51
|
Design, Synthesis, and Evaluation of 2,4-Di(piperidine-substituted-anilino)pyrimidine Derivatives as ALK Inhibitors. B KOREAN CHEM SOC 2015. [DOI: 10.1002/bkcs.10357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
52
|
Wu D, Jin G, Zhao D, Zhang Y, Zhao J, Yu H. [Methodology of Establishing and Identifying NCI-H2228/Crizotinib-resistant Cell Lines In Vitro]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 18:330-9. [PMID: 26104888 PMCID: PMC5999905 DOI: 10.3779/j.issn.1009-3419.2015.06.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
背景与目的 小分子靶向药物发生耐药的机制及寻找克服耐药的手段是目前提高临床疗效需要迫切解决的问题。本研究探讨采用不同方法建立对Crizotinib耐药的非小细胞肺癌NCI-H2228/Crizotinib细胞株的可行性及鉴定分析,为深入研究Crizotinib耐药发生的机制并寻找克服耐药的手段提供实验基础和理论依据。 方法 采用逐步增加药物浓度和化学诱变剂处理NCI-H2228细胞,诱导细胞对Crizotinib耐药。MTT法检测亲本细胞和耐药细胞的50%抑制浓度(50% inhibitory concentration, IC50)和群体倍增时间。RT-PCR和Western blot实验检测棘皮动物微管相关蛋白样4-间变性淋巴瘤激酶(echinoderm microtubule-associated protein like 4-anaplastic lymph kinase, EML4-ALK)基因表达。对耐药细胞和亲本细胞的EML4-ALK基因全长测序并对比分析发生耐药的机制。 结果 逐步增加药物浓度的方法耗时过长,细胞恢复生长缓慢,不能有效诱导NCI-H2228细胞对Crizotinib耐药;化学诱变剂ENU可以在短时间内诱导NCI-H2228细胞对Crizotinib耐药[IC50=(3.810±1.100)μmol/L,P=0.002, 9,vs亲本细胞]。耐药细胞EML4-ALK基因发生点突变的频率高于亲本细胞。 结论 化学诱变剂诱导细胞耐药操作简便,可有效缩短实验流程,为深入研究耐药发生机制,寻找克服靶向药物耐药的手段提供了前期技术方法和实验依据。
Collapse
Affiliation(s)
- Di Wu
- Tumor Center, No.1 Hospital of Jilin University, Changchun 130012, China
| | - Guihua Jin
- School of Life Sciences, Northeast Normal University, Changchun 130024, China
| | - Dawei Zhao
- Department of Breast Tumor, Jilin Province Tumor Hospital, Changchun 130012, China
| | - Yue Zhang
- School of Life Sciences, Northeast Normal University, Changchun 130024, China
| | - Jing Zhao
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun 130062, China
| | - Hong Yu
- Cell Biology Laboratory, Jilin Province Tumor Institute, Changchun 130012, China
| |
Collapse
|
53
|
Crescenzo R, Inghirami G. Anaplastic lymphoma kinase inhibitors. Curr Opin Pharmacol 2015; 23:39-44. [PMID: 26051994 DOI: 10.1016/j.coph.2015.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 05/10/2015] [Accepted: 05/15/2015] [Indexed: 11/30/2022]
Abstract
The anaplastic lymphoma kinase (ALK) gene is a member of the insulin receptor superfamily and it has been associated with more than twenty distinct chimera, including established drivers of several human cancers. Multiple clinical trials have proven that the pharmacological inhibition of ALK signaling leads to remarkable clinical improvement and improves the quality of life of ALK+ cancer patients. Crizotinib was the first ALKi to achieve approval from the Food and Drug Administration, although additional compounds are now moving into diversified clinical trials.
Collapse
Affiliation(s)
- Ramona Crescenzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10021, USA; Department of Molecular Biotechnology and Health Science, Center for Experimental Research and Medical Studies (CeRMS), University of Torino, Torino 10126, Italy
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10021, USA; Department of Molecular Biotechnology and Health Science, Center for Experimental Research and Medical Studies (CeRMS), University of Torino, Torino 10126, Italy; Department of Pathology, NYU Cancer Center, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
54
|
Amin AD, Rajan SS, Liang WS, Pongtornpipat P, Groysman MJ, Tapia EO, Peters TL, Cuyugan L, Adkins J, Rimsza LM, Lussier YA, Puvvada SD, Schatz JH. Evidence Suggesting That Discontinuous Dosing of ALK Kinase Inhibitors May Prolong Control of ALK+ Tumors. Cancer Res 2015; 75:2916-27. [PMID: 26018086 DOI: 10.1158/0008-5472.can-14-3437] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 05/01/2015] [Indexed: 01/30/2023]
Abstract
The anaplastic lymphoma kinase (ALK) is chromosomally rearranged in a subset of certain cancers, including 2% to 7% of non-small cell lung cancers (NSCLC) and ∼70% of anaplastic large cell lymphomas (ALCL). The ALK kinase inhibitors crizotinib and ceritinib are approved for relapsed ALK(+) NSCLC, but acquired resistance to these drugs limits median progression-free survival on average to ∼10 months. Kinase domain mutations are detectable in 25% to 37% of resistant NSCLC samples, with activation of bypass signaling pathways detected frequently with or without concurrent ALK mutations. Here we report that, in contrast to NSCLC cells, drug-resistant ALCL cells show no evidence of bypassing ALK by activating alternate signaling pathways. Instead, drug resistance selected in this setting reflects upregulation of ALK itself. Notably, in the absence of crizotinib or ceritinib, we found that increased ALK signaling rapidly arrested or killed cells, allowing a prolonged control of drug-resistant tumors in vivo with the administration of discontinuous rather than continuous regimens of drug dosing. Furthermore, even when drug resistance mutations were detected in the kinase domain, overexpression of the mutant ALK was toxic to tumor cells. We confirmed these findings derived from human ALCL cells in murine pro-B cells that were transformed to cytokine independence by ectopic expression of an activated NPM-ALK fusion oncoprotein. In summary, our results show how ALK activation functions as a double-edged sword for tumor cell viability, with potential therapeutic implications.
Collapse
Affiliation(s)
| | - Soumya S Rajan
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona
| | - Winnie S Liang
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona. Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona
| | | | - Matthew J Groysman
- Undergraduate Biology Research Program, University of Arizona, Tucson, Arizona
| | - Edgar O Tapia
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona
| | - Tara L Peters
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona
| | - Lori Cuyugan
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona. Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Jonathan Adkins
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona. Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Lisa M Rimsza
- Department of Pathology, University of Arizona, Tucson, Arizona
| | - Yves A Lussier
- BIO5 Institute, University of Arizona, Tucson, Arizona. Department of Medicine, University of Arizona, Tucson, Arizona. Statistics Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona
| | - Soham D Puvvada
- Department of Medicine, University of Arizona, Tucson, Arizona
| | - Jonathan H Schatz
- BIO5 Institute, University of Arizona, Tucson, Arizona. Department of Medicine, University of Arizona, Tucson, Arizona. Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona.
| |
Collapse
|
55
|
Lovisa F, Cozza G, Cristiani A, Cuzzolin A, Albiero A, Mussolin L, Pillon M, Moro S, Basso G, Rosolen A, Bonvini P. ALK kinase domain mutations in primary anaplastic large cell lymphoma: consequences on NPM-ALK activity and sensitivity to tyrosine kinase inhibitors. PLoS One 2015; 10:e0121378. [PMID: 25874976 PMCID: PMC4395299 DOI: 10.1371/journal.pone.0121378] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 01/31/2015] [Indexed: 12/18/2022] Open
Abstract
ALK inhibitor crizotinib has shown potent antitumor activity in children with refractory Anaplastic Large Cell Lymphoma (ALCL) and the opportunity to include ALK inhibitors in first-line therapies is oncoming. However, recent studies suggest that crizotinib-resistance mutations may emerge in ALCL patients. In the present study, we analyzed ALK kinase domain mutational status of 36 paediatric ALCL patients at diagnosis to identify point mutations and gene aberrations that could impact on NPM-ALK gene expression, activity and sensitivity to small-molecule inhibitors. Amplicon ultra-deep sequencing of ALK kinase domain detected 2 single point mutations, R335Q and R291Q, in 2 cases, 2 common deletions of exon 23 and 25 in all the patients, and 7 splicing-related INDELs in a variable number of them. The functional impact of missense mutations and INDELs was evaluated. Point mutations were shown to affect protein kinase activity, signalling output and drug sensitivity. INDELs, instead, generated kinase-dead variants with dominant negative effect on NPM-ALK kinase, in virtue of their capacity of forming non-functional heterocomplexes. Consistently, when co-expressed, INDELs increased crizotinib inhibitory activity on NPM-ALK signal processing, as demonstrated by the significant reduction of STAT3 phosphorylation. Functional changes in ALK kinase activity induced by both point mutations and structural rearrangements were resolved by molecular modelling and dynamic simulation analysis, providing novel insights into ALK kinase domain folding and regulation. Therefore, these data suggest that NPM-ALK pre-therapeutic mutations may be found at low frequency in ALCL patients. These mutations occur randomly within the ALK kinase domain and affect protein activity, while preserving responsiveness to crizotinib.
