901
|
Birkbak NJ, Hiley CT, Swanton C. Evolutionary Precision Medicine: A Role for Repeat Epidermal Growth Factor Receptor Analysis in ALK-Rearranged Lung Adenocarcinoma? J Clin Oncol 2015; 33:3681-3. [PMID: 26417000 DOI: 10.1200/jco.2015.63.2976] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024] Open
Affiliation(s)
- Nicolai Juul Birkbak
- University College London Cancer Institute and The Francis Crick Institute, London, United Kingdom
| | - Crispin T Hiley
- University College London Cancer Institute and The Francis Crick Institute, London, United Kingdom
| | - Charles Swanton
- University College London Cancer Institute and The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
902
|
Salgia R. Diagnostic challenges in non-small-cell lung cancer: an integrated medicine approach. Future Oncol 2015; 11:489-500. [PMID: 25675128 DOI: 10.2217/fon.14.275] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The discovery of diverse driver mutations in lung cancer has heralded a new era of personalized medicine in thoracic oncology, with targeted therapies approved for specific subgroups of patients. The increasing number of patient subgroups that may respond to targeted therapy has resulted in a greater reliance upon effective and increasingly complex diagnostics, which must be interpreted in an interactive multidisciplinary forum. This review discusses the molecular diagnostics available and under development for established and emerging targets, and how these may be integrated into current treatment algorithms. The roles of the pulmonologist, interventional radiologist, thoracic surgeon and molecular pathologist are discussed, and their interactions with the medical oncologist, and/or thoracic surgeon and radiation oncologist in making individual treatment decisions.
Collapse
|
903
|
Wang L, Lin Y, Cai Q, Long H, Zhang Y, Rong T, Ma G, Liang Y. Detection of rearrangement of anaplastic lymphoma kinase (ALK) and mutation of epidermal growth factor receptor (EGFR) in primary pulmonary lymphoepithelioma-like carcinoma. J Thorac Dis 2015; 7:1556-62. [PMID: 26543602 DOI: 10.3978/j.issn.2072-1439.2015.05.11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Primary pulmonary lymphoepithelioma-like carcinoma (LELC) is a distinct rare subtype of lung cancer. The prevalence of anaplastic lymphoma kinase (ALK) rearrangement and epidermal growth factor receptor (EGFR) mutation in primary pulmonary LELC had not been thoroughly investigated. METHODS We investigated a cohort of 42 patients with primary pulmonary LELC and genotyped for ALK rearrangement and EGFR mutation. ALK rearrangement was detected by fluorescence in situ hybridization (FISH). EGFR mutational analysis of exons 18 through 21 was analyzed by TaqMan real-time polymerase chain reaction (PCR). RESULTS Epstein-Barr virus-encoded RNAs (EBERs) showed positive signals in all 42 patients. By immunohistochemistry staining, all patients demonstrated positive expression of CK5/6 and P63, but almost all patients were negative for TTF-1 (34/34, 100%) or CK7 (34/35, 97.1%). None of the 42 patients had ALK rearrangement. Of 42 patients tested, only one patient (2.4%) harbored L858R mutation and gefitinib was applied to this case, however no objective response was observed and the progression free survival (PFS) time was only 1 month. CONCLUSIONS Primary pulmonary LELC is a unique histological subtype of lung cancer. ALK rearrangement and EGFR mutation are lack and they may not be the oncogenic driver gene in pulmonary LELC. Future efforts should be made to explore other oncogenic driver gene to guide targeted therapy in this rare disease to determine the optimal treatment.
Collapse
Affiliation(s)
- Liang Wang
- 1 Department of Hematologic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China ; 2 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 ; 3 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 ; 4 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
| | - Yongbin Lin
- 1 Department of Hematologic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China ; 2 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 ; 3 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 ; 4 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
| | - Qingqing Cai
- 1 Department of Hematologic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China ; 2 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 ; 3 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 ; 4 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
| | - Hao Long
- 1 Department of Hematologic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China ; 2 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 ; 3 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 ; 4 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
| | - Yu Zhang
- 1 Department of Hematologic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China ; 2 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 ; 3 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 ; 4 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
| | - Tiehua Rong
- 1 Department of Hematologic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China ; 2 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 ; 3 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 ; 4 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
| | - Guowei Ma
- 1 Department of Hematologic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China ; 2 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 ; 3 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 ; 4 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
| | - Ying Liang
- 1 Department of Hematologic Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China ; 2 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 ; 3 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 ; 4 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
| |
Collapse
|
904
|
Slater J, Shields L, Racette RJ, Juzwishin D, Coppes M. L'émergence de la thérapeutique de précision: de nouveaux défis et de nouvelles possibilités pour les leaders en santé du Canada. Healthc Manage Forum 2015; 28:S40-S46. [PMID: 26487732 DOI: 10.1177/0840470415607256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
À l'ère de la médecine personnalisée et de la médecine de précision, l'approche envers les soins est en rapide mutation. Les cliniciens exigent de plus en plus d'information génétique et moléculaire et les patients s'attendent à la fournir pour la prévention, le dépistage, le diagnostic, le pronostic, la promotion de la santé et le traitement d'un nombre croissant de maladies. Les leaders en santé canadiens doivent comprendre les changements nécessaires liés à ces technologies perturbantes et ouvrir la voie. Le présent article s'attarde sur la thérapeutique de précision, mais contient également de l'information générale sur les concepts et la terminologie liés à la médecine personnalisée et à la médecine de précision. Il explore également le leadership en santé canadien et les problèmes liés au système qui peuvent nuire à leur mise en œuvre. L'article vise à inspirer, informer et mobiliser les leaders en santé canadiens à amorcer un dialogue sur les transformations nécessaires pour préparer le système de santé à profiter des bienfaits de la thérapeutique de précision.
Collapse
Affiliation(s)
- Jim Slater
- Diagnostic Services Manitoba, Winnipeg (Manitoba) Canada
| | - Laura Shields
- Hoffmann-La Roche Limitée, Mississauga (Ontario) Canada
| | - Ray J Racette
- Collège canadien des leaders en santé, Ottawa (Ontario) Canada
| | | | | |
Collapse
|
905
|
Slater J, Shields L, Racette RJ, Juzwishin D, Coppes M. The emergence of precision therapeutics: New challenges and opportunities for Canada's health leaders. Healthc Manage Forum 2015; 28:S33-S39. [PMID: 26487734 DOI: 10.1177/0840470415604771] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In the era of personalized and precision medicine, the approach to healthcare is quickly changing. Genetic and other molecular information are being increasingly demanded by clinicians and expected by patients for prevention, screening, diagnosis, prognosis, health promotion, and treatment of an increasing number of conditions. As a result of these developments, Canadian health leaders must understand and be prepared to lead the necessary changes associated with these disruptive technologies. This article focuses on precision therapeutics but also provides background on the concepts and terminology related to personalized and precision medicine and explores Canadian health leadership and system issues that may pose barriers to their implementation. The article is intended to inspire, educate, and mobilize Canadian health leaders to initiate dialogue around the transformative changes necessary to ready the healthcare system to realize the benefits of precision therapeutics.
Collapse
Affiliation(s)
- Jim Slater
- Diagnostic Services Manitoba, Winnipeg, Manitoba, Canada.
| | - Laura Shields
- Hoffmann La Roche Limited, Mississauga, Ontario, Canada
| | - Ray J Racette
- Canadian College of Health Leaders, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
906
|
Clinical impact of sequential treatment with ALK-TKIs in patients with advanced ALK-positive non-small cell lung cancer: Results of a multicenter analysis. Lung Cancer 2015; 90:255-60. [DOI: 10.1016/j.lungcan.2015.09.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 09/09/2015] [Accepted: 09/12/2015] [Indexed: 12/20/2022]
|
907
|
Abstract
The purpose of our review is to summarize the clinical activity of oral targeted agents against brain metastases. This includes BRAF inhibitors (dabrafenib and vemurafenib), human epidermal growth factor receptor inhibitors (lapatinib, gefitinib, erlotinib, and afatinib), multi-kinase angiogenesis inhibitors (sorafenib, sunitinib, pazopanib, and vandetanib), and ALK/c-MET (crizotinib) and ALK/IGF-1 (ceritinib) inhibitors. Effective systemic therapies are needed for long-term benefit in brain metastases and documentation of intracranial activity for many therapies is poor. Our review provides a summary of the literature with pertinent data for clinicians. This is needed as subjects with brain metastases are often prevented from enrolling in clinical trials and investigations focused on systemic therapies for brain metastases are rare.
