951
|
Chuang JC, Neal JW, Niu XM, Wakelee HA. Adjuvant therapy for EGFR mutant and ALK positive NSCLC: Current data and future prospects. Lung Cancer 2015; 90:1-7. [PMID: 26275476 DOI: 10.1016/j.lungcan.2015.07.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 07/27/2015] [Indexed: 10/23/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) against targetable mutations such as epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) are highly effective in treating advanced stage lung cancers harboring such mutations. Questions remain, however, about whether these agents can improve cure rates for early stage lung cancers in the adjuvant setting. Here, we examine the current data and ongoing trials addressing this issue.
Collapse
Affiliation(s)
- Jody C Chuang
- Division of Hematology and Oncology, Stanford Hospital & Clinics, Stanford, CA, USA
| | - Joel W Neal
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiao-Min Niu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 Huaihai West Road, Shanghai 200030, PR China
| | - Heather A Wakelee
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
952
|
Kim HJ, Lee KY, Kim YW, Choi YJ, Lee JE, Choi CM, Baek IJ, Rho JK, Lee JC. P-glycoprotein confers acquired resistance to 17-DMAG in lung cancers with an ALK rearrangement. BMC Cancer 2015. [PMID: 26219569 PMCID: PMC4517346 DOI: 10.1186/s12885-015-1543-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Because anaplastic lymphoma kinase (ALK) is dependent on Hsp90 for protein stability, Hsp90 inhibitors are effective in controlling growth of lung cancer cells with ALK rearrangement. We investigated the mechanism of acquired resistance to 17-(Dimethylaminoethylamino)-17-demethoxygeldanamycin (17-DMAG), a geldanamycin analogue Hsp90 inhibitor, in H3122 and H2228 non-small cell lung cancer cell lines with ALK rearrangement. METHODS Resistant cell lines (H3122/DR-1, H3122/DR-2 and H2228/DR) were established by repeated exposure to increasing concentrations of 17-DMAG. Mechanisms for resistance by either NAD(P)H/quinone oxidoreductase 1 (NQO1), previously known as a factor related to 17-DMAG resistance, or P-glycoprotein (P-gp; ABCB1/MDR1) were queried using RT-PCR, western blot analysis, chemical inhibitors, the MTT cell proliferation/survival assay, and cellular efflux of rhodamine 123. RESULTS The resistant cells showed no cross-resistance to AUY922 or ALK inhibitors, suggesting that ALK dependency persists in cells with acquired resistance to 17-DMAG. Although expression of NQO1 was decreased in H3122/DR-1 and H3122/DR-2, NQO1 inhibition by dicumarol did not affect the response of parental cells (H2228 and H3122) to 17-DMAG. Interestingly, all resistant cells showed the induction of P-gp at the protein and RNA levels, which was associated with an increased efflux of the P-gp substrate rhodamine 123 (Rho123). Transfection with siRNA directed against P-gp or treatment with verapamil, an inhibitor of P-gp, restored the sensitivity to the drug in all cells with acquired resistance to 17-DMAG. Furthermore, we also observed that the growth-inhibitory effect of 17-DMAG was decreased in A549/PR and H460/PR cells generated to over-express P-gp by long-term exposure to paclitaxel, and these cells recovered their sensitivity to 17-DMAG through the inhibition of P-gp. CONCLUSION P-gp over-expression is a possible mechanism of acquired resistance to 17-DMAG in cells with ALK rearrangement.
Collapse
Affiliation(s)
- Hee Joung Kim
- Department of Internal Medicine, Konkuk University Medical Center, Seoul, South Korea.
| | - Kye Young Lee
- Department of Internal Medicine, Konkuk University Medical Center, Seoul, South Korea.
| | - Young Whan Kim
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Seoul National University College of Medicine, Seoul, South Korea.
| | - Yun Jung Choi
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea.
| | - Jung-Eun Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea.
| | - Chang Min Choi
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea. .,Department of Oncology, Asan Medical Center, College of Medicine, University of Ulsan, 86 Asanbyeongwon-gil, Songpa-gu, Seoul, 138-736, South Korea.
| | - In-Jeoung Baek
- Asan Institute for Life Sciences, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea.
| | - Jin Kyung Rho
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea. .,Asan Institute for Life Sciences, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea.
| | - Jae Cheol Lee
- Department of Oncology, Asan Medical Center, College of Medicine, University of Ulsan, 86 Asanbyeongwon-gil, Songpa-gu, Seoul, 138-736, South Korea.
| |
Collapse
|
953
|
Doebele RC, Davis LE, Vaishnavi A, Le AT, Estrada-Bernal A, Keysar S, Jimeno A, Varella-Garcia M, Aisner DL, Li Y, Stephens PJ, Morosini D, Tuch BB, Fernandes M, Nanda N, Low JA. An Oncogenic NTRK Fusion in a Patient with Soft-Tissue Sarcoma with Response to the Tropomyosin-Related Kinase Inhibitor LOXO-101. Cancer Discov 2015. [PMID: 26216294 DOI: 10.1158/2159-8290.cd-15-0443] [Citation(s) in RCA: 312] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
UNLABELLED Oncogenic TRK fusions induce cancer cell proliferation and engage critical cancer-related downstream signaling pathways. These TRK fusions occur rarely, but in a diverse spectrum of tumor histologies. LOXO-101 is an orally administered inhibitor of the TRK kinase and is highly selective only for the TRK family of receptors. Preclinical models of LOXO-101 using TRK-fusion-bearing human-derived cancer cell lines demonstrate inhibition of the fusion oncoprotein and cellular proliferation in vitro, and tumor growth in vivo. The tumor of a 41-year-old woman with soft-tissue sarcoma metastatic to the lung was found to harbor an LMNA-NTRK1 gene fusion encoding a functional LMNA-TRKA fusion oncoprotein as determined by an in situ proximity ligation assay. In a phase I study of LOXO-101 (ClinicalTrials.gov no. NCT02122913), this patient's tumors underwent rapid and substantial tumor regression, with an accompanying improvement in pulmonary dyspnea, oxygen saturation, and plasma tumor markers. SIGNIFICANCE TRK fusions have been deemed putative oncogenic drivers, but their clinical significance remained unclear. A patient with a metastatic soft-tissue sarcoma with an LMNA-NTRK1 fusion had rapid and substantial tumor regression with a novel, highly selective TRK inhibitor, LOXO-101, providing the first clinical evidence of benefit from inhibiting TRK fusions.
Collapse
Affiliation(s)
| | - Lara E Davis
- Oregon Health and Science University Knight Cancer Institute, Portland, Oregon
| | | | - Anh T Le
- University of Colorado Cancer Center, Aurora, Colorado
| | | | | | | | | | - Dara L Aisner
- University of Colorado Cancer Center, Aurora, Colorado
| | - Yali Li
- Foundation Medicine, Cambridge, Massachusetts
| | | | | | | | | | - Nisha Nanda
- Loxo Oncology, South San Francisco, California
| | | |
Collapse
|
954
|
Abstract
Discovery and translational research has led to the identification of a series of “cancer drivers”—genes that, when mutated or otherwise misregulated, can drive malignancy. An increasing number of drugs that directly target such drivers have demonstrated activity in clinical trials and are shaping a new landscape for molecularly targeted cancer therapies. Such therapies rely on molecular and genetic diagnostic tests to detect the presence of a biomarker that predicts response. Here, we highlight some of the key discoveries bringing precision oncology to cancer patients. Large-scale “omics” approaches as well as modern, hypothesis-driven science in both academic and industry settings have significantly contributed to the field. Based on these insights, we discuss current challenges and how to foster future biomedical innovation in cancer drug discovery and development.
Collapse
Affiliation(s)
- Margit A Huber
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany
| | - Norbert Kraut
- Oncology Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| |
Collapse
|
955
|
Abstract
With the widespread availability of biological antitumor drugs, the current scene of chemotherapies is changing. New chemotherapy agents, such as crizotinib, an inhibitor of anaplastic lymphoma kinase (ALK) and ROS1, usually used in pretreated advanced ALK-positive non-small-cell lung carcinoma, are more often used, and a description of the onset of side effects with suggestions for their management could be of interest for physicians. We describe a case of diffuse and aggressive renal polycystosis induced by crizotinib, which regressed after therapy, which could be of interest considering its wide extension and disappearance after the end of treatment. We also suggest some considerations from the literature and from the case reported that could be helpful in the management of this condition, which is known to be caused by crizotinib treatment.