Collapse
Affiliation(s)
- Federica Lovisa
- Clinica di Oncoematologia Pediatrica di Padova, Azienda Ospedaliera-Università di Padova, Padua, Italy
| | - Giorgio Cozza
- Dipartimento di Scienze Biomediche, Università di Padova, Padua, Italy
| | - Andrea Cristiani
- Dipartimento di Scienze del Farmaco, Università di Padova, Padua, Italy
| | - Alberto Cuzzolin
- Dipartimento di Scienze del Farmaco, Università di Padova, Padua, Italy
| | | | - Lara Mussolin
- Clinica di Oncoematologia Pediatrica di Padova, Azienda Ospedaliera-Università di Padova, Padua, Italy; Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Marta Pillon
- Clinica di Oncoematologia Pediatrica di Padova, Azienda Ospedaliera-Università di Padova, Padua, Italy
| | - Stefano Moro
- Dipartimento di Scienze del Farmaco, Università di Padova, Padua, Italy
| | - Giuseppe Basso
- Clinica di Oncoematologia Pediatrica di Padova, Azienda Ospedaliera-Università di Padova, Padua, Italy
| | - Angelo Rosolen
- Clinica di Oncoematologia Pediatrica di Padova, Azienda Ospedaliera-Università di Padova, Padua, Italy
| | - Paolo Bonvini
- Clinica di Oncoematologia Pediatrica di Padova, Azienda Ospedaliera-Università di Padova, Padua, Italy; Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| |
Collapse
|
56
|
Popper HH, Ryska A, Tímár J, Olszewski W. Molecular testing in lung cancer in the era of precision medicine. Transl Lung Cancer Res 2015; 3:291-300. [PMID: 25806314 DOI: 10.3978/j.issn.2218-6751.2014.10.01] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 10/08/2014] [Indexed: 12/16/2022]
Abstract
The clinical expectations how pathologists should submit lung cancer diagnosis have changed dramatically. Until mid 90-ties a clear separation between small cell lung carcinoma (SCLC) and non-small cell lung carcinoma (NSCLC) was mostly sufficient. With the invention of antiangiogenic treatment a differentiation between squamous and non-squamous NSCLC was requested. When epidermal growth factor receptor (EGFR) mutation was detected in patients with pulmonary adenocarcinomas and subsequent specific treatment with tyrosine kinase inhibitors (TKIs) was invented, sub-classification of NSCLC and molecular analysis of the tumor tissue for mutations was asked for. Pathologists no longer submit just a diagnosis, but instead are involved in a multidisciplinary team for lung cancer patient management. After EGFR several other driver genes such as echinoderm microtubule associated protein like 4-AL-Kinase 1 (EML4-ALK1), c-ros oncogene 1, receptor tyrosine kinase (ROS1), discoidin domain receptor tyrosine kinase 2 (DDR2), fibroblast growth factor receptor 1 (FGFR1) were discovered, and more to come. Due to new developments in bronchology (EUS, EBUS) the amount of tissue submitted for diagnosis and molecular analysis is decreasing, however, the genes to be analyzed are increasing. Many of these driver gene aberrations are inversions or translocations and thus require FISH analysis. Each of these analyses requires a certain amount of tumor cells or one to two tissue sections from an already limited amount of tissues or cells. In this respect new genetic test systems have been introduced such as next generation sequencing, which enables not only to detect multiple mutations in different genes, but also amplifications and fusion genes. As soon as these methods have been validated for routine molecular analysis this will enable the analysis of multiple genetic changes simultaneously. In this review we will focus on genetic aberrations in NSCLC, resistance to new target therapies, and also to methodological requirements for a meaningful evaluation of lung cancer tissue and cells.
Collapse
Affiliation(s)
- Helmut H Popper
- 1 Research Unit for Molecular Lung & Pleura Pathology, Institute of Pathology, Medical University of Graz, Austria ; 2 The Fingerland Department of Pathology, Charles University Faculty of Medicine and Faculty Hospital in Hradec Kralove, Czech Republic ; 3 Department of Pathology, Semmelweis University, Budapest, Molecular Oncology Research Group, HAS-SU, Budapest, Hungary ; 4 Department of Pathology, Cancer Center, 5 Roentgen Str. 02-781 Warsaw, Poland
| | - Ales Ryska
- 1 Research Unit for Molecular Lung & Pleura Pathology, Institute of Pathology, Medical University of Graz, Austria ; 2 The Fingerland Department of Pathology, Charles University Faculty of Medicine and Faculty Hospital in Hradec Kralove, Czech Republic ; 3 Department of Pathology, Semmelweis University, Budapest, Molecular Oncology Research Group, HAS-SU, Budapest, Hungary ; 4 Department of Pathology, Cancer Center, 5 Roentgen Str. 02-781 Warsaw, Poland
| | - József Tímár
- 1 Research Unit for Molecular Lung & Pleura Pathology, Institute of Pathology, Medical University of Graz, Austria ; 2 The Fingerland Department of Pathology, Charles University Faculty of Medicine and Faculty Hospital in Hradec Kralove, Czech Republic ; 3 Department of Pathology, Semmelweis University, Budapest, Molecular Oncology Research Group, HAS-SU, Budapest, Hungary ; 4 Department of Pathology, Cancer Center, 5 Roentgen Str. 02-781 Warsaw, Poland
| | - Wlodzimierz Olszewski
- 1 Research Unit for Molecular Lung & Pleura Pathology, Institute of Pathology, Medical University of Graz, Austria ; 2 The Fingerland Department of Pathology, Charles University Faculty of Medicine and Faculty Hospital in Hradec Kralove, Czech Republic ; 3 Department of Pathology, Semmelweis University, Budapest, Molecular Oncology Research Group, HAS-SU, Budapest, Hungary ; 4 Department of Pathology, Cancer Center, 5 Roentgen Str. 02-781 Warsaw, Poland
| |
Collapse
|
57
|
Morán T, Quiroga V, Gil MDLL, Vilà L, Pardo N, Carcereny E, Capdevila L, Muñoz-Mármol AM, Rosell R. Targeting EML4-ALK driven non-small cell lung cancer (NSCLC). Transl Lung Cancer Res 2015; 2:128-41. [PMID: 25806224 DOI: 10.3978/j.issn.2218-6751.2013.03.04] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 03/07/2013] [Indexed: 01/18/2023]
Affiliation(s)
- Teresa Morán
- Medical Oncology Department, Hospital Universitari Germans Trias i Pujol, Institut Català d'Oncologia-Badalona, Universitat Autònoma de Barcelona, Carretera de Canyet s/n, 08916 Badalona, Barcelona, Spain
| | - Vanesa Quiroga
- Medical Oncology Department, Hospital Universitari Germans Trias i Pujol, Institut Català d'Oncologia-Badalona, Universitat Autònoma de Barcelona, Carretera de Canyet s/n, 08916 Badalona, Barcelona, Spain
| | - María de Los Llanos Gil
- Medical Oncology Department, Hospital Universitari Germans Trias i Pujol, Institut Català d'Oncologia-Badalona, Universitat Autònoma de Barcelona, Carretera de Canyet s/n, 08916 Badalona, Barcelona, Spain
| | - Laia Vilà
- Medical Oncology Department, Hospital Universitari Germans Trias i Pujol, Institut Català d'Oncologia-Badalona, Universitat Autònoma de Barcelona, Carretera de Canyet s/n, 08916 Badalona, Barcelona, Spain
| | - Nuria Pardo
- Medical Oncology Department, Hospital Universitari Germans Trias i Pujol, Institut Català d'Oncologia-Badalona, Universitat Autònoma de Barcelona, Carretera de Canyet s/n, 08916 Badalona, Barcelona, Spain
| | - Enric Carcereny
- Medical Oncology Department, Hospital Universitari Germans Trias i Pujol, Institut Català d'Oncologia-Badalona, Universitat Autònoma de Barcelona, Carretera de Canyet s/n, 08916 Badalona, Barcelona, Spain
| | - Laia Capdevila
- Medical Oncology Department, Hospital Universitari Germans Trias i Pujol, Institut Català d'Oncologia-Badalona, Universitat Autònoma de Barcelona, Carretera de Canyet s/n, 08916 Badalona, Barcelona, Spain
| | - Ana M Muñoz-Mármol
- Medical Oncology Department, Hospital Universitari Germans Trias i Pujol, Institut Català d'Oncologia-Badalona, Universitat Autònoma de Barcelona, Carretera de Canyet s/n, 08916 Badalona, Barcelona, Spain
| | - Rafael Rosell
- Medical Oncology Department, Hospital Universitari Germans Trias i Pujol, Institut Català d'Oncologia-Badalona, Universitat Autònoma de Barcelona, Carretera de Canyet s/n, 08916 Badalona, Barcelona, Spain
| |
Collapse
|
58
|
Wang P, Cai J, Chen J, Ji M. Synthesis and anticancer activities of ceritinib analogs modified in the terminal piperidine ring. Eur J Med Chem 2015; 93:1-8. [DOI: 10.1016/j.ejmech.2015.01.056] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 01/26/2015] [Accepted: 01/27/2015] [Indexed: 10/24/2022]
|
59
|
Song A, Kim TM, Kim DW, Kim S, Keam B, Lee SH, Heo DS. Molecular Changes Associated with Acquired Resistance to Crizotinib in ROS1-Rearranged Non-Small Cell Lung Cancer. Clin Cancer Res 2015; 21:2379-87. [PMID: 25688157 DOI: 10.1158/1078-0432.ccr-14-1350] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 01/28/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Although ROS1-rearranged non-small cell lung cancer (NSCLC) is sensitive to crizotinib, development of resistance is inevitable. Here, we identified molecular alterations in crizotinib-resistant tumors from two NSCLC patients with the CD74-ROS1 rearrangement, and in HCC78 cells harboring SLC34A2-ROS1 that showed resistance to crizotinib (HCC78CR cells). EXPERIMENTAL DESIGN ROS1 kinase domain mutations were examined in fresh tumor tissues from two NSCLC patients and HCC78CR1-3 cells by direct sequencing. Ba/F3 cells expressing ROS1 secondary mutations were constructed to evaluate resistance to crizotinib. An upregulated pathway was identified using phospho-receptor tyrosine kinase array, EGFR signaling antibody array, and RNA sequencing (RNA-seq). Cell proliferation and ROS1 downstream signaling pathways were compared between HCC78 and HCC78CR1-3 cells. RESULTS The ROS1 G2032R mutation was identified in crizotinib-resistant tumors from one patient. Furthermore, HCC78CR1 and CR2 cells harbored a novel ROS1 L2155S mutation (73.3% and 76.2%, respectively). ROS1 G2032R and L2155S mutations conferred resistance to crizotinib in Ba/F3 cells. Evidence of epithelial-to-mesenchymal transition with downregulated E-cadherin and upregulated vimentin was observed in HCC78CR1-2 cells and in the other patient. RNA-seq and EGFR signaling antibody array revealed that the EGFR pathway was significantly upregulated in HCC78CR3 versus HCC78 cells. Cells with the ROS1 mutation and upregulated EGFR were sensitive to foretinib, an inhibitor of c-MET, VEGFR2, and ROS1 and irreversible EGFR tyrosine kinase inhibitors plus crizotinib, respectively. CONCLUSIONS Molecular changes associated with acquired crizotinib resistance in ROS1-rearranged NSCLC are heterogeneous, including ROS1 tyrosine kinase mutations, EGFR activation, and epithelial-to-mesenchymal transition.
