapeutic targets for anticancer therapy. Targets such as epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) gene rearrangements have successfully been targeted with appropriate tyrosine kinase inhibitors (TKIs). Other driver mutations such as ROS, MET, RET, BRAF have also been investigated with targeted agents with some success in the early phase clinical setting. Novel strategies in the field of immune-oncology have also led to the development of inhibitors of cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed death-1 receptor (PD-1), which are important pathways in allowing cancer cells to escape detection by the immune system. These inhibitors have been successfully tried in NSCLC and also now bring the exciting possibility of long term responses in advanced NSCLC. In this review recent data on novel targets and therapeutic strategies and their future prospects are discussed.
Collapse
|
258
|
Uguen A, Marcorelles P, De Braekeleer M. Searching for ROS1 Rearrangements in Lung Cancer by Fluorescent In Situ Hybridization: The Importance of Probe Design. J Thorac Oncol 2015. [DOI: 10.1097/jto.0000000000000605] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
259
|
Zhu Q, Zhan P, Zhang X, Lv T, Song Y. Clinicopathologic characteristics of patients with ROS1 fusion gene in non-small cell lung cancer: a meta-analysis. Transl Lung Cancer Res 2015. [PMID: 26207220 DOI: 10.3978/j.issn.2218-6751.2015.05.01] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The receptor tyrosine kinase ROS1 is a driver gene in the non-small cell lung cancer (NSCLC) with promising target treatment potential. The clinical features of NSCLC patients harboring ROS1 fusion gene were not fully understood due to small-to-modest sample sizes of these association studies. METHODS We systematically searched PubMed, EMBASE, and the Cochrane Central Register of Controlled Trials (CENTRAL) from their inception to March 31, 2015. We analyzed the association between ROS1 fusion genes and four common clinical variables, i.e., gender, smoking status, pathological type and clinical stage. RESULTS Eighteen studies consisting of 9,898 NSCLC patients were included in this meta-analysis. Pooled results showed that significantly higher rate of ROS1 fusion gene was detected in female NSCLC patients (OR =1.54, 95% CI: 1.02-2.34, P=0.042), patients without a smoking history (OR =3.27, 95% CI: 1.44-7.45, P=0.005), patients with adenocarcinomas NSCLC (OR =10.24, 95% CI: 5.13-20.40, P<0.001), and patients with an advanced clinical stages III-IV (OR =2.57, 95% CI: 1.78-3.71, P<0.001). The pooled prevalence of ROS1 fusion gene was 2.4% (95% CI: 1.8-3.1%) in adenocarcinoma and a significantly lower (0.2%) in non-adenocarcinoma tumors. CONCLUSIONS ROS1 rearrangement was more prevalent in female patients, patients without a smoking history, patients with adenocarcinoma, and patients on more advanced stages (stages III to IV).
Collapse
Affiliation(s)
- Qingqing Zhu
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medical, Nanjing 210002, China ; 2 Department of Cardiology, Drum Tower Hospital, Nanjing University School of Medical, Nanjing 210002, China
| | - Ping Zhan
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medical, Nanjing 210002, China ; 2 Department of Cardiology, Drum Tower Hospital, Nanjing University School of Medical, Nanjing 210002, China
| | - Xinlin Zhang
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medical, Nanjing 210002, China ; 2 Department of Cardiology, Drum Tower Hospital, Nanjing University School of Medical, Nanjing 210002, China
| | - Tangfeng Lv
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medical, Nanjing 210002, China ; 2 Department of Cardiology, Drum Tower Hospital, Nanjing University School of Medical, Nanjing 210002, China
| | - Yong Song
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medical, Nanjing 210002, China ; 2 Department of Cardiology, Drum Tower Hospital, Nanjing University School of Medical, Nanjing 210002, China
| |
Collapse
|
260
|
Targeted therapies for patients with advanced NSCLC harboring wild-type EGFR: what's new and what's enough. CHINESE JOURNAL OF CANCER 2015; 34:310-9. [PMID: 26187152 PMCID: PMC4593374 DOI: 10.1186/s40880-015-0036-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/25/2015] [Indexed: 01/05/2023]
Abstract
Historically, non-small cell lung cancer (NSCLC) is divided into squamous and nonsquamous subtypes based on histologic features. With a growing number of oncogenic drivers being identified in squamous and nonsquamous NSCLC, this malignancy has been recently divided into several distinct subtypes according to the specific molecular alterations. This new paradigm has substantially highlighted the treatment of advanced NSCLC, shifting it from standard chemotherapy according to specific histologic subtypes to targeted therapy according to specific oncogenic drivers. The application of epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) in NSCLC patients harboring activating EGFR mutations has been a representative model of precise medicine in the treatment of NSCLC. As the role of EGFR-TKIs in routine management of patients with advanced NSCLC has been well established, this review provides an overview of alternative targeted therapy in the treatment of NSCLC, including EGFR-TKIs for patients with wild-type EGFR NSCLC, as well as other targeted agents either clinical available or in early- to late-stage development.
Collapse
|
261
|
Kumar M, Ernani V, Owonikoko TK. Biomarkers and targeted systemic therapies in advanced non-small cell lung cancer. Mol Aspects Med 2015; 45:55-66. [PMID: 26187108 DOI: 10.1016/j.mam.2015.06.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 06/24/2015] [Indexed: 01/15/2023]
Abstract
The last decade has witnessed significant growth in therapeutic options for patients diagnosed with lung cancer. This is due in major part to our improved technological ability to interrogate the genomics of cancer cells, which has enabled the development of biologically rational anticancer agents. The recognition that lung cancer is not a single disease entity dates back many decades to the histological subclassification of malignant neoplasms of the lung into subcategories of small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). While SCLC continues to be regarded as a single histologic and therapeutic category, the NSCLC subset has undergone additional subcategorizations with distinct management algorithms for specific histologic and molecular subtypes. The defining characteristics of these NSCLC subtypes have evolved into important tools for prognosis and for predicting the likelihood of benefit when patients are treated with anticancer agents.
Collapse
Affiliation(s)
- Mukesh Kumar
- Department of Hematology & Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Vinicius Ernani
- Department of Hematology & Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Taofeek K Owonikoko
- Department of Hematology & Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
262
|
Genestreti G, Grossi F, Genova C, Burgio MA, Bongiovanni A, Gavelli G, Bartolotti M, Di Battista M, Cavallo G, Brandes AA. Third- and further-line therapy in advanced non-small-cell lung cancer patients: an overview. Future Oncol 2015; 10:2081-96. [PMID: 25396779 DOI: 10.2217/fon.14.96] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) treatment has led to improved efficacy and compliance due to individual tailoring of the therapeutic options and the use of strategies based on both clinical characteristics and histological and biological features of the disease. In nonsquamous NSCLC, novel agents, such as pemetrexed and bevacizumab, have improved survival in the first-line setting. Maintenance therapy with pemetrexed and erlotinib resulted in improved progression-free survival compared with second-line therapy at disease progression. In the second-line setting, pemetrexed improves survival in nonsquamous NSCLC compared with docetaxel, and erlotinib has shown a survival benefit compared with best supportive care in patients who did not previously receive an EGF receptor inhibitor. Although the benefit of first- and second-line treatment over best supportive care alone has been firmly established, the role of further-line treatment remains controversial. This article summarizes the state-of-the-art treatments in this setting.
Collapse
Affiliation(s)
- Giovenzio Genestreti
- Department of Medical Oncology, Bellaria Hospital - IRCCS Institute of Neurological Sciences, Azienda USL, Via Altura 3, 47841 Bologna, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
263
|
Fang B, Mehran RJ, Heymach JV, Swisher SG. Predictive biomarkers in precision medicine and drug development against lung cancer. CHINESE JOURNAL OF CANCER 2015; 34:295-309. [PMID: 26134262 PMCID: PMC4593363 DOI: 10.1186/s40880-015-0028-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/04/2015] [Indexed: 02/06/2023]
Abstract
The molecular characterization of various cancers has shown that cancers with the same origins, histopathologic diagnoses, and clinical stages can be highly heterogeneous in their genetic and epigenetic alterations that cause tumorigenesis. A number of cancer driver genes with functional abnormalities that trigger malignant transformation and that are required for the survival of cancer cells have been identified. Therapeutic agents targeting some of these cancer drivers have been successfully developed, resulting in substantial improvements in clinical symptom amelioration and outcomes in a subset of cancer patients. However, because such therapeutic drugs often benefit only a limited number of patients, the successes of clinical development and applications rely on the ability to identify those patients who are sensitive to the targeted therapies. Thus, biomarkers that can predict treatment responses are critical for the success of precision therapy for cancer patients and of anticancer drug development. This review discusses the molecular heterogeneity of lung cancer pathogenesis; predictive biomarkers for precision medicine in lung cancer therapy with drugs targeting epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-ros oncogene 1 receptor tyrosine kinase (ROS1), and immune checkpoints; biomarkers associated with resistance to these therapeutics; and approaches to identify predictive biomarkers in anticancer drug development. The identification of predictive biomarkers during anticancer drug development is expected to greatly facilitate such development because it will increase the chance of success or reduce the attrition rate. Additionally, such identification will accelerate the drug approval process by providing effective patient stratification strategies in clinical trials to reduce the sample size required to demonstrate clinical benefits.