Collapse
|
956
|
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
|
957
|
Ahluwalia MS, Rich JN. Growth Factor Receptor Fusions Predict Therapeutic Sensitivity. Clin Cancer Res 2015; 21:3105-7. [PMID: 25995339 DOI: 10.1158/1078-0432.ccr-15-0458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 04/02/2015] [Indexed: 11/16/2022]
Abstract
Dysregulated growth factor pathways promote tumor growth in many cancers, but receptor-targeting strategies frequently offer limited benefit despite activation by receptor overexpression or amplification. In contrast, tumors harboring growth factor receptor fusions display exquisite dependence on receptor activity, providing predictive markers for patient response to inform precise oncology treatment.
Collapse
Affiliation(s)
- Manmeet S Ahluwalia
- Burkhardt Brain Tumor Neuro-Oncology Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio.
| | - Jeremy N Rich
- Burkhardt Brain Tumor Neuro-Oncology Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio. Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
958
|
Ge L, Shi R. Progress of EGFR-TKI and ALK/ROS1 inhibitors in advanced non-small cell lung cancer. Int J Clin Exp Med 2015; 8:10330-10339. [PMID: 26379824 PMCID: PMC4565207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 07/11/2015] [Indexed: 06/05/2023]
Abstract
To discuss the mechanism and clinical application of EGFR-TKI and ALK/ROS1 inhibitors in non-small cell lung cancer (NSCLC), we reviewed recent available data mainly from PubMed. We found that chemotherapy, progression-free survival (PFS), objective response rate (ORR), and quality of life of patients with advanced NSCLC can be greatly improved in these drugs medication compared with conventional chemotherapy. Though many questions like resistance to EGFR-TKI and ALK/ROS1 inhibitors exist, molecular targeted therapy is an important therapeutic method for the management of NSCLC. The role of molecule targeted therapy in the initiation and development of NSCLC deserves further study.
Collapse
Affiliation(s)
- Liangqing Ge
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
- Department of Cardiovascular Medicine, The first people’s hospital of Changde cityChangde 415000, Hunan, China
| | - Ruizheng Shi
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| |
Collapse
|
959
|
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
|
960
|
Zou HY, Friboulet L, Kodack DP, Engstrom LD, Li Q, West M, Tang RW, Wang H, Tsaparikos K, Wang J, Timofeevski S, Katayama R, Dinh DM, Lam H, Lam JL, Yamazaki S, Hu W, Patel B, Bezwada D, Frias RL, Lifshits E, Mahmood S, Gainor JF, Affolter T, Lappin PB, Gukasyan H, Lee N, Deng S, Jain RK, Johnson TW, Shaw AT, Fantin VR, Smeal T. PF-06463922, an ALK/ROS1 Inhibitor, Overcomes Resistance to First and Second Generation ALK Inhibitors in Preclinical Models. Cancer Cell 2015; 28:70-81. [PMID: 26144315 PMCID: PMC4504786 DOI: 10.1016/j.ccell.2015.05.010] [Citation(s) in RCA: 354] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 03/16/2015] [Accepted: 05/18/2015] [Indexed: 01/15/2023]
Abstract
We report the preclinical evaluation of PF-06463922, a potent and brain-penetrant ALK/ROS1 inhibitor. Compared with other clinically available ALK inhibitors, PF-06463922 displayed superior potency against all known clinically acquired ALK mutations, including the highly resistant G1202R mutant. Furthermore, PF-06463922 treatment led to regression of EML4-ALK-driven brain metastases, leading to prolonged mouse survival, in a superior manner. Finally, PF-06463922 demonstrated high selectivity and safety margins in a variety of preclinical studies. These results suggest that PF-06463922 will be highly effective for the treatment of patients with ALK-driven lung cancers, including those who relapsed on clinically available ALK inhibitors because of secondary ALK kinase domain mutations and/or brain metastases.
Collapse
Affiliation(s)
- Helen Y Zou
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Luc Friboulet
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - David P Kodack
- Department of Radiation Oncology, Edwin L. Steele Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lars D Engstrom
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Qiuhua Li
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Melissa West
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Ruth W Tang
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Hui Wang
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Konstantinos Tsaparikos
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Jinwei Wang
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Sergei Timofeevski
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Ryohei Katayama
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Dac M Dinh
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Hieu Lam
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Justine L Lam
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Shinji Yamazaki
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Wenyue Hu
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Bhushankumar Patel
- Department of Radiation Oncology, Edwin L. Steele Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Divya Bezwada
- Department of Radiation Oncology, Edwin L. Steele Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Rosa L Frias
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Eugene Lifshits
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Sidra Mahmood
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Justin F Gainor
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Timothy Affolter
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Patrick B Lappin
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Hovhannes Gukasyan
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Nathan Lee
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Shibing Deng
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Rakesh K Jain
- Department of Radiation Oncology, Edwin L. Steele Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ted W Johnson
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Alice T Shaw
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Valeria R Fantin
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA
| | - Tod Smeal
- Pfizer World Wide Research and Development, 10724 Science Center Drive, San Diego, CA 92121, USA.
| |
Collapse
|
961
|
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
|
962
|
Cystic Brain Metastases in NSCLC Harboring the EML4-ALK Translocation after Treatment with Crizotinib. J Thorac Oncol 2015; 10:1116-7. [DOI: 10.1097/jto.0000000000000409] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
963
|
Congrès de l’association américaine de recherche contre le cancer — AACR 2015. ONCOLOGIE 2015. [DOI: 10.1007/s10269-015-2535-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
964
|
Jamal-Hanjani M, Lee SM. Lung cancer. Lung Cancer Manag 2015. [DOI: 10.2217/lmt.15.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Mariam Jamal-Hanjani Mariam Jamal-Hanjani is a Specialist Registrar in Medical Oncology. She studied physics and then medicine at University College London (UCL), and is currently pursuing her PhD in non-small-cell lung cancer tumor evolution and intratumor heterogeneity at the Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute.
Collapse
Affiliation(s)
- Mariam Jamal-Hanjani
- University College London Hospitals NHS Trust, UCL Cancer Institute & CRUK Lung Cancer Centre of Excellence, 250 Euston Road, London, NW1 2PT, UK
| | - Siow-Ming Lee
- University College London Hospitals NHS Trust, UCL Cancer Institute & CRUK Lung Cancer Centre of Excellence, 250 Euston Road, London, NW1 2PT, UK
| |
Collapse
|
965
|
[Is chemotherapy still an option in oncogene-addicted non-small cell lung cancer? No]. Bull Cancer 2015; 102:S96-9. [PMID: 26118885 DOI: 10.1016/s0007-4551(15)31225-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 04/09/2015] [Indexed: 11/21/2022]
Abstract
With the emergence of molecular targeted therapies in the management of non-small cell lung cancer, the role of conventional chemotherapy can be questioned. This article presents the key arguments against the use of cytotoxics in presence of a targetable alteration.
Collapse
|
966
|
Oser MG, Niederst MJ, Sequist LV, Engelman JA. Transformation from non-small-cell lung cancer to small-cell lung cancer: molecular drivers and cells of origin. Lancet Oncol 2015; 16:e165-72. [PMID: 25846096 DOI: 10.1016/s1470-2045(14)71180-5] [Citation(s) in RCA: 710] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lung cancer is the most common cause of cancer deaths worldwide. The two broad histological subtypes of lung cancer are small-cell lung cancer (SCLC), which is the cause of 15% of cases, and non-small-cell lung cancer (NSCLC), which accounts for 85% of cases and includes adenocarcinoma, squamous-cell carcinoma, and large-cell carcinoma. Although NSCLC and SCLC are commonly thought to be different diseases owing to their distinct biology and genomic abnormalities, the idea that these malignant disorders might share common cells of origin has been gaining support. This idea has been supported by the unexpected findings that a subset of NSCLCs with mutated EGFR return as SCLC when resistance to EGFR tyrosine kinase inhibitors develops. Additionally, other case reports have described the coexistence of NSCLC and SCLC, further challenging the commonly accepted view of their distinct lineages. Here, we summarise the published clinical observations and biology underlying tumours with combined SCLC and NSCLC histology and cancers that transform from adenocarcinoma to SCLC. We also discuss pre-clinical studies pointing to common potential cells of origin, and speculate how the distinct paths of differentiation are determined by the genomics of each disease.