Collapse
|
908
|
Hrustanovic G, Bivona TG. RAS-MAPK signaling influences the efficacy of ALK-targeting agents in lung cancer. Mol Cell Oncol 2015; 3:e1091061. [PMID: 27308613 DOI: 10.1080/23723556.2015.1091061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 08/31/2015] [Accepted: 08/31/2015] [Indexed: 10/22/2022]
Abstract
ALK (anaplastic lymphoma kinase) inhibitors are approved in for ALK gene rearrangement positive (ALK+) lung cancer, but resistance remains a challenge. We discovered that RAS-RAF-MEK-ERK signaling controls the ALK inhibitor response in ALK+ lung cancer and is critical for ALK inhibitor resistance. Upfront ALK-MEK inhibitor polytherapy may enhance response and forestall resistance.
Collapse
Affiliation(s)
- Gorjan Hrustanovic
- Division of Hematology and Medical Oncology, University of California San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Trever G Bivona
- Division of Hematology and Medical Oncology, University of California San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
909
|
Gottesman MM, Lavi O, Hall MD, Gillet JP. Toward a Better Understanding of the Complexity of Cancer Drug Resistance. Annu Rev Pharmacol Toxicol 2015; 56:85-102. [PMID: 26514196 DOI: 10.1146/annurev-pharmtox-010715-103111] [Citation(s) in RCA: 255] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Resistance to anticancer drugs is a complex process that results from alterations in drug targets; development of alternative pathways for growth activation; changes in cellular pharmacology, including increased drug efflux; regulatory changes that alter differentiation pathways or pathways for response to environmental adversity; and/or changes in the local physiology of the cancer, such as blood supply, tissue hydrodynamics, behavior of neighboring cells, and immune system response. All of these specific mechanisms are facilitated by the intrinsic hallmarks of cancer, such as tumor cell heterogeneity, redundancy of growth-promoting pathways, increased mutation rate and/or epigenetic alterations, and the dynamic variation of tumor behavior in time and space. Understanding the relative contribution of each of these factors is further complicated by the lack of adequate in vitro models that mimic clinical cancers. Several strategies to use current knowledge of drug resistance to improve treatment of cancer are suggested.
Collapse
Affiliation(s)
- Michael M Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892; , ,
| | - Orit Lavi
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892; , ,
| | - Matthew D Hall
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892; , ,
| | - Jean-Pierre Gillet
- Laboratory of Molecular Cancer Biology, Molecular Physiology Research Unit-URPhyM, Namur Research Institute for Life Sciences (NARILIS), Faculty of Medicine, University of Namur, B-5000 Namur, Belgium;
| |
Collapse
|
910
|
Patel SP, Schwaederle M, Daniels GA, Fanta PT, Schwab RB, Shimabukuro KA, Kesari S, Piccioni DE, Bazhenova LA, Helsten TL, Lippman SM, Parker BA, Kurzrock R. Molecular inimitability amongst tumors: implications for precision cancer medicine in the age of personalized oncology. Oncotarget 2015; 6:32602-9. [PMID: 26418953 PMCID: PMC4741715 DOI: 10.18632/oncotarget.5289] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/04/2015] [Indexed: 01/01/2023] Open
Abstract
Tumor sequencing has revolutionized oncology, allowing for detailed interrogation of the molecular underpinnings of cancer at an individual level. With this additional insight, it is increasingly apparent that not only do tumors vary within a sample (tumor heterogeneity), but also that each patient's individual tumor is a constellation of unique molecular aberrations that will require an equally unique personalized therapeutic regimen. We report here the results of 439 patients who underwent Clinical Laboratory Improvement Amendment (CLIA)-certified next generation sequencing (NGS) across histologies. Among these patients, 98.4% had a unique molecular profile, and aside from three primary brain tumor patients with a single genetic lesion (IDH1 R132H), no two patients within a given histology were molecularly identical. Additionally, two sets of patients had identical profiles consisting of two mutations in common and no other anomalies. However, these profiles did not segregate by histology (lung adenocarcinoma-appendiceal cancer (KRAS G12D and GNAS R201C), and lung adenocarcinoma-liposarcoma (CDK4 and MDM2 amplification pairs)). These findings suggest that most advanced tumors are molecular singletons within and between histologies, and that tumors that differ in histology may still nonetheless exhibit identical molecular portraits, albeit rarely.
Collapse
Affiliation(s)
- Sandip P. Patel
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Maria Schwaederle
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Gregory A. Daniels
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Paul T. Fanta
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Richard B. Schwab
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Kelly A. Shimabukuro
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Santosh Kesari
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - David E. Piccioni
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Lyudmila A. Bazhenova
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Teresa L. Helsten
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Scott M. Lippman
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Barbara A. Parker
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| |
Collapse
|
911
|
赵 静, 张 坤, 张 力, 王 红. [Clinical Efficacy of Crizotinib in Advanced ALK Positive
Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 18:616-20. [PMID: 26483333 PMCID: PMC6000091 DOI: 10.3779/j.issn.1009-3419.2015.10.03] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 07/26/2015] [Indexed: 11/05/2022]
Abstract
BACKGROUND The aim of this study is to explore clinical efficacy of crizotinib in advanced anaplastic lymphoma kinase (ALK) positive non-small cell lung cancer. METHODS patients with advanced non-small cell lung cancer habouring ALK positive were randomly divided into crizotinib group (n=14) and chemotherapy group (n=14). Patients in the crizotinib group were receive oral treatment with crizotinib (250 mg) twice daily. Patients in the chemotherapy group were administrated docetaxel injection (75 mg/m2) every three weeks and every patient was treated at least 3 period. Then clinical efficacy was observed after 12 mo followed-up. RESULTS Effective rate of patients in the crizotinib group was 64.29%. It was significantly higher than that of the chemotherapy group (21.43%)(P=0.026). The stable rate of patients in the crizotinib group was 85.71%. It was significantly higher than that of the chemotherapy group 40.86% (χ2=5.600, P=0.018). Median progression free survival (PFS) of the crizotinib group was 7.0 mo. It was longer than that of the chemotherapy group (4.0 mo)(P=0.002). CONCLUSIONS Crizotinib is superior to standard chemotherapy in patients with previously treated, advanced ALK positive non-small cell lung cancer. The median PFS of patients is shorter. It can improve the quality of life about patients.
.
