101
|
Kauffmann-Guerrero D, Kahnert K, Kumbrink J, Syunyaeva Z, Tufman A, Huber RM. Successful Treatment of a Patient With NSCLC Harboring an EGFR Mutation and a Concomitant Met Exon 14 Skipping Mutation Combining Afatinib and Crizotinib. Clin Lung Cancer 2018; 20:59-62. [PMID: 30341016 DOI: 10.1016/j.cllc.2018.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/26/2018] [Accepted: 09/15/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Diego Kauffmann-Guerrero
- Department of Internal Medicine V, University of Munich (LMU), Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany.
| | - Kathrin Kahnert
- Department of Internal Medicine V, University of Munich (LMU), Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Jörg Kumbrink
- Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University, Munich, Germany; German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Zulfiya Syunyaeva
- Department of Internal Medicine V, University of Munich (LMU), Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Amanda Tufman
- Department of Internal Medicine V, University of Munich (LMU), Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Rudolf M Huber
- Department of Internal Medicine V, University of Munich (LMU), Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
102
|
Mignard X, Ruppert AM, Antoine M, Vasseur J, Girard N, Mazières J, Moro-Sibilot D, Fallet V, Rabbe N, Thivolet-Bejui F, Rouquette I, Lantuejoul S, Cortot A, Saffroy R, Cadranel J, Lemoine A, Wislez M. c-MET Overexpression as a Poor Predictor of MET Amplifications or Exon 14 Mutations in Lung Sarcomatoid Carcinomas. J Thorac Oncol 2018; 13:1962-1967. [PMID: 30149144 DOI: 10.1016/j.jtho.2018.08.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 07/01/2018] [Accepted: 08/10/2018] [Indexed: 10/28/2022]
Abstract
INTRODUCTION MNNG HOS transforming gene (MET) abnormalities such as amplification and exon 14 mutations may be responsive to targeted therapies. They are prevalent in lung sarcomatoid carcinomas (LSCs) and must be diagnosed as efficiently as possible. Hypothetically, c-MET overexpression by immunohistochemistry (IHC) may prove effective as a screening test for MET abnormalities. METHODS Tissue samples were obtained from consecutive patients with a resected LSC in four oncologic centers. IHC was performed using the SP44 antibody (Ventana, Tucson, Arizona) and evaluated using the MetMab score and H-score. Fluorescence in situ hybridization was applied with the dual color probe set from Zytovision (Clinisciences, Nanterre, France). True MET amplification was diagnosed when MET gene copy number was 5 or greater and the ratio between MET gene copy number and chromosome 7 number was greater than 2. All MET exon 14 alterations including those affecting splice sites occurring within splice donor and acceptor sites were detected in the routine molecular testing on genetic platforms. RESULTS A total of 81 LSCs were included. Fourteen (17%) exhibited positive IHC using the MetMab score and 15 (18.5%) using the H-score. MET amplification was detected in six tumors (8.5%) and MET exon 14 mutation in five (6%). A weak positive correlation between IHC and fluorescence in situ hybridization was found (r = 0.27, p = 0.0001). IHC sensitivity for MET amplification was 50%, with a specificity of 83%, positive predictive value of 21.4%, and negative predictive value of 94.7%. IHC sensitivity for MET exon 14 mutations was 20%, with a specificity of 83%, positive predictive value of 7%, and negative predictive value of 94%. CONCLUSION IHC is not a relevant screening tool for MET abnormalities in LSC.