Collapse
Affiliation(s)
- Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Reza J Mehran
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - John V Heymach
- Department of Thoracic and Head/Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Stephen G Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
264
|
Wu S, Wang J, Zhou L, Su D, Liu Y, Liang X, Zhang S, Zeng X. Clinicopathological characteristics and outcomes of ROS1-rearranged patients with lung adenocarcinoma without EGFR, KRAS mutations and ALK rearrangements. Thorac Cancer 2015; 6:413-20. [PMID: 26273395 PMCID: PMC4511318 DOI: 10.1111/1759-7714.12191] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/08/2014] [Indexed: 11/28/2022] Open
Abstract
Background c-ros oncogene 1 (ROS1) rearrangement presents one of the newest molecular targets in non-small cell lung cancer (NSCLC). ROS1 rearrangement is predominantly found in adenocarcinoma cases and is exclusive to other oncogenes, such as epidermal growth factor receptor (EGFR), Kirsten rat sarcoma viral oncogene homolog (KRAS), and anaplastic lymphoma kinase (ALK). The aim of this study was to investigate the clinicopathological characteristics and outcomes of ROS1-rearranged patients with lung adenocarcinoma without EGFR and KRAS mutations and ALK rearrangements. Methods Wild-type EGFR/KRAS/ALK patients with lung adenocarcinoma were selected from Beijing Chest Hospital. Specimens were conducted in tissue microarrays. ROS1 rearrangement was screened using fluorescence in situ hybridization. Results Our study included 127 patients with lung adenocarcinoma without EGFR and KRAS mutations and ALK rearrangements. ROS1 rearrangement was detected in five (3.9%) of the 127 patients. Compared with ROS1-negative patients, the positive rate of ROS1 in female patients was significantly higher than in male patients (9.8% vs. 0.0%, P = 0.009). There were no differences in age, smoking status, stage or histological subtype between ROS1-positive and ROS1-negative patients. No significant difference in survival was detected between the ROS1-positive and ROS1-negative patients. Conclusions ROS1 rearrangement is a rare subset of lung adenocarcinoma. In 127 patients with lung adenocarcinoma, 3.9% of ROS1-positive patients with wild-type EGFR/KRAS/ALK were found.
Collapse
Affiliation(s)
- Shafei Wu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College Beijing, China
| | - Jinghui Wang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute Beijing, China
| | - Lijuan Zhou
- Department of Pathology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute Beijing, China
| | - Dan Su
- Department of Pathology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute Beijing, China
| | - Yuanyuan Liu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College Beijing, China
| | - Xiaolong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College Beijing, China
| | - Shucai Zhang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute Beijing, China
| | - Xuan Zeng
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College Beijing, China
| |
Collapse
|
265
|
McCoach CE, Doebele RC. The minority report: targeting the rare oncogenes in NSCLC. Curr Treat Options Oncol 2015; 15:644-57. [PMID: 25228144 DOI: 10.1007/s11864-014-0310-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OPINION STATEMENT Lung cancer is still responsible for the highest number of cancer deaths worldwide. Despite this fact, significant progress has been made in the treatment of non-small cell lung cancer (NSCLC). Specifically, efforts to identify and treat genetic alterations (gene mutations, gene fusions, gene amplification events, etc.) that result in oncogenic drivers are now standard of care (EGFR and ALK) or an intense area of research. The most prevalent oncogenic drivers have likely already been identified; thus, there is now a focus on subgroups of tumors with less common genetic alterations. Interestingly, as we explore these less common mutations, we are discovering that many occur across other tumor types (i.e., non-lung cancer), further justifying their study. Furthermore, many studies have demonstrated that by searching broadly for multiple genetic alterations in large subsets of patients they are able to identify potentially targetable alterations in the majority of patients. Although individually, the rare oncogenic drivers subgroups may seem to occur too infrequently to justify their exploration, the fact that the majority of patients with NSCLC harbor a potentially actionable driver mutation within their tumors and the fact that different types of cancers often have the same oncogenic driver justifies this approach.
Collapse
Affiliation(s)
- Caroline E McCoach
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, 12801 E. 17th Avenue, Aurora, CO, 80045, USA
| | | |
Collapse
|
266
|
Daga A, Ansari A, Patel S, Mirza S, Rawal R, Umrania V. Current Drugs and Drug Targets in Non-Small Cell Lung Cancer: Limitations and Opportunities. Asian Pac J Cancer Prev 2015; 16:4147-56. [DOI: 10.7314/apjcp.2015.16.10.4147] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
267
|
Rothschild SI. Targeted Therapies in Non-Small Cell Lung Cancer-Beyond EGFR and ALK. Cancers (Basel) 2015; 7:930-49. [PMID: 26018876 PMCID: PMC4491691 DOI: 10.3390/cancers7020816] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/07/2015] [Accepted: 05/13/2015] [Indexed: 01/30/2023] Open
Abstract
Systemic therapy for non-small cell lung cancer (NSCLC) has undergone a dramatic paradigm shift over the past decade. Advances in our understanding of the underlying biology of NSCLC have revealed distinct molecular subtypes. A substantial proportion of NSCLC depends on oncogenic molecular aberrations (so-called "driver mutations") for their malignant phenotype. Personalized therapy encompasses the strategy of matching these subtypes with effective targeted therapies. EGFR mutations and ALK translocation are the most effectively targeted oncogenes in NSCLC. EGFR mutations and ALK gene rearrangements are successfully being targeted with specific tyrosine kinase inhibitors. The number of molecular subgroups of NSCLC continues to grow. The scope of this review is to discuss recent data on novel molecular targets as ROS1, BRAF, KRAS, HER2, c-MET, RET, PIK3CA, FGFR1 and DDR2. Thereby the review will focus on therapeutic strategies targeting these aberrations. Moreover, the emerging challenge of acquired resistance to initially effective therapies will be discussed.
Collapse
Affiliation(s)
- Sacha I Rothschild
- Medical Oncology, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland.
| |
Collapse
|
268
|
Clinical and the prognostic characteristics of lung adenocarcinoma patients with ROS1 fusion in comparison with other driver mutations in East Asian populations. J Thorac Oncol 2015; 9:1171-9. [PMID: 25157770 DOI: 10.1097/jto.0000000000000232] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The prevalence, demographic features, and clinical outcomes of lung adenocarcinoma patients with novel ROS1 oncogenic rearrangement in East Asian populations are not clear. This study aimed to investigate the clinical and prognostic characteristics of lung adenocarcinoma in patients with ROS1 fusion compared with other driver mutations. METHODS Multiplex reverse transcription-polymerase chain reaction was used to detect the ROS1 fusion gene in lung adenocarcinoma cases. Immunohistochemistry was used to confirm the expression of ROS1. The demographic data and clinical outcomes of patients with the ROS1 fusion gene were compared with those of patients without the ROS1 fusion gene, including those with the EGFR mutation, EML4-ALK fusion, KRAS mutation, and quadruple-negative patients. RESULTS Of 492 patients with lung adenocarcinoma, 12 (2.4%) had the ROS1 fusion gene. Their median age was 45.0 years, significantly younger than that of the ROS1 fusion-negative cohorts (p < 0.001). Acinar (including cribriform) and solid patterns were the two most common histologic subtypes in the ROS1 fusion tumors (7 of 12, 58.3%) and were predominantly seen in CD74-ROS1 fusion tumors (66.7%). There was no significant survival difference between the ROS1 fusion-positive and ROS1 fusion-negative cohorts in surgical group, but ROS1 fusion-positive patients might have worse outcomes than EGFR-mutant patients in the stage IV group. CONCLUSIONS The ROS1 fusion gene can be successfully detected in East Asian patients with lung adenocarcinoma using multiplex reverse transcription-polymerase chain reaction. These patients tend to be younger and have characteristic histologic subtypes. Due to the small number of ROS1 fusion patients, the prognostic value of ROS1 fusion need further studies to confirm.
Collapse
|
269
|
Fu S, Liang Y, Lin YB, Wang F, Huang MY, Zhang ZC, Wang J, Cen WJ, Shao JY. The Frequency and Clinical Implication of ROS1 and RET Rearrangements in Resected Stage IIIA-N2 Non-Small Cell Lung Cancer Patients. PLoS One 2015; 10:e0124354. [PMID: 25905642 PMCID: PMC4408029 DOI: 10.1371/journal.pone.0124354] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 02/27/2015] [Indexed: 11/19/2022] Open
Abstract
To evaluate the frequency and clinicopathological features of ROS1 and RET rearrangements in N2 node positive stage IIIA (IIIA-N2) non-small cell lung cancer (NSCLC) patients, we retrospectively screened 204 cases with a tissue microarray (TMA) panel by fluorescent in situ hybridization (FISH), and confirmed by direct sequencing and immunohistochemistry (IHC). The relationship between ROS1 or RET rearrangements, clinicopathological features, and prognostic factors were analyzed in resected stage IIIA-N2 NSCLC. Of the 204 cases, 4 cases were confirmed with ROS1 rearrangement, but no RET rearrangement was detected. All 4 ROS1-rearranged cases were adenocarcinomas. The predominant pathological type was acinar pattern in ROS1-rearranged tumors, except for 1 case harboring a mixture acinar and mucous tumor cells. Variants of ROS1 rearrangement were SDC4-ROS1 (E2:E32), SDC4-ROS1 (E4:E32) and SDC4-ROS1 (E4:E34). There was no significant association between ROS1 rearrangement and clinicopathological characteristics. In this cohort, multivariate analysis for overall survival (OS) indicated that squamous cell carcinoma and lobectomy were independent predictors of poor prognosis; R0 surgical resection and non-pleural invasion were independent predictors of good prognosis. In resected stage IIIA-N2 NSCLC patients, ROS1-rearranged cases tended to occur in younger patients with adenocarcinomas. The prognosis of resected stage IIIA-N2 is generally considered poor, but patients with ROS1 rearrangement will benefit from the targeted therapy.