Collapse
Affiliation(s)
- Ahnah Song
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Tae Min Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea. Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong-Wan Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea. Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Soyeon Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Bhumsuk Keam
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea. Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea. Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dae Seog Heo
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea. Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
60
|
Ou SHI, Greenbowe J, Khan ZU, Azada MC, Ross JS, Stevens PJ, Ali SM, Miller VA, Gitlitz B. I1171 missense mutation (particularly I1171N) is a common resistance mutation in ALK-positive NSCLC patients who have progressive disease while on alectinib and is sensitive to ceritinib. Lung Cancer 2015; 88:231-4. [PMID: 25736571 DOI: 10.1016/j.lungcan.2015.02.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 02/04/2015] [Accepted: 02/04/2015] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Acquired resistance mutations to anaplastic lymphoma kinase (ALK) inhibitors such as crizotinib and alectinib have been documented in non-small cell lung cancer (NSCLC) patients harboring ALK rearrangement (ALK+). Of note I1171T/N/S mutations in the ALK kinase domain have recently been described by several groups to confer resistance to alectinib, a second-generation ALK inhibitor. Additionally one of these reports demonstrated one ALK+ NSCLC patient harboring an I1171T acquired mutation has responded to ceritinib, another second-generation ALK inhibitor. MATERIALS AND METHODS We reported the presence of an ALK I1171N resistance mutation from comprehensive genomic profiling from a liver biopsy of a progressing metastatic lesion in an ALK+ patient on alectinib after an initial partial response. The patient then responded to ceritinib 750 mg orally once daily but required dose reduction to 600 mg once daily. She initially had grade 3 elevation of liver enzymes from crizotinib necessitating the original switch to alectinib but experienced no transaminase elevations with alectinib or ceritinib. CONCLUSIONS This is the fifth patient case to date demonstrating that ALK I1171 mutation confers resistance to alectinib and the second reported case of ALK I1171 mutation being sensitivity to ceritinib. Substitutions of isoleucine at amino acid 1171 in the ALK kinase domain may distinguish which second generation ALK inhibitor will be effective after crizotinib failure. This case also provides evidence that transaminase elevations is likely a unique adverse event associated with crizotinib and unlikely a "class" effect involving all ALK inhibitors.
Collapse
Affiliation(s)
- Sai-Hong Ignatius Ou
- Department of Medicine, Division of Hematology-Oncology, University of California Irvine School of Medicine, Orange, CA 92868, United States; Chao Family Comprehensive Cancer Center, University of California Irvine Medical Center, Orange, CA 92868, United States.
| | - Joel Greenbowe
- Foundation Medicine Inc., 150 Second Street, Cambridge, MA 02141, United States
| | - Ziad U Khan
- Department of Medicine, Division of Hematology-Oncology, University of California Irvine School of Medicine, Orange, CA 92868, United States; Chao Family Comprehensive Cancer Center, University of California Irvine Medical Center, Orange, CA 92868, United States
| | - Michele C Azada
- Department of Medicine, Division of Hematology-Oncology, University of California Irvine School of Medicine, Orange, CA 92868, United States; Chao Family Comprehensive Cancer Center, University of California Irvine Medical Center, Orange, CA 92868, United States
| | - Jeffrey S Ross
- Foundation Medicine Inc., 150 Second Street, Cambridge, MA 02141, United States; Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, NY 12208, United States
| | - Phil J Stevens
- Foundation Medicine Inc., 150 Second Street, Cambridge, MA 02141, United States
| | - Siraj M Ali
- Foundation Medicine Inc., 150 Second Street, Cambridge, MA 02141, United States
| | - Vincent A Miller
- Foundation Medicine Inc., 150 Second Street, Cambridge, MA 02141, United States
| | - Barbara Gitlitz
- USC Norris Comprehensive Cancer Center and Hospital, Department of Medicine, Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, United States
| |
Collapse
|
61
|
Next-generation sequencing reveals a Novel NSCLC ALK F1174V mutation and confirms ALK G1202R mutation confers high-level resistance to alectinib (CH5424802/RO5424802) in ALK-rearranged NSCLC patients who progressed on crizotinib. J Thorac Oncol 2015; 9:549-53. [PMID: 24736079 DOI: 10.1097/jto.0000000000000094] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Acquired secondary mutations in the anaplastic lymphoma kinase (ALK) gene have been identified in ALK-rearranged (ALK+) non-small-cell lung cancer (NSCLC) patients who developed disease progression while on crizotinib treatment. Here, we identified a novel secondary acquired NSCLC ALK F1174V mutation by comprehensive next-generation sequencing in one ALK+ NSCLC patient who progressed on crizotinib after a prolonged partial response to crizotinib. In a second case, we identified a secondary acquired ALK G1202R, which also confers resistance to alectinib (CH5424802/RO5424802), a second-generation ALK inhibitor that can inhibit ALK gatekeeper L1196M mutation in vitro. ALK G1202R is located at the solvent front of the ALK kinase domain and exhibits a high level of resistance to all other ALK inhibitors currently in clinical development in vitro. Comprehensive genomic profiling of resistant tumor is increasingly important in tailoring treatment decisions after disease progression on crizotinib in ALK+ NSCLC given the promise of second-generation ALK inhibitors and other therapeutic strategies.
Collapse
|
62
|
Takeoka K, Okumura A, Maesako Y, Akasaka T, Ohno H. Crizotinib resistance in acute myeloid leukemia with inv(2)(p23q13)/RAN binding protein 2 (RANBP2) anaplastic lymphoma kinase (ALK) fusion and monosomy 7. Cancer Genet 2015; 208:85-90. [PMID: 25766836 DOI: 10.1016/j.cancergen.2015.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 12/30/2014] [Accepted: 01/12/2015] [Indexed: 12/16/2022]
Abstract
This is the first report on the development of a p.G1269A mutation within the kinase domain (KD) of ALK after crizotinib treatment in RANBP2-ALK acute myeloid leukemia (AML). An elderly woman with AML with an inv(2)(p23q13)/RANBP2-ALK and monosomy 7 was treated with crizotinib. After a short-term hematological response and the restoration of normal hematopoiesis, she experienced a relapse of AML. Fluorescence in situ hybridization using the ALK break-apart probe confirmed the inv(2)(p23q13), while G-banded karyotyping revealed the deletion of a segment of the short arm of chromosome 1 [del(1)(p13p22)] after crizotinib therapy. The ALK gene carried a heterozygous mutation at the nucleotide position g.716751G>C within exon 25, causing the p.G1269A amino acid substitution within the ALK-KD. Reverse transcriptase PCR revealed that the mutated ALK allele was selectively transcribed and the mutation occurred in the ALK allele rearranged with RANBP2. As both the del(1)(p13p22) at the cytogenetic level and p.G1269A at the nucleotide level newly appeared after crizotinib treatment, it is likely that they were secondarily acquired alterations involved in crizotinib resistance. Although secondary genetic abnormalities in ALK are most frequently described in non-small cell lung cancers harboring an ALK alteration, this report suggests that an ALK-KD mutation can occur independently of the tumor cell type or fusion partner after crizotinib treatment.
Collapse
MESH Headings
- Aged
- Anaplastic Lymphoma Kinase
- Chromosome Deletion
- Chromosome Inversion
- Chromosomes, Human, Pair 2
- Chromosomes, Human, Pair 7
- Crizotinib
- Drug Resistance, Neoplasm
- Female
- Humans
- In Situ Hybridization, Fluorescence
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Molecular Chaperones/genetics
- Nuclear Pore Complex Proteins/genetics
- Protein Kinase Inhibitors/therapeutic use
- Pyrazoles/therapeutic use
- Pyridines/therapeutic use
- Receptor Protein-Tyrosine Kinases/genetics
- Recombinant Fusion Proteins/genetics
Collapse
Affiliation(s)
- Kayo Takeoka
- Tenri Institute of Medical Research, Tenri Hospital, Nara, Japan
| | - Atsuko Okumura
- Tenri Institute of Medical Research, Tenri Hospital, Nara, Japan
| | | | | | - Hitoshi Ohno
- Tenri Institute of Medical Research, Tenri Hospital, Nara, Japan; Department of Hematology, Tenri Hospital, Nara, Japan.
| |
Collapse
|
63
|
Cappuzzo F, Moro-Sibilot D, Gautschi O, Boleti E, Felip E, Groen HJM, Germonpré P, Meldgaard P, Arriola E, Steele N, Fox J, Schnell P, Engelsberg A, Wolf J. Management of crizotinib therapy for ALK-rearranged non-small cell lung carcinoma: an expert consensus. Lung Cancer 2014; 87:89-95. [PMID: 25576294 DOI: 10.1016/j.lungcan.2014.12.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 12/08/2014] [Accepted: 12/14/2014] [Indexed: 11/30/2022]
Abstract
Within 4 years of the discovery of anaplastic lymphoma kinase (ALK) rearrangements in non-small cell lung cancer (NSCLC), the ALK inhibitor crizotinib gained US and European approval for the treatment of advanced ALK-positive NSCLC. This was due to the striking response data observed with crizotinib in phase I and II trials in patients with ALK-positive NSCLC, as well as the favorable tolerability and safety profile observed. Recently published phase III data established crizotinib as a new standard of care for this NSCLC molecular subset. A consequence of such rapid approval, however, is the limited clinical experience and relative paucity of information concerning optimal therapy management. In this review, we discuss the development of crizotinib and the clinical relevance of its safety profile, examining crizotinib-associated adverse events in detail and making specific management recommendations. Crizotinib-associated adverse events were mostly mild to moderate in severity in clinical studies, and appropriate monitoring and supportive therapies are considered effective in avoiding the need for dose interruption or reduction in most cases. Therapy management of patients following disease progression on crizotinib is also discussed. Based on available clinical data, it is evident that patients may have prolonged benefit from crizotinib after Response Evaluation Criteria in Solid Tumors-defined disease progression, and crizotinib should be continued for as long as the patient derives benefit.
Collapse
Affiliation(s)
| | - Denis Moro-Sibilot
- Hôpital A. Michallon, Centre Hospitalier Universitaire, Grenoble, France
| | - Oliver Gautschi
- Department of Oncology, Luzerner Kantonsspital, Luzern, Switzerland
| | | | - Enriqueta Felip
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Harry J M Groen
- University of Groningen and University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, The Netherlands
| | | | - Peter Meldgaard
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Nicola Steele
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Jesme Fox
- The Roy Castle Lung Cancer Foundation, Liverpool, United Kingdom
| | | | | | - Jürgen Wolf
- Department of Internal Medicine, Centre for Integrated Oncology, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
64
|
Identification of a Novel HIP1-ALK Fusion Variant in Non–Small-Cell Lung Cancer (NSCLC) and Discovery of ALK I1171 (I1171N/S) Mutations in Two ALK-Rearranged NSCLC Patients with Resistance to Alectinib. J Thorac Oncol 2014; 9:1821-5. [DOI: 10.1097/jto.0000000000000368] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
65
|
Bresler SC, Weiser DA, Huwe PJ, Park JH, Krytska K, Ryles H, Laudenslager M, Rappaport EF, Wood AC, McGrady PW, Hogarty MD, London WB, Radhakrishnan R, Lemmon MA, Mossé YP. ALK mutations confer differential oncogenic activation and sensitivity to ALK inhibition therapy in neuroblastoma. Cancer Cell 2014; 26:682-94. [PMID: 25517749 PMCID: PMC4269829 DOI: 10.1016/j.ccell.2014.09.019] [Citation(s) in RCA: 252] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 07/13/2014] [Accepted: 09/25/2014] [Indexed: 02/01/2023]
Abstract
Genetic studies have established anaplastic lymphoma kinase (ALK), a cell surface receptor tyrosine kinase, as a tractable molecular target in neuroblastoma. We describe comprehensive genomic, biochemical, and computational analyses of ALK mutations across 1,596 diagnostic neuroblastoma samples. ALK tyrosine kinase domain mutations occurred in 8% of samples--at three hot spots and 13 minor sites--and correlated significantly with poorer survival in high- and intermediate-risk neuroblastoma. Biochemical and computational studies distinguished oncogenic (constitutively activating) from nononcogenic mutations and allowed robust computational prediction of their effects. The mutated variants also showed differential in vitro crizotinib sensitivities. Our studies identify ALK genomic status as a clinically important therapeutic stratification tool in neuroblastoma and will allow tailoring of ALK-targeted therapy to specific mutations.