Collapse
Affiliation(s)
- Xavier Mignard
- Sorbonne University, GRC n°04, Theranoscan, F-75252, Paris, France
| | - Anne-Marie Ruppert
- Sorbonne University, GRC n°04, Theranoscan, F-75252, Paris, France; AP-HP, GH HUEP, Tenon Hospital, Chest Department and Thoracic Oncology, F-75970, Paris, France
| | - Martine Antoine
- Sorbonne University, GRC n°04, Theranoscan, F-75252, Paris, France; AP-HP, GH HUEP, Tenon Hospital, Cytology and Pathology Department, F-75970, Paris, France
| | - Julie Vasseur
- AP-HP, GH Paris-Sud, Paul Brousse Hospital, Department of Biochiemistry and Oncogenetics, Platform Oncomolpath/INCa, Villejuif, France
| | - Nicolas Girard
- Respiratory Medicine and Thoracic Oncology Service, Louis Pradel Hospital, Hospices Civils de Lyon, Lyon, France
| | | | | | - Vincent Fallet
- AP-HP, GH HUEP, Tenon Hospital, Chest Department and Thoracic Oncology, F-75970, Paris, France
| | - Nathalie Rabbe
- AP-HP, GH HUEP, Tenon Hospital, Chest Department and Thoracic Oncology, F-75970, Paris, France
| | | | - Isabelle Rouquette
- Toulouse Universitary Cancer Institute, IUCT-Oncopole, Pathology Department, Toulouse, France
| | - Sylvie Lantuejoul
- Biopathology Department, Léon Bérard Center, Unicancer Grenoble, France; Grenoble Alpes University, Grenoble, France
| | - Alexis Cortot
- CHU Lille, Thoracic Oncology Department, Univ. Lille, Siric ONCOLille, Lille, France
| | - Raphaël Saffroy
- AP-HP, GH Paris-Sud, Paul Brousse Hospital, Department of Biochiemistry and Oncogenetics, Platform Oncomolpath/INCa, Villejuif, France
| | - Jacques Cadranel
- Sorbonne University, GRC n°04, Theranoscan, F-75252, Paris, France; AP-HP, GH HUEP, Tenon Hospital, Chest Department and Thoracic Oncology, F-75970, Paris, France
| | - Antoinette Lemoine
- AP-HP, GH Paris-Sud, Paul Brousse Hospital, Department of Biochiemistry and Oncogenetics, Platform Oncomolpath/INCa, Villejuif, France
| | - Marie Wislez
- Sorbonne University, GRC n°04, Theranoscan, F-75252, Paris, France; AP-HP, GH HUEP, Tenon Hospital, Chest Department and Thoracic Oncology, F-75970, Paris, France.
| |
Collapse
|
103
|
Zhu YC, Wang WX, Xu CW, Zhuang W, Song ZB, Du KQ, Chen G, Lv TF, Song Y. A novel co-existing ZCCHC8-ROS1 and de-novo MET amplification dual driver in advanced lung adenocarcinoma with a good response to crizotinib. Cancer Biol Ther 2018; 19:1097-1101. [PMID: 30095326 PMCID: PMC6301800 DOI: 10.1080/15384047.2018.1491506] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/22/2018] [Accepted: 06/17/2018] [Indexed: 01/06/2023] Open
Abstract
In non-small cell lung cancer (NSCLC), driver gene alterations, such as EGFR, ALK, MET, and ROS1, are usually mutually exclusive. Few clinical cases with co-existing ROS1 fusion and de-novo MET amplification have been reported. In addition, the efficacy of crizotinib in Chinese patients with driver co-existing alterations is uncertain. A 65-year-old female was diagnosed with lung adenocarcinoma metastatic to the brain. She had sufficient tumor tissue for detection of the target gene; however, common driver gene mutations, such as EGFR-wild and ALK-negative, were not initially detected. The patient was ultimately shown to have both ZCCHC8-ROS1 and de-novo MET gene amplification through next-generation sequencing with sensitivity to the targeted therapy of crizotinib. Unfortunately, the progression-free survival was only 6 months in length. We report here the first patient with co-existing ROS1 fusion and de-novo MET amplification to receive crizotinib in China. Treatment of our patient was effective with targeted therapy based on a precise diagnosis. Advanced or metastatic NSCLC patients with co-existing ROS1 fusion and de-novo MET amplification are sensitive to crizotinib. These uncommon driver gene mutations may be missed using the current first-generation detection assay. We must be aware of the incidence of concomitant ROS1 fusion and de-novo MET amplification because NSCLC patients could benefit from targeted therapy.