Collapse
Affiliation(s)
- Sha Fu
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Ying Liang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yong-Bin Lin
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Fang Wang
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Ma-Yan Huang
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Zi-Chen Zhang
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Jing Wang
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Wen-Jian Cen
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Jian-Yong Shao
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
- * E-mail:
| |
Collapse
|
270
|
Kohno T, Nakaoku T, Tsuta K, Tsuchihara K, Matsumoto S, Yoh K, Goto K. Beyond ALK-RET, ROS1 and other oncogene fusions in lung cancer. Transl Lung Cancer Res 2015; 4:156-64. [PMID: 25870798 DOI: 10.3978/j.issn.2218-6751.2014.11.11] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 10/28/2014] [Indexed: 01/08/2023]
Abstract
Fusions of the RET and ROS1 protein tyrosine kinase oncogenes with several partner genes were recently identified as new targetable genetic aberrations in cases of non-small cell lung cancer (NSCLC) lacking activating EGFR, KRAS, ALK, BRAF, or HER2 oncogene aberrations. RET and ROS1 fusion-positive tumors are mainly observed in young, female, and/or never smoking patients. Studies based on in vitro and in vivo (i.e., mouse) models and studies of several fusion-positive patients indicate that inhibiting the kinase activity of the RET and ROS1 fusion proteins is a promising therapeutic strategy. Accordingly, there are several ongoing clinical trials aimed at examining the efficacy of tyrosine kinase inhibitors (TKIs) against RET and ROS1 proteins in patients with fusion-positive lung cancer. Other gene fusions (NTRK1, NRG1, and FGFR1/2/3) that are targetable by existing TKIs have also been identified in NSCLCs. Options for personalized lung cancer therapy will be increased with the help of multiplex diagnosis systems able to detect multiple druggable gene fusions.
Collapse
Affiliation(s)
- Takashi Kohno
- 1 Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan ; 2 Division of Translational Research, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo and Chiba, Japan ; 3 Division of Thoracic Oncology, National Cancer Center Hospital East, Chiba, Japan ; 4 Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Takashi Nakaoku
- 1 Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan ; 2 Division of Translational Research, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo and Chiba, Japan ; 3 Division of Thoracic Oncology, National Cancer Center Hospital East, Chiba, Japan ; 4 Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Koji Tsuta
- 1 Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan ; 2 Division of Translational Research, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo and Chiba, Japan ; 3 Division of Thoracic Oncology, National Cancer Center Hospital East, Chiba, Japan ; 4 Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Katsuya Tsuchihara
- 1 Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan ; 2 Division of Translational Research, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo and Chiba, Japan ; 3 Division of Thoracic Oncology, National Cancer Center Hospital East, Chiba, Japan ; 4 Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Shingo Matsumoto
- 1 Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan ; 2 Division of Translational Research, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo and Chiba, Japan ; 3 Division of Thoracic Oncology, National Cancer Center Hospital East, Chiba, Japan ; 4 Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Kiyotaka Yoh
- 1 Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan ; 2 Division of Translational Research, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo and Chiba, Japan ; 3 Division of Thoracic Oncology, National Cancer Center Hospital East, Chiba, Japan ; 4 Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Koichi Goto
- 1 Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan ; 2 Division of Translational Research, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo and Chiba, Japan ; 3 Division of Thoracic Oncology, National Cancer Center Hospital East, Chiba, Japan ; 4 Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
271
|
Saito M, Shimada Y, Shiraishi K, Sakamoto H, Tsuta K, Totsuka H, Chiku S, Ichikawa H, Kato M, Watanabe SI, Yoshida T, Yokota J, Kohno T. Development of Lung Adenocarcinomas with Exclusive Dependence on Oncogene Fusions. Cancer Res 2015; 75:2264-71. [DOI: 10.1158/0008-5472.can-14-3282] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 03/13/2015] [Indexed: 11/16/2022]
|
272
|
Zhou C. Lung cancer molecular epidemiology in China: recent trends. Transl Lung Cancer Res 2015; 3:270-9. [PMID: 25806311 DOI: 10.3978/j.issn.2218-6751.2014.09.01] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/01/2014] [Indexed: 12/19/2022]
Abstract
Lung cancer is both the most common diagnosed cancer and the leading cause of cancer related deaths in China. During the past three decades, the incidence and mortality of lung cancer in China are increasing rapidly. According to data from National Central Cancer Registry (NCCR) in 2010, the crude incidence of lung cancer in China was 46.08 per 100,000 population (61.86 per 100,000 men and 29.54 per 100,000 women), with an estimated over 600,000 new diagnosed lung cancer patients (416,333 males and 189,613 females). Meanwhile, the crude mortality of lung cancer in China was 37.00 per 100,000 population (50.04 per 100,000 men and 23.33 per 100,000 women). Consistent with the change in developed countries, adenocarcinoma has become the most predominant histological subtype of lung cancer in China. For the majority advanced non-small-cell lung cancer (NSCLC) patients, especially patients with adenocarcinoma, targeted therapy became increasing important in the treatment. Chinese researcher have done a lot work in terms of lung cancer molecular epidemiology, therefore, in this review, we further summarized the epidemiology of driver genes in NSCLC, hoping to help clinicians to better screen certain driver genes in China for treatment decisions.
Collapse
Affiliation(s)
- Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| |
Collapse
|
273
|
Shan L, Lian F, Guo L, Qiu T, Ling Y, Ying J, Lin D. Detection of ROS1 gene rearrangement in lung adenocarcinoma: comparison of IHC, FISH and real-time RT-PCR. PLoS One 2015; 10:e0120422. [PMID: 25742289 PMCID: PMC4351102 DOI: 10.1371/journal.pone.0120422] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 01/07/2015] [Indexed: 01/05/2023] Open
Abstract
Aims To compare fluorescence in situ hybridization (FISH), immunohistochemistry (IHC) and quantitative real-time reverse transcription-PCR (qRT-PCR) assays for detection of ROS1 fusion in a large number of ROS1-positive lung adenocatcinoma (ADC) patients. Methods Using IHC analysis, sixty lung ADCs including 16 cases with ROS1 protein expression and 44 cases without ROS1 expression were selected for this study. The ROS1 fusion status was examined by FISH and qRT-PCR assay. Results Among 60 cases, 16 (26.7%), 13 (21.7%) and 20 (33.3%) cases were ROS1 positive revealed by IHC, FISH and qRT-PCR, respectively. Using FISH as a standard method for ROS1 fusion detection, the sensitivity and specificity of IHC were 100% and 93.6%, respectively. Three IHC-positive cases, which showed FISH negative, were demonstrated with ROS1 fusion by qRT-PCR analysis. The sensitivity and specificity of qRT-PCR for detection for ROS1 fusion were 100% and 85.1%, respectively. The total concordance rate between IHC and qRT-PCR were 93.3%. Conclusion IHC is a reliable and rapid screening tool in routine pathologic laboratories for the identification of suitable candidates for ROS1-targeted therapy. Some ROS1 IHC-positive but FISH-negative cases did harbor the translocation events and may benefit from crizotinib.
Collapse
Affiliation(s)
- Ling Shan
- Department of Pathology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Fang Lian
- Department of Pathology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lei Guo
- Department of Pathology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Tian Qiu
- Department of Pathology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yun Ling
- Department of Pathology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jianming Ying
- Department of Pathology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- * E-mail: (DL); (JY)
| | - Dongmei Lin
- Department of Pathology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- * E-mail: (DL); (JY)
| |
Collapse
|
274
|
Haghgoo SM, Allameh A, Mortaz E, Garssen J, Folkerts G, Barnes PJ, Adcock IM. Pharmacogenomics and targeted therapy of cancer: Focusing on non-small cell lung cancer. Eur J Pharmacol 2015; 754:82-91. [PMID: 25725115 DOI: 10.1016/j.ejphar.2015.02.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 02/11/2015] [Accepted: 02/17/2015] [Indexed: 12/20/2022]
Abstract
Recent studies have been established high degree of genetic diversity in solid organ tumors among individuals and even between individual tumor cells. This intratumor and intertumor genetic diversity results in a heterogeneous tumor with unique characteristics which potentially allows effective drug therapy. The goal of pharmacogenomics is to elucidate the genetic network(s) that underlie drug efficacy and drug resistance. Advances in targeted and personalized therapy play an increasingly important role in many common cancers, notably lung cancer, due to the high incidence, prevalence, mortality and the greater tendency towards drug resistance seen in these patients. Non-small cell lung cancer (NSCLC) is characterized by mutations in the epidermal growth factor receptor (EGFR) and or downstream kinase pathways. This has led to the development of highly selective monoclonal antibodies and EGFR tyrosine kinase inhibitors (EGFR-TKIs) to prevent cancer initiation, proliferation, differentiation, angiogenesis, survival, and invasion. However, resistance to many of these new treatments is induced and further pharmacogenomic analysis has revealed mutations associated with increased or reduced drug efficacy. Combinations of kinase inhibitors or potentially the targeting of cancer stem cells may further increase the success of pharmacogenomics in treating patients with lung cancer.