Collapse
Affiliation(s)
- 静 赵
- />100071 北京,中国人民解放军军事医学科学院附属医院肺部肿瘤科Department of Lung Oncology, Affiliated Hospital of the PLA Military Academy of Medical Sciences, Beijing 100071, China
| | - 坤 张
- />100071 北京,中国人民解放军军事医学科学院附属医院肺部肿瘤科Department of Lung Oncology, Affiliated Hospital of the PLA Military Academy of Medical Sciences, Beijing 100071, China
| | - 力予 张
- />100071 北京,中国人民解放军军事医学科学院附属医院肺部肿瘤科Department of Lung Oncology, Affiliated Hospital of the PLA Military Academy of Medical Sciences, Beijing 100071, China
| | - 红 王
- />100071 北京,中国人民解放军军事医学科学院附属医院肺部肿瘤科Department of Lung Oncology, Affiliated Hospital of the PLA Military Academy of Medical Sciences, Beijing 100071, China
| |
Collapse
|
912
|
Masters GA, Temin S, Azzoli CG, Giaccone G, Baker S, Brahmer JR, Ellis PM, Gajra A, Rackear N, Schiller JH, Smith TJ, Strawn JR, Trent D, Johnson DH. Systemic Therapy for Stage IV Non-Small-Cell Lung Cancer: American Society of Clinical Oncology Clinical Practice Guideline Update. J Clin Oncol 2015; 33:3488-515. [PMID: 26324367 PMCID: PMC5019421 DOI: 10.1200/jco.2015.62.1342] [Citation(s) in RCA: 380] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE To provide evidence-based recommendations to update the American Society of Clinical Oncology guideline on systemic therapy for stage IV non-small-cell lung cancer (NSCLC). METHODS An Update Committee of the American Society of Clinical Oncology NSCLC Expert Panel based recommendations on a systematic review of randomized controlled trials from January 2007 to February 2014. RESULTS This guideline update reflects changes in evidence since the previous guideline. RECOMMENDATIONS There is no cure for patients with stage IV NSCLC. For patients with performance status (PS) 0 to 1 (and appropriate patient cases with PS 2) and without an EGFR-sensitizing mutation or ALK gene rearrangement, combination cytotoxic chemotherapy is recommended, guided by histology, with early concurrent palliative care. Recommendations for patients in the first-line setting include platinum-doublet therapy for those with PS 0 to 1 (bevacizumab may be added to carboplatin plus paclitaxel if no contraindications); combination or single-agent chemotherapy or palliative care alone for those with PS 2; afatinib, erlotinib, or gefitinib for those with sensitizing EGFR mutations; crizotinib for those with ALK or ROS1 gene rearrangement; and following first-line recommendations or using platinum plus etoposide for those with large-cell neuroendocrine carcinoma. Maintenance therapy includes pemetrexed continuation for patients with stable disease or response to first-line pemetrexed-containing regimens, alternative chemotherapy, or a chemotherapy break. In the second-line setting, recommendations include docetaxel, erlotinib, gefitinib, or pemetrexed for patients with nonsquamous cell carcinoma; docetaxel, erlotinib, or gefitinib for those with squamous cell carcinoma; and chemotherapy or ceritinib for those with ALK rearrangement who experience progression after crizotinib. In the third-line setting, for patients who have not received erlotinib or gefitinib, treatment with erlotinib is recommended. There are insufficient data to recommend routine third-line cytotoxic therapy. Decisions regarding systemic therapy should not be made based on age alone. Additional information can be found at http://www.asco.org/guidelines/nsclc and http://www.asco.org/guidelineswiki.
Collapse
Affiliation(s)
- Gregory A Masters
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| | - Sarah Temin
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| | - Christopher G Azzoli
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| | - Giuseppe Giaccone
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| | - Sherman Baker
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| | - Julie R Brahmer
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| | - Peter M Ellis
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| | - Ajeet Gajra
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| | - Nancy Rackear
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| | - Joan H Schiller
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| | - Thomas J Smith
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| | - John R Strawn
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| | - David Trent
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| | - David H Johnson
- Gregory A. Masters, Helen F. Graham Cancer Center, Newark, DE; Sarah Temin, American Society of Clinical Oncology, Alexandria; Sherman Baker Jr, Virginia Commonwealth University; David Trent, Virginia Cancer Center, Richmond, VA; Christopher G. Azzoli, Massachusetts General Hospital Cancer Center, Boston, MA; Giuseppe Giaccone, Lombardi Cancer Center, Georgetown University, Washington, DC; Julie R. Brahmer and Thomas J. Smith, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Peter M. Ellis, Juravinski Cancer Centre, Hamilton, Ontario, Canada; Ajeet Gajra, Upstate Medical University, Syracuse, NY; Nancy Rackear, Uniting Against Lung Cancer, Fort Lauderdale, FL; Joan H. Schiller, University of Texas Southwestern; David H. Johnson, University of Texas Southwestern Medical Center at Dallas, Dallas; and John R. Strawn, patient representative, Houston, TX
| |
Collapse
|
913
|
Hollingsworth SJ. Precision medicine in oncology drug development: a pharma perspective. Drug Discov Today 2015; 20:1455-63. [PMID: 26482740 DOI: 10.1016/j.drudis.2015.10.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 09/30/2015] [Accepted: 10/12/2015] [Indexed: 11/30/2022]
Abstract
A rapid expansion in precision medicine founded on the potential for durable clinical benefit through matching a drug to a predictive marker used to select patients has driven the development of targeted drugs with accompanied companion diagnostics for patient selection. Oncology has been at the forefront, with the improvements in patient survival notable. Increasing numbers of molecular subgroups require an equally increasing number (and new generation) of highly selective agents targeting inevitably lower incidence molecular segments, posing significant challenges for drug development. Innovative trial designs (umbrella or basket studies) are emerging as patient-centric approaches and public-private partnerships, cross-industry, government and non-profit sector collaborations are enabling implementation. Success will require continued innovation, new paradigms in oncology drug development and market approval and continued collaboration.
Collapse
Affiliation(s)
- Simon J Hollingsworth
- AstraZeneca, Innovative Medicines & Early Development - Oncology, Unit 310 - Darwin Building, Cambridge Science Park, Milton Road, Cambridge CB4 0FZ, UK.
| |
Collapse
|
914
|
Mologni L. Current and future treatment of anaplastic lymphoma kinase-rearranged cancer. World J Clin Oncol 2015; 6:104-8. [PMID: 26468446 PMCID: PMC4600184 DOI: 10.5306/wjco.v6.i5.104] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 07/16/2015] [Accepted: 08/20/2015] [Indexed: 02/06/2023] Open
Abstract
Aberrant forms of the anaplastic lymphoma kinase (ALK) are involved in the pathogenesis of several types of cancer, including anaplastic large cell lymphoma, non-small-cell lung cancer (NSCLC), inflammatory myofibroblastic tumors, colorectal cancer, neuroblastoma and others. In general, the ALK catalytic domain is rearranged and fused to a dimerization domain encoded by an unrelated gene. Less frequently, full-length ALK is activated by point mutations. The common theme is unregulated firing of ALK downstream signalling, leading to uncontrolled cell division and increased cell survival. ALK-driven tumors can be treated with Crizotinib, an orally available dual ALK/MET inhibitor, currently approved for advanced ALK-positive NSCLCs. Crizotinib-treated patients achieve high response rates, with an excellent toxicity profile. However, drug-resistant disease often develops, particularly in NSCLC patients. The processes leading to drug resistance include both ALK-dependent (point mutations or gene amplification), as well as ALK-independent mechanisms, which are here briefly discussed. Recently, Ceritinib has been approved for Crizotinib-refractory NSCLC, further extending patients' survival, but resistance again emerged. Novel ALK kinase inhibitors are currently under clinical development, showing great promise for improved efficacy in drug-resistance disease. It is opinion of the author that drug-resistance is likely to arise under any treatment, due to intrinsic heterogeneity and adaptability of cancer. To prevent or delay this phenomenon, we need to treat less advanced disease, with drugs that are rapidly effective in order not to allow enough time for tumor evolution, and we want to have more and more drugs with non-overlapping resistance profiles, for subsequent lines of targeted therapy. Finally, the use of drug combinations may exponentially decrease the chances of resistance.
Collapse
|
915
|
Vergoulidou M. More than a Decade of Tyrosine Kinase Inhibitors in the Treatment of Solid Tumors: What We Have Learned and What the Future Holds. Biomark Insights 2015; 10:33-40. [PMID: 26483606 PMCID: PMC4599592 DOI: 10.4137/bmi.s22436] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 08/02/2015] [Accepted: 08/05/2015] [Indexed: 12/11/2022] Open
Abstract
The use of tyrosine kinase inhibitors (TKIs) in the treatment of solid tumors is the expected standard of care for many types of tumors. Since the description of signal transduction pathways, followed by the development of small molecules designed to inhibit those pathways, there has been significant improvement not only in progression-free survival and overall survival but also in aiming toward chemotherapy-free treatment of solid tumors to maximize quality of life. This article reviews available TKIs and discusses toxicity, dosing, and resistance.
Collapse
Affiliation(s)
- Maria Vergoulidou
- Department of Haematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charitè Medical University, Berlin, Germany
| |
Collapse
|
916
|
Weber B, Liu M, Sobkin P, Morris SW, Hout D, van der Westhuizen N, Tonseth RP, Saltman DL. Successful treatment of hepatic oligometastases with stereotactic ablative radiotherapy and radiofrequency ablation in an anaplastic lymphoma kinase fusion-positive lung cancer patient. J Med Radiat Sci 2015; 63:67-70. [PMID: 27087977 PMCID: PMC4775829 DOI: 10.1002/jmrs.144] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/18/2015] [Accepted: 08/28/2015] [Indexed: 01/08/2023] Open
Abstract
Local ablative therapy with stereotactic ablative radiotherapy may improve survival in oncogene‐addicted lung cancer patients with extracranial oligometastatic disease treated with targeted therapies. There is limited data on the use of radiofrequency ablation (RFA) in this same setting. We present a case of an anaplastic lymphoma kinase (ALK)‐positive lung cancer patient with hepatic oligometastatic progression who was successfully treated with both stereotactic ablative radiation and RFA while continuing with an ALK inhibitor.