Collapse
Affiliation(s)
- Matthew G Oser
- Massachusetts General Hospital Cancer Center, Department of Medicine and Harvard Medical School, Boston, MA 02114, USA
| | - Matthew J Niederst
- Massachusetts General Hospital Cancer Center, Department of Medicine and Harvard Medical School, Boston, MA 02114, USA
| | - Lecia V Sequist
- Massachusetts General Hospital Cancer Center, Department of Medicine and Harvard Medical School, Boston, MA 02114, USA
| | - Jeffrey A Engelman
- Massachusetts General Hospital Cancer Center, Department of Medicine and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
967
|
Abstract
Ceritinib is an oral anaplastic lymphoma kinase (ALK) inhibitor developed by Novartis for the treatment of tumours characterised by genetic abnormalities in ALK. ALK is a member of the insulin receptor family of tyrosine kinases that can become oncogenic when fused to other proteins. Ceritinib has been approved in the US under 'Breakthrough Therapy' designation for the second-line treatment of ALK-positive non-small cell lung cancer (NSCLC). Regulatory submissions have also been made in the EU and other countries. Phase III development is ongoing worldwide to evaluate ceritinib both as a first- and second-line therapy for ALK-positive NSCLC. This article summarizes the milestones in the development of ceritinib leading to this first approval for the treatment of patients with ALK-positive metastatic NSCLC who have progressed on or are intolerant to crizotinib.
Collapse
|
968
|
Treatment of ALK-Rearranged Non-Small Cell Lung Cancer: Recent Progress and Future Directions. Drugs 2015; 75:1059-70. [DOI: 10.1007/s40265-015-0415-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
969
|
Gainor JF, Tan DSW, De Pas T, Solomon BJ, Ahmad A, Lazzari C, de Marinis F, Spitaleri G, Schultz K, Friboulet L, Yeap BY, Engelman JA, Shaw AT. Progression-Free and Overall Survival in ALK-Positive NSCLC Patients Treated with Sequential Crizotinib and Ceritinib. Clin Cancer Res 2015; 21:2745-52. [PMID: 25724526 PMCID: PMC4470734 DOI: 10.1158/1078-0432.ccr-14-3009] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 02/08/2015] [Indexed: 01/01/2023]
Abstract
PURPOSE Anaplastic lymphoma kinase (ALK) rearrangements are important therapeutic targets in non-small cell lung cancer (NSCLC) that confer sensitivity to the ALK inhibitors crizotinib and ceritinib. To determine the outcome of sequential treatment with crizotinb and ceritinib, we retrospectively evaluated a cohort of ALK-positive patients treated with both agents. EXPERIMENTAL DESIGN We identified 73 ALK-positive NSCLC patients treated with crizotinib followed by ceritinib at four institutions. Medical records were reviewed to determine overall survival (OS) and progression-free survival (PFS) on crizotinib and ceritinib. RESULTS Among 73 ALK-positive patients, the median PFS (mPFS) on crizotinib was 8.2 months [95% confidence interval (CI), 7.4-10.6]. The median interval from crizotinib discontinuation to initiation of ceritinib was 25 days (range, 1-694). The mPFS on ceritinib was 7.8 months (6.5-9.1). Among 53 patients with no interval therapies between crizotinib and ceritinib, the mPFS on ceritinib was similar at 7.8 months (5.4-9.8). The median combined PFS for sequential treatment with crizotinib and ceritinib was 17.4 months (15.5-19.4). Among 23 patients who underwent post-crizotinib/pre-ceritinib biopsies, there was no difference in PFS on ceritinib between patients with or without ALK resistance mutations (mPFS 5.8 vs. 6.5 months, respectively; P = 0.510). In the overall study population, median OS was 49.4 months (35.5-63.1). CONCLUSIONS Ceritinib has significant antitumor activity in ALK-positive NSCLC-even when crizotinib immediately precedes treatment with ceritinib (median combined PFS 17.0 months). Additional studies are necessary to further define the impact of specific ALK resistance mutations on duration of response to ceritinib.
Collapse
Affiliation(s)
- Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Center and Genome Institute of Singapore, Singapore, Singapore
| | - Tomasso De Pas
- Department of Medicine, Istituto Europeo di Oncologia, Milan, Italy
| | - Benjamin J Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Aziah Ahmad
- Division of Medical Oncology, National Cancer Center and Genome Institute of Singapore, Singapore, Singapore
| | - Chiara Lazzari
- Department of Medicine, Istituto Europeo di Oncologia, Milan, Italy
| | | | | | - Katherine Schultz
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Luc Friboulet
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Beow Y Yeap
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jeffrey A Engelman
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Alice T Shaw
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
970
|
Santarpia M, Gil N, Rosell R. Strategies to overcome resistance to tyrosine kinase inhibitors in non-small-cell lung cancer. Expert Rev Clin Pharmacol 2015; 8:461-77. [PMID: 26068305 DOI: 10.1586/17512433.2015.1055252] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The use of molecularly targeted agents has dramatically improved the prognosis of defined subsets of patients with non-small-cell lung cancer harboring somatically activated oncogenes, such as mutant EGFR or rearranged ALK. However, after initial marked responses to EGFR or ALK tyrosine kinase inhibitors (TKIs), almost all patients inevitably progress due to development of acquired resistance. Multiple molecular mechanisms of resistance have been identified; the best characterized are secondary mutations in the tyrosine kinase domain of the oncogene, such as T790M in EGFR and L1196M in ALK, which prevent target inhibition by the corresponding TKI. Other mechanisms include copy number gain of the ALK fusion gene and the activation of bypass signaling pathways that can maintain downstream proliferation and survival signals despite inhibition of the original drug target. Here, the authors provide an overview of the known mechanisms of resistance to TKIs and outline the therapeutic strategies, including new investigational agents and targeted therapies combinations, that have been developed to overcome resistance.
Collapse
Affiliation(s)
- Mariacarmela Santarpia
- Medical Oncology Unit, Human Pathology Department, University of Messina, Messina, Italy
| | | | | |
Collapse
|
971
|
Okuma Y, Tanaka Y, Kamei T, Hosomi Y, Okamura T. Alectinib for choroidal metastasis in a patient with crizotinib-resistant ALK rearranged positive non-small cell lung cancer. Onco Targets Ther 2015; 8:1321-5. [PMID: 26082648 PMCID: PMC4461085 DOI: 10.2147/ott.s83179] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Choroidal metastasis is rare in cancer patients. Small molecules of molecular targeted agents for lung cancer with actionable mutations were reported to be palliated for symptoms caused by choroidal metastasis. Visual disturbance by choroidal metastasis significantly decreases quality of life during the patient’s remaining lifespan; therefore, radiotherapy or laser photocoagulation is proposed with consensus. However, improvement in survival with matched molecular targeted agents for oncogenic driver mutations reminds us to also be concerned with late treatment toxicities. A 30-year-old female patient previously treated with crizotinib harboring ALK rearranged non-small cell lung cancer complained of visual disturbance, fever, and bone pains undergoing anti-PD-1 antibody treatment. A decreased proportion of ALK fusion was demonstrated by fluorescence in situ hybridization in liver metastasis compared to the primary site in a chemo-naïve state. She was diagnosed with low vision, choroidal metastasis and retinal detachment. Therefore, she started alectinib treatment and both her ocular and systemic symptoms were palliated in a week. Later, she temporarily discontinued alectinib because of skin rash although the choroidal metastasis and retinal detachment resolved and she regained low vision completely at 2 weeks. She obtained partial response with alectinib for more than 5 months after recovering from skin rash.