Collapse
Affiliation(s)
- You-cai Zhu
- Department of Thoracic Disease Diagnosis and Treatment Center Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, People’s Republic of China
| | - Wen-xian Wang
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People’s Republic of China
| | - Chun-wei Xu
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, Fujian, People’s Republic of China
| | - Wu Zhuang
- Department of Medical Thoracic Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, Fujian, People’s Republic of China
| | - Zheng-bo Song
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People’s Republic of China
| | - Kai-qi Du
- Department of Thoracic Disease Diagnosis and Treatment Center Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, People’s Republic of China
| | - Gang Chen
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, Fujian, People’s Republic of China
| | - Tang-feng Lv
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Jiangsu, People’s Republic of China
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Jiangsu, People’s Republic of China
| |
Collapse
|
104
|
Dudnik E, Bshara E, Grubstein A, Fridel L, Shochat T, Roisman LC, Ilouze M, Rozenblum AB, Geva S, Zer A, Rotem O, Allen AM, Peled N. Rare targetable drivers (RTDs) in non-small cell lung cancer (NSCLC): Outcomes with immune check-point inhibitors (ICPi). Lung Cancer 2018; 124:117-124. [PMID: 30268448 DOI: 10.1016/j.lungcan.2018.07.044] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Efficacy of immune check-point inhibitors (ICPi) in NSCLC with rare targetable drivers (RTDs) is largely unknown. MATERIALS AND METHODS Consecutive patients with NSCLC and RTDs (non-EGFR/ALK, n-82) were selected from the Davidoff Cancer Center database. ORR, PFS, OS with ICPi, OS since advanced disease diagnosis, TMB, MSI, and PD-L1 expression were analyzed; uni- and multivariate PFS and OS analyses were done. OS with ICPi was compared between the RTD cohort and the non-selected NSCLC cohort (n-278). RESULTS Of 50 tumors tested, 32%, 38%, and 30% were associated with ≥50%, 1-49% and <1% PD-L1 expression, respectively. Median TMB (n-48) comprised 4 muts/Mb (0-57); TMB ≥ 10 muts/Mb was seen in 19% of tumors. Both TMB and PD-L1 expression varied across different RTDs. All the 47 tumors were MSI stable. ORR with ICPi (n-44) was 16%, median PFS was 3.2 months (95% CI, 2.6-5.0), median OS was 16.2 months (95% CI, 8.4-NR). No correlation was seen between OS with ICPi and PD-L1 expression (p > 0.4), TMB (p > 0.8), or RTD type (p > 0.3). In the multivariate analysis, ECOG PS (p-0.005), targeted agents exposure (p-0.005), and ICPi exposure (p-0.04) were the only variables which correlated with OS since advanced disease diagnosis. Median OS since advanced disease diagnosis comprised 32 months (95% CI, 19.9-44.9) and 13 months (95% CI, 6.6-15.9) for patients who were and were not exposed to ICPi, respectively (log-rank test-6.3; p-0.01). In the inter-cohort comparison, for patients matched for ECOG PS (0/1), median OS with ICPi comprised 17.5 months (95% CI, 8.1-NR) and 8.6 months (95% CI, 6.7-NR) for RTD and non-selected patients, respectively (log-rank test-2.4, p-0.1). CONCLUSION In NSCLC with RTD, ICPi have favorable efficacy and independent impact on OS. NSCLC with RTD is associated with MSI stable status and variable levels of PD-L1 expression and TMB; their predictive value remains to be determined.
Collapse
Affiliation(s)
- Elizabeth Dudnik
- Thoracic Cancer Service, Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, Petah Tikva 49100, Israel.
| | - Elias Bshara
- Sackler Faculty of Medicine, Tel Aviv University, POB 39040 Ramat Aviv, Tel Aviv 69978, Israel
| | - Ahuva Grubstein
- Department of Diagnostic Radiology, Rabin Medical Center, Beilinson Campus, Petah Tikva 49100, Israel
| | - Ludmila Fridel
- Department of Pathology, Rabin Medical Center, Beilinson Campus, Petah Tikva 49100, Israel
| | - Tzippy Shochat
- Statistical Consulting Unit, Rabin Medical Center, Beilinson Campus, Petah Tikva 49100, Israel
| | - Laila C Roisman
- Cancer Institute, Soroka University Medical Center, Beer-Sheva 84101, Israel
| | - Maya Ilouze
- Thoracic Cancer Service, Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, Petah Tikva 49100, Israel
| | | | - Smadar Geva
- Sackler Faculty of Medicine, Tel Aviv University, POB 39040 Ramat Aviv, Tel Aviv 69978, Israel
| | - Alona Zer
- Thoracic Cancer Service, Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, Petah Tikva 49100, Israel
| | - Ofer Rotem
- Thoracic Cancer Service, Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, Petah Tikva 49100, Israel
| | - Aaron M Allen