Collapse
Affiliation(s)
- Britta Weber
- Department of Oncology Aarhus University Hospital Aarhus Denmark
| | - Mitchell Liu
- Department of Radiation Oncology BC Cancer Agency Vancouver British Columbia Canada
| | - Paul Sobkin
- Department of Radiology Island Health Victoria British Columbia Canada
| | | | - David Hout
- Insight Genetics, Inc. Nashville Tennessee USA
| | | | - R Petter Tonseth
- Department of Functional Imaging BC Cancer Agency Vancouver British Columbia Canada
| | - David L Saltman
- Department of Medical Oncology BC Cancer Agency Victoria British Columbia Canada
| |
Collapse
|
917
|
Guo L, Zhang H, Shao W, Chen B. Crizotinib as a personalized alternative for targeted anaplastic lymphoma kinase rearrangement in previously treated patients with non-small-cell lung cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:5491-7. [PMID: 26491259 PMCID: PMC4599072 DOI: 10.2147/dddt.s91988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Crizotinib, the first clinically designed and synthesized as a tyrosine kinase inhibitor targeting mesenchymal–epithelial transition factor, indicating marked anticancer activity in patients with advanced, anaplastic lymphoma kinase-positive non-small-cell lung cancer, was approved by the US Food and Drug Administration in 2011. In this review, we focus on the efficacy of crizotinib compared with chemotherapy in advanced anaplastic lymphoma kinase-positive lung cancer and present the role of crizotinib as a personalized alternative in previously treated patients with non-small-cell lung cancer.
Collapse
Affiliation(s)
- Liting Guo
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), The Affiliated Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Haijun Zhang
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), The Affiliated Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Weiwei Shao
- Department of Pathology, the First People's Hospital of Yancheng, Yancheng, Jiangsu, People's Republic of China
| | - Baoan Chen
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), The Affiliated Zhongda Hospital, Medical School of Southeast University, Nanjing
| |
Collapse
|
918
|
Targeting brain metastases in ALK-rearranged non-small-cell lung cancer. Lancet Oncol 2015; 16:e510-21. [DOI: 10.1016/s1470-2045(15)00013-3] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 05/19/2015] [Accepted: 05/26/2015] [Indexed: 01/04/2023]
|
919
|
Voena C, Menotti M, Mastini C, Di Giacomo F, Longo DL, Castella B, Merlo MEB, Ambrogio C, Wang Q, Minero VG, Poggio T, Martinengo C, D'Amico L, Panizza E, Mologni L, Cavallo F, Altruda F, Butaney M, Capelletti M, Inghirami G, Jänne PA, Chiarle R. Efficacy of a Cancer Vaccine against ALK-Rearranged Lung Tumors. Cancer Immunol Res 2015; 3:1333-1343. [PMID: 26419961 DOI: 10.1158/2326-6066.cir-15-0089] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/23/2015] [Indexed: 01/14/2023]
Abstract
Non-small cell lung cancer (NSCLC) harboring chromosomal rearrangements of the anaplastic lymphoma kinase (ALK) gene is treated with ALK tyrosine kinase inhibitors (TKI), but the treatment is successful for only a limited amount of time; most patients experience a relapse due to the development of drug resistance. Here, we show that a vaccine against ALK induced a strong and specific immune response that both prophylactically and therapeutically impaired the growth of ALK-positive lung tumors in mouse models. The ALK vaccine was efficacious also in combination with ALK TKI treatment and significantly delayed tumor relapses after TKI suspension. We found that lung tumors containing ALK rearrangements induced an immunosuppressive microenvironment, regulating the expression of PD-L1 on the surface of lung tumor cells. High PD-L1 expression reduced ALK vaccine efficacy, which could be restored by administration of anti-PD-1 immunotherapy. Thus, combinations of ALK vaccine with TKIs and immune checkpoint blockade therapies might represent a powerful strategy for the treatment of ALK-driven NSCLC.
Collapse
Affiliation(s)
- Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Matteo Menotti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Cristina Mastini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Filomena Di Giacomo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Dario Livio Longo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Molecular Imaging Center, University of Torino, Torino, Italy
| | - Barbara Castella
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Maria Elena Boggio Merlo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Chiara Ambrogio
- Molecular Oncology Program, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Qi Wang
- Department of Pathology, Children's Hospital Harvard Medical School, Boston, MA 02115, USA
| | - Valerio Giacomo Minero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Teresa Poggio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Cinzia Martinengo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Lucia D'Amico
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Elena Panizza
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Luca Mologni
- Department of Health Sciences, University of Milano-Bicocca, Milano, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Fiorella Altruda
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Mohit Butaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Marzia Capelletti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Giorgio Inghirami
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Pasi A Jänne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Belfer Institute for Applied Cancer Science, Dana Farber Cancer Institute, Boston, MA 02115, USA
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy.,Department of Pathology, Children's Hospital Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
920
|
El-Osta H, Shackelford R. Personalized treatment options for ALK-positive metastatic non-small-cell lung cancer: potential role for Ceritinib. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2015; 8:145-54. [PMID: 26622190 PMCID: PMC4638315 DOI: 10.2147/pgpm.s71100] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The fusion of echinoderm microtubule-associated protein-like 4 with the anaplastic lymphoma kinase (EML4-ALK) is found in 3%–7% of non-small-cell lung cancer (NSCLC) cases and confers sensitivity to crizotinib, the first United States Food and Drug Administration (FDA)-approved ALK inhibitor drug. Although crizotinib has an excellent initial therapeutic effect, acquired resistance to this drug invariably develops within the first year of treatment. Resistance may involve secondary gatekeeper mutations within the ALK gene interfering with crizotinib–ALK interactions, or compensatory activation of aberrant bypass signaling pathways. New therapeutic strategies to overcome crizotinib resistance are needed. Ceritinib, a second-generation ALK inhibitor, overcomes several crizotinib-resistant ALK mutations and has demonstrated efficacy against tumor growth in several in vitro and in vivo preclinical models of crizotinib resistance. Notably, the dose-escalation Phase I ASCEND-1 trial has shown a marked activity of ceritinib in both crizotinib-naïve and crizotinib-resistant ALK-rearranged lung cancer. The overall response rate was 58% in a subgroup of patients with ALK-rearranged late-stage NSCLC. Drug discontinuation rate due to toxicity was 10%. The standard dose was established at 750 mg daily. This paper outlines the pathogenesis and treatment of ALK-positive lung cancer, focuses on the preclinical and clinical results surrounding the accelerated FDA approval of ceritinib for the treatment of ALK-positive metastatic NSCLC patients who have progressed on/or are crizotinib intolerant, and discusses the potential efforts seeking to maximize ceritinib efficacy and expand its usage to other indications in cancer therapy.
Collapse
Affiliation(s)
- Hazem El-Osta
- Department of Medicine, Feist-Weiller Cancer Center, Louisiana State University Health Science Center-Shreveport, Shreveport, LA, USA
| | - Rodney Shackelford
- Department of Pathology, Feist-Weiller Cancer Center, Louisiana State University Health Science Center-Shreveport, Shreveport, LA, USA
| |
Collapse
|
921
|
Devarakonda S, Morgensztern D, Govindan R. Genomic alterations in lung adenocarcinoma. Lancet Oncol 2015; 16:e342-51. [PMID: 26149886 DOI: 10.1016/s1470-2045(15)00077-7] [Citation(s) in RCA: 288] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/07/2015] [Accepted: 02/09/2015] [Indexed: 12/11/2022]
Abstract
Treatment for non-small-cell lung cancer is evolving from the use of cytotoxic chemotherapy to personalised treatment based on molecular alterations. This past decade has witnessed substantial progress in the treatment of patients with EGFR mutations and ALK rearrangements, and it is now possible to study complex genomic alterations in cancer using next-generation sequencing. Sequencing data from large-scale consortia, such as The Cancer Genome Atlas, as well as several independent groups, have helped identify novel drivers and potentially targetable alterations in lung adenocarcinomas. These data clearly suggest that lung adenocarcinoma is associated with distinct genomic alterations compared with other lung cancer subtypes, and highlight the widespread molecular heterogeneity that underlies the disease. In this Review, we discuss some of the key findings from genomic studies of lung adenocarcinoma.