Collapse
Affiliation(s)
- Yusuke Okuma
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious diseases Center Komagome Hospital, The Jikei University School of Medicine, Tokyo, Japan ; Division of Oncology, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuichiro Tanaka
- Department of Ophthalmology, Tokyo Metropolitan Cancer and Infectious diseases Center Komagome Hospital, Tokyo, Japan
| | - Tina Kamei
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious diseases Center Komagome Hospital, The Jikei University School of Medicine, Tokyo, Japan
| | - Yukio Hosomi
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious diseases Center Komagome Hospital, The Jikei University School of Medicine, Tokyo, Japan
| | - Tatsuru Okamura
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious diseases Center Komagome Hospital, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
972
|
CsToth I, Meert AP, Sculier JP, Berghmans T. [Renal cysts - A novel complication of crizotinib treatment for lung cancer]. Rev Mal Respir 2015; 32:956-8. [PMID: 26033699 DOI: 10.1016/j.rmr.2015.01.007] [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] [Received: 01/02/2015] [Accepted: 01/24/2015] [Indexed: 11/30/2022]
Abstract
We report the case of a woman with an ALK positive lung adenocarcinoma, who developed bilateral complex renal cysts 17 months after the introduction of treatment with crizotinib. Clinical investigation led to the conclusion that the cysts were due to anticancer drug. Regression of the renal cysts was observed one month after cessation of the crizotinib. This case illustrates that specific and little known toxicities can occur with these novel molecules which have entered use for the management of lung cancer.
Collapse
Affiliation(s)
- I CsToth
- Service des soins intensifs et urgences oncologiques, clinique d'oncologie thoracique, institut Jules-Bordet, centre des tumeurs de l'Université Libre de Bruxelles, rue, Héger-Bordet, 1, 1000 Bruxelles, Belgique
| | - A-P Meert
- Service des soins intensifs et urgences oncologiques, clinique d'oncologie thoracique, institut Jules-Bordet, centre des tumeurs de l'Université Libre de Bruxelles, rue, Héger-Bordet, 1, 1000 Bruxelles, Belgique
| | - J-P Sculier
- Service des soins intensifs et urgences oncologiques, clinique d'oncologie thoracique, institut Jules-Bordet, centre des tumeurs de l'Université Libre de Bruxelles, rue, Héger-Bordet, 1, 1000 Bruxelles, Belgique
| | - T Berghmans
- Service des soins intensifs et urgences oncologiques, clinique d'oncologie thoracique, institut Jules-Bordet, centre des tumeurs de l'Université Libre de Bruxelles, rue, Héger-Bordet, 1, 1000 Bruxelles, Belgique.
| |
Collapse
|
973
|
Chau NG, Lorch JH. Exceptional Responders Inspire Change: Lessons for Drug Development From the Bedside to the Bench and Back. Oncologist 2015; 20:699-701. [PMID: 26040619 DOI: 10.1634/theoncologist.2014-0476] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 04/15/2015] [Indexed: 11/17/2022] Open
Affiliation(s)
- Nicole G Chau
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Jochen H Lorch
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
974
|
Brosseau S, Oulkhouir Y, Naltet C, Zalcman G. Résistances aux thérapeutiques ciblées et stratégies des lignes ultérieures dans les CBNPC métastatiques. Bull Cancer 2015; 102:S27-33. [DOI: 10.1016/s0007-4551(15)31215-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 04/09/2015] [Indexed: 11/26/2022]
|
975
|
Amin AD, Rajan SS, Liang WS, Pongtornpipat P, Groysman MJ, Tapia EO, Peters TL, Cuyugan L, Adkins J, Rimsza LM, Lussier YA, Puvvada SD, Schatz JH. Evidence Suggesting That Discontinuous Dosing of ALK Kinase Inhibitors May Prolong Control of ALK+ Tumors. Cancer Res 2015; 75:2916-27. [PMID: 26018086 DOI: 10.1158/0008-5472.can-14-3437] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 05/01/2015] [Indexed: 01/30/2023]
Abstract
The anaplastic lymphoma kinase (ALK) is chromosomally rearranged in a subset of certain cancers, including 2% to 7% of non-small cell lung cancers (NSCLC) and ∼70% of anaplastic large cell lymphomas (ALCL). The ALK kinase inhibitors crizotinib and ceritinib are approved for relapsed ALK(+) NSCLC, but acquired resistance to these drugs limits median progression-free survival on average to ∼10 months. Kinase domain mutations are detectable in 25% to 37% of resistant NSCLC samples, with activation of bypass signaling pathways detected frequently with or without concurrent ALK mutations. Here we report that, in contrast to NSCLC cells, drug-resistant ALCL cells show no evidence of bypassing ALK by activating alternate signaling pathways. Instead, drug resistance selected in this setting reflects upregulation of ALK itself. Notably, in the absence of crizotinib or ceritinib, we found that increased ALK signaling rapidly arrested or killed cells, allowing a prolonged control of drug-resistant tumors in vivo with the administration of discontinuous rather than continuous regimens of drug dosing. Furthermore, even when drug resistance mutations were detected in the kinase domain, overexpression of the mutant ALK was toxic to tumor cells. We confirmed these findings derived from human ALCL cells in murine pro-B cells that were transformed to cytokine independence by ectopic expression of an activated NPM-ALK fusion oncoprotein. In summary, our results show how ALK activation functions as a double-edged sword for tumor cell viability, with potential therapeutic implications.
Collapse
Affiliation(s)
| | - Soumya S Rajan
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona
| | - Winnie S Liang
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona. Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona
| | | | - Matthew J Groysman
- Undergraduate Biology Research Program, University of Arizona, Tucson, Arizona
| | - Edgar O Tapia
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona
| | - Tara L Peters
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona
| | - Lori Cuyugan
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona. Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Jonathan Adkins
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona. Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Lisa M Rimsza
- Department of Pathology, University of Arizona, Tucson, Arizona
| | - Yves A Lussier
- BIO5 Institute, University of Arizona, Tucson, Arizona. Department of Medicine, University of Arizona, Tucson, Arizona. Statistics Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona
| | - Soham D Puvvada
- Department of Medicine, University of Arizona, Tucson, Arizona
| | - Jonathan H Schatz
- BIO5 Institute, University of Arizona, Tucson, Arizona. Department of Medicine, University of Arizona, Tucson, Arizona. Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona.
| |
Collapse
|
976
|
Abstract
Cancer is a close second to heart disease for cause of death in the USA, and could soon surpass heart disease as the population ages and the incidence of cancer continues to increase. While heart disease can be addressed through behavior modification and education (e.g., smoking cessation, dietary changes, exercises that promote cardiovascular fitness), pharmacology and improved surgical devices and methods, cancer ultimately requires improved and novel drug treatments to bring mortality rates down. In 2014, the US FDA approved 17 drugs and/or drug combinations in 12 disease sites for a total of 19 indications in melanoma, hematologic malignancies, gastrointestinal carcinoma, non-small-cell lung cancer, gynecologic malignancies and lymphoma/lymphoproliferative disorders.
Collapse
Affiliation(s)
- Juliet E Wolford
- The Chao Family NCI-Designated Comprehensive Cancer Center, University of California, Irvine Medical Center, 101 The City Drive South, Bldg 56, Orange, CA 92868, USA
| | - Krishnansu S Tewari
- The Chao Family NCI-Designated Comprehensive Cancer Center, University of California, Irvine Medical Center, 101 The City Drive South, Bldg 56, Orange, CA 92868, USA
| |
Collapse
|
977
|
Abstract
Lung cancer is one of the most frequently diagnosed cancers and is the leading cause of cancer-related death worldwide. Non-small-cell lung cancer (NSCLC), a heterogeneous class of tumours, represents approximately 85% of all new lung cancer diagnoses. Tobacco smoking remains the main risk factor for developing this disease, but radon exposure and air pollution also have a role. Most patients are diagnosed with advanced-stage disease owing to inadequate screening programmes and late onset of clinical symptoms; consequently, patients have a very poor prognosis. Several diagnostic approaches can be used for NSCLC, including X-ray, CT and PET imaging, and histological examination of tumour biopsies. Accurate staging of the cancer is required to determine the optimal management strategy, which includes surgery, radiochemotherapy, immunotherapy and targeted approaches with anti-angiogenic monoclonal antibodies or tyrosine kinase inhibitors if tumours harbour oncogene mutations. Several of these driver mutations have been identified (for example, in epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK)), and therapy continues to advance to tackle acquired resistance problems. Also, palliative care has a central role in patient management and greatly improves quality of life. For an illustrated summary of this Primer, visit: http://go.nature.com/rWYFgg.