- Thoracic Cancer Service, Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, Petah Tikva 49100, Israel; Sackler Faculty of Medicine, Tel Aviv University, POB 39040 Ramat Aviv, Tel Aviv 69978, Israel
| | - Nir Peled
- Cancer Institute, Soroka University Medical Center, Beer-Sheva 84101, Israel; Ben Gurion University of Negev, POB 653 Beer-Sheva 8410501, Israel
| |
Collapse
|
105
|
Hypoxia leads to decreased autophosphorylation of the MET receptor but promotes its resistance to tyrosine kinase inhibitors. Oncotarget 2018; 9:27039-27058. [PMID: 29930749 PMCID: PMC6007473 DOI: 10.18632/oncotarget.25472] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 05/08/2018] [Indexed: 12/16/2022] Open
Abstract
The receptor tyrosine kinase MET and its ligand, the Hepatocyte Growth Factor/Scattor Factor (HGF/SF), are essential to the migration, morphogenesis, and survival of epithelial cells. In addition, dysregulation of MET signaling has been shown to promote tumor progression and invasion in many cancers. Therefore, HGF/SF and MET are major targets for chemotherapies. Improvement of targeted therapies requires a perfect understanding of tumor microenvironment that strongly modifies half-life, bio-accessibility and thus, efficacy of treatments. In particular, hypoxia is a crucial microenvironmental phenomenon promoting invasion and resistance to treatments. Under hypoxia, MET auto-phosphorylation resulting from ligand stimulation or from receptor overexpression is drastically decreased within minutes of oxygen deprivation but is quickly reversible upon return to normoxia. Besides a decreased phosphorylation of its proximal adaptor GAB1 under hypoxia, activation of the downstream kinases Erk and Akt is maintained, while still being dependent on MET receptor. Consistently, several cellular responses induced by HGF/SF, including motility, morphogenesis, and survival are effectively induced under hypoxia. Interestingly, using a semi-synthetic ligand, we show that HGF/SF binding to MET is strongly impaired during hypoxia but can be quickly restored upon reoxygenation. Finally, we show that two MET-targeting tyrosine kinase inhibitors (TKIs) are less efficient on MET signalling under hypoxia. Like MET loss of phosphorylation, this hypoxia-induced resistance to TKIs is reversible under normoxia. Thus, although hypoxia does not affect downstream signaling or cellular responses induced by MET, it causes immediate resistance to TKIs. These results may prove useful when designing and evaluation of MET-targeted therapies against cancer.
Collapse
|
106
|
Basilico C, Modica C, Maione F, Vigna E, Comoglio PM. Targeting the MET oncogene by concomitant inhibition of receptor and ligand via an antibody-"decoy" strategy. Int J Cancer 2018; 143:1774-1785. [PMID: 29693242 DOI: 10.1002/ijc.31550] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 02/07/2018] [Accepted: 03/01/2018] [Indexed: 12/18/2022]
Abstract
MET, a master gene sustaining "invasive growth," is a relevant target for cancer precision therapy. In the vast majority of tumors, wild-type MET behaves as a "stress-response" gene and relies on the ligand (HGF) to sustain cell "scattering," invasive growth and apoptosis protection (oncogene "expedience"). In this context, concomitant targeting of MET and HGF could be crucial to reach effective inhibition. To test this hypothesis, we combined an anti-MET antibody (MvDN30) inducing "shedding" (i.e., removal of MET from the cell surface), with a "decoy" (i.e., the soluble extracellular domain of the MET receptor) endowed with HGF-sequestering ability. To avoid antibody/decoy interaction-and subsequent neutralization-we identified a single aminoacid in the extracellular domain of MET-lysine 842-that is critical for MvDN30 binding and engineered the corresponding recombinant decoyMET (K842E). DecoyMETK842E retains the ability to bind HGF with high affinity and inhibits HGF-induced MET phosphorylation. In HGF-dependent cellular models, MvDN30 antibody and decoyMETK842E used in combination cooperate in restraining invasive growth, and synergize in blocking cancer cell "scattering." The antibody and the decoy unbridle apoptosis of colon cancer stem cells grown in vitro as spheroids. In a preclinical model, built by orthotopic transplantation of a human pancreatic carcinoma in SCID mice engineered to express human HGF, concomitant treatment with antibody and decoy significantly reduces metastatic spread. The data reported indicate that vertical targeting of the MET/HGF axis results in powerful inhibition of ligand-dependent MET activation, providing proof of concept in favor of combined target therapy of MET "expedience."