Collapse
Affiliation(s)
- Siddhartha Devarakonda
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel Morgensztern
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA; Alvin J Siteman Cancer Center, St Louis, MO, USA
| | - Ramaswamy Govindan
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA; Alvin J Siteman Cancer Center, St Louis, MO, USA.
| |
Collapse
|
922
|
Devarakonda S, Ganesh B, Mann J, Govindan R. Crizotinib: an orphan drug for treating non-small-cell lung cancer. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1086334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
923
|
Gandhi S, Chen H, Zhao Y, Dy GK. First-line treatment of advanced ALK-positive non-small-cell lung cancer. LUNG CANCER-TARGETS AND THERAPY 2015; 6:71-82. [PMID: 28210152 PMCID: PMC5217519 DOI: 10.2147/lctt.s63491] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Non-small-cell lung cancer (NSCLC) is one of the leading causes of cancer deaths, both within the US and worldwide. There have been major treatment advances in NSCLC over the past decade with the discovery of molecular drivers of NSCLC, which has ushered in an era of personalized medicine. There are several actionable genetic aberrations in NSCLC, such as epidermal growth factor receptor and anaplastic lymphoma kinase (ALK). In 3%-7% of NSCLC, a chromosomal inversion event in chromosome 2 leads to fusion of a portion of the ALK gene with the echinoderm microtubule-associated protein-like 4 (EML4) gene. The constitutive activation of the ALK fusion oncogene renders it vulnerable to therapeutic intervention. This review focuses on the first-line treatment of advanced ALK-positive NSCLC using ALK inhibitors. Crizotinib was the first agent proven to be efficacious as first-line treatment for ALK-positive NSCLC. However, acquired resistance inevitably develops. The central nervous system is a sanctuary site that represents a common site for disease progression as well. Hence, more potent, selective next-generation ALK inhibitors that are able to cross the blood-brain barrier have been developed for treatment against crizotinib-resistant ALK-positive NSCLC and are also currently being evaluated for first-line therapy as well. In this review, we provide summary of the clinical experience with these drugs in the treatment of ALK-positive NSCLC.
Collapse
Affiliation(s)
- Shipra Gandhi
- Department of Internal Medicine, State University of New York
| | - Hongbin Chen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Yujie Zhao
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Grace K Dy
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
924
|
Xie J, Zhang X. The Impact of Genomic Profiling for Novel Cancer Therapy--Recent Progress in Non-Small Cell Lung Cancer. J Genet Genomics 2015; 43:3-10. [PMID: 26842989 DOI: 10.1016/j.jgg.2015.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 09/02/2015] [Accepted: 09/07/2015] [Indexed: 12/31/2022]
Abstract
There is high expectation for significant improvements in cancer patient care after completion of the human genome project in 2003. Through pains-taking analyses of genomic profiles in cancer patients, a number of targetable gene alterations have been discovered, with some leading to novel therapies, such as activating mutations of EGFR, BRAF and ALK gene fusions. As a result, clinical management of cancer through targeted therapy has finally become a reality for a subset of cancers, such as lung adenocarcinomas and melanomas. In this review, we summarize how gene mutation discovery leads to new treatment strategies using non-small cell lung cancer (NSCLC) as an example. We also discuss possible future implications of cancer genome analyses.
Collapse
Affiliation(s)
- Jingwu Xie
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; The Simon Cancer Center and The Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Xiaoli Zhang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; The Simon Cancer Center and The Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
925
|
Jiang T, Ren S, Zhou C. Role of circulating-tumor DNA analysis in non-small cell lung cancer. Lung Cancer 2015; 90:128-34. [PMID: 26415994 DOI: 10.1016/j.lungcan.2015.09.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 09/13/2015] [Indexed: 01/04/2023]
Abstract
The discovery of actionable driver mutations such as epidermal growth factor receptor (EGFR) and microtubule-associated protein-like 4 anaplastic lymphoma kinase (EML4-ALK) and their highly responses to EGFR and ALK tyrosine kinase inhibitors (TKIs) in patients with advanced non-small-cell lung cancer (NSCLC) allowed precise medicine into reality. However, a substantial part of patients still have no sufficient tissue to perform genomic analysis. As a promising noninvasive biomarker and potential surrogate for the entire tumor genome, circulating tumor DNA (ctDNA) has been applied to the detection of driver gene mutations and epigenetic alteration and monitoring of tumor burden, acquired resistance, tumor heterogeneity and early diagnosis. Since precise therapy is a strategy that optimal therapy is decided based on simultaneous tumor genome information, ctDNA, as a liquid biopsy, may help to perform dynamic genetic surveillance. In this paper we will perspectively discuss the biology and identification of ctDNA in the blood of NSCLC patients and its clinical applications in patient management.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, PR China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, PR China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, PR China.
| |
Collapse
|
926
|
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]
|
927
|
|
928
|
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
|
929
|
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
|
930
|
Kort A, Sparidans RW, Wagenaar E, Beijnen JH, Schinkel AH. Brain accumulation of the EML4-ALK inhibitor ceritinib is restricted by P-glycoprotein (P-GP/ABCB1) and breast cancer resistance protein (BCRP/ABCG2). Pharmacol Res 2015; 102:200-7. [PMID: 26361725 DOI: 10.1016/j.phrs.2015.09.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/04/2015] [Accepted: 09/04/2015] [Indexed: 11/27/2022]
Abstract
We aimed to clarify the roles of the multidrug transporters ABCB1 and ABCG2 in oral availability and brain accumulation of ceritinib, an oral anaplastic lymphoma kinase (ALK) inhibitor used to treat metastatic non-small cell lung cancer (NSCLC) after progression on crizotinib. Importantly, NSCLC is prone to form brain metastases. Transport of ceritinib by human (h) ABCB1 or hABCG2 or mouse (m) Abcg2 was assessed in vitro. To study the single and combined roles of Abcb1a/1b and Abcg2 in ceritinib disposition in vivo, we used appropriate knockout mouse strains. Ceritinib was very efficiently transported by hABCB1, and efficiently by hABCG2 and mAbcg2 in vitro, and transport was specifically inhibited by the ABCB1 inhibitor zosuquidar and ABCG2 inhibitor Ko143, respectively. Absorption and 24-h oral availability were not significantly affected by the absence of Abcb1 and/or Abcg2, but the brain concentrations were greatly increased (>38-fold) in Abcb1a/1b(-/-) mice at 3 and 24h after oral administration of 20mg/kg ceritinib. The brain concentrations increased another ∼ 3-fold (to >90-fold) in Abcb1a/1b;Abcg2(-/-) mice, indicating that there was a significant additional effect of Abcg2-mediated transport of ceritinib as well in vivo. Overall, brain accumulation, but not the 24-h oral availability of ceritinib were profoundly restricted by Abcb1a/1b and Abcg2, with Abcb1a/1b being the dominant efflux protein. Our data suggest that coadministration of ceritinib with a dual ABCB1 and ABCG2 inhibitor may improve treatment of brain (micro) metastases positioned behind a functionally intact blood-brain barrier, and possibly also of tumors resistant to ceritinib due to ABCB1 or ABCG2 overexpression.