Collapse
|
978
|
Burns MW, Kim ES. Profile of ceritinib in the treatment of ALK+ metastatic non-small-cell lung cancer. LUNG CANCER-TARGETS AND THERAPY 2015; 6:35-42. [PMID: 28210149 PMCID: PMC5217515 DOI: 10.2147/lctt.s69114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Lung cancer has become one of the leading causes of death in both men and women in the United States, with approximately 230,000 new cases and 160,000 deaths each year. Approximately 80% of lung cancer patients are diagnosed with non-small-cell lung cancer (NSCLC), a subset of epithelial lung cancers that are generally insensitive to chemotherapy. An estimated 3%–7% of NSCLC patients harbor tumors containing anaplastic lymphoma kinase (ALK) gene rearrangement as an oncogenic driver. Subsequent development of the first-generation tyrosine kinase inhibitor crizotinib demonstrated substantial initial ALK+-tumor regression, yet ultimately displayed resistance in treated patients. The recently approved tyrosine kinase inhibitor ceritinib has been shown to be an effective antitumor agent against crizotinib-naïve and -resistant ALK+-NSCLC patients. In this review, we will provide an overview of biology and management of ALK+-NSCLC with a special focus on clinical development of ceritinib.
Collapse
Affiliation(s)
- Mark W Burns
- Wilmot Cancer Center, University of Rochester, Rochester, NY, USA
| | - Eric S Kim
- Wilmot Cancer Center, University of Rochester, Rochester, NY, USA
| |
Collapse
|
979
|
Toyokawa G, Seto T. Updated Evidence on the Mechanisms of Resistance to ALK Inhibitors and Strategies to Overcome Such Resistance: Clinical and Preclinical Data. Oncol Res Treat 2015; 38:291-8. [DOI: 10.1159/000430852] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/21/2015] [Indexed: 11/19/2022]
|
980
|
FDA-approved small-molecule kinase inhibitors. Trends Pharmacol Sci 2015; 36:422-39. [PMID: 25975227 DOI: 10.1016/j.tips.2015.04.005] [Citation(s) in RCA: 728] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 04/02/2015] [Accepted: 04/08/2015] [Indexed: 02/07/2023]
Abstract
Kinases have emerged as one of the most intensively pursued targets in current pharmacological research, especially for cancer, due to their critical roles in cellular signaling. To date, the US FDA has approved 28 small-molecule kinase inhibitors, half of which were approved in the past 3 years. While the clinical data of these approved molecules are widely presented and structure-activity relationship (SAR) has been reported for individual molecules, an updated review that analyzes all approved molecules and summarizes current achievements and trends in the field has yet to be found. Here we present all approved small-molecule kinase inhibitors with an emphasis on binding mechanism and structural features, summarize current challenges, and discuss future directions in this field.
Collapse
|
981
|
Refining the treatment of NSCLC according to histological and molecular subtypes. Nat Rev Clin Oncol 2015; 12:511-26. [PMID: 25963091 DOI: 10.1038/nrclinonc.2015.90] [Citation(s) in RCA: 231] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In the past decade, the characterization of non-small-cell lung cancer (NSCLC) into subtypes based on genotype and histology has resulted in dramatic improvements in disease outcome in select patient subgroups. In particular, molecularly targeted agents that inhibit EGFR or ALK are approved for the treatment of NSCLC harbouring genetic alterations in the genes encoding these proteins. Although acquired resistance usually limits the duration of response to these therapies, a number of new agents have proven effective at tackling specific resistance mechanisms to first-generation inhibitors. Large initiatives are starting to address the role of biomarker-driven targeted therapy in squamous lung cancers, and in the adjuvant setting. Immunotherapy undeniably holds great promise and our understanding of subsets of NSCLC based on patterns of immune response is continuing to evolve. In addition, efforts are underway to identify rare genomic subsets through genomic screening, functional studies, and molecular characterization of exceptional responders. This Review provides an overview of the key developments in the treatment of NSCLC, and discusses potential strategies to further optimize therapy by targeting disease subtypes.
Collapse
|
982
|
CT Radiogenomic Characterization of EGFR, K-RAS, and ALK Mutations in Non-Small Cell Lung Cancer. Eur Radiol 2015; 26:32-42. [PMID: 25956936 DOI: 10.1007/s00330-015-3814-0] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 03/24/2015] [Accepted: 04/21/2015] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To assess the association between CT features and EGFR, ALK, KRAS mutations in non-small cell lung cancer. METHODS Patients undergoing chest CT and testing for the above gene mutations were included. Qualitative evaluation of CTs included: lobe; lesion diameter; shape; margins; ground-glass opacity; density; cavitation; air bronchogram; pleural thickening; intratumoral necrosis; nodules in tumour lobe; nodules in non-tumour lobes; pleural retraction; location; calcifications; emphysema; fibrosis; pleural contact; pleural effusion. Statistical analysis was performed to assess association of features with each gene mutation. ROC curves for gene mutations were drawn; the corresponding area under the curve was calculated. P-values <0.05 were considered significant. RESULTS Of 285 patients, 60/280 (21.43 %) were positive for EGFR mutation; 31/270 (11.48 %) for ALK rearrangement; 64/240 (26.67 %) for KRAS mutation. EGFR mutation was associated with air bronchogram, pleural retraction, females, non-smokers, small lesion size, and absence of fibrosis. ALK rearrangements were associated with age and pleural effusion. KRAS mutation was associated with round shape, nodules in non-tumour lobes, and smoking. CONCLUSIONS This study disclosed associations between CT features and alterations of EGFR (air bronchogram, pleural retraction, small lesion size, absence of fibrosis), ALK (pleural effusion) and KRAS (round lesion shape, nodules in non-tumour lobes). KEY POINTS Air bronchogram, pleural retraction, small size relate to EGFR mutation in NSCLC. Pleural effusion and younger age relate to ALK mutation. Round lesion shape, nodules in non-tumour lobes relate to KRAS mutation.
Collapse
|
983
|
Lampaki S, Lazaridis G, Zarogoulidis K, Kioumis I, Papaiwannou A, Tsirgogianni K, Karavergou A, Tsiouda T, Karavasilis V, Yarmus L, Darwiche K, Freitag L, Sakkas A, Kantzeli A, Baka S, Hohenforst-Schmidt W, Zarogoulidis P. Defining the role of tyrosine kinase inhibitors in early stage non-small cell lung cancer. J Cancer 2015; 6:568-74. [PMID: 26000049 PMCID: PMC4439943 DOI: 10.7150/jca.11893] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 04/14/2015] [Indexed: 01/17/2023] Open
Abstract
Historical, the non-small cell lung cancer (NSCLC) was as a united disease entity and the chemotherapy to the metastatic cancer had limited results. Recent studies for the metastatic non-small cell lung cancer led to the ascertainment that the NSCLC does not constitute exclusively a disease entity, but different neoplasms guided from different molecular paths, different biological behavior and at extension requires different confrontation. Thus the new direction for the therapeutic approach of NSCLC is henceforth the most individualized approach based on the activated molecular paths of tumor. Distinct subtypes of NSCLC are driven by a specific genetic alteration, like EGFR, ALK, ROS1 or BRAF mutations, and these genetic alterations are sensitized to the inhibition of specific oncogenic pathways. The benefit from the use of tyrosine kinase inhibitors in patients with EGFR mutations it was confirmed by six randomized studies of phase III that investigated the role of gefitinib, erlotinib and afatinib. In these studies the response rates vary in the impressive percentages from 55% to 86% and were connected with a remarkable median progression free survival of approximately 8 to 13 months, and with better quality of life compared to that of chemotherapy. In early stages NSCLC is needed the individualization of systemic treatment in order to reduce toxicity that is observed in the classic chemotherapy and to impact outcome. The role of EGFR TKI's has been evaluated in the adjuvant chemotherapy in early stage resected NSCLC. The data from these studies suggest that adjuvant TKI therapy might not increase the overall survival, but delay the recurrences. Prospective trials restricted to EGFR or ALK driven NSCLC subsets potentially offering the opportunity for a definitive answer in early disease adjuvant setting (ALCHEMIST) or as induction treatment before stage III chemo-radiotherapy (RTOG 1210/Alliance 31101), are ongoing. Ongoing prospective trials may offer the opportunity for a definitive answer of the role of tyrosine kinase inhibitors in induction treatment before chemo-radiotherapy or in early disease adjuvant therapy.