Collapse
Affiliation(s)
| | - Chiara Modica
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Turin, Torino, Italy
| | - Federica Maione
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Elisa Vigna
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Turin, Torino, Italy
| | | |
Collapse
|
107
|
Liu J, Cui S, Pan F, Ni Y, Zhong H, Xiong L, Jin B, Chu T, Gu A, Jiang L. Feasibility of continuing crizotinib therapy after RECIST-PD in advanced non-small cell lung cancer patients with ALK/ ROS-1 mutations. J Cancer 2018; 9:1863-1869. [PMID: 29805713 PMCID: PMC5968775 DOI: 10.7150/jca.24950] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/02/2018] [Indexed: 12/11/2022] Open
Abstract
Objectives: To study whether ongoing clinical benefits of continuing anaplastic lymphoma kinase (ALK) and c-ros oncogene 1 (ROS1) inhibition are achieved by crizotinib treatment post progressive disease (PD) in advanced non-small cell lung cancer (NSCLC) patients harboring ALK/ROS1 mutations. Materials and methods: Demographic and clinicopathologic parameters were collected from 38 patients who continued crizotinib therapy beyond Response Evaluation Criteria in Solid Tumors (RECIST)-defined PD and analyzed. After adjusting for potential confounding factors, factors influencing the time from RECIST-PD to crizotinib discontinuation (progress-free survival 2, PFS2) were analyzed. Results: The median time from first dose treatment to RECIST-PD (PFS1) was 9.6 months (95% CI 5.6-13.6 months). The estimated median PFS2 was 5.9 months (95% CI 0.1-11.7 months). Six- and twelve-month crizotinib treatment probabilities after initial PD were 42.1% (95% CI 25.7-58.6%) and 21.1% (95% CI 7.5-34.6%), respectively. Patients who demonstrated RECIST-PD due to new lesions had a longer median PFS2 compared to patients who were attested to enlargement of original lesions (10.0 versus 2.4 months, p = 0.009). The median PFS2 was numerically longer among patients who received local therapy compared to those who did not receive local therapy, however the difference was not significant (9.9 versus 4.2 months, p = 0.094). Multivariable Cox regression analysis showed that only the progression pattern [new lesions versus enlargement of original lesions, HR = 0.329 (95% CI 0.138-0.782), p = 0.012] remained an independent prognostic factor of PFS2. Conclusion: Continuation of crizotinib therapy after RECIST-PD in Chinese NSCLC patients with positive ALK/ROS1 mutations is feasible in clinical practice.
Collapse
Affiliation(s)
- Jun Liu
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shaohua Cui
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Pan
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yiqian Ni
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hua Zhong
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Liwen Xiong
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Jin
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Tianqing Chu
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Aiqin Gu
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Liyan Jiang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
108
|
Chen C, He Z, Xie D, Zheng L, Zhao T, Zhang X, Cheng D. Molecular Mechanism Behind the Resistance of the G1202R-Mutated Anaplastic Lymphoma Kinase to the Approved Drug Ceritinib. J Phys Chem B 2018; 122:4680-4692. [PMID: 29648831 DOI: 10.1021/acs.jpcb.8b02040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Anaplastic lymphoma kinase (ALK) has been regarded as an essential target for the treatment of nonsmall cell lung cancer (NSCLC). However, the emergence of the G1202R solvent front mutation that confers resistance to the drugs was reported for the first as well as the second generation ALK inhibitors. It was thought that the G1202R solvent front mutation might hinder the drug binding. In this study, a different fact could be clarified by multiple molecular modeling methodologies through a structural analogue of ceritinib (compound 10, Cpd-10) that is reported to be a potent inhibitor against the G1202R mutation. Herein, molecular docking, accelerated molecular dynamics (aMD) simulations in conjunction with principal component analysis (PCA), and free energy map calculations were used to produce reasonable and representative initial conformations for the conventional MD simulations. Compared with Cpd-10, the binding specificity of ceritinib between ALK wild-type (ALKWT) and ALK G1202R (ALKG1202R) are primarily controlled by the conformational change of the P-loop- and A-loop-induced energetic redistributions, and the variation is nonpolar interactions, as indicated by conventional MD simulations, PCA, dynamic cross-correlation map (DCCM) analysis, and free energy calculations. Furthermore, the umbrella sampling (US) simulations were carried out to make clear the principle of the dissociation processes of ceritinib and Cpd-10 toward ALKWT and ALKG1202R. The calculation results suggest that Cpd-10 has similar dissociation processes from both ALKWT and ALKG1202R, but ceritinib is more easily dissociated from ALKG1202R than from ALKWT, thus less residence time is responsible for the ceritinib resistance. Our results suggest that both the binding specificity and the drug residence time should be emphasized in rational drug design to overcome the G1202R solvent front mutation of ALK resistance.