Collapse
Affiliation(s)
- Scott C Bresler
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Graduate Group in Biochemistry and Molecular Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Daniel A Weiser
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Peter J Huwe
- Graduate Group in Biochemistry and Molecular Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jin H Park
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Graduate Group in Biochemistry and Molecular Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kateryna Krytska
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Hannah Ryles
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Marci Laudenslager
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eric F Rappaport
- Nucleic Acid Core Facility, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Andrew C Wood
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Patrick W McGrady
- Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL 32611, USA
| | - Michael D Hogarty
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Wendy B London
- Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL 32611, USA; Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Ravi Radhakrishnan
- Graduate Group in Biochemistry and Molecular Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark A Lemmon
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Graduate Group in Biochemistry and Molecular Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Yaël P Mossé
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
66
|
Garner AP, Gozgit JM, Anjum R, Vodala S, Schrock A, Zhou T, Serrano C, Eilers G, Zhu M, Ketzer J, Wardwell S, Ning Y, Song Y, Kohlmann A, Wang F, Clackson T, Heinrich MC, Fletcher JA, Bauer S, Rivera VM. Ponatinib inhibits polyclonal drug-resistant KIT oncoproteins and shows therapeutic potential in heavily pretreated gastrointestinal stromal tumor (GIST) patients. Clin Cancer Res 2014; 20:5745-5755. [PMID: 25239608 DOI: 10.1158/1078-0432.ccr-14-1397] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE KIT is the major oncogenic driver of gastrointestinal stromal tumors (GIST). Imatinib, sunitinib, and regorafenib are approved therapies; however, efficacy is often limited by the acquisition of polyclonal secondary resistance mutations in KIT, with those located in the activation (A) loop (exons 17/18) being particularly problematic. Here, we explore the KIT-inhibitory activity of ponatinib in preclinical models and describe initial characterization of its activity in patients with GIST. EXPERIMENTAL DESIGN The cellular and in vivo activities of ponatinib, imatinib, sunitinib, and regorafenib against mutant KIT were evaluated using an accelerated mutagenesis assay and a panel of engineered and GIST-derived cell lines. The ponatinib-KIT costructure was also determined. The clinical activity of ponatinib was examined in three patients with GIST previously treated with all three FDA-approved agents. RESULTS In engineered and GIST-derived cell lines, ponatinib potently inhibited KIT exon 11 primary mutants and a range of secondary mutants, including those within the A-loop. Ponatinib also induced regression in engineered and GIST-derived tumor models containing these secondary mutations. In a mutagenesis screen, 40 nmol/L ponatinib was sufficient to suppress outgrowth of all secondary mutants except V654A, which was suppressed at 80 nmol/L. This inhibitory profile could be rationalized on the basis of structural analyses. Ponatinib (30 mg daily) displayed encouraging clinical activity in two of three patients with GIST. CONCLUSION Ponatinib possesses potent activity against most major clinically relevant KIT mutants and has demonstrated preliminary evidence of activity in patients with refractory GIST. These data strongly support further evaluation of ponatinib in patients with GIST.
Collapse
Affiliation(s)
| | | | - Rana Anjum
- ARIAD Pharmaceuticals, Inc, Cambridge, MA
| | | | | | | | - Cesar Serrano
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Grant Eilers
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Meijun Zhu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Julia Ketzer
- Sarcoma Center, Dept. of Medical Oncology, West German Cancer Center, University of Duisburg-Essen, Essen, Germany
| | | | - Yaoyu Ning
- ARIAD Pharmaceuticals, Inc, Cambridge, MA
| | | | | | - Frank Wang
- ARIAD Pharmaceuticals, Inc, Cambridge, MA
| | | | - Michael C Heinrich
- Portland VA Medical Center and OHSU Knight Cancer Institute, Portland, Oregon
| | - Jonathan A Fletcher
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sebastian Bauer
- Sarcoma Center, Dept. of Medical Oncology, West German Cancer Center, University of Duisburg-Essen, Essen, Germany
| | | |
Collapse
|
67
|
Katayama R, Friboulet L, Koike S, Lockerman EL, Khan TM, Gainor JF, Iafrate AJ, Takeuchi K, Taiji M, Okuno Y, Fujita N, Engelman JA, Shaw AT. Two novel ALK mutations mediate acquired resistance to the next-generation ALK inhibitor alectinib. Clin Cancer Res 2014; 20:5686-96. [PMID: 25228534 DOI: 10.1158/1078-0432.ccr-14-1511] [Citation(s) in RCA: 229] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
PURPOSE The first-generation ALK tyrosine kinase inhibitor (TKI) crizotinib is a standard therapy for patients with ALK-rearranged non-small cell lung cancer (NSCLC). Several next-generation ALK-TKIs have entered the clinic and have shown promising activity in crizotinib-resistant patients. As patients still relapse even on these next-generation ALK-TKIs, we examined mechanisms of resistance to the next-generation ALK-TKI alectinib and potential strategies to overcome this resistance. EXPERIMENTAL DESIGN We established a cell line model of alectinib resistance, and analyzed a resistant tumor specimen from a patient who had relapsed on alectinib. We developed Ba/F3 models harboring alectinib-resistant ALK mutations and evaluated the potency of other next-generation ALK-TKIs in these models. We tested the antitumor activity of the next-generation ALK-TKI ceritinib in the patient with acquired resistance to alectinib. To elucidate structure-activity relationships of ALK mutations, we performed computational thermodynamic simulation with MP-CAFEE. RESULTS We identified a novel V1180L gatekeeper mutation from the cell line model and a second novel I1171T mutation from the patient who developed resistance to alectinib. Both ALK mutations conferred resistance to alectinib as well as to crizotinib, but were sensitive to ceritinib and other next-generation ALK-TKIs. Treatment of the patient with ceritinib led to a marked response. Thermodynamics simulation suggests that both mutations lead to distinct structural alterations that decrease the binding affinity with alectinib. CONCLUSIONS We have identified two novel ALK mutations arising after alectinib exposure that are sensitive to other next-generation ALK-TKIs. The ability of ceritinib to overcome alectinib-resistance mutations suggests a potential role for sequential therapy with multiple next-generation ALK-TKIs.
Collapse
Affiliation(s)
- Ryohei Katayama
- Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts. Department of Medicine, Harvard Medical School, Boston, Massachusetts. Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Luc Friboulet
- Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts. Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Sumie Koike
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Elizabeth L Lockerman
- Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts. Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Tahsin M Khan
- Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts
| | - Justin F Gainor
- Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts. Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - A John Iafrate
- Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts. Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Kengo Takeuchi
- Pathology Project for Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makoto Taiji
- Processor Research Team, RIKEN Advanced Institute of Computational Sciences 6F, Kobe, Hyogo, Japan
| | - Yasushi Okuno
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naoya Fujita
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Jeffrey A Engelman
- Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts. Department of Medicine, Harvard Medical School, Boston, Massachusetts.
| | - Alice T Shaw
- Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts. Department of Medicine, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
68
|
Politi K, Gettinger S. Perfect ALKemy: optimizing the use of ALK-directed therapies in lung cancer. Clin Cancer Res 2014; 20:5576-8. [PMID: 25228532 DOI: 10.1158/1078-0432.ccr-14-2306] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mutations in ALK are a common mechanism of acquired resistance to small molecule ALK inhibitors in ALK-rearranged lung cancer. Different mutants exhibit differential sensitivity to ALK inhibitors. Matching the mutational profile of the tumor with the appropriate ALK inhibitor is likely to be important to maximize benefit for patients.
Collapse
Affiliation(s)
- Katerina Politi
- Departments of Pathology and Medicine (Section of Medical Oncology), Yale University School of Medicine and Yale Cancer Center, New Haven, Connecticut.
| | - Scott Gettinger
- Medicine (Section of Medical Oncology), Yale University School of Medicine and Yale Cancer Center, New Haven, Connecticut.
| |
Collapse
|
69
|
Alifrangis CC, McDermott U. Reading between the lines; understanding drug response in the post genomic era. Mol Oncol 2014; 8:1112-9. [PMID: 24957465 PMCID: PMC5528621 DOI: 10.1016/j.molonc.2014.05.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 05/27/2014] [Indexed: 12/22/2022] Open
Abstract
Following the fanfare of initial, often dramatic, success with small molecule inhibitors in the treatment of defined genomic subgroups, it can be argued that the extension of targeted therapeutics to the majority of patients with solid cancers has stalled. Despite encouraging FDA approval rates, the attrition rates of these compounds remains high in early stage clinical studies, with single agent studies repeatedly showing poor efficacy In striking contrast, our understanding of the complexity of solid neoplasms has increased in huge increments, following the publication of large-scale genomic and transcriptomic datasets from large collaborations such as the International Cancer Genome Consortium (ICGC http://www.icgc.org/) and The Cancer Genome Atlas (TCGA http://cancergenome.nih.gov/). However, there remains a clear disconnect between these rich datasets describing the genomic complexity of cancer, including both intra- and inter-tumour heterogeneity, and what a treating oncologist can consider to be a clinically "actionable" mutation profile. Our understanding of these data is in its infancy and we still find difficulties ascribing characteristics to tumours that consistently predict therapeutic response for the majority of small molecule inhibitors. This article will seek to explore the recent studies of the patterns and impact of mutations in drug resistance, and demonstrate how we may use this data to reshape our thinking about biological pathways, critical dependencies and their therapeutic interruption.