Collapse
Affiliation(s)
- Sofia Lampaki
- 1. Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - George Lazaridis
- 2. Department of Medical Oncology, "G. Papageorgiou" University Hospital, Thessaloniki, Greece, Nea Eukarpia
| | - Konstantinos Zarogoulidis
- 1. Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Kioumis
- 1. Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Antonis Papaiwannou
- 1. Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Katerina Tsirgogianni
- 1. Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anastasia Karavergou
- 1. Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Theodora Tsiouda
- 3. Pulmonary Department, "Theageneio Anticancer" Hospital, Thessaloniki, Greece, Alexander Simeonidi 2
| | - Vasilis Karavasilis
- 3. Pulmonary Department, "Theageneio Anticancer" Hospital, Thessaloniki, Greece, Alexander Simeonidi 2
| | - Lonny Yarmus
- 4. Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, U.S.A
| | - Kaid Darwiche
- 5. Department of Interventional Pulmonology, Ruhrlandklinik, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Lutz Freitag
- 5. Department of Interventional Pulmonology, Ruhrlandklinik, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Antonios Sakkas
- 6. Department of Internal Medicine, "Evangelisches Krankenhaus", Mülheim, Germany
| | - Angeliki Kantzeli
- 7. Department of Internal Medicine, "Helios Klinikum", Wuppertal, Germany
| | - Sofia Baka
- 8. Oncology Department, "Interbalkan" European Medical Center, Thessaloniki, Greece
| | | | - Paul Zarogoulidis
- 1. Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
984
|
Niu FY, Wu YL. Personalized treatment strategies for non-small-cell lung cancer in Chinese patients: the role of crizotinib. Onco Targets Ther 2015; 8:999-1007. [PMID: 25999733 PMCID: PMC4427450 DOI: 10.2147/ott.s64664] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Anaplastic lymphoma kinase (ALK) rearrangement is an oncogene targeted with approved drugs second to epidermal growth factor receptor (EGFR) in lung cancer. Crizotinib was developed and introduced into clinical practice rapidly and successfully after the discovery of ALK rearrangement in non-small-cell lung cancer. Chinese and other Asian patients treated with crizotinib seem to have lower toxicity and higher efficacy compared with other ethnicities. Crizotinib showed potent antitumor activity and manageable toxicity in mesenchymal-epithelial transition factor (c-Met)/ROS1-positive non-small-cell lung cancer patients, but prospective clinical trials are still needed to confirm its efficacy and safety. Crizotinib appears to be effective against tumors originating from various organs that harbor ALK abnormalities. In the near future, we would classify the tumors by their genetic information beyond organs, such as ALKoma, EGFRoma, and RAFoma, and a single compound could be used for many different types of cancer in different organs. The major challenge of the widespread use of crizotinib in clinical practice is establishing convenient diagnostic techniques for the detection of ALK/c-Met/ROS1. In the present study, we reviewed the application of crizotinib in Chinese patients.
Collapse
Affiliation(s)
- Fei-Yu Niu
- Graduate School, Southern Medical University, Guangzhou, People’s Republic of China
- Guangdong Lung Cancer Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
| |
Collapse
|
985
|
Kim ES. The Future of Molecular Medicine: Biomarkers, BATTLEs, and Big Data. Am Soc Clin Oncol Educ Book 2015:22-7. [PMID: 25993137 DOI: 10.14694/edbook_am.2015.35.22] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Edward S. Kim
- From the Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC
| |
Collapse
|
986
|
Califano R, Abidin A, Tariq NUA, Economopoulou P, Metro G, Mountzios G. Beyond EGFR and ALK inhibition: Unravelling and exploiting novel genetic alterations in advanced non small-cell lung cancer. Cancer Treat Rev 2015; 41:401-11. [DOI: 10.1016/j.ctrv.2015.03.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 03/03/2015] [Accepted: 03/22/2015] [Indexed: 02/07/2023]
|
987
|
|
988
|
Beyond the Vascular Endothelial Growth Factor Axis: Update on Role of Imaging in Nonantiangiogenic Molecular Targeted Therapies in Oncology. AJR Am J Roentgenol 2015; 204:919-32. [DOI: 10.2214/ajr.14.12876] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
989
|
Gross S, Rahal R, Stransky N, Lengauer C, Hoeflich KP. Targeting cancer with kinase inhibitors. J Clin Invest 2015; 125:1780-9. [PMID: 25932675 DOI: 10.1172/jci76094] [Citation(s) in RCA: 315] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Kinase inhibitors have played an increasingly prominent role in the treatment of cancer and other diseases. Currently, more than 25 oncology drugs that target kinases have been approved, and numerous additional therapeutics are in various stages of clinical evaluation. In this Review, we provide an in-depth analysis of activation mechanisms for kinases in cancer, highlight recent successes in drug discovery, and demonstrate the clinical impact of selective kinase inhibitors. We also describe the substantial progress that has been made in designing next-generation inhibitors to circumvent on-target resistance mechanisms, as well as ongoing strategies for combining kinase inhibitors in the clinic. Last, there are numerous prospects for the discovery of novel kinase targets, and we explore cancer immunotherapy as a new and promising research area for studying kinase biology.
Collapse
|
990
|
Mechanisms of Acquired Resistance to ALK Inhibitors and the Rationale for Treating ALK-positive Lung Cancer. Cancers (Basel) 2015; 7:763-83. [PMID: 25941796 PMCID: PMC4491683 DOI: 10.3390/cancers7020763] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 04/14/2015] [Accepted: 04/21/2015] [Indexed: 12/24/2022] Open
Abstract
The discovery of an echinoderm microtubule-associated protein-like 4 (EML4)-anaplastic lymphoma kinase (ALK) fusion gene led to improved clinical outcomes in patients with lung cancer after the development of the first ALK-targeting agent, crizotinib. Some second-generation ALK tyrosine kinase inhibitors (TKIs), which might be more potent than crizotinib or effective on crizotinib-resistant patients, have been developed. Although these ALK-TKIs show an excellent response initially, most patients eventually acquire resistance. Therefore, careful consideration of the resistance mechanisms might lead to superior therapeutic strategies. Here, we summarize the history of ALK-TKIs and their underlying resistance mechanisms in both the preclinical and clinical settings. In addition, we discuss potential future treatment strategies in ALK-TKI-naïve and -resistant patients with lung cancer harboring the EML4-ALK fusion gene.
Collapse
|
991
|
Duchemann B, Friboulet L, Besse B. Therapeutic management of ALK+ nonsmall cell lung cancer patients. Eur Respir J 2015; 46:230-42. [PMID: 25929953 DOI: 10.1183/09031936.00236414] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 03/14/2015] [Indexed: 02/04/2023]
Abstract
With therapeutic approaches based on oncogene addiction offering significant anticancer benefit, the identification of anaplastic lymphoma kinase (ALK) rearrangements is a key aspect of the management of lung cancers. The EML4-ALK gene fusion is detected in 4-8% of all lung cancers, predominantly in light smokers or nonsmokers. Crizotinib, the first agent to be approved in this indication, is associated with a median progression-free survival of 10.9 months when given as first-line treatment and 7.7 months when administered after chemotherapy. Median overall survival with crizotinib in the second-line setting is 20.3 months. Second-generation ALK inhibitors are currently being evaluated, with early studies giving impressive results, notably in patients resistant to crizotinib or with brain metastases. Among available chemotherapies, pemetrexed appears to be particularly active in this population. Despite this progress, several questions remain unanswered. What detection strategies should be favoured? What underlies the mechanisms of resistance and what options are available to overcome them? What are the best approaches for progressing patients? This review provides an overview of current data in the literature and addresses these questions.