Collapse
Affiliation(s)
- Chaohong Chen
- Department of Thoracic Cardiovascular , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou 325000 , China
| | - Zhifeng He
- Department of Thoracic Cardiovascular , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou 325000 , China
| | - Deyao Xie
- Department of Thoracic Cardiovascular , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou 325000 , China
| | - Liangcheng Zheng
- Department of Thoracic Cardiovascular , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou 325000 , China
| | - Tianhao Zhao
- Department of Thoracic Cardiovascular , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou 325000 , China
| | - Xinbo Zhang
- Department of Thoracic Cardiovascular , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou 325000 , China
| | - Dezhi Cheng
- Department of Thoracic Cardiovascular , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou 325000 , China
| |
Collapse
|
109
|
Guo J, Guo L, Sun L, Wu Z, Ye J, Liu J, Zuo Q. Capture-based ultra-deep sequencing in plasma ctDNA reveals the resistance mechanism of ALK inhibitors in a patient with advanced ALK-positive NSCLC. Cancer Biol Ther 2018; 19:359-363. [PMID: 29400604 DOI: 10.1080/15384047.2018.1433496] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Anaplastic lymphoma kinase (ALK) is a validated molecular target in non-small-cell lung cancer (NSCLC). However, the clinical benefits of ALK inhibitors are almost universally limited by the emergence of drug resistance. METHODS We monitored the plasma circulating tumor DNA (ctDNA) using captured-based ultra-deep sequencing analysis of one patient with metastatic ALK-positive NSCLC who had received therapies including first-, second- and third-generation ALK inhibitors. Functional in vitro studies were further undertaken to elucidate the mechanism of resistance. RESULTS ALK T1151Sins mutation was detected when the patient developed resistance to ceritinib, and undetectable when she responded to lorlatinib. MET amplification was present when the tumor developed resistance to lorlatinib, and reduced when the patient received combination therapy of lorlatinib with crizotinib, which corresponded to clinical radiologic responses. In addition, further functional in vitro studies demonstrated that ALK harboring the T1151Sins mutation, while conferring resistance to ceritinib, was inhibited by lorlatinib. CONCLUSIONS Clinical evidence and in vitro validation revealed the clinical usefulness of captured-base ultra-deep sequencing on longitudinal plasma ctDNA in revealing the underlying resistance mechanism and guiding the precise administration of ALK inhibitors in patients with advanced ALK-positive NSCLC.
Collapse
Affiliation(s)
- Jing Guo
- a Department of Oncology , Nanfang Hospital, Southern Medical University , Guangzhou , Guangdong Province , China
| | - Lihong Guo
- a Department of Oncology , Nanfang Hospital, Southern Medical University , Guangzhou , Guangdong Province , China
| | - Li Sun
- a Department of Oncology , Nanfang Hospital, Southern Medical University , Guangzhou , Guangdong Province , China
| | - Zhenzhen Wu
- a Department of Oncology , Nanfang Hospital, Southern Medical University , Guangzhou , Guangdong Province , China
| | - Junyi Ye
- b Burning Rock Biotech , Guangzhou , Guangdong Province , China
| | - Jing Liu
- b Burning Rock Biotech , Guangzhou , Guangdong Province , China
| | - Qiang Zuo
- a Department of Oncology , Nanfang Hospital, Southern Medical University , Guangzhou , Guangdong Province , China
| |
Collapse
|
110
|
Ilie M, Szafer-Glusman E, Hofman V, Long-Mira E, Suttmann R, Darbonne W, Butori C, Lalvée S, Fayada J, Selva E, Yu W, Marquette CH, Shames DS, Punnoose E, Hofman P. Expression of MET in circulating tumor cells correlates with expression in tumor tissue from advanced-stage lung cancer patients. Oncotarget 2018; 8:26112-26121. [PMID: 28212540 PMCID: PMC5432243 DOI: 10.18632/oncotarget.15345] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 01/28/2017] [Indexed: 11/25/2022] Open
Abstract