Collapse
Affiliation(s)
- Anita Kort
- Division of Molecular Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands; Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Rolf W Sparidans
- Division of Pharmacoepidemiology & Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584CG Utrecht, The Netherlands
| | - Els Wagenaar
- Division of Molecular Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Jos H Beijnen
- Division of Pharmacoepidemiology & Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584CG Utrecht, The Netherlands; Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands; Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Alfred H Schinkel
- Division of Molecular Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands.
| |
Collapse
|
931
|
Structural insight into selectivity and resistance profiles of ROS1 tyrosine kinase inhibitors. Proc Natl Acad Sci U S A 2015; 112:E5381-90. [PMID: 26372962 DOI: 10.1073/pnas.1515281112] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Oncogenic ROS1 fusion proteins are molecular drivers in multiple malignancies, including a subset of non-small cell lung cancer (NSCLC). The phylogenetic proximity of the ROS1 and anaplastic lymphoma kinase (ALK) catalytic domains led to the clinical repurposing of the Food and Drug Administration (FDA)-approved ALK inhibitor crizotinib as a ROS1 inhibitor. Despite the antitumor activity of crizotinib observed in both ROS1- and ALK-rearranged NSCLC patients, resistance due to acquisition of ROS1 or ALK kinase domain mutations has been observed clinically, spurring the development of second-generation inhibitors. Here, we profile the sensitivity and selectivity of seven ROS1 and/or ALK inhibitors at various levels of clinical development. In contrast to crizotinib's dual ROS1/ALK activity, cabozantinib (XL-184) and its structural analog foretinib (XL-880) demonstrate a striking selectivity for ROS1 over ALK. Molecular dynamics simulation studies reveal structural features that distinguish the ROS1 and ALK kinase domains and contribute to differences in binding site and kinase selectivity of the inhibitors tested. Cell-based resistance profiling studies demonstrate that the ROS1-selective inhibitors retain efficacy against the recently reported CD74-ROS1(G2032R) mutant whereas the dual ROS1/ALK inhibitors are ineffective. Taken together, inhibitor profiling and stringent characterization of the structure-function differences between the ROS1 and ALK kinase domains will facilitate future rational drug design for ROS1- and ALK-driven NSCLC and other malignancies.
Collapse
|
932
|
Abstract
In recent years, many personalized treatments have been developed for NSCLC (non-small-cell lung cancer) patients. Among these, gefitinib, erlotinib, and afatinib are selective epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors for patients with EGFR gene mutations, while crizotinib and ceritinib are two new tyrosine kinase inhibitors directed against the echinoderm microtubule-like protein 4-anaplastic lymphoma kinase translocation. The possibility of these new molecules being used to treat patients without adenocarcinoma histology is notably small. For example, EGFR mutations and anaplastic lymphoma kinase fusion gene rearrangement are rare in patients with squamous cell carcinoma (generally <1%). Additionally, the benefit of targeted treatment approaches in patients with small-cell lung cancer histology is limited. All of these findings highlight the distinctive nature of adenocarcinoma of the lung among all lung cancer subtypes. Unfortunately, to date, less than 15% of patients with adenocarcinoma of the lung are ideal candidates for these targeted therapies.
Collapse
Affiliation(s)
- Giandomenico Roviello
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
933
|
Maione P, Sacco PC, Sgambato A, Casaluce F, Rossi A, Gridelli C. Overcoming resistance to targeted therapies in NSCLC: current approaches and clinical application. Ther Adv Med Oncol 2015; 7:263-73. [PMID: 26327924 DOI: 10.1177/1758834015595048] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The discovery that a number of aberrant tumorigenic processes and signal transduction pathways are mediated by druggable protein kinases has led to a revolutionary change in nonsmall cell lung cancer (NSCLC) treatment. Epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) are the targets of several tyrosine kinase inhibitors (TKIs), some of them approved for treatment and others currently in clinical development. First-generation agents offer, in target populations, a substantial improvement of outcomes compared with standard chemotherapy in the treatment of advanced NSCLC. Unfortunately, drug resistance develops after initial benefit through a variety of mechanisms. Novel generation EGFR and ALK inhibitors are currently in advanced clinical development and are producing encouraging results in patients with acquired resistance to previous generation agents. The search for new drugs or strategies to overcome the TKI resistance in patients with EGFR mutations or ALK rearrangements is to be considered a priority for the improvement of outcomes in the treatment of advanced NSCLC.
Collapse
Affiliation(s)
- Paolo Maione
- Division of Medical Oncology, 'S. G. Moscati' Hospital, Contrada Amoretta, Avellino, 83100, Italy
| | | | - Assunta Sgambato
- Department of Clinical and Experimental Medicine, Second University of Naples, Naples, Italy
| | - Francesca Casaluce
- Department of Clinical and Experimental Medicine, Second University of Naples, Naples, Italy
| | - Antonio Rossi
- Division of Medical Oncology, 'S. G. Moscati' Hospital, Avellino, Italy
| | - Cesare Gridelli
- Division of Medical Oncology, 'S. G. Moscati' Hospital, Avellino, Italy
| |
Collapse
|
934
|
Paoletti X, Ezzalfani M, Le Tourneau C. Statistical controversies in clinical research: requiem for the 3 + 3 design for phase I trials. Ann Oncol 2015; 26:1808-1812. [PMID: 26088197 PMCID: PMC4551156 DOI: 10.1093/annonc/mdv266] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/18/2015] [Accepted: 06/02/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND More than 95% of published phase I trials have used the 3 + 3 design to identify the dose to be recommended for phase II trials. However, the statistical community agrees on the limitations of the 3 + 3 design compared with model-based approaches. Moreover, the mechanisms of action of targeted agents strongly challenge the hypothesis that the maximum tolerated dose constitutes the optimal dose, and more outcomes including clinical and biological activity increasingly need to be taken into account to identify the optimal dose. PATIENTS AND METHODS We review key elements from clinical publications and from the statistical literature to show that the 3 + 3 design lacks the necessary flexibility to address the challenges of targeted agents. RESULTS The design issues raised by expansion cohorts, new definitions of dose-limiting toxicity and trials of combinations are not easily addressed by the 3 + 3 design or its extensions. CONCLUSIONS Alternative statistical proposals have been developed to make a better use of the complex data generated by phase I trials. Their applications require a close collaboration between all actors of early phase clinical trials.
Collapse
Affiliation(s)
- X Paoletti
- Biostatistics and Epidemiology Department, Gustave Roussy, Villejuif; INSERM U1018, CESP, Paris-Sud University, Villejuif.
| | - M Ezzalfani
- INSERM/Institut Curie/Mines ParisTech U900, Paris
| | - C Le Tourneau
- INSERM/Institut Curie/Mines ParisTech U900, Paris; Department of Medical Oncology, Clinical Trial Unit, Institut Curie, Paris & Saint-Cloud, France
| |
Collapse
|
935
|
Hrustanovic G, Olivas V, Pazarentzos E, Tulpule A, Asthana S, Blakely CM, Okimoto RA, Lin L, Neel DS, Sabnis A, Flanagan J, Chan E, Varella-Garcia M, Aisner DL, Vaishnavi A, Ou SHI, Collisson EA, Ichihara E, Mack PC, Lovly CM, Karachaliou N, Rosell R, Riess JW, Doebele RC, Bivona TG. RAS-MAPK dependence underlies a rational polytherapy strategy in EML4-ALK-positive lung cancer. Nat Med 2015; 21:1038-47. [PMID: 26301689 PMCID: PMC4734742 DOI: 10.1038/nm.3930] [Citation(s) in RCA: 236] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 07/23/2015] [Indexed: 12/20/2022]
Abstract
One strategy for combating cancer-drug resistance is to deploy rational polytherapy up front that suppresses the survival and emergence of resistant tumor cells. Here we demonstrate in models of lung adenocarcinoma harboring the oncogenic fusion of ALK and EML4 that the GTPase RAS-mitogen-activated protein kinase (MAPK) pathway, but not other known ALK effectors, is required for tumor-cell survival. EML4-ALK activated RAS-MAPK signaling by engaging all three major RAS isoforms through the HELP domain of EML4. Reactivation of the MAPK pathway via either a gain in the number of copies of the gene encoding wild-type K-RAS (KRAS(WT)) or decreased expression of the MAPK phosphatase DUSP6 promoted resistance to ALK inhibitors in vitro, and each was associated with resistance to ALK inhibitors in individuals with EML4-ALK-positive lung adenocarcinoma. Upfront inhibition of both ALK and the kinase MEK enhanced both the magnitude and duration of the initial response in preclinical models of EML4-ALK lung adenocarcinoma. Our findings identify RAS-MAPK dependence as a hallmark of EML4-ALK lung adenocarcinoma and provide a rationale for the upfront inhibition of both ALK and MEK to forestall resistance and improve patient outcomes.