Collapse
Affiliation(s)
- Constantine C Alifrangis
- Cambridge Institute of Medical Research, University of Cambridge, Cambridge, UK; Cancer Genome Project, Wellcome Trust Sanger Institute, Cambridge, UK; Dept of Medical Oncology, Charing Cross Hospital, London, UK.
| | - Ultan McDermott
- Cancer Genome Project, Wellcome Trust Sanger Institute, Cambridge, UK
| |
Collapse
|
70
|
Toyokawa G, Seto T. ALK Inhibitors: What Is the Best Way to Treat Patients With ALK+ Non–Small-Cell Lung Cancer? Clin Lung Cancer 2014; 15:313-9. [DOI: 10.1016/j.cllc.2014.05.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 04/09/2014] [Accepted: 05/19/2014] [Indexed: 01/30/2023]
|
71
|
Toyokawa G, Seto T. Anaplastic lymphoma kinase rearrangement in lung cancer: its biological and clinical significance. Respir Investig 2014; 52:330-8. [PMID: 25453376 DOI: 10.1016/j.resinv.2014.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 06/04/2014] [Accepted: 06/25/2014] [Indexed: 12/30/2022]
Abstract
Anaplastic lymphoma kinase (ALK) has been found to fuse with other partners, such as echinoderm microtubule-associated protein-like 4 (EML4), leading to potent malignant transformation in lung cancer, specifically non-small-cell lung cancer (NSCLC). The frequency of the ALK rearrangement in patients with NSCLC is reported to be 4-7%, and the rearrangement is frequently observed in relatively younger patients, non- or light smokers and those with adenocarcinoma histology without other genetic disorders, such as mutations of the epidermal growth factor receptor gene. Crizotinib, which is a first-in-class ALK tyrosine kinase inhibitor (TKI), was shown to be effective and well tolerated in ALK-positive NSCLC patients by a single-arm phase I study. Furthermore, a phase III randomized study demonstrated the superiority of crizotinib to standard chemotherapy (pemetrexed or docetaxel) in the treatment of NSCLC patients harboring the ALK rearrangement who had received one prior platinum-based chemotherapy. However, the mechanisms of resistance to crizotinib are major concerns when administering crizotinib to ALK-positive NSCLC patients, and they include second mutations and a gain in the copy number of the ALK gene, activation of other oncogenes, etc. Treatment strategies to overcome these mechanisms of resistance have been developed, including the use of second-generation ALK inhibitors, such as alectinib and ceritinib, heat shock protein 90 inhibitors and so on. In this article, we review the pre-clinical and clinical data regarding the biologal and clinical significance of the ALK rearrangement in lung cancer.
Collapse
Affiliation(s)
- Gouji Toyokawa
- Department of Thoracic Oncology, National Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka 811-1395, Japan
| | - Takashi Seto
- Department of Thoracic Oncology, National Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka 811-1395, Japan.
| |
Collapse
|
72
|
Spaans JN, Goss GD. Drug resistance to molecular targeted therapy and its consequences for treatment decisions in non-small-cell lung cancer. Front Oncol 2014; 4:190. [PMID: 25101246 PMCID: PMC4107955 DOI: 10.3389/fonc.2014.00190] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/07/2014] [Indexed: 01/10/2023] Open
Abstract
Our ability to detect and directly target the oncogenic alterations responsible for tumor proliferation has contributed significantly to the management of lung cancer in the last decade. The therapeutic efficacy of molecularly targeted therapy is, however, mainly limited to patients harboring certain genetic mutations and is generally short-lived. Herein, we review primary and secondary drug resistance using the most well-studied of the molecularly targeted agents, the tyrosine kinase inhibitors targeting the epidermal growth factor (EGF) receptor, and the anaplastic lymphoma kinase (ALK) rearrangement, the current limitations of targeted therapies and their consequences on the management of patients with lung cancer.
Collapse
Affiliation(s)
| | - Glenwood D Goss
- Ottawa Hospital Research Institute , Ottawa, ON , Canada ; Ottawa Hospital Cancer Centre , Ottawa, ON , Canada ; Department of Medicine, University of Ottawa , Ottawa, ON , Canada
| |
Collapse
|
73
|
Esfahani K, Agulnik JS, Cohen V. A Systemic Review of Resistance Mechanisms and Ongoing Clinical Trials in ALK-Rearranged Non-Small Cell Lung Cancer. Front Oncol 2014; 4:174. [PMID: 25101240 PMCID: PMC4104550 DOI: 10.3389/fonc.2014.00174] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/19/2014] [Indexed: 12/17/2022] Open
Abstract
The identification of oncogenic driver mutations in non-small cell lung cancer (NSCLC) has led to a paradigm shift and the development of specific molecular treatments. Tumors harboring a rearranged EML4–ALK fusion oncogene are highly sensitive to therapy with ALK-targeted inhibitors. Crizotinib is the first approved treatment for advanced lung tumors containing this genetic abnormality. In this mini review, we discuss the existing data on crizotinib as well as ongoing trials involving this medication. A brief overview of the known resistance mechanisms to crizotinib will also be presented followed by a summary of the ongoing trials involving next-generation ALK-inhibitors or other targeted therapies in patients with ALK+ NSCLC.
Collapse
Affiliation(s)
- Khashayar Esfahani
- Department of Oncology, Segal Cancer Center, Sir Mortimer B. Davis Jewish General Hospital , Montreal, QC , Canada
| | - Jason Scott Agulnik
- Division of Pulmonary Diseases, Department of Oncology, Peter Brojde Cancer Center , Montreal, QC , Canada
| | - Victor Cohen
- Department of Oncology, Segal Cancer Center, Sir Mortimer B. Davis Jewish General Hospital , Montreal, QC , Canada
| |
Collapse
|
74
|
Abstract
Recent advances in the understanding of the complex biology of non-small cell lung carcinoma (NSCLC), particularly activation of oncogenes by mutation, translocation and amplification, have provided new treatment targets for this disease, and allowed the identification of subsets of NSCLC tumors, mostly with adenocarcinoma histology, having unique molecular profiles that can predict response to targeted therapy. The identification of specific genetic and molecular targetable abnormalities using tumor tissue and cytology specimens followed by the administration of a specific inhibitor to the target, are the basis of personalized lung cancer treatment. In this new paradigm, the role of a precise pathology diagnosis of lung cancer and the proper handling of tissue and cytology samples for molecular testing is becoming increasingly important. These changes have posed multiple new challenges for pathologists to adequately integrate routine histopathology analysis and molecular testing into the clinical pathology practice for tumor diagnosis and subsequent selection of the most appropriate therapy.
Collapse
Affiliation(s)
- Junya Fujimoto
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas; Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
75
|
3D-QSAR and molecular fragment replacement study on diaminopyrimidine and pyrrolotriazine ALK inhibitors. J Mol Struct 2014. [DOI: 10.1016/j.molstruc.2014.03.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
76
|
ALK inhibitors and advanced non-small cell lung cancer (Review). Int J Oncol 2014; 45:499-508. [DOI: 10.3892/ijo.2014.2475] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 05/16/2014] [Indexed: 11/05/2022] Open
|
77
|
Song Z, Yang Y, Liu Z, Peng X, Guo J, Yang X, Wu K, Ai J, Ding J, Geng M, Zhang A. Discovery of novel 2,4-diarylaminopyrimidine analogues (DAAPalogues) showing potent inhibitory activities against both wild-type and mutant ALK kinases. J Med Chem 2014; 58:197-211. [PMID: 24785465 DOI: 10.1021/jm5005144] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We have developed a series of new 2,4-diarylaminopyrimidine analogues (DAAPalogues) bearing a flexible amino acid side chain, different from the majority of the literature reported ALK inhibitors that often possess a structurally constrained arylpiperazine fragment or its equivalents in the solvent-interaction region. Extensive structural elaboration led to compound 15 possessing IC50 values of 2.7 and 15.3 nM, respectively, in the ALK wild-type and gate-keeper mutant L1196M enzymatic assays. This compound not only showed high proliferative inhibition against ALK-addicted cells across different oncogenic forms but also effectively suppressed several ALK secondary mutant cells, including the gate-keeper L1196M and F1174L. Significant antitumor efficacy was achieved in the ALK-driven SUP-M2 xenograft model.
Collapse
Affiliation(s)
- Zilan Song
- CAS Key Laboratory of Receptor Research, Synthetic Organic & Medicinal Chemistry Laboratory, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , Shanghai 201203, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Hayes SA, Hudson AL, Clarke SJ, Molloy MP, Howell VM. From mice to men: GEMMs as trial patients for new NSCLC therapies. Semin Cell Dev Biol 2014; 27:118-27. [PMID: 24718320 DOI: 10.1016/j.semcdb.2014.04.002] [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] [Received: 03/31/2014] [Accepted: 04/01/2014] [Indexed: 01/05/2023]
Abstract
Given the large socio-economic burden of cancer, there is an urgent need for in vivo animal cancer models that can provide a rationale for personalised therapeutic regimens that are translatable to the clinic. Recent developments in establishing mouse models that closely resemble human lung cancers involve the application of genetically engineered mouse models (GEMMs) for use in drug efficacy studies or to guide patient therapy. Here, we review recent applications of GEMMs in non-small cell lung cancer research for drug development and their potential in aiding biomarker discovery and understanding of biological mechanisms behind clinical outcomes and drug interactions.
Collapse
Affiliation(s)
- Sarah A Hayes
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales, Australia; Department of Medical Oncology, Royal North Shore Hospital, University of Sydney, St. Leonards, New South Wales, Australia
| | - Amanda L Hudson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales, Australia; Department of Medical Oncology, Royal North Shore Hospital, University of Sydney, St. Leonards, New South Wales, Australia
| | - Stephen J Clarke
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales, Australia; Department of Medical Oncology, Royal North Shore Hospital, University of Sydney, St. Leonards, New South Wales, Australia
| | - Mark P Molloy
- Australian Proteome Analysis Facility (APAF), Macquarie University, Sydney, Australia; Department of Chemistry & Biomolecular Sciences, Macquarie University, Sydney, Australia
| | - Viive M Howell
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales, Australia; Department of Medical Oncology, Royal North Shore Hospital, University of Sydney, St. Leonards, New South Wales, Australia.
| |
Collapse
|
79
|
Friboulet L, Li N, Katayama R, Lee CC, Gainor JF, Crystal AS, Michellys PY, Awad MM, Yanagitani N, Kim S, Pferdekamper AC, Li J, Kasibhatla S, Sun F, Sun X, Hua S, McNamara P, Mahmood S, Lockerman EL, Fujita N, Nishio M, Harris JL, Shaw AT, Engelman JA. The ALK inhibitor ceritinib overcomes crizotinib resistance in non-small cell lung cancer. Cancer Discov 2014; 4:662-673. [PMID: 24675041 DOI: 10.1158/2159-8290.cd-13-0846] [Citation(s) in RCA: 629] [Impact Index Per Article: 62.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
UNLABELLED Non-small cell lung cancers (NSCLC) harboring anaplastic lymphoma kinase (ALK) gene rearrangements invariably develop resistance to the ALK tyrosine kinase inhibitor (TKI) crizotinib. Herein, we report the first preclinical evaluation of the next-generation ALK TKI, ceritinib (LDK378), in the setting of crizotinib resistance. An interrogation of in vitro and in vivo models of acquired resistance to crizotinib, including cell lines established from biopsies of patients with crizotinib-resistant NSCLC, revealed that ceritinib potently overcomes crizotinib-resistant mutations. In particular, ceritinib effectively inhibits ALK harboring L1196M, G1269A, I1171T, and S1206Y mutations, and a cocrystal structure of ceritinib bound to ALK provides structural bases for this increased potency. However, we observed that ceritinib did not overcome two crizotinib-resistant ALK mutations, G1202R and F1174C, and one of these mutations was identified in 5 of 11 biopsies from patients with acquired resistance to ceritinib. Altogether, our results demonstrate that ceritinib can overcome crizotinib resistance, consistent with clinical data showing marked efficacy of ceritinib in patients with crizotinib-resistant disease. SIGNIFICANCE The second-generation ALK inhibitor ceritinib can overcome several crizotinib-resistant mutations and is potent against several in vitro and in vivo laboratory models of acquired resistance to crizotinib. These findings provide the molecular basis for the marked clinical activity of ceritinib in patients with ALK-positive NSCLC with crizotinib-resistant disease. Cancer Discov; 4(6); 662-73. ©2014 AACR. See related commentary by Ramalingam and Khuri, p. 634 This article is highlighted in the In This Issue feature, p. 621.
Collapse
Affiliation(s)
- Luc Friboulet
- Massachusetts General Hospital Cancer Center, Boston, MA 02129, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Nanxin Li
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121
| | - Ryohei Katayama
- Massachusetts General Hospital Cancer Center, Boston, MA 02129, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.,Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, JAPAN
| | - Christian C Lee
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121
| | - Justin F Gainor
- Massachusetts General Hospital Cancer Center, Boston, MA 02129, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Adam S Crystal
- Massachusetts General Hospital Cancer Center, Boston, MA 02129, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | | | - Mark M Awad
- Massachusetts General Hospital Cancer Center, Boston, MA 02129, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Noriko Yanagitani
- Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, JAPAN
| | - Sungjoon Kim
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121
| | | | - Jie Li
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121
| | | | - Frank Sun
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121
| | - Xiuying Sun
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121
| | - Su Hua
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121
| | - Peter McNamara
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121
| | - Sidra Mahmood
- Massachusetts General Hospital Cancer Center, Boston, MA 02129, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth L Lockerman
- Massachusetts General Hospital Cancer Center, Boston, MA 02129, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Naoya Fujita
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, JAPAN
| | - Makoto Nishio
- Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, JAPAN
| | - Jennifer L Harris
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121
| | - Alice T Shaw
- Massachusetts General Hospital Cancer Center, Boston, MA 02129, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey A Engelman
- Massachusetts General Hospital Cancer Center, Boston, MA 02129, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
80
|
Iwama E, Okamoto I, Harada T, Takayama K, Nakanishi Y. Development of anaplastic lymphoma kinase (ALK) inhibitors and molecular diagnosis in ALK rearrangement-positive lung cancer. Onco Targets Ther 2014; 7:375-85. [PMID: 24623980 PMCID: PMC3949762 DOI: 10.2147/ott.s38868] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The fusion of echinoderm microtubule-associated protein-like 4 with anaplastic lymphoma kinase (ALK) was identified as a transforming gene for lung cancer in 2007. This genetic rearrangement accounts for 2%–5% of non-small-cell lung cancer (NSCLC) cases, occurring predominantly in younger individuals with adenocarcinoma who are never- or light smokers. A small-molecule tyrosine-kinase inhibitor of ALK, crizotinib, was rapidly approved by the US Food and Drug Administration on the basis of its pronounced clinical activity in patients with ALK rearrangement-positive NSCLC. Next-generation ALK inhibitors, such as alectinib, LDK378, and AP26113, are also being developed in ongoing clinical trials. In addition, the improvement and validation of methods for the detection of ALK rearrangement in NSCLC patients will be key to the optimal clinical use of ALK inhibitors. We here summarize recent progress in the development of new ALK inhibitors and in the molecular diagnosis of ALK rearrangement-positive NSCLC.
Collapse
Affiliation(s)
- Eiji Iwama
- Department of Comprehensive Clinical Oncology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan ; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Isamu Okamoto
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka, Japan
| | - Taishi Harada
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Takayama
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoichi Nakanishi
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan ; Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka, Japan
| |
Collapse
|
81
|
Activating mutations in ALK kinase domain confer resistance to structurally unrelated ALK inhibitors in NPM-ALK-positive anaplastic large-cell lymphoma. J Cancer Res Clin Oncol 2014; 140:589-98. [PMID: 24509625 PMCID: PMC3949014 DOI: 10.1007/s00432-014-1589-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/17/2014] [Indexed: 11/18/2022]
Abstract
Purpose Crizotinib, the first FDA-approved ALK inhibitor, showed significant antitumor activity in young patients with anaplastic large-cell lymphoma (ALCL) frequently displaying ALK rearrangement. However, long-term therapeutic benefits of crizotinib are limited due to development of drug resistance. CH5424802—more potent and selective ALK inhibitor—comprises a good candidate for second-line treatment in crizotinib-relapsed patients. The aim of this study was to determine possible mechanisms of resistance to ALK inhibitors that can appear in ALCL patients. Methods ALK+ ALCL cell lines resistant to crizotinib (Karpas299CR) and to CH5424802 (Karpas299CHR) were established by long-term exposure of Karpas299 cells to these inhibitors. Next, alterations in their sensitivity to ALK, HSP90 and mTOR inhibitors were investigated by cell viability and BrdU incorporation assays and immunoblot analysis. Results cDNA sequencing of ALK kinase domain revealed activating mutations—I1171T in Karpas299CR and F1174C in Karpas299CHR. The resistant cells displayed diminished sensitivity to structurally unrelated ALK inhibitors—crizotinib, CH5424802 and TAE684. Nevertheless, CH5424802 and TAE684 were still more potent against the resistant cells than crizotinib. Moreover, Karpas299CR and Karpas299CHR cells remained sensitive to HSP90 or mTOR inhibitors. Conclusions Resistance mediated by activating mutations in ALK kinase domain may emerge in ALCL patients during ALK inhibitors treatment. However, more potent second-generation ALK inhibitors, HSP90 or mTOR inhibitors may represent an effective therapy for relapsed ALK+ ALCL patients. Electronic supplementary material The online version of this article (doi:10.1007/s00432-014-1589-3) contains supplementary material, which is available to authorized users.
Collapse
|
82
|
Huang Q, Johnson TW, Bailey S, Brooun A, Bunker KD, Burke BJ, Collins MR, Cook AS, Cui JJ, Dack KN, Deal JG, Deng YL, Dinh D, Engstrom LD, He M, Hoffman J, Hoffman RL, Johnson PS, Kania RS, Lam H, Lam JL, Le PT, Li Q, Lingardo L, Liu W, Lu MW, McTigue M, Palmer CL, Richardson PF, Sach NW, Shen H, Smeal T, Smith GL, Stewart AE, Timofeevski S, Tsaparikos K, Wang H, Zhu H, Zhu J, Zou HY, Edwards MP. Design of Potent and Selective Inhibitors to Overcome Clinical Anaplastic Lymphoma Kinase Mutations Resistant to Crizotinib. J Med Chem 2014; 57:1170-87. [DOI: 10.1021/jm401805h] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Qinhua Huang
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Ted W. Johnson
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Simon Bailey
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Alexei Brooun
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Kevin D. Bunker
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Benjamin J. Burke
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Michael R. Collins
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Andrew S. Cook
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - J. Jean Cui
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Kevin N. Dack
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Judith G. Deal
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Ya-Li Deng
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Dac Dinh
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Lars D. Engstrom
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Mingying He
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Jacqui Hoffman
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Robert L. Hoffman
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Patrick S. Johnson
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Robert S. Kania
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Hieu Lam
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Justine L. Lam
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Phuong T. Le
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Qiuhua Li
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Laura Lingardo
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Wei Liu
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Melissa West Lu
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Michele McTigue
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Cynthia L. Palmer
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Paul F. Richardson
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Neal W. Sach
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Hong Shen
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Tod Smeal
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Graham L. Smith
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Albert E. Stewart
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Sergei Timofeevski
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Konstantinos Tsaparikos
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Hui Wang
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Huichun Zhu
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Jinjiang Zhu
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Helen Y. Zou
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Martin P. Edwards
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| |
Collapse
|
83
|
Lee HJ, Latif M, Choe H, Ali I, Lee HK, Yang EH, Yun JI, Chae CH, Jung JK, Kim HR, Lee CO, Park CH, Lee K. ALK inhibitors of bis-ortho-alkoxy-para-piperazinesubstituted-pyrimidines and -triazines for cancer treatment. Arch Pharm Res 2014; 37:1130-8. [DOI: 10.1007/s12272-013-0323-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 12/23/2013] [Indexed: 01/15/2023]
|
84
|
The resistance tetrad: amino acid hotspots for kinome-wide exploitation of drug-resistant protein kinase alleles. Methods Enzymol 2014; 548:117-46. [PMID: 25399644 DOI: 10.1016/b978-0-12-397918-6.00005-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Acquired resistance to targeted kinase inhibitors is a well-documented clinical problem that is potentially fatal for patients to whom a suitable back-up is not available. However, protein kinase alleles that promote resistance to inhibitors can be exploited experimentally as gold-standards for "on"- and "off"-target validation strategies and constitute a powerful resource for assessing the ability of new or combined therapies to override resistance. Clinical resistance to kinase inhibitors is an evident in all tyrosine kinase-driven malignancies, where high rates of mutation drive tumor evolution toward the insidious drug-resistant (DR) state through a variety of mechanisms. Unfortunately, this problem is likely to intensify in the future as the number of target kinases, approved inhibitors, and clinical indications increase. To empower the analysis of resistance in kinases, we have validated a bioinformatic, structural, and cellular workflow for designing and evaluating resistance at key mutational hotspots among kinome members. In this chapter, we discuss how mutation of amino acids in the gatekeeper and hinge-loop regions (collectively termed the "resistance tetrad") and the DFG motif represent an effective approach for generating panels of DR kinase alleles for chemical genetics and biological target validation.
Collapse
|
85
|
MET and EGFR mutations identified in ALK-rearranged pulmonary adenocarcinoma: molecular analysis of 25 ALK-positive cases. J Thorac Oncol 2013; 8:574-81. [PMID: 23449277 DOI: 10.1097/jto.0b013e318287c395] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Oncogenic ALK kinase activity associated with ALK gene rearrangement is the target of crizotinib, an ALK inhibitor recently approved by the Food and Drug Administration for the treatment of ALK-rearranged (ALK+) non-small cell lung cancers. ALK+ status is generally thought to be mutually exclusive of epidermal growth factor receptor (EGFR) and KRAS mutations. However, the mutation status of other genes is not widely known in ALK+ tumors. The aim of this study is to survey for mutations involving other genes in 25 ALK+ cases confirmed by fluorescent in situ hybridization. METHODS Using the DNA extracted from formalin-fixed paraffin-embedded tumor samples, a MassArray-based Lung Cancer Mutations Screening Panel was performed to test for 179 individual mutations in 10 genes, including EGFR, KRAS, BRAF, ERBB2, JAK2, AKT1, AKT2, KIT, MET and PIK3CA, which have been implicated in lung carcinogenesis and/or considered as potential therapeutic targets. RESULTS Five of 25 ALK+ cases showed additional genetic abnormalities, which were verified by gene sequencing. One patient had EGFR del L747-S752. The remaining four mutations were in the MET gene: MET N375S (n = 2) and MET R988C (n = 2). No MET amplification was found by fluorescent in situ hybridization in the four cases with MET mutation. No mutations were detected in the other genes tested. CONCLUSIONS In summary, additional mutations were found in 20% of ALK+ cases involving two of the 10 genes tested. Our study highlights that EGFR mutation can be present in ALK+ tumors, though uncommon. Clinical implication of MET mutation in our cases is uncertain and further study is needed.
Collapse
|
86
|
Chen Z, Akbay E, Mikse O, Tupper T, Cheng K, Wang Y, Tan X, Altabef A, Woo SA, Chen L, Reibel JB, Janne PA, Sharpless NE, Engelman JA, Shapiro GI, Kung AL, Wong KK. Co-clinical trials demonstrate superiority of crizotinib to chemotherapy in ALK-rearranged non-small cell lung cancer and predict strategies to overcome resistance. Clin Cancer Res 2013; 20:1204-1211. [PMID: 24327273 DOI: 10.1158/1078-0432.ccr-13-1733] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE To extend the results of a phase III trial in patients with non-small cell lung cancer with adenocarcinomas harboring EML4-ALK fusion. EXPERIMENTAL DESIGN We conducted a co-clinical trial in a mouse model comparing the ALK inhibitor crizotinib to the standard-of-care cytotoxic agents docetaxel or pemetrexed. RESULTS Concordant with the clinical outcome in humans, crizotinib produced a substantially higher response rate compared with chemotherapy, associated with significantly longer progression-free survival. Overall survival was also prolonged in crizotinib- compared with chemotherapy-treated mice. Pemetrexed produced superior overall survival compared with docetaxel, suggesting that this agent may be the preferred chemotherapy in the ALK population. In addition, in the EML4-ALK-driven mouse lung adenocarcinoma model, HSP90 inhibition can overcome both primary and acquired crizotinib resistance. Furthermore, HSP90 inhibition, as well as the second-generation ALK inhibitor TAE684, demonstrated activity in newly developed lung adenocarcinoma models driven by crizotinib-insensitive EML4-ALK L1196M or F1174L. CONCLUSIONS Our findings suggest that crizotinib is superior to standard chemotherapy in ALK inhibitor-naïve disease and support further clinical investigation of HSP90 inhibitors and second-generation ALK inhibitors in tumors with primary or acquired crizotinib resistance.
Collapse
Affiliation(s)
- Zhao Chen
- Department of Medicine, Harvard Medical School, Boston MA 02115.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115.,Ludwig Center at Dana-Farber/Harvard Cancer Center, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Esra Akbay
- Department of Medicine, Harvard Medical School, Boston MA 02115.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115.,Ludwig Center at Dana-Farber/Harvard Cancer Center, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Oliver Mikse
- Department of Medicine, Harvard Medical School, Boston MA 02115.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115.,Ludwig Center at Dana-Farber/Harvard Cancer Center, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Tanya Tupper
- Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Katherine Cheng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115.,Ludwig Center at Dana-Farber/Harvard Cancer Center, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Yuchuan Wang
- Department of Imaging, Dana-Farber Cancer Institute, Boston, MA 02115.,Department of Radiology, Brigham and Women's Hospital, Boston, MA 02115
| | - Xiaohong Tan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Abigail Altabef
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Sue-Ann Woo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Liang Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Jacob B Reibel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Pasi A Janne
- Department of Medicine, Harvard Medical School, Boston MA 02115.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115.,Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Norman E Sharpless
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC 27599
| | - Jeffrey A Engelman
- Department of Medicine, Harvard Medical School, Boston MA 02115.,Department of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, MA 02114
| | - Geoffrey I Shapiro
- Department of Medicine, Harvard Medical School, Boston MA 02115.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115.,Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115.,Early Drug Development Center, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Andrew L Kung
- Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, MA 02115.,Department of Pediatric Oncology, Dana-Farber Cancer Institute and Children's Hospital, Boston, MA 02115
| | - Kwok-Kin Wong
- Department of Medicine, Harvard Medical School, Boston MA 02115.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115.,Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115.,Ludwig Center at Dana-Farber/Harvard Cancer Center, Dana-Farber Cancer Institute, Boston, MA 02115
| |
Collapse
|
87
|
Foretinib is a potent inhibitor of oncogenic ROS1 fusion proteins. Proc Natl Acad Sci U S A 2013; 110:19519-24. [PMID: 24218589 DOI: 10.1073/pnas.1319583110] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The rapidly growing recognition of the role of oncogenic ROS1 fusion proteins in the malignant transformation of multiple cancers, including lung adenocarcinoma, cholangiocarcinoma, and glioblastoma, is driving efforts to develop effective ROS1 inhibitors for use as molecularly targeted therapy. Using a multidisciplinary approach involving small molecule screening in combination with in vitro and in vivo tumor models, we show that foretinib (GSK1363089) is a more potent ROS1 inhibitor than crizotinib (PF-02341066), an ALK/ROS inhibitor currently in clinical evaluation for lung cancer patients harboring ROS1 rearrangements. Whereas crizotinib has demonstrated promising early results in patients with ROS1-rearranged non-small-cell lung carcinoma, recently emerging clinical evidence suggests that patients may develop crizotinib resistance due to acquired point mutations in the kinase domain of ROS1, thus necessitating identification of additional potent ROS1 inhibitors for therapeutic intervention. We confirm that the ROS1(G2032R) mutant, recently reported in clinical resistance to crizotinib, retains foretinib sensitivity at concentrations below safe, clinically achievable levels. Furthermore, we use an accelerated mutagenesis screen to preemptively identify mutations in the ROS1 kinase domain that confer resistance to crizotinib and demonstrate that these mutants also remain foretinib sensitive. Taken together, our data strongly suggest that foretinib is a highly effective ROS1 inhibitor, and further clinical investigation to evaluate its potential therapeutic benefit for patients with ROS1-driven malignancies is warranted.
Collapse
|
88
|
Novel 2,4-dianilino-5-fluoropyrimidine derivatives possessing ALK inhibitory activities. Arch Pharm Res 2013; 37:873-81. [DOI: 10.1007/s12272-013-0247-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Accepted: 09/17/2013] [Indexed: 12/22/2022]
|
89
|
Gainor JF, Shaw AT. Emerging paradigms in the development of resistance to tyrosine kinase inhibitors in lung cancer. J Clin Oncol 2013; 31:3987-96. [PMID: 24101047 DOI: 10.1200/jco.2012.45.2029] [Citation(s) in RCA: 265] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The success of tyrosine kinase inhibitors (TKIs) in select patients with non-small-cell lung cancer (NSCLC) has transformed management of the disease, placing new emphasis on understanding the molecular characteristics of tumor specimens. It is now recognized that genetic alterations in the epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) define two unique subtypes of NSCLC that are highly responsive to genotype-directed TKIs. Despite this initial sensitivity, however, the long-term effectiveness of such therapies is universally limited by the development of resistance. Identifying the mechanisms underlying this resistance is an area of intense, ongoing investigation. In this review, we provide an overview of recent experience in the field, focusing on results from preclinical resistance models and studies of patient-derived, TKI-resistant tumor specimens. Although diverse TKI resistance mechanisms have been identified within EGFR-mutant and ALK-positive patients, we highlight common principles of resistance shared between these groups. These include the development of secondary mutations in the kinase target, gene amplification of the primary oncogene, and upregulation of bypass signaling tracts. In EGFR-mutant and ALK-positive patients alike, acquired resistance may also be a dynamic and multifactorial process that may necessitate the use of treatment combinations. We believe that insights into the mechanisms of TKI resistance in patients with EGFR mutations or ALK rearrangements may inform the development of novel treatment strategies in NSCLC, which may also be generalizable to other kinase-driven malignancies.
Collapse
Affiliation(s)
- Justin F Gainor
- From the Massachusetts General Hospital Cancer Center, Boston, MA
| | | |
Collapse
|
90
|
Lazzari C, Spitaleri G, Catania C, Barberis M, Noberasco C, Santarpia M, Delmonte A, Toffalorio F, Conforti F, De Pas TM. Targeting ALK in patients with advanced non small cell lung cancer: biology, diagnostic and therapeutic options. Crit Rev Oncol Hematol 2013; 89:358-65. [PMID: 24156959 DOI: 10.1016/j.critrevonc.2013.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 07/14/2013] [Accepted: 09/17/2013] [Indexed: 10/26/2022] Open
Abstract
The discovery of EML4-ALK fusion gene in a subgroup of patients with lung adenocarcinoma led to the development of a new class of agents, the ALK inhibitors, and dramatically improved the clinical outcome of these patients. The striking results from clinical trials with crizotinib, the first ALK inhibitor evaluated, allowed the accelerated approval of crizotinib from the USA Food and Drug Administration (FDA). Despite the high initial results, patients acquire resistance to crizotinib, and different next generation ALK kinase inhibitors have been developed. In the current review, we will analyze the biology of EML4-ALK gene, the acquired resistance mechanisms to crizotinib, the therapeutic strategies, currently under evaluation, designed to overcome crizotinib resistance, and the open issues that need to be addressed in order to improve outcome in ALK+ Non Small Cell Lung Cancer (NSCLC) patients.
Collapse
Affiliation(s)
- Chiara Lazzari
- European Institute of Oncology, Division of Thoracic Oncology, Italy.
| | | | - Chiara Catania
- European Institute of Oncology, Division of Thoracic Oncology, Italy
| | | | | | | | - Angelo Delmonte
- European Institute of Oncology, Division of Thoracic Oncology, Italy
| | | | - Fabio Conforti
- European Institute of Oncology, Division of Thoracic Oncology, Italy
| | | |
Collapse
|
91
|
Heterogeneity of genetic changes associated with acquired crizotinib resistance in ALK-rearranged lung cancer. J Thorac Oncol 2013; 8:415-22. [PMID: 23344087 DOI: 10.1097/jto.0b013e318283dcc0] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Anaplastic lymphoma kinase (ALK)-rearranged non-small-cell lung cancer (NSCLC) is markedly sensitive to the ALK inhibitor crizotinib. However, acquired resistance to crizotinib is inevitable through several mechanisms. Therefore, this study was conducted to identify genetic alterations associated with crizotinib resistance. METHODS Tumor samples were derived from seven ALK-positive NSCLC patients who showed acquired resistance to crizotinib, and these patients were analyzed for ALK, EGFR, and KRAS mutations and ALK and EGFR gene amplifications. In vitro cytotoxicity of crizotinib and ALK downstream signals were compared between crizotinib-naive and -resistant NSCLC cells. RESULTS After a median duration of 6 months (range, 4-12 months), seven ALK-positive NSCLC patients developed acquired resistance to crizotinib. Three patients harbored secondary ALK mutations, including one patient with both mutations: L1196M (n = 2) and G1269A (n = 2). Of note, one patient displayed ALK gene copy number gain (4.1-fold increase compared with the pre-crizotinib specimen) and EGFR L858R mutation with high polysomy. The amphiregulin concentration was high in the supernatant fluid from five patients with malignant pleural effusion (116.4-18934.0 pg/ml). SNU-2535 cells derived from a patient who harbored the G1269 mutation were resistant to crizotinib treatment similar to H3122 CR1 cells. L1196M and G1269A mutant clones were less sensitive to crizotinib and ALK downstream signals were ineffectively suppressed in these clones. CONCLUSIONS Genetic changes associated with crizotinib resistance are heterogeneous in ALK-rearranged NSCLC patients who respond to crizotinib and subsequently develop resistance.
Collapse
|
92
|
Tang J, Karhinen L, Xu T, Szwajda A, Yadav B, Wennerberg K, Aittokallio T. Target inhibition networks: predicting selective combinations of druggable targets to block cancer survival pathways. PLoS Comput Biol 2013; 9:e1003226. [PMID: 24068907 PMCID: PMC3772058 DOI: 10.1371/journal.pcbi.1003226] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 08/01/2013] [Indexed: 01/11/2023] Open
Abstract
A recent trend in drug development is to identify drug combinations or multi-target agents that effectively modify multiple nodes of disease-associated networks. Such polypharmacological effects may reduce the risk of emerging drug resistance by means of attacking the disease networks through synergistic and synthetic lethal interactions. However, due to the exponentially increasing number of potential drug and target combinations, systematic approaches are needed for prioritizing the most potent multi-target alternatives on a global network level. We took a functional systems pharmacology approach toward the identification of selective target combinations for specific cancer cells by combining large-scale screening data on drug treatment efficacies and drug-target binding affinities. Our model-based prediction approach, named TIMMA, takes advantage of the polypharmacological effects of drugs and infers combinatorial drug efficacies through system-level target inhibition networks. Case studies in MCF-7 and MDA-MB-231 breast cancer and BxPC-3 pancreatic cancer cells demonstrated how the target inhibition modeling allows systematic exploration of functional interactions between drugs and their targets to maximally inhibit multiple survival pathways in a given cancer type. The TIMMA prediction results were experimentally validated by means of systematic siRNA-mediated silencing of the selected targets and their pairwise combinations, showing increased ability to identify not only such druggable kinase targets that are essential for cancer survival either individually or in combination, but also synergistic interactions indicative of non-additive drug efficacies. These system-level analyses were enabled by a novel model construction method utilizing maximization and minimization rules, as well as a model selection algorithm based on sequential forward floating search. Compared with an existing computational solution, TIMMA showed both enhanced prediction accuracies in cross validation as well as significant reduction in computation times. Such cost-effective computational-experimental design strategies have the potential to greatly speed-up the drug testing efforts by prioritizing those interventions and interactions warranting further study in individual cancer cases.
Collapse
Affiliation(s)
- Jing Tang
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
93
|
Park CH, Choe H, Jang IY, Kwon SY, Latif M, Lee HK, Lee HJ, Yang EH, Yun JI, Chae CH, Cho SY, Choi SU, Ha JD, Jung H, Kim HR, Kim P, Lee CO, Yun CS, Lee K. Novel bis-ortho-alkoxy-para-piperazinesubstituted-2,4-dianilinopyrimidines (KRCA-0008) as potent and selective ALK inhibitors for anticancer treatment. Bioorg Med Chem Lett 2013; 23:6192-6. [PMID: 24095090 DOI: 10.1016/j.bmcl.2013.08.090] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 08/19/2013] [Accepted: 08/23/2013] [Indexed: 10/26/2022]
Abstract
The synthesis of bis-ortho-alkoxy-para-piperazinesubstituted-2,4-dianilinopyrimidines is described and their structure-activity-relationship to anaplastic lymphoma kinase (ALK) is presented. KRCA-0008 is selective and potent to ALK and Ack1, and displays drug-like properties without hERG liability. KRCA-0008 demonstrates in vivo efficacy comparable to Crizotinib in xenograft mice model.
Collapse
Affiliation(s)
- Chi Hoon Park
- Bio-Organic Science Division, Korea Research Institute of Chemical Technology, PO Box 107, Daejeon 305-600, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Crizotinib in the Treatment of Non–Small-Cell Lung Cancer. Clin Lung Cancer 2013; 14:473-80. [DOI: 10.1016/j.cllc.2013.04.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 04/15/2013] [Accepted: 04/16/2013] [Indexed: 12/19/2022]
|
95
|
Weng L, Zhang L, Peng Y, Huang RS. Pharmacogenetics and pharmacogenomics: a bridge to individualized cancer therapy. Pharmacogenomics 2013; 14:315-24. [PMID: 23394393 DOI: 10.2217/pgs.12.213] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In the past decade, advances in pharmacogenetics and pharmacogenomics (PGx) have gradually unveiled the genetic basis of interindividual differences in drug responses. A large portion of these advances have been made in the field of anticancer therapy. Currently, the US FDA has updated the package inserts of approximately 30 anticancer agents to include PGx information. Given the complexity of this genetic information (e.g., tumor mutation and gene overexpression, chromosomal translocation and germline variations), as well as the variable level of scientific evidence, the FDA recommendation and potential action needed varies among drugs. In this review, we have highlighted some of these PGx discoveries for their scientific values and utility in improving therapeutic efficacy and reducing side effects. Furthermore, examples are also provided for the role of PGx in new anticancer drug development by revealing novel druggable targets.
Collapse
Affiliation(s)
- Liming Weng
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
96
|
Mechanisms of resistance to EGFR tyrosine kinase inhibitors gefitinib/erlotinib and to ALK inhibitor crizotinib. Lung Cancer 2013; 81:328-336. [PMID: 23809060 DOI: 10.1016/j.lungcan.2013.05.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/24/2013] [Accepted: 05/29/2013] [Indexed: 01/15/2023]
Abstract
The discovery of several molecular alterations that underlie non-small cell lung cancer (NSCLC) pathogenesis has led to the development of targeted therapies. In particular, gefitinib and erlotinib have become the standard of care in patients harboring epidermal growth factor receptor mutations, while crizotinib showed an impressive efficacy in patients with ALK-positive NSCLC. Nevertheless, the occurrence of clinical resistance limits the long term results of these novel agents. The identification of the molecular mechanisms responsible for acquired resistance to targeted therapy is crucial in order to pursue the creation of rational strategies to overcome resistance. In the current review, we will focus on the acquired resistance mechanisms to EGFR-TKIs and crizotinib and the therapeutic strategies currently under study to overcome resistance.
Collapse
|
97
|
Koudelakova V, Kneblova M, Trojanec R, Drabek J, Hajduch M. Non-small cell lung cancer - genetic predictors. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2013; 157:125-36. [DOI: 10.5507/bp.2013.034] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 04/25/2013] [Indexed: 01/14/2023] Open
|
98
|
Sechler M, Cizmic AD, Avasarala S, Van Scoyk M, Brzezinski C, Kelley N, Bikkavilli RK, Winn RA. Non-small-cell lung cancer: molecular targeted therapy and personalized medicine - drug resistance, mechanisms, and strategies. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2013; 6:25-36. [PMID: 23690695 PMCID: PMC3656464 DOI: 10.2147/pgpm.s26058] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Targeted therapies for cancer bring the hope of specific treatment, providing high efficacy and in some cases lower toxicity than conventional treatment. Although targeted therapeutics have helped immensely in the treatment of several cancers, like chronic myelogenous leukemia, colon cancer, and breast cancer, the benefit of these agents in the treatment of lung cancer remains limited, in part due to the development of drug resistance. In this review, we discuss the mechanisms of drug resistance and the current strategies used to treat lung cancer. A better understanding of these drug-resistance mechanisms could potentially benefit from the development of a more robust personalized medicine approach for the treatment of lung cancer.
Collapse
Affiliation(s)
- Marybeth Sechler
- Division of Pulmonary Sciences and Critical Care, University of Colorado, Aurora, CO, USA ; Program in Cancer Biology, University of Colorado, Aurora, CO, USA
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Sang J, Acquaviva J, Friedland JC, Smith DL, Sequeira M, Zhang C, Jiang Q, Xue L, Lovly CM, Jimenez JP, Shaw AT, Doebele RC, He S, Bates RC, Camidge DR, Morris SW, El-Hariry I, Proia DA. Targeted inhibition of the molecular chaperone Hsp90 overcomes ALK inhibitor resistance in non-small cell lung cancer. Cancer Discov 2013; 3:430-43. [PMID: 23533265 DOI: 10.1158/2159-8290.cd-12-0440] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
UNLABELLED EML4-ALK gene rearrangements define a unique subset of patients with non-small cell lung carcinoma (NSCLC), and the clinical success of the anaplastic lymphoma kinase (ALK) inhibitor crizotinib in this population has become a paradigm for molecularly targeted therapy. Here, we show that the Hsp90 inhibitor ganetespib induced loss of EML4-ALK expression and depletion of multiple oncogenic signaling proteins in ALK-driven NSCLC cells, leading to greater in vitro potency, superior antitumor efficacy, and prolonged animal survival compared with results obtained with crizotinib. In addition, combinatorial benefit was seen when ganetespib was used with other targeted ALK agents both in vitro and in vivo. Importantly, ganetespib overcame multiple forms of crizotinib resistance, including secondary ALK mutations, consistent with activity seen in a patient with crizotinib-resistant NSCLC. Cancer cells driven by ALK amplification and oncogenic rearrangements of ROS1 and RET kinase genes were also sensitive to ganetespib exposure. Taken together, these results highlight the therapeutic potential of ganetespib for ALK-driven NSCLC. SIGNIFICANCE In addition to direct kinase inhibition, pharmacologic blockade of the molecular chaperone Hsp90 is emerging as a promising approach for treating tumors driven by oncogenic rearrangements of ALK. The bioactivity profi le of ganetespib presented here underscores a new therapeutic opportunity to target ALK and overcome multiple mechanisms of resistance in patients with ALK-positive NSCLC.
Collapse
Affiliation(s)
- Jim Sang
- Synta Pharmaceuticals Corp, Lexington, MA 02421, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Wu D, Yu H, Li J. [Mechanisms of resistance to EML4-ALK inhibitors in non-small cell lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2013; 16:48-53. [PMID: 23327874 PMCID: PMC6000462 DOI: 10.3779/j.issn.1009-3419.2013.01.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Di Wu
- Department of Medical Oncology, Jilin Province Tumor Hospital, Changchun 130012, China;Tumor Center, First Hospital of Jilin University, Changchun 130021, China
| | | | | |
Collapse
|