Given the difficulty in obtaining adequate tissue in NSCLC, we investigated the utility of circulating tumor cells (CTCs) for MET status assessment in NSCLC patients. We used two platforms for CTC capture, and assessed MET expression in CTCs and matched-bronchial biopsies in patients with advanced-stage III/IV lung adenocarcinoma. Baseline peripheral blood was collected from 256 advanced-stage III/IV NSCLC patients from Genentech clinical trials, and from 106 patients with advanced-stage III/IV lung adenocarcinoma treated at the Department of Pneumology, Pasteur Hospital, Nice. CTCs were enriched using CellSearch (Genentech), or ISET technologies (Pasteur Hospital). MET expression was evaluated by immunofluorescence on CellSearch, and by immunocytochemistry on ISET-enriched CTCs and on matched FFPE tissue sections (Pasteur Hospital). CTCs were detected in 83 of 256 (32%) patients evaluated on CellSearch, with 30 samples (12%) exhibiting ≥ 5 CTCs/7.5 ml blood. On ISET, CTC were observed in 80 of 106 patients (75%), and 79 patients (75%) exhibited ≥ 5 CTCs/4 ml blood. MET expression on ISET CTCs was positive in 72% of cases, and the MET expression on matched-patient tissue was positive in 65% patients using the Onartuzumab IHC scoring algorithm (93% concordance). Quantification of MET expression using H-scores showed strong correlation between MET expression in tissue and CTCs (Spearman correlation, 0.93). MET status in CTCs isolated on ISET filters from blood samples of advanced-stage NSCLC patients correlated strongly with MET status in tumor tissue, illustrating the potential for using CTCs as a non-invasive, real-time biopsy to determine MET status of patients entering clinical trials.
Collapse
Affiliation(s)
- Marius Ilie
- Laboratory of Clinical and Experimental Pathology and Liquid Biopsy Laboratory, Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France.,Institute for Research on Cancer and Ageing, Nice (IRCAN), INSERM U1081/UMR CNRS 7284, Team 3, Antoine Lacassagne Cancer Center, Nice, France
| | - Edith Szafer-Glusman
- Department of Oncology Biomarker Development and Oncology Clinical Development, Genentech, Inc, South San Francisco, California, USA
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology and Liquid Biopsy Laboratory, Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France.,Institute for Research on Cancer and Ageing, Nice (IRCAN), INSERM U1081/UMR CNRS 7284, Team 3, Antoine Lacassagne Cancer Center, Nice, France.,Nice Hospital-Related Biobank (BB 0025-00033), Pasteur Hospital, Nice, France
| | - Elodie Long-Mira
- Laboratory of Clinical and Experimental Pathology and Liquid Biopsy Laboratory, Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France.,Institute for Research on Cancer and Ageing, Nice (IRCAN), INSERM U1081/UMR CNRS 7284, Team 3, Antoine Lacassagne Cancer Center, Nice, France
| | - Rebecca Suttmann
- Department of Oncology Biomarker Development and Oncology Clinical Development, Genentech, Inc, South San Francisco, California, USA
| | - Walter Darbonne
- Department of Oncology Biomarker Development and Oncology Clinical Development, Genentech, Inc, South San Francisco, California, USA
| | - Catherine Butori
- Laboratory of Clinical and Experimental Pathology and Liquid Biopsy Laboratory, Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France
| | - Salomé Lalvée
- Laboratory of Clinical and Experimental Pathology and Liquid Biopsy Laboratory, Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France
| | - Julien Fayada
- Nice Hospital-Related Biobank (BB 0025-00033), Pasteur Hospital, Nice, France
| | - Eric Selva
- Nice Hospital-Related Biobank (BB 0025-00033), Pasteur Hospital, Nice, France
| | - Wei Yu
- Department of Oncology Biomarker Development and Oncology Clinical Development, Genentech, Inc, South San Francisco, California, USA
| | | | - David S Shames
- Department of Oncology Biomarker Development and Oncology Clinical Development, Genentech, Inc, South San Francisco, California, USA
| | - Elizabeth Punnoose
- Department of Oncology Biomarker Development and Oncology Clinical Development, Genentech, Inc, South San Francisco, California, USA
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology and Liquid Biopsy Laboratory, Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France.,Institute for Research on Cancer and Ageing, Nice (IRCAN), INSERM U1081/UMR CNRS 