Collapse
Affiliation(s)
- Gorjan Hrustanovic
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Victor Olivas
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Evangelos Pazarentzos
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Asmin Tulpule
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Saurabh Asthana
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Collin M Blakely
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Ross A Okimoto
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Luping Lin
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Dana S Neel
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Amit Sabnis
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Jennifer Flanagan
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Elton Chan
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Marileila Varella-Garcia
- Department of Medicine, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
- Department of Pathology, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
| | - Dara L Aisner
- Department of Pathology, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
| | - Aria Vaishnavi
- Department of Medicine, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
| | - Sai-Hong I Ou
- Division of Hematology-Oncology, University of California Irvine School of Medicine, Orange, California, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, California, USA
| | - Eric A Collisson
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Eiki Ichihara
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Philip C Mack
- University of California Davis School of Medicine
- Comprehensive Cancer Center, Sacramento, California, USA
| | - Christine M Lovly
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Niki Karachaliou
- Cancer Biology and Precision Medicine Program Catalan Institute of Oncology Hospital Germans Trias i Pujol Badalona, Barcelona, Spain
| | - Rafael Rosell
- Cancer Biology and Precision Medicine Program Catalan Institute of Oncology Hospital Germans Trias i Pujol Badalona, Barcelona, Spain
| | - Jonathan W Riess
- University of California Davis School of Medicine
- Comprehensive Cancer Center, Sacramento, California, USA
| | - Robert C Doebele
- Department of Medicine, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
| | - Trever G Bivona
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| |
Collapse
|
936
|
Gridelli C, Balducci L, Ciardiello F, Di Maio M, Felip E, Langer C, Lilenbaum RC, Perrone F, Senan S, de Marinis F. Treatment of Elderly Patients With Non–Small-Cell Lung Cancer: Results of an International Expert Panel Meeting of the Italian Association of Thoracic Oncology. Clin Lung Cancer 2015; 16:325-33. [DOI: 10.1016/j.cllc.2015.02.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/18/2015] [Accepted: 02/26/2015] [Indexed: 12/29/2022]
|
937
|
Kang GA, Lee M, Song D, Lee HK, Ahn S, Park CH, Lee CO, Yun CS, Jung H, Kim P, Ha JD, Cho SY, Kim HR, Hwang JY. Synthesis and evaluation of novel 2,4-diaminopyrimidines bearing bicyclic aminobenzazepines for anaplastic lymphoma kinase (ALK) inhibitor. Bioorg Med Chem Lett 2015; 25:3992-8. [DOI: 10.1016/j.bmcl.2015.07.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/02/2015] [Accepted: 07/04/2015] [Indexed: 10/23/2022]
|
938
|
Cortinovis DL, Canova S, Bidoli P. What are the options for non-small-cell lung cancer patients post second-line therapy? Future Oncol 2015; 11:2379-82. [DOI: 10.2217/fon.15.159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Diego Luigi Cortinovis
- Department of Oncology, San Gerardo Hospital, Monza, Via Pergolesi 33, 20900, Monza, Italy
| | - Stefania Canova
- Department of Oncology, San Gerardo Hospital, Monza, Via Pergolesi 33, 20900, Monza, Italy
| | - Paolo Bidoli
- Department of Oncology, San Gerardo Hospital, Monza, Via Pergolesi 33, 20900, Monza, Italy
| |
Collapse
|
939
|
Liao BC, Lin CC, Shih JY, Yang JCH. Treating patients with ALK-positive non-small cell lung cancer: latest evidence and management strategy. Ther Adv Med Oncol 2015; 7:274-90. [PMID: 26327925 PMCID: PMC4543853 DOI: 10.1177/1758834015590593] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Rearrangements in anaplastic lymphoma kinase (ALK) gene and echinoderm microtubule-associated protein-like 4 (EML4) gene were first described in a small portion of patients with non-small cell lung cancer (NSCLC) in 2007. Fluorescence in situ hybridization is used as the diagnostic test for detecting an EML4-ALK rearrangement. Crizotinib, an ALK inhibitor, is effective in treating advanced ALK-positive NSCLC, and the US Food and Drug Administration approved it for treating ALK-positive NSCLC in 2011. Several mechanisms of acquired resistance to crizotinib have recently been reported. Second-generation ALK inhibitors were designed to overcome these resistance mechanisms. Two of them, ceritinib and alectinib, were approved in 2014 for advanced ALK-positive NSCLC in the US and Japan, respectively. Heat shock protein 90 (Hsp90) inhibitors also showed activity against ALK-positive NSCLC. Here we review the recent development of crizotinib, ceritinib, alectinib and other second-generation ALK inhibitors as well as Hsp90 inhibitors. We also discuss management strategies for advanced ALK-positive NSCLC.
Collapse
Affiliation(s)
- Bin-Chi Liao
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Chi Lin
- Department of Oncology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, Taiwan
| | - Jin-Yuan Shih
- Department of Internal Medicine, National Taiwan University Hospital, Taiwan Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - James Chih-Hsin Yang
- Department of Oncology, National Taiwan University Hospital, Taiwan Graduate Institute of Oncology and Cancer Research Center, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
940
|
Targeted Therapy for Brain Metastases in EGFR-Mutated and ALK-Rearranged Non-Small-Cell Lung Cancer. J Thorac Oncol 2015; 10:1268-1278. [DOI: 10.1097/jto.0000000000000615] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
941
|
Abstract
The therapeutic targeting of anaplastic lymphoma kinase (ALK) has been a burgeoning area of research since 2007 when ALK fusions were initially identified in patients with non-small cell lung cancer. The field has rapidly progressed through development of the first-generation ALK inhibitor, crizotinib, to an understanding of mechanisms of acquired resistance to crizotinib and is currently witnessing an explosion in the development of next-generation ALK inhibitors such as ceritinib, alectinib, PF-06463922, AP26113, X-396, and TSR-011. As with most targeted therapies, acquired resistance appears to be an inevitable outcome. Current preclinical and clinical studies are focused on the development of rational therapeutic strategies, including novel ALK inhibitors, as well as rational combination therapies to maximize disease control by delaying or overcoming acquired therapeutic resistance. This review summarizes the existing clinical data and ongoing research pertaining to the clinical application of ALK inhibitors in patients with non-small cell lung cancer.
Collapse
Affiliation(s)
- Wade T. Iams
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Christine M. Lovly
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN 37232, USA
| |
Collapse
|
942
|
Teuwen LA, Van den Mooter T, Dirix L. Management of pulmonary toxicity associated with targeted anticancer therapies. Expert Opin Drug Metab Toxicol 2015; 11:1695-707. [PMID: 26293379 DOI: 10.1517/17425255.2015.1080687] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Targeted anticancer therapies act by interfering with defined molecular entities and/or biologic pathways. Because of their more specific mechanism of action, adverse events (AEs) on healthy tissues are intended to be minimal, resulting in a different toxicity profile from that observed with conventional cytotoxic chemotherapy. Pulmonary AEs are rare but potentially life-threatening and it is, therefore, critical to recognize early on and manage appropriately. AREAS COVERED In this review, we aim to offer an overview of both more frequent and rare pulmonary AEs caused by targeted anticancer therapies and discuss possible treatment algorithms. Anti-vascular endothelial growth factor, anti-human epidermal growth factor receptor and anti-CD20 therapy will be reviewed, as well as immune checkpoint inhibitors, anaplastic lymphoma kinase inhibitors and mammalian target of rapamycin inhibitors. EXPERT OPINION Novel agents used in the treatment of cancer have specific side-effects, the result of allergic reactions, on-target and off-target effects. Clinical syndromes associated with pulmonary toxicity vary from bronchospasms, hypersensitivity reactions, pneumonitis, acute respiratory distress, lung bleeding, pleural effusion to pneumothorax. Knowledge of risk factors, a high index of suspicion and a complete diagnostic work-up are essential for limiting the risk of these events becoming life threatening. The development of treatment algorithms is extremely helpful in managing these events. It is probable that these toxicities will be even more frequent with the introduction of combination therapies with the obvious challenge of discerning the responsible agent.