Collapse
Affiliation(s)
- Boris Duchemann
- Dept of Medical Oncology, Hopital Avicenne, Bobigny, France Paris 13 University, Paris, France
| | - Luc Friboulet
- Dept of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Benjamin Besse
- Dept of Cancer Medicine, Gustave Roussy, Villejuif, France Paris-Sud University, Paris, France
| |
Collapse
|
992
|
Sequist LV, Soria JC, Goldman JW, Wakelee HA, Gadgeel SM, Varga A, Papadimitrakopoulou V, Solomon BJ, Oxnard GR, Dziadziuszko R, Aisner DL, Doebele RC, Galasso C, Garon EB, Heist RS, Logan J, Neal JW, Mendenhall MA, Nichols S, Piotrowska Z, Wozniak AJ, Raponi M, Karlovich CA, Jaw-Tsai S, Isaacson J, Despain D, Matheny SL, Rolfe L, Allen AR, Camidge DR. Rociletinib in EGFR-mutated non-small-cell lung cancer. N Engl J Med 2015; 372:1700-9. [PMID: 25923550 DOI: 10.1056/nejmoa1413654] [Citation(s) in RCA: 520] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Non-small-cell lung cancer (NSCLC) with a mutation in the gene encoding epidermal growth factor receptor (EGFR) is sensitive to approved EGFR inhibitors, but resistance develops, mediated by the T790M EGFR mutation in most cases. Rociletinib (CO-1686) is an EGFR inhibitor active in preclinical models of EGFR-mutated NSCLC with or without T790M. METHODS In this phase 1-2 study, we administered rociletinib to patients with EGFR-mutated NSCLC who had disease progression during previous treatment with an existing EGFR inhibitor. In the expansion (phase 2) part of the study, patients with T790M-positive disease received rociletinib at a dose of 500 mg twice daily, 625 mg twice daily, or 750 mg twice daily. Key objectives were assessment of safety, side-effect profile, pharmacokinetics, and preliminary antitumor activity of rociletinib. Tumor biopsies to identify T790M were performed during screening. Treatment was administered in continuous 21-day cycles. RESULTS A total of 130 patients were enrolled. The first 57 patients to be enrolled received the free-base form of rociletinib (150 mg once daily to 900 mg twice daily). The remaining patients received the hydrogen bromide salt (HBr) form (500 mg twice daily to 1000 mg twice daily). A maximum tolerated dose (the highest dose associated with a rate of dose-limiting toxic effects of less than 33%) was not identified. The only common dose-limiting adverse event was hyperglycemia. In an efficacy analysis that included patients who received free-base rociletinib at a dose of 900 mg twice daily or the HBr form at any dose, the objective response rate among the 46 patients with T790M-positive disease who could be evaluated was 59% (95% confidence interval [CI], 45 to 73), and the rate among the 17 patients with T790M-negative disease who could be evaluated was 29% (95% CI, 8 to 51). CONCLUSIONS Rociletinib was active in patients with EGFR-mutated NSCLC associated with the T790M resistance mutation. (Funded by Clovis Oncology; ClinicalTrials.gov number, NCT01526928.).
Collapse
Affiliation(s)
- Lecia V Sequist
- From Massachusetts General Hospital (L.V.S., R.S.H., J.L., Z.P.), Harvard Medical School (L.V.S., G.R.O., R.S.H., Z.P.), and Dana-Farber Cancer Institute (G.R.O.) - all in Boston; the Drug Development Department, Université Paris-Sud, Gustave Roussy Cancer Campus (J.-C.S.), and Institut Gustave Roussy (A.V.), Villejuif - both in France; the David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.W.G., E.B.G., M.A.M., S.N.), and Stanford Cancer Institute, Stanford University, Stanford (H.A.W., J.W.N.) - both in California; the Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit (S.M.G., C.G., A.J.W.); University of Texas M.D. Anderson Cancer Center, Houston (V.P.); the Peter MacCallum Cancer Centre, Melbourne, VIC, Australia (B.J.S.); the Medical University of Gdansk, Gdansk, Poland (R.D.); the University of Colorado (D.L.A., D.R.C.) and University of Colorado Cancer Center (R.C.D.) - both in Aurora; and Clovis Oncology, San Francisco (M.R., C.A.K., S.J.-T., S.L.M., A.R.A.), Boulder, CO (J.I., D.D.), and Cambridge, United Kingdom (L.R.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
993
|
Gelsomino F, Rossi G, Tiseo M. Clinical implications and future perspectives in testing non-small cell lung cancer (NSCLC) for anaplastic lymphoma kinase (ALK) gene rearrangements. J Thorac Dis 2015; 7:220-3. [PMID: 25922694 DOI: 10.3978/j.issn.2072-1439.2015.01.32] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 12/16/2014] [Indexed: 01/19/2023]
Affiliation(s)
- Francesco Gelsomino
- 1 Department of Medical Oncology, Medical Oncology Unit 1, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy ; 2 Section of Pathologic Anatomy, University Hospital Policlinico of Modena, Modena, Italy ; 3 Division of Medical Oncology, University Hospital of Parma, Parma, Italy
| | - Giulio Rossi
- 1 Department of Medical Oncology, Medical Oncology Unit 1, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy ; 2 Section of Pathologic Anatomy, University Hospital Policlinico of Modena, Modena, Italy ; 3 Division of Medical Oncology, University Hospital of Parma, Parma, Italy
| | - Marcello Tiseo
- 1 Department of Medical Oncology, Medical Oncology Unit 1, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy ; 2 Section of Pathologic Anatomy, University Hospital Policlinico of Modena, Modena, Italy ; 3 Division of Medical Oncology, University Hospital of Parma, Parma, Italy
| |
Collapse
|
994
|
Ke EE, Zhou Q, Wu YL. Emerging paradigms in targeted treatments for Asian patients with NSCLC. Expert Opin Pharmacother 2015; 16:1167-76. [DOI: 10.1517/14656566.2015.1040391] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
995
|
Heuckmann JM, Thomas RK. A new generation of cancer genome diagnostics for routine clinical use: overcoming the roadblocks to personalized cancer medicine. Ann Oncol 2015; 26:1830-1837. [PMID: 25899787 DOI: 10.1093/annonc/mdv184] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 04/07/2015] [Indexed: 12/22/2022] Open
Abstract
The identification of 'druggable' kinase gene alterations has revolutionized cancer treatment in the last decade by providing new and successfully targetable drug targets. Thus, genotyping tumors for matching the right patients with the right drugs have become a clinical routine. Today, advances in sequencing technology and computational genome analyses enable the discovery of a constantly growing number of genome alterations relevant for clinical decision making. As a consequence, several technological approaches have emerged in order to deal with these rapidly increasing demands for clinical cancer genome analyses. Here, we describe challenges on the path to the broad introduction of diagnostic cancer genome analyses and the technologies that can be applied to overcome them. We define three generations of molecular diagnostics that are in clinical use. The latest generation of these approaches involves deep and thus, highly sensitive sequencing of all therapeutically relevant types of genome alterations-mutations, copy number alterations and rearrangements/fusions-in a single assay. Such approaches therefore have substantial advantages (less time and less tissue required) over PCR-based methods that typically have to be combined with fluorescence in situ hybridization for detection of gene amplifications and fusions. Since these new technologies work reliably on routine diagnostic formalin-fixed, paraffin-embedded specimens, they can help expedite the broad introduction of personalized cancer therapy into the clinic by providing comprehensive, sensitive and accurate cancer genome diagnoses in 'real-time'.
Collapse
Affiliation(s)
| | - R K Thomas
- Department of Translational Genomics, Medical Faculty, University of Cologne, Cologne, Germany.
| |
Collapse
|
996
|
Hata AN, Engelman JA, Faber AC. The BCL2 Family: Key Mediators of the Apoptotic Response to Targeted Anticancer Therapeutics. Cancer Discov 2015; 5:475-87. [PMID: 25895919 DOI: 10.1158/2159-8290.cd-15-0011] [Citation(s) in RCA: 462] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/27/2015] [Indexed: 12/23/2022]
Abstract
UNLABELLED The ability of cancer cells to suppress apoptosis is critical for carcinogenesis. The BCL2 family proteins comprise the sentinel network that regulates the mitochondrial or intrinsic apoptotic response. Recent advances in our understanding of apoptotic signaling pathways have enabled methods to identify cancers that are "primed" to undergo apoptosis, and have revealed potential biomarkers that may predict which cancers will undergo apoptosis in response to specific therapies. Complementary efforts have focused on developing novel drugs that directly target antiapoptotic BCL2 family proteins. In this review, we summarize the current knowledge of the role of BCL2 family members in cancer development and response to therapy, focusing on targeted therapeutics, recent progress in the development of apoptotic biomarkers, and therapeutic strategies designed to overcome deficiencies in apoptosis. SIGNIFICANCE Apoptosis, long known to be important for response to conventional cytotoxic chemotherapy, has more recently been shown to be essential for the efficacy of targeted therapies. Approaches that increase the likelihood of a cancer to undergo apoptosis following therapy may help improve targeted treatment strategies. Cancer Discov; 5(5); 475-87. ©2015 AACR.
Collapse
Affiliation(s)
- Aaron N Hata
- Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts. Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Jeffrey A Engelman
- Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts. Department of Medicine, Harvard Medical School, Boston, Massachusetts.
| | - Anthony C Faber
- Virginia Commonwealth University Philips Institute for Oral Health Research, School of Dentistry and Massey Cancer Center, Richmond, Virginia.
| |
Collapse
|
997
|
Kanaan Z, Kloecker GH, Paintal A, Perez CA. Novel targeted therapies for resistant ALK-rearranged non-small-cell lung cancer: ceritinib and beyond. Onco Targets Ther 2015; 8:885-92. [PMID: 25945060 PMCID: PMC4408973 DOI: 10.2147/ott.s67262] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality in both sexes, accounting for over one quarter of cancer deaths. Non-small-cell lung cancer (NSCLC) comprises 85%-90% of lung cancer diagnoses and despite advances in multimodality therapies, 5-year survival rates remain dismal with a median survival for patients with metastatic disease of 1 year. The positive outcomes of targeted therapies against the kinase domain of epidermal growth factor receptor in NSCLC triggered consistent efforts to identify the so-called driver mutations as other potential targets. Anaplastic large-cell kinase (ALK) gene rearrangements were identified and targeted resulting in promising response rates in early studies. Unfortunately, most of the patients treated with crizotinib, the first-generation ALK inhibitor, progressed within 9 months. Ceritinib is a second-generation ALK inhibitor that has demonstrated activity in crizotinib-resistant patients, becoming a promising treatment option in this population. Furthermore, additional novel ALK inhibitors and agents targeting alternative pathways have been recruited to rechallenge this evasive disease post-crizotinib resistance.
Collapse
Affiliation(s)
- Zeyad Kanaan
- Division of Medical Oncology and Hematology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Goetz H Kloecker
- Division of Medical Oncology and Hematology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Ajit Paintal
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Cesar A Perez
- Division of Medical Oncology and Hematology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| |
Collapse
|
998
|
Scagliotti GV, Bironzo P, Vansteenkiste JF. Addressing the unmet need in lung cancer: The potential of immuno-oncology. Cancer Treat Rev 2015; 41:465-75. [PMID: 25936526 DOI: 10.1016/j.ctrv.2015.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/19/2015] [Accepted: 04/02/2015] [Indexed: 12/15/2022]
Abstract
Chemotherapy is currently the standard of care for non-oncogene-driven advanced non-small cell lung cancer (NSCLC). Due to improvements in chemotherapeutic choices and supportive care, patients currently typically undergo multiple lines of chemotherapy as their disease progresses. Although treatments have improved over recent years, limited benefits are seen, especially in patients receiving later-line chemotherapy, as response rates can be low, response duration short and survival poor. Furthermore, only a small percentage of patients derive benefit from later-line therapy, with most experiencing deteriorating quality of life and significant toxicities. More recently, molecular targeted therapies have provided improvements in outcomes. However, these treatments only offer a clear benefit in subsets of tumours harbouring the appropriate genomic alteration (mutation, amplification, translocation). Most of the genomic abnormalities susceptible to therapeutic intervention are detected in adenocarcinoma, mainly in never smokers, while alterations in the genome of other histological subtypes are known but specific agents targeting these alterations have yet to be developed. Thus, the therapeutic management of these subtypes represents an ongoing challenge. Recent advances in immunotherapy have highlighted the potential of immuno-oncology based treatments for NSCLC, offering the potential to provide durable responses and outcomes regardless of histology or mutation status. This review discusses the current unmet medical needs in NSCLC, the limits of current first-line and later-line chemotherapy and targeted agents, and the emergence of new therapeutic strategies.
Collapse
Affiliation(s)
- G V Scagliotti
- University of Torino, Department of Oncology, Orbassano, Torino, Italy.
| | - P Bironzo
- University of Torino, Department of Oncology, Orbassano, Torino, Italy
| | - J F Vansteenkiste
- Respiratory Oncology Unit and Trial Unit, Department of Pulmonology, University Hospitals KU Leuven, Leuven, Belgium
| |
Collapse
|
999
|
Abstract
For patients with advanced cancers there has been a concerted effort to transition from a generic treatment paradigm to one based on tumor-specific biologic, and patient-specific clinical characteristics. This approach, known as precision therapy has been made possible owing to widespread availability and a reduction in the cost of cutting-edge technologies that are used to study the genomic, proteomic, and metabolic attributes of individual tumors. This review traces the evolution of precision therapy for lung cancer from the identification of molecular subsets of the disease to the development and approval of tyrosine kinase, as well as immune checkpoint inhibitors for lung cancer therapy. Challenges of the precision therapy era including the emergence of acquired resistance, identification of untargetable mutations, and the effect on clinical trial design are discussed. We conclude by highlighting newer applications for the concept of precision therapy.
Collapse
Affiliation(s)
- Arun Rajan
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - David S Schrump
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
1000
|
Herbst RS, Gandara DR, Hirsch FR, Redman MW, LeBlanc M, Mack PC, Schwartz LH, Vokes E, Ramalingam SS, Bradley JD, Sparks D, Zhou Y, Miwa C, Miller VA, Yelensky R, Li Y, Allen JD, Sigal EV, Wholley D, Sigman CC, Blumenthal GM, Malik S, Kelloff GJ, Abrams JS, Blanke CD, Papadimitrakopoulou VA. Lung Master Protocol (Lung-MAP)-A Biomarker-Driven Protocol for Accelerating Development of Therapies for Squamous Cell Lung Cancer: SWOG S1400. Clin Cancer Res 2015; 21:1514-24. [PMID: 25680375 PMCID: PMC4654466 DOI: 10.1158/1078-0432.ccr-13-3473] [Citation(s) in RCA: 281] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/12/2015] [Indexed: 11/16/2022]
Abstract
The Lung Master Protocol (Lung-MAP, S1400) is a groundbreaking clinical trial designed to advance the efficient development of targeted therapies for squamous cell carcinoma (SCC) of the lung. There are no approved targeted therapies specific to advanced lung SCC, although The Cancer Genome Atlas project and similar studies have detected a significant number of somatic gene mutations/amplifications in lung SCC, some of which are targetable by investigational agents. However, the frequency of these changes is low (5%-20%), making recruitment and study conduct challenging in the traditional clinical trial setting. Here, we describe our approach to development of a biomarker-driven phase II/II multisubstudy "Master Protocol," using a common platform (next-generation DNA sequencing) to identify actionable molecular abnormalities, followed by randomization to the relevant targeted therapy versus standard of care.
Collapse
Affiliation(s)
- Roy S Herbst
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut.
| | - David R Gandara
- University of California Davis Cancer Center, Sacramento, California
| | - Fred R Hirsch
- Department of Medical Oncology, University of Colorado Health Sciences Center, Aurora, Colorado
| | - Mary W Redman
- SWOG Statistical Center; Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Michael LeBlanc
- SWOG Statistical Center; Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Philip C Mack
- University of California Davis Cancer Center, Sacramento, California
| | - Lawrence H Schwartz
- Department of Radiology, Columbia University Medical College, New York, New York
| | - Everett Vokes
- Department of Medicine, University of Chicago Medicine and Biologic Sciences, Chicago, Illinois
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia
| | - Jeffrey D Bradley
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Yang Zhou
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut
| | | | | | | | - Yali Li
- Foundation Medicine, Cambridge, Massachusetts
| | - Jeff D Allen
- Friends of Cancer Research, Washington, District of Columbia
| | - Ellen V Sigal
- Friends of Cancer Research, Washington, District of Columbia
| | - David Wholley
- Foundation for the National Institutes of Health, Bethesda, Maryland
| | | | - Gideon M Blumenthal
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Shakun Malik
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Gary J Kelloff
- Cancer Imaging Program, National Cancer Institute, Bethesda, Maryland
| | - Jeffrey S Abrams
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - Charles D Blanke
- SWOG Group Chair's Office; Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | | |
Collapse
|