Collapse
Affiliation(s)
- Seyyed Mortaza Haghgoo
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Esmaeil Mortaz
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK; Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Chronic Respiratory Diseases Research Center and National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Masih Daneshvari Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands; Nutricia Research, Immunology, Utrecht, The Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Peter J Barnes
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Ian M Adcock
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
275
|
Mazières J, Zalcman G, Crinò L, Biondani P, Barlesi F, Filleron T, Dingemans AMC, Léna H, Monnet I, Rothschild SI, Cappuzzo F, Besse B, Thiberville L, Rouvière D, Dziadziuszko R, Smit EF, Wolf J, Spirig C, Pecuchet N, Leenders F, Heuckmann JM, Diebold J, Milia JD, Thomas RK, Gautschi O. Crizotinib therapy for advanced lung adenocarcinoma and a ROS1 rearrangement: results from the EUROS1 cohort. J Clin Oncol 2015; 33:992-9. [PMID: 25667280 DOI: 10.1200/jco.2014.58.3302] [Citation(s) in RCA: 279] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Approximately 1% of lung adenocarcinomas are driven by oncogenic ROS1 rearrangement. Crizotinib is a potent inhibitor of both ROS1 and ALK kinase domains. PATIENTS AND METHODS In the absence of a prospective clinical trial in Europe, we conducted a retrospective study in centers that tested for ROS1 rearrangement. Eligible patients had stage IV lung adenocarcinoma, had ROS1 rearrangement according to fluorescent in situ hybridization, and had received crizotinib therapy through an individual off-label use. Best response was assessed locally using RECIST (version 1.1). All other data were analyzed centrally. RESULTS We identified 32 eligible patients. One patient was excluded because next-generation sequencing was negative for ROS1 fusion. Median age was 50.5 years, 64.5% of patients were women, and 67.7% were never-smokers. Thirty patients were evaluable for progression-free survival (PFS), and 29 patients were evaluable for best response. We observed four patients with disease progression, two patients with stable disease, and objective response in 24 patients, including five complete responses (overall response rate, 80%; disease control rate, 86.7%). Median PFS was 9.1 months, and the PFS rate at 12 months was 44%. No unexpected adverse effects were observed. Twenty-six patients received pemetrexed (either alone or in combination with platinum and either before or after crizotinib) and had a response rate of 57.7% and a median PFS of 7.2 months. CONCLUSION Crizotinib was highly active at treating lung cancer in patients with a ROS1 rearrangement, suggesting that patients with lung adenocarcinomas should be tested for ROS1. Prospective clinical trials with crizotinib and other ROS1 inhibitors are ongoing or planned.
Collapse
Affiliation(s)
- Julien Mazières
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany.
| | - Gérard Zalcman
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Lucio Crinò
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Pamela Biondani
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Fabrice Barlesi
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Thomas Filleron
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Anne-Marie C Dingemans
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Hervé Léna
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Isabelle Monnet
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Sacha I Rothschild
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Federico Cappuzzo
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Benjamin Besse
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Luc Thiberville
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Damien Rouvière
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Rafal Dziadziuszko
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Egbert F Smit
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Jurgen Wolf
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Christian Spirig
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Nicolas Pecuchet
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Frauke Leenders
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Johannes M Heuckmann
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Joachim Diebold
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Julie D Milia
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Roman K Thomas
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| | - Oliver Gautschi
- Julien Mazières, Damien Rouvière, and Julie D. Milia, Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier; Thomas Filleron, Institut Universitaire du Cancer, Toulouse; Gérard Zalcman, Centre Hospitalier Universitaire, Université de Caen-Basse Normandie, Caen; Pamela Biondani and Benjamin Besse, Gustave Roussy, Villejuif; Fabrice Barlesi, Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille; Hervé Léna, Centre Hospitalier Universitaire, Rennes; Isabelle Monnet, Centre Hospitalier Intercommunal de Créteil, Créteil; Luc Thiberville, Centre Hospitalier Universitaire, Rouen; Nicolas Pecuchet, Institut Curie, Paris, France; Lucio Crinò, Medica-Azienda Ospedaliera, Perugia; Federico Cappuzzo, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy; Anne-Marie C. Dingemans, Maastricht University Medical Center, Maastricht; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Sacha I. Rothschild, University Hospital Basel, Medical Oncology, Basel; Christian Spirig, Klinik St Anna; Joachim Diebold and Oliver Gautschi, Cantonal Hospital Luzern, Lucerne, Switzerland; Rafal Dziadziuszko, Gdansk Medical University, Gdansk, Poland; Jurgen Wolf and Roman K. Thomas, Center of Integrated Oncology Köln-Bonn, University Hospital Cologne, University of Cologne; Frauke Leenders and Roman K. Thomas, University of Cologne; Johannes M. Heuckmann, Blackfield AG, Cologne, Germany
| |
Collapse
|
276
|
Lopez-Chavez A, Thomas A, Rajan A, Raffeld M, Morrow B, Kelly R, Carter CA, Guha U, Killian K, Lau CC, Abdullaev Z, Xi L, Pack S, Meltzer PS, Corless CL, Sandler A, Beadling C, Warrick A, Liewehr DJ, Steinberg SM, Berman A, Doyle A, Szabo E, Wang Y, Giaccone G. Molecular profiling and targeted therapy for advanced thoracic malignancies: a biomarker-derived, multiarm, multihistology phase II basket trial. J Clin Oncol 2015; 33:1000-7. [PMID: 25667274 DOI: 10.1200/jco.2014.58.2007] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
PURPOSE We conducted a basket clinical trial to assess the feasibility of such a design strategy and to independently evaluate the effects of multiple targeted agents against specific molecular aberrations in multiple histologic subtypes concurrently. PATIENTS AND METHODS We enrolled patients with advanced non-small-cell lung cancer (NSCLC), small-cell lung cancer, and thymic malignancies who underwent genomic characterization of oncogenic drivers. Patients were enrolled onto a not-otherwise-specified arm and treated with standard-of-care therapies or one of the following five biomarker-matched treatment groups: erlotinib for EGFR mutations; selumetinib for KRAS, NRAS, HRAS, or BRAF mutations; MK2206 for PIK3CA, AKT, or PTEN mutations; lapatinib for ERBB2 mutations or amplifications; and sunitinib for KIT or PDGFRA mutations or amplification. RESULTS Six hundred forty-seven patients were enrolled, and 88% had their tumors tested for at least one gene. EGFR mutation frequency was 22.1% in NSCLC, and erlotinib achieved a response rate of 60% (95% CI, 32.3% to 83.7%). KRAS mutation frequency was 24.9% in NSCLC, and selumetinib failed to achieve its primary end point, with a response rate of 11% (95% CI, 0% to 48%). Completion of accrual to all other arms was not feasible. In NSCLC, patients with EGFR mutations had the longest median survival (3.51 years; 95% CI, 2.89 to 5.5 years), followed by those with ALK rearrangements (2.94 years; 95% CI, 1.66 to 4.61 years), those with KRAS mutations (2.3 years; 95% CI, 2.3 to 2.17 years), those with other genetic abnormalities (2.17 years; 95% CI, 1.3 to 2.74 years), and those without an actionable mutation (1.85 years; 95% CI, 1.61 to 2.13 years). CONCLUSION This basket trial design was not feasible for many of the arms with rare mutations, but it allowed the study of the genetics of less common malignancies.
Collapse
Affiliation(s)
- Ariel Lopez-Chavez
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Anish Thomas
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Arun Rajan
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Mark Raffeld
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Betsy Morrow
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Ronan Kelly
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Corey Allan Carter
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Udayan Guha
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Keith Killian
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Christopher C Lau
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Zied Abdullaev
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Liqiang Xi
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Svetlana Pack
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Paul S Meltzer
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Christopher L Corless
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Alan Sandler
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Carol Beadling
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Andrea Warrick
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - David J Liewehr
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Seth M Steinberg
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Arlene Berman
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Austin Doyle
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Eva Szabo
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Yisong Wang
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Giuseppe Giaccone
- Ariel Lopez-Chavez, Anish Thomas, Arun Rajan, Mark Raffeld, Betsy Morrow, Ronan Kelly, Corey Allan Carter, Udayan Guha, Keith Killian, Christopher C. Lau, Zied Abdullaev, Liqiang Xi, Svetlana Pack, Paul S. Meltzer, David J. Liewehr, Seth M. Steinberg, Arlene Berman, Eva Szabo, Yisong Wang, and Giuseppe Giaccone, National Cancer Institute; Austin Doyle, Cancer Therapy Evaluation Program, Bethesda, MD; Ariel Lopez-Chavez, Christopher L. Corless, Alan Sandler, Carol Beadling, and Andrea Warrick, Knight Cancer Institute, Oregon Health and Science University, Portland, OR; Ariel Lopez-Chavez, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; and Yisong Wang and Giuseppe Giaccone, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC.
| |
Collapse
|
277
|
Solomon B. Validating ROS1 rearrangements as a therapeutic target in non-small-cell lung cancer. J Clin Oncol 2015; 33:972-4. [PMID: 25667277 DOI: 10.1200/jco.2014.59.8334] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Benjamin Solomon
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| |
Collapse
|
278
|
Coexistence of three variants involving two different fusion partners of ROS1 including a novel variant of ROS1 fusions in lung adenocarcinoma: a case report. J Thorac Oncol 2015; 9:e43-6. [PMID: 24828671 DOI: 10.1097/jto.0000000000000118] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
279
|
Pulmonary adenocarcinoma with signet ring cell features: a comprehensive study from 3 distinct patient cohorts. Am J Surg Pathol 2015; 38:1681-8. [PMID: 25007143 DOI: 10.1097/pas.0000000000000280] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Comprehensive biological characteristics of pulmonary adenocarcinomas with signet ring cell features (SRC⁺) are not well known. Herein, we systematically evaluated clinical and molecular features of SRC⁺ cases with particular attention to smoking status. Surgically treated lung adenocarcinomas (n=763) with follow-up ≥5 years in 3 cohorts were reviewed: all patients in 2006 to 2007 ("all-comers," n=222; 168 ever-smokers), a never-smoker cohort (n=266), and a cohort of ever-smokers (n=275). SRC⁺ tumors had ≥10% of SRCs agreed by 2 pathologists. SRC⁺ cases were tested for rearrangement of ALK and ROS1, as well as 187 known mutations in 10 oncogenes including EGFR, KRAS, BRAF, ERBB2, JAK2, AKT1, AKT2, KIT, MET, and PIK3CA. Overall, 53 of 763 cases (7%) were SRC⁺. In the 2006 to 2007 "all comer" cohort, 9% were SRC⁺. In the never-smoker cohort, 9% were SRC⁺. In the smoker cohort, 3% were SRC⁺. Univariable analysis showed that SRC⁺ never-smokers had shorter overall and disease-free survival (P=0.006 and 0.0004, respectively), but the significance faded in the multivariable analysis. For the other 2 cohorts, crude 5-year survival was decreased by 6% to 27% in SRC⁺ cases without reaching statistical significance. In SRC⁺ tumors, KRAS mutation was most common (29%), followed by ALK (26%), EGFR (18%), ROS1 (6%), BRAF (6%), and PIK3CA (3%). In summary, SRC⁺ tumors in never-smokers had a worse survival by univariable analysis only. SRC⁺ cases seemed enriched for ALK⁺ and ROS1⁺, and other mutations were generally in keeping with the patient's smoking status.
Collapse
|
280
|
Abdelazem AZ, Al-Sanea MM, Park BS, Park HM, Yoo KH, Sim T, Park JB, Lee SH, Lee SH. Synthesis and biological evaluation of new pyrazol-4-ylpyrimidine derivatives as potential ROS1 kinase inhibitors. Eur J Med Chem 2015; 90:195-208. [DOI: 10.1016/j.ejmech.2014.11.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 11/10/2014] [Accepted: 11/11/2014] [Indexed: 10/24/2022]
|
281
|
Saber A, van der Wekken A, Hiltermann TJ, Kok K, van den Berg A, Groen HJ. Genomic aberrations guiding treatment of non-small cell lung cancer patients. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.ctrc.2015.03.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
282
|
Li H, Pan Y, Wang R, Li Y, Sun Y, Chen H. Response to crizotinib observed in metastatic mediastinum lymph node from a non-small cell lung cancer patient harboring EZR-ROS1 fusion. J Cancer Res Clin Oncol 2015; 141:185-7. [PMID: 25230898 DOI: 10.1007/s00432-014-1821-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 08/31/2014] [Indexed: 10/24/2022]
Affiliation(s)
- Hang Li
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China
| | | | | | | | | | | |
Collapse
|
283
|
Morgensztern D, Campo MJ, Dahlberg SE, Doebele RC, Garon E, Gerber DE, Goldberg SB, Hammerman PS, Heist R, Hensing T, Horn L, Ramalingam SS, Rudin CM, Salgia R, Sequist L, Shaw AT, Simon GR, Somaiah N, Spigel DR, Wrangle J, Johnson D, Herbst RS, Bunn P, Govindan R. Molecularly targeted therapies in non-small-cell lung cancer annual update 2014. J Thorac Oncol 2015; 10:S1-63. [PMID: 25535693 PMCID: PMC4346098 DOI: 10.1097/jto.0000000000000405] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There have been significant advances in the understanding of the biology and treatment of non-small-cell lung cancer (NSCLC) during the past few years. A number of molecularly targeted agents are in the clinic or in development for patients with advanced NSCLC. We are beginning to understand the mechanisms of acquired resistance after exposure to tyrosine kinase inhibitors in patients with oncogene addicted NSCLC. The advent of next-generation sequencing has enabled to study comprehensively genomic alterations in lung cancer. Finally, early results from immune checkpoint inhibitors are very encouraging. This review summarizes recent advances in the area of cancer genomics, targeted therapies, and immunotherapy.
Collapse
Affiliation(s)
- Daniel Morgensztern
- Department of Medical Oncology, Washington University School of Medicine, Saint Louis, MO
| | - Meghan J. Campo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA
| | - Suzanne E. Dahlberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston MA
| | - Robert C. Doebele
- Department of Medical Oncology, University of Colorado School of Medicine and University of Colorado Cancer Center, Aurora, CO
| | - Edward Garon
- UCLA Santa Monica Hematology Oncology, Santa Monica, CA
| | - David E. Gerber
- Division of Hematology-Oncology, Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Sarah B. Goldberg
- Department of Medical Oncology, Yale School of Medicine and Cancer Center, New Haven, CT
| | | | - Rebecca Heist
- Department of Oncology, Massachusetts General Hospital, Boston, MA
| | - Thomas Hensing
- Department of Oncology, The University of Chicago Medicine, Chicago, IL
| | - Leora Horn
- Division of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN
| | - Suresh S. Ramalingam
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA
| | | | - Ravi Salgia
- Department of Oncology, The University of Chicago Medicine, Chicago, IL
| | - Lecia Sequist
- Department of Oncology, Massachusetts General Hospital, Boston, MA
| | - Alice T. Shaw
- Department of Oncology, Massachusetts General Hospital, Boston, MA
| | - George R. Simon
- Division of Hematology-Oncology, Medical University of South Carolina, Charleston, SC
| | - Neeta Somaiah
- Division of Hematology-Oncology, Medical University of South Carolina, Charleston, SC
| | | | - John Wrangle
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - David Johnson
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Roy S. Herbst
- Department of Medical Oncology, Yale School of Medicine and Cancer Center, New Haven, CT
| | - Paul Bunn
- Division of Medical Oncology, University of Colorado Denver School of Medicine, Denver, CO
| | - Ramaswamy Govindan
- Department of Medical Oncology, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
284
|
Cao Y, Xiao G, Qiu X, Ye S, Lin T. Efficacy and safety of crizotinib among Chinese EML4-ALK-positive, advanced-stage non-small cell lung cancer patients. PLoS One 2014; 9:e114008. [PMID: 25501361 PMCID: PMC4264693 DOI: 10.1371/journal.pone.0114008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/15/2014] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION We report the efficacy and safety of crizotinib treatment among Chinese patients with advanced-stage NSCLC. METHODS We retrospectively analyzed patients with EML4-ALK positive advanced NSCLC who were treated with crizotinib from May 2012 to Aug 2013. Baseline clinical parameters, treatment protocol, response to therapy and survival were noted. The primary goal was to evaluate the efficacy of crizotinib in patients who were previously treated patients or who had poor ECOG performance status (PS). RESULTS Forty patients were evaluable for safety and efficacy. Median age was 43 years, 100% had adenocarcinoma and stage IV disease, and 42.5% were female. Six patients received frontline treatment with crizotinib, 17 patients had 1 prior treatment, and 17 patients had more than 2 lines of prior treatment. Patients received a median of 5 cycles of treatment (range 1-15 cycles). After the first cycle, 92.5% (37/40) patients archived partial remission (PR). At the end of the follow-up period, the overall PR rate was 70% (28/40), and progression of disease (PD) occurred in 30% of patients (12/40). The median PFS was 28 weeks (95% CI 15.4 to 40.5 weeks), and median OS was 40 weeks (95% CI 38.6 to 49.3 weeks). The most frequent treatment-related AEs were vomiting (47.5%), vision disorder (27.5%) and increased ALT/AST (42%); most toxicities were Grade 1/2. Observed treatment-related Grade 3/4 AEs included increased ALT/AST (10%) and vomiting (5%). The EML4-ALK fusion rate and number of prior chemotherapy cycles did not appear to significantly affect the efficacy of crizotinib. However, PS 0-2 patients had improved PFS (50 weeks vs. 24 weeks, p = 0.015). CONCLUSIONS Crizotinib was safe, well-tolerated, and effective in Chinese patients with pre-treated ALK-rearranged NSCLC. QOL was improved and PS appears to have an effect on the efficacy of crizotinib, but prior treatment and ALK fusion rate do not.
Collapse
Affiliation(s)
- Yabing Cao
- Department of Oncology, Kiang Wu Hospital, Estrada do Repouso, Macau, China, 999999
- * E-mail:
| | - Guangli Xiao
- Department of Oncology, Kiang Wu Hospital, Estrada do Repouso, Macau, China, 999999
| | - Xibin Qiu
- Department of Oncology, Kiang Wu Hospital, Estrada do Repouso, Macau, China, 999999
| | - Sheng Ye
- Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, P.R. China, 510080
| | - Tongyu Lin
- Cancer Center of Sun Yat-sen University, State Key Laboratory of Oncology in Southern China, Department of Medical Oncology, No. 651 Dongfeng Road East, Guangzhou, Guangdong, P.R. China, 510060
| |
Collapse
|
285
|
Jurmeister P, Lenze D, Berg E, Mende S, Schäper F, Kellner U, Herbst H, Sers C, Budczies J, Dietel M, Hummel M, von Laffert M. Parallel screening for ALK, MET and ROS1 alterations in non-small cell lung cancer with implications for daily routine testing. Lung Cancer 2014; 87:122-9. [PMID: 25534130 DOI: 10.1016/j.lungcan.2014.11.018] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/08/2014] [Accepted: 11/30/2014] [Indexed: 10/24/2022]
Abstract
OBJECTIVES ALK, MET and ROS1 are prognostic and predictive markers in NSCLC, which need to be implemented in daily routine. To evaluate different detection approaches and scoring systems for optimal stratification of patients eligible for mutation testing in the future, we screened a large and unselected cohort of NSCLCs for all three alterations. MATERIAL AND METHODS Using tissue microarrays, 473 surgically resected NSCLCs were tested for ALK and MET expression by IHC and genomic alterations in the ALK, MET and ROS1 gene by FISH. For MET IHC, two different criteria (MetMAb and H-score), for MET FISH, three different scoring systems (UCCC, Cappuzzo, PathVysion) were investigated. RESULTS ALK and ROS1 positivity was seen in 2.6% and 1.3% of all ADCs, respectively, but not in pure SCCs. One ROS1 translocated tumor showed additional ROS1 amplification. MET IHC+/FISH+ cases were found in both histological subtypes (8.6% in all NSCLCs; 10.6% in ADCs; 5.0% in SCCs) and were associated with pleural invasion, lymphatic vessel invasion and lymph node metastasis. MET altered ADCs more frequently showed a papillary growth pattern. Whereas ALK testing revealed homogenous results in IHC and FISH, we saw discordant results for MET in about 10% of cases. Both METIHC scoring systems revealed almost identical results. We did not encounter any combined FISH positivity for ALK, MET or ROS1. However, three ALK positive cases harbored MET overexpression. CONCLUSION In daily routine, IHC could support FISH in the identification of ALK altered NSCLCs. Further research is needed to assess the role of discordant MET results by means of IHC and FISH as well as the relevance of tumors with an increased ROS1 gene copy number.
Collapse
Affiliation(s)
- Philipp Jurmeister
- Institute of Pathology, Campus Charité Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Dido Lenze
- Institute of Pathology, Campus Charité Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Erika Berg
- Institute of Pathology, Campus Charité Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Stefanie Mende
- Institute of Pathology, Campus Charité Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Frank Schäper
- Pathology-Berlin, Bioptisches Institut Gemeinschaftspraxis für Pathologie, Lindenberger Weg 27, 13125 Berlin, Germany
| | - Udo Kellner
- Institute of Pathology, Johannes Wesling Klinikum Minden, Hans-Nolte-Straße 1, 32429 Minden, Germany
| | - Hermann Herbst
- Institute of Pathology, Vivantes Klinikum Berlin, Oranienburger Straße 285, 13437 Berlin, Germany
| | - Christine Sers
- Institute of Pathology, Campus Charité Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jan Budczies
- Institute of Pathology, Campus Charité Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Manfred Dietel
- Institute of Pathology, Campus Charité Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Michael Hummel
- Institute of Pathology, Campus Charité Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Maximilian von Laffert
- Institute of Pathology, Campus Charité Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
286
|
Ou SHI. Republished: lung cancer in never-smokers. Does smoking history matter in the era of molecular diagnostics and targeted therapy? Postgrad Med J 2014; 90:228-35. [PMID: 24643262 DOI: 10.1136/postgradmedj-2012-201296rep] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Lung cancer in never-smokers was recognised as a distinct clinical entity around the mid-2000s because these patients tended to be Asian women and diagnosed at a younger age with a preponderance of adenocarcinoma and better survival outcome despite a more advanced stage of presentation. It was soon discovered that lung cancer in never-smokers had a higher prevalence of activating EGFR mutations and we tend to classify lung cancer by smoking status for screening purpose. With the discoveries of many actionable driver mutations such as activating EGFR mutations and ALK rearrangement in adenocarcinoma of the lung we have switched to classifying non-small cell lung cancer into different individual molecular subgroups based on the presence of a dominant driver mutation. Although many actionable driver mutations are found in never-smokers with adenocarcinoma, this review will summarise that a substantial proportion of patients with these actionable driver mutations had a previous smoking history. Alternatively among the driver mutations that are associated with smoking history, a fair amount of these patients were never-smokers. Thus smoking status should not be used as a screen strategy for identifying driver mutations in clinical practice. Finally smoking history may have predictive and/or prognostic significance within individual molecular subgroups and identifying the difference according to smoking history may help optimise future targeted therapy.
Collapse
|
287
|
Zer A, Leighl N. Promising Targets and Current Clinical Trials in Metastatic Non-Squamous NSCLC. Front Oncol 2014; 4:329. [PMID: 25505733 PMCID: PMC4243502 DOI: 10.3389/fonc.2014.00329] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/31/2014] [Indexed: 11/24/2022] Open
Abstract
Lung adenocarcinoma is the most common subtype of lung cancer today. With the discovery of epidermal growth factor receptor (EGFR) mutations, anaplastic lymphoma kinase (ALK) rearrangements, and effective targeted therapy, personalized medicine has become a reality for patients with lung adenocarcinoma. Here, we review potential additional targets and novel therapies of interest in lung adenocarcinoma including targets within the cell surface (receptor tyrosine kinases EGFR, human epidermal growth factor receptor 2, RET, ROS1, mesenchymal-epidermal transition, TRK), targets in intracellular signal transduction (ALK, RAS-RAF-MEK, PI3K-AKT-PTEN, WNT), nuclear targets such as poly-ADP ribose polymerase, heat shock protein 90, and histone deacetylase, and selected pathways in the tumor environment. With the evolving ability to identify specific molecular aberrations in patient tumors in routine practice, our ability to further personalize therapy in lung adenocarcinoma is rapidly expanding.
Collapse
Affiliation(s)
- Alona Zer
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Natasha Leighl
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
288
|
Affiliation(s)
- Kathryn A Gold
- From the Department of Thoracic/Head and Neck Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston
| |
Collapse
|
289
|
Shaw AT, Ou SHI, Bang YJ, Camidge DR, Solomon BJ, Salgia R, Riely GJ, Varella-Garcia M, Shapiro GI, Costa DB, Doebele RC, Le LP, Zheng Z, Tan W, Stephenson P, Shreeve SM, Tye LM, Christensen JG, Wilner KD, Clark JW, Iafrate AJ. Crizotinib in ROS1-rearranged non-small-cell lung cancer. N Engl J Med 2014; 371:1963-1971. [PMID: 25264305 PMCID: PMC4264527 DOI: 10.1056/nejmoa1406766] [Citation(s) in RCA: 1488] [Impact Index Per Article: 135.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Chromosomal rearrangements of the gene encoding ROS1 proto-oncogene receptor tyrosine kinase (ROS1) define a distinct molecular subgroup of non-small-cell lung cancers (NSCLCs) that may be susceptible to therapeutic ROS1 kinase inhibition. Crizotinib is a small-molecule tyrosine kinase inhibitor of anaplastic lymphoma kinase (ALK), ROS1, and another proto-oncogene receptor tyrosine kinase, MET. METHODS We enrolled 50 patients with advanced NSCLC who tested positive for ROS1 rearrangement in an expansion cohort of the phase 1 study of crizotinib. Patients were treated with crizotinib at the standard oral dose of 250 mg twice daily and assessed for safety, pharmacokinetics, and response to therapy. ROS1 fusion partners were identified with the use of next-generation sequencing or reverse-transcriptase-polymerase-chain-reaction assays. RESULTS The objective response rate was 72% (95% confidence interval [CI], 58 to 84), with 3 complete responses and 33 partial responses. The median duration of response was 17.6 months (95% CI, 14.5 to not reached). Median progression-free survival was 19.2 months (95% CI, 14.4 to not reached), with 25 patients (50%) still in follow-up for progression. Among 30 tumors that were tested, we identified 7 ROS1 fusion partners: 5 known and 2 novel partner genes. No correlation was observed between the type of ROS1 rearrangement and the clinical response to crizotinib. The safety profile of crizotinib was similar to that seen in patients with ALK-rearranged NSCLC. CONCLUSIONS In this study, crizotinib showed marked antitumor activity in patients with advanced ROS1-rearranged NSCLC. ROS1 rearrangement defines a second molecular subgroup of NSCLC for which crizotinib is highly active. (Funded by Pfizer and others; ClinicalTrials.gov number, NCT00585195.).
Collapse
Affiliation(s)
- Alice T Shaw
- From the Massachusetts General Hospital Cancer Center (A.T.S., L.P.L., Z.Z., J.W.C., A.J.I.), Dana-Farber Cancer Institute (G.I.S.), and Beth Israel Deaconess Medical Center (D.B.C.) - all in Boston; University of California at Irvine, Irvine (S.-H.I.O.), and Pfizer Oncology, La Jolla (W.T., S.M.S., L.M.T., J.G.C., K.D.W.) - both in California; Seoul National University Hospital, Seoul, South Korea (Y.-J.B.); University of Colorado, Aurora (D.R.C., M.V.-G., R.C.D.); Peter MacCallum Cancer Centre, Melbourne, VIC, Australia (B.J.S.); University of Chicago, Chicago (R.S.); Memorial Sloan Kettering Cancer Center, New York (G.J.R.); Karolinska Institutet, Stockholm (Z.Z.); and Rho, Chapel Hill, NC (P.S.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
290
|
[Targeted therapies in non-small cell lung cancer in 2014]. Rev Mal Respir 2014; 32:182-92. [PMID: 25704901 DOI: 10.1016/j.rmr.2014.08.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 08/06/2014] [Indexed: 11/21/2022]
Abstract
For several years, the identification of molecular sequencing alterations has considerably changed the perception and treatment of non-small cell lung cancer (NSCLC). These alterations have been defined as "driver mutations", such as mutations in EGFR and EML4-ALK fusion gene, and are highly sensitive to specific therapies. Other targets have also been identified recently. Personalized medicine is now a reality for patients with advanced NSCLC on the basis of routine screening for EGFR, HER2, KRAS, BRAF, PI3KCA mutations and EML4-ALK rearrangement. This article describes identified biomarkers, available targeted therapies, and the main clinical research approaches in NSCLC.
Collapse
|
291
|
Kodama T, Tsukaguchi T, Satoh Y, Yoshida M, Watanabe Y, Kondoh O, Sakamoto H. Alectinib shows potent antitumor activity against RET-rearranged non-small cell lung cancer. Mol Cancer Ther 2014; 13:2910-8. [PMID: 25349307 DOI: 10.1158/1535-7163.mct-14-0274] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Alectinib/CH5424802 is a known inhibitor of anaplastic lymphoma kinase (ALK) and is being evaluated in clinical trials for the treatment of ALK fusion-positive non-small cell lung cancer (NSCLC). Recently, some RET and ROS1 fusion genes have been implicated as driver oncogenes in NSCLC and have become molecular targets for antitumor agents. This study aims to explore additional target indications of alectinib by testing its ability to inhibit the activity of kinases other than ALK. We newly verified that alectinib inhibited RET kinase activity and the growth of RET fusion-positive cells by suppressing RET phosphorylation. In contrast, alectinib hardly inhibited ROS1 kinase activity unlike other ALK/ROS1 inhibitors such as crizotinib and LDK378. It also showed antitumor activity in mouse models of tumors driven by the RET fusion. In addition, alectinib showed kinase inhibitory activity against RET gatekeeper mutations (RET V804L and V804M) and blocked cell growth driven by the KIF5B-RET V804L and V804M. Our results suggest that alectinib is effective against RET fusion-positive tumors. Thus, alectinib might be a therapeutic option for patients with RET fusion-positive NSCLC.
Collapse
Affiliation(s)
- Tatsushi Kodama
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Japan
| | | | - Yasuko Satoh
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Japan
| | - Miyuki Yoshida
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Japan
| | | | - Osamu Kondoh
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Japan
| | - Hiroshi Sakamoto
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Japan.
| |
Collapse
|
292
|
Boyle TA, Masago K, Ellison KE, Yatabe Y, Hirsch FR. ROS1 immunohistochemistry among major genotypes of non-small-cell lung cancer. Clin Lung Cancer 2014; 16:106-11. [PMID: 25467930 PMCID: PMC4770803 DOI: 10.1016/j.cllc.2014.10.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/14/2014] [Accepted: 10/21/2014] [Indexed: 12/15/2022]
Abstract
Identification of ROS1 rearrangements in patients with lung cancer allows them to benefit from targeted therapy. We compared immunohistochemistry (IHC) with more cumbersome methods such as fluorescence in situ hybridization and reverse transcriptase polymerase chain reaction for identification of ROS1 rearrangements in patients with lung adenocarcinoma (n = 33). Our results showed that IHC is a sensitive (100%) and specific (100%) method to identify ROS1 rearrangements in patients with lung cancer.
Collapse
Affiliation(s)
- Theresa A Boyle
- Department of Medical Oncology and Pathology, University of Colorado, Aurora, CO
| | - Katsuhiro Masago
- Division of Integrated Oncology, Institute of Biomedical Research and Innovation, Kobe, Japan; Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Kim E Ellison
- Department of Medical Oncology and Pathology, University of Colorado, Aurora, CO
| | - Yasushi Yatabe
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Fred R Hirsch
- Department of Medical Oncology and Pathology, University of Colorado, Aurora, CO.
| |
Collapse
|
293
|
Zhang N, Yang JJ, Zhang XC, Xie Z, Wang BC, Tu HY, Jiang BY, Wu YL. Responses to crizotinib in a patient with c-ros oncogene 1, receptor tyrosine kinase-positive advanced lung adenocarcinoma: A case report. Oncol Lett 2014; 8:2624-2626. [PMID: 25364439 PMCID: PMC4214434 DOI: 10.3892/ol.2014.2571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 07/08/2014] [Indexed: 11/13/2022] Open
Abstract
Rearrangements to the c-ros oncogene 1, receptor tyrosine kinase (ROS1) gene are reported in 1–2% of lung adenocarcinomas. These rearrangements are associated with a response to the small-molecule tyrosine kinase inhibitor crizotinib. ROS1 rearrangements can be detected using fluorescence in situ hybridization (FISH), which is considered the gold standard technique in detecting ROS1 rearrangements, and determining whether a patient would respond well to crizotinib treatment. However, FISH is an expensive and time-consuming assay, requiring specialized microscopy equipment and some level of technical expertise. The present report describes the case of a patient with advanced lung adenocarcinoma, who was identified to be negative for ROS-1 rearrangements by FISH, but positive by immunohistochemistry (IHC). The health of the patient improved following treatment with crizotinib. These results indicate that IHC assay could be an alternative option for the detection of ROS1 gene rearrangements.
Collapse
Affiliation(s)
- Na Zhang
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China ; Second Clinical Medial Committee, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Xu-Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Zhi Xie
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Bin-Chao Wang
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Hai-Yan Tu
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Ben-Yuan Jiang
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
294
|
Peraldo Neia C, Cavalloni G, Balsamo A, Venesio T, Napoli F, Sassi F, Martin V, Frattini M, Aglietta M, Leone F. Screening for the FIG-ROS1 fusion in biliary tract carcinomas by nested PCR. Genes Chromosomes Cancer 2014; 53:1033-40. [PMID: 25231053 DOI: 10.1002/gcc.22212] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 08/08/2014] [Indexed: 01/30/2023] Open
Abstract
ROS1 rearrangements have been detected in a variety of tumors and are considered as suitable targets of anticancer therapies. We developed a new, quick, specific, and sensitive PCR test to screen for the FIG-ROS1 fusion and applied it to a series of Italian patients with bile duct carcinoma (BTC). Formalin-fixed, paraffin-embedded tissues, derived from 65 Italian BTC patients, and six cell lines were analyzed by nested PCR to investigate the prevalence of a previously reported FIG-ROS1 fusion. The specificity and sensitivity of nested PCR were investigated in FIG-ROS1 positive U118MG cells in reconstitution experiments with peripheral blood mononuclear cells. We found that six out of 65 (9%) BTC patients were positive for the FIG-ROS1 fusion, comprising two out of 14 (14%) gallbladder carcinoma (GBC) patients and four out of 25 (16%) extrahepatic cholangiocarcinoma (ECC) patients. None of the 26 intrahepatic cholangiocarcinoma cases harbored the FIG-ROS1 fusion. All the cell lines were negative for this variant. In conclusion, 14-16% of GBC and ECC were positive for FIG-ROS1. This may have clinical implications, since these patients will potentially benefit from the treatment with specific ROS1 inhibitors.
Collapse
Affiliation(s)
- Caterina Peraldo Neia
- Department of Oncology, University of Turin Medical School, IRCCS-Candiolo, Turin, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
295
|
Kerr KM, Bubendorf L, Edelman MJ, Marchetti A, Mok T, Novello S, O'Byrne K, Stahel R, Peters S, Felip E. Second ESMO consensus conference on lung cancer: pathology and molecular biomarkers for non-small-cell lung cancer. Ann Oncol 2014; 25:1681-1690. [PMID: 24718890 DOI: 10.1093/annonc/mdu145] [Citation(s) in RCA: 215] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023] Open
Abstract
To complement the existing treatment guidelines for all tumour types, ESMO organises consensus conferences to focus on specific issues in each type of tumour. The Second ESMO Consensus Conference on Lung Cancer was held on 11-12 May 2013 in Lugano. A total of 35 experts met to address several questions on management of patients with non-small-cell lung cancer (NSCLC) in each of four areas: pathology and molecular biomarkers, early stage disease, locally advanced disease and advanced (metastatic) disease. For each question, recommendations were made including reference to the grade of recommendation and level of evidence. This consensus paper focuses on recommendations for pathology and molecular biomarkers in relation to the diagnosis of lung cancer, primarily non-small-cell carcinomas.
Collapse
Affiliation(s)
- K M Kerr
- Department of Pathology, Aberdeen Royal Infirmary and Aberdeen University Medical School, Aberdeen, UK.
| | - L Bubendorf
- Institute for Pathology, University Hospital Basel, Basel, Switzerland
| | - M J Edelman
- University of New Mexico Cancer Center, Albuquerque, USA
| | - A Marchetti
- Center of Predictive Molecular Medicine, Center of Excellence on Ageing, University-Foundation, Chieti, Italy
| | - T Mok
- Department of Clinical Oncology, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin New Territories, Hong Kong, China
| | - S Novello
- Thoracic Oncology Unit, Department of Oncology, University of Turin, Azienda Ospedaliero-Universitaria San Luigi Orbassano, Italy
| | - K O'Byrne
- Trinity College, Dublin, Ireland; Queensland University of Technology, Brisbane, Australia
| | - R Stahel
- Clinic of Oncology, University Hospital Zürich, Zürich
| | - S Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - E Felip
- Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| |
Collapse
|
296
|
Saito M, Shiraishi K, Matsumoto K, Schetter AJ, Ogata-Kawata H, Tsuchiya N, Kunitoh H, Nokihara H, Watanabe SI, Tsuta K, Kumamoto K, Takenoshita S, Yokota J, Harris CC, Kohno T. A three-microRNA signature predicts responses to platinum-based doublet chemotherapy in patients with lung adenocarcinoma. Clin Cancer Res 2014; 20:4784-93. [PMID: 25142144 DOI: 10.1158/1078-0432.ccr-14-1096] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
PURPOSE To examine the clinical utility of intratumor microRNAs (miRNA) as a biomarker for predicting responses to platinum-based doublet chemotherapy in patients with recurring lung adenocarcinoma (LADC). EXPERIMENTAL DESIGN The expression of miRNAs was examined in LADC tissues surgically resected from patients treated with platinum-based doublet chemotherapy at the time of LADC recurrence. Microarray-based screening of 904 miRNAs followed by quantitative reverse transcription-PCR-based verification in 40 test cohort samples, including 16 (40.0%) responders, was performed to identify miRNAs that are differentially expressed in chemotherapy responders and nonresponders. Differential expression was confirmed in a validation cohort (n = 63 samples), including 18 (28.6%) responders. An miRNA signature that predicted responses to platinum-based doublet chemotherapy was identified and its accuracy was examined by principal component and support vector machine analyses. Genotype data for the TP53-Arg72Pro polymorphism, which is associated with responses to platinum-based doublet chemotherapy, were subsequently incorporated into the prediction analysis. RESULTS A signature comprising three miRNAs (miR1290, miR196b, and miR135a*) enabled the prediction of a chemotherapeutic response (rather than progression-free and overall survival) with high accuracy in both the test and validation cohorts (82.5% and 77.8%). Examination of the latter was performed using miRNAs extracted from archived formalin-fixed paraffin-embedded tissues. Combining this miRNA signature with the TP53-Arg72Pro polymorphism genotype marginally improved the predictive power. CONCLUSION The three-miRNA signature in surgically resected primary LADC tissues may by clinically useful for predicting responsiveness to platinum-based doublet chemotherapy in patients with LADC recurrence.
Collapse
Affiliation(s)
- Motonobu Saito
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan. Department of Organ Regulatory Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kenji Matsumoto
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Setagaya, Tokyo, Japan
| | - Aaron J Schetter
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Hiroko Ogata-Kawata
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Naoto Tsuchiya
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Hideo Kunitoh
- Department of Medical Oncology, Japanese Red Cross Medical Center, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroshi Nokihara
- Division of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shun-Ichi Watanabe
- Division of Thoracic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Koji Tsuta
- Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Kensuke Kumamoto
- Department of Organ Regulatory Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Seiichi Takenoshita
- Department of Organ Regulatory Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Jun Yokota
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan. Cancer Genome Biology, Institute of Predictive and Personalized Medicine of Cancer, Barcelona, Spain
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan.
| |
Collapse
|
297
|
Crizotinib: a review of its use in the treatment of anaplastic lymphoma kinase-positive, advanced non-small cell lung cancer. Drugs 2014; 73:2031-51. [PMID: 24288180 DOI: 10.1007/s40265-013-0142-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Crizotinib (Xalkori(®)) is an orally active, small molecule inhibitor of multiple receptor tyrosine kinases, including anaplastic lymphoma kinase (ALK), c-Met/hepatocyte growth factor receptor and c-ros oncogene 1. In the EU, crizotinib has been conditionally approved for the treatment of adults with previously treated, ALK-positive, advanced non-small cell lung cancer (NSCLC). This approval has been based on objective response rate and tolerability data from two ongoing phase I/II studies (PROFILE 1001 and PROFILE 1005); these results have been substantiated and extended by findings from an ongoing phase III study (PROFILE 1007) in patients with ALK-positive, advanced NSCLC who had received one prior platinum-based regimen. Those treated with crizotinib experienced significant improvements in progression-free survival, objective response rate, lung cancer symptoms and global quality of life, as compared with those treated with standard second-line chemotherapy (pemetrexed or docetaxel). The relative survival benefit with crizotinib is unclear, however, as the data are still immature and likely to be confounded by the high cross-over rate among chemotherapy recipients. Crizotinib treatment was generally well tolerated in the three PROFILE studies, with liver transaminase elevations and neutropenia being the most common grade 3 or 4 adverse events. Crizotinib is the standard of care in terms of the treatment of patients with ALK-positive, advanced NSCLC; while the current EU approval is for second (or subsequent)-line use only, the first-line use of the drug is being evaluated in ongoing phase III studies. Key issues relating to the use of crizotinib in clinical practice include identifying the small subset of eligible patients, the almost inevitable development of resistance and the high cost of treatment.
Collapse
|
298
|
Abstract
Non-small-cell lung cancers (NSCLCs), the most common lung cancers, are known to have diverse pathological features. During the past decade, in-depth analyses of lung cancer genomes and signalling pathways have further defined NSCLCs as a group of distinct diseases with genetic and cellular heterogeneity. Consequently, an impressive list of potential therapeutic targets was unveiled, drastically altering the clinical evaluation and treatment of patients. Many targeted therapies have been developed with compelling clinical proofs of concept; however, treatment responses are typically short-lived. Further studies of the tumour microenvironment have uncovered new possible avenues to control this deadly disease, including immunotherapy.
Collapse
Affiliation(s)
- Zhao Chen
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA. [2]
| | - Christine M Fillmore
- 1] Stem Cell Program, Boston Children's Hospital, Boston, Massachusetts 02115, USA. [2] Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA. [3] Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA. [4]
| | - Peter S Hammerman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | - Carla F Kim
- 1] Stem Cell Program, Boston Children's Hospital, Boston, Massachusetts 02115, USA. [2] Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA. [3] Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Kwok-Kin Wong
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA. [2] Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA. [3] Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| |
Collapse
|
299
|
Chen Z, Fillmore CM, Hammerman PS, Kim CF, Wong KK. Non-small-cell lung cancers: a heterogeneous set of diseases. Nat Rev Cancer 2014; 14:535-46. [PMID: 25056707 PMCID: PMC5712844 DOI: 10.1038/nrc3775] [Citation(s) in RCA: 1370] [Impact Index Per Article: 124.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Non-small-cell lung cancers (NSCLCs), the most common lung cancers, are known to have diverse pathological features. During the past decade, in-depth analyses of lung cancer genomes and signalling pathways have further defined NSCLCs as a group of distinct diseases with genetic and cellular heterogeneity. Consequently, an impressive list of potential therapeutic targets was unveiled, drastically altering the clinical evaluation and treatment of patients. Many targeted therapies have been developed with compelling clinical proofs of concept; however, treatment responses are typically short-lived. Further studies of the tumour microenvironment have uncovered new possible avenues to control this deadly disease, including immunotherapy.
Collapse
Affiliation(s)
- Zhao Chen
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA. [2]
| | - Christine M Fillmore
- 1] Stem Cell Program, Boston Children's Hospital, Boston, Massachusetts 02115, USA. [2] Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA. [3] Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA. [4]
| | - Peter S Hammerman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | - Carla F Kim
- 1] Stem Cell Program, Boston Children's Hospital, Boston, Massachusetts 02115, USA. [2] Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA. [3] Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Kwok-Kin Wong
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA. [2] Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA. [3] Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| |
Collapse
|
300
|
Zhao W, Damodaran S, Villalona-Calero MA. From single-gene to multiplex analysis in lung cancer, challenges and accomplishments: a review of a single institution's experience. Lung Cancer Manag 2014; 4:315-324. [PMID: 25580159 DOI: 10.2217/lmt.14.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Molecular selection has led to the successful use of novel tyrosine kinase inhibitors (TKIs) in non-small-cell lung cancers (NSCLCs). For instance, mutations in EGFR and translocations and fusions in ALK render tumor cells sensitive to some TKIs, leading to substantial clinical benefits. Molecular testing such as DNA sequencing or fragment analysis following PCR, and evaluation of copy number and gene positioning by FISH, have been developed and used clinically to identify mutations/fusions. Meanwhile, TKIs to target actionable mutations/fusions in several other oncogenes are being evaluated. High-throughput sequencing can provide therapy-predictive information as well as identify novel targetable genomic alterations. In this article, we report our experience enabling single-gene testing, and our evolution to panel-based next-generation sequencing.
Collapse
Affiliation(s)
- Weiqiang Zhao
- Departments of Internal Medicine & Pathology, The Ohio State University, 320 W, 10th Avenue, Columbus, OH 43210, USA ; Comprehensive Cancer Center, The Ohio State University, 320 W, 10th Avenue, Columbus, OH 43210, USA
| | - Senthilkumar Damodaran
- Departments of Internal Medicine & Pathology, The Ohio State University, 320 W, 10th Avenue, Columbus, OH 43210, USA ; Comprehensive Cancer Center, The Ohio State University, 320 W, 10th Avenue, Columbus, OH 43210, USA
| | - Miguel A Villalona-Calero
- Departments of Internal Medicine & Pathology, The Ohio State University, 320 W, 10th Avenue, Columbus, OH 43210, USA ; Comprehensive Cancer Center, The Ohio State University, 320 W, 10th Avenue, Columbus, OH 43210, USA
| |
Collapse
|