Collapse
Affiliation(s)
- Laure-Anne Teuwen
- a 1 Sint-Augustinus, Resident in Internal Medicine , Oosterveldlaan 24, 2610 Wilrijk-Antwerp, Belgium
| | - Tom Van den Mooter
- b 2 Sint-Augustinus, Resident in Medical Oncology , Oosterveldlaan 24, 2610 Wilrijk-Antwerp, Belgium
| | - Luc Dirix
- c 3 Sint-Augustinus, Medical Oncology , Oosterveldlaan 24, 2610 Wilrijk-Antwerp, Belgium +32 34 433 737 ; +32 34 430 09 ;
| |
Collapse
|
943
|
RESPONSE OF UVEAL METASTASES TO ALK INHIBITORS IN ALK-POSITIVE NON-SMALL-CELL LUNG CANCER. Retin Cases Brief Rep 2015; 10:37-40. [PMID: 26263240 DOI: 10.1097/icb.0000000000000190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE To present a case of uveal metastases in a patient with anaplastic lymphoma kinase (ALK)-positive non-small-cell lung cancer and the response to systemic ALK inhibitors. METHODS A retrospective case report. A 75-year-old nonsmoker who has ALK-positive left upper bronchus adenocarcinoma-developed uveal metastases during his course of treatment. RESULTS Initially, the patient's disseminated malignancy showed a significant response to crizotinib. However, because the bone metastases started progressing again and he developed bilateral ocular metastases, he was switched to ceritinib. After initiation of ceritinib therapy, the uveal melanomas have shown a significant decrease in thickness and no new lesions have developed. CONCLUSION ALK inhibitors are an effective first-line treatment in patients with ALK-positive uveal metastases secondary to ALK-positive non-small-cell lung cancer. Despite the fact that crizotinib, a first-generation ALK inhibitor, is initially effective in dealing with non-small-cell lung cancer and its metastases, resistance to it seems to develop on a regular basis. A second-generation ALK inhibitor, such as ceritinib, is also effective in overcoming this resistance in treating those with ALK-positive uveal metastases.
Collapse
|
944
|
Bironzo P, Mele T, Novello S. Achievements in targeted therapies. Lung Cancer 2015. [DOI: 10.1183/2312508x.10010714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
945
|
Adam V, Dooms C, Vansteenkiste J. Lung cancer at the intensive care unit: The era of targeted therapy. Lung Cancer 2015; 89:218-21. [DOI: 10.1016/j.lungcan.2015.05.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/11/2015] [Indexed: 11/30/2022]
|
946
|
Kaczmar J, Mehra R. The efficacy of ceritinib in patients with ALK-positive non-small cell lung cancer. Ther Adv Respir Dis 2015; 9:236-41. [PMID: 26229087 DOI: 10.1177/1753465815597834] [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: 01/23/2023] Open
Abstract
Research over the last decade has determined that the gene rearrangement involving the anaplastic lymphoma kinase (ALK) gene is an oncogenic driver in approximately 5% of patients with non-small cell lung carcinoma (NSCLC). This review describes the discovery of the ALK translocation, development of ALK directed therapy, and acquired resistance to ALK directed therapy with a focus on the clinical data and efficacy of the most recently approved ALK inhibitor, ceritinib.
Collapse
Affiliation(s)
- John Kaczmar
- Fox Chase Cancer Center, 333 Cottman Avenue Philadelphia, PA 19111, USA
| | - Ranee Mehra
- Fox Chase Cancer Center, 333 Cottman Avenue Philadelphia, PA 19111, USA
| |
Collapse
|
947
|
Berghoff AS, Preusser M. The future of targeted therapies for brain metastases. Future Oncol 2015; 11:2315-27. [DOI: 10.2217/fon.15.127] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Brain metastases (BM) are an increasing challenge in the management of patients with advanced cancer. Treatment options for BM are limited and mainly focus on the application of local therapies. Systemic therapies including targeted therapies are only poorly investigated, as patients with BM were frequently excluded from clinical trials. Several targeted therapies have shown promising activity in patients with BM. In the present review we discuss existing and emerging targeted therapies for the most frequent BM primary tumor types. We focus on challenges in the conduction of clinical trials on targeted therapies in BM patients such as patient selection, combination with radiotherapy, the obstacles of the blood–brain barrier and the definition of study end points.
Collapse
Affiliation(s)
- Anna S Berghoff
- Department for Medicine I, Comprehensive Cancer Center Central Nervous System Unit (CCC-CNS), Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center – CNS Tumors Unit, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Department for Medicine I, Comprehensive Cancer Center Central Nervous System Unit (CCC-CNS), Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center – CNS Tumors Unit, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
948
|
Guérin A, Sasane M, Wakelee H, Zhang J, Culver K, Dea K, Nitulescu R, Galebach P, Macalalad AR. Treatment, overall survival, and costs in patients with ALK-positive non-small-cell lung cancer after crizotinib monotherapy. Curr Med Res Opin 2015; 31:1587-97. [PMID: 26029864 DOI: 10.1185/03007995.2015.1057115] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Limited post-crizotinib treatment options for ALK-positive non-small cell lung cancer (NSCLC) might lead to poor survival and high economic burden. OBJECTIVE To evaluate real-world treatment patterns, overall survival (OS), and costs following crizotinib discontinuation. METHODS This study used chart review and claims data. First, 27 participating US oncologists reviewed medical records of ALK-positive NSCLC patients who discontinued crizotinib monotherapy and reported patient demographic and clinical information, including post-crizotinib treatment and mortality. OS was estimated using Kaplan-Meier analyses. Second, three large administrative US claims databases were pooled. NSCLC patients were selected if they discontinued crizotinib monotherapy. Post-crizotinib costs were analyzed separately for patients who did or did not discontinue antineoplastic treatment after crizotinib monotherapy. All data were collected prior to ceritinib approval for this patient population. RESULTS A total of 119 ALK-positive NSCLC patients discontinued crizotinib monotherapy. Upon discontinuation, 42% had no additional antineoplastic treatment and 13% received radiation therapy only. The median OS post-crizotinib was 61 days; patients with brain metastases had shorter OS than those who did not (44 vs. 69 days, P = 0.018), and patients without further antineoplastic treatment had shorter OS than those who did (17 vs. 180 days, P < 0.001). From claims data, 305 ALK-positive NSCLC patients discontinued crizotinib monotherapy. After discontinuation, 72% had no additional antineoplastic treatment. Among patients who continued antineoplastic treatment, monthly healthcare costs averaged $22,160, driven by pharmacy ($9202), inpatient ($6419), and outpatient radiotherapy ($2888) and imaging ($1179) costs. Among patients who discontinued any antineoplastic treatment, monthly healthcare costs averaged $3423, mostly driven by inpatient costs ($2074). CONCLUSIONS After crizotinib monotherapy, most patients either received radiotherapy only or discontinued antineoplastic treatment altogether. OS after discontinuing crizotinib was poor and shorter among those with brain metastases than without, and among those without subsequent antineoplastic treatment than with. Patients who continued antineoplastic treatment incurred substantial healthcare costs.
Collapse
Affiliation(s)
- A Guérin
- a a Analysis Group Inc. , Boston , MA , United States
| | | | | | | | | | | | | | | | | |
Collapse
|
949
|
Kelleher FC. Similitude and evolution of treatment algorithms. Curr Med Res Opin 2015; 31:1583-5. [PMID: 26086695 DOI: 10.1185/03007995.2015.1062749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Fergal C Kelleher
- a Department of Medical Oncology , St. Vincent's University Hospital , Dublin , Ireland
| |
Collapse
|
950
|
Spira A, Halmos B, Powell CA. Update in Lung Cancer 2014. Am J Respir Crit Care Med 2015; 192:283-94. [PMID: 26230235 PMCID: PMC4584253 DOI: 10.1164/rccm.201504-0756up] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/15/2015] [Indexed: 12/14/2022] Open
Abstract
In the past 2 years, lung cancer research and clinical care have advanced significantly. Advancements in the field have improved outcomes and promise to lead to further reductions in deaths from lung cancer, the leading cause of cancer death worldwide. These advances include identification of new molecular targets for personalized targeted therapy, validation of molecular signatures of lung cancer risk in smokers, progress in lung tumor immunotherapy, and implementation of population-based lung cancer screening with chest computed tomography in the United States. In this review, we highlight recent research in these areas and challenges for the future.
Collapse
Affiliation(s)
- Avrum Spira
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts
| | - Balazs Halmos
- Department of Medicine, Columbia University Medical Center, New York, New York; and
| | - Charles A. Powell
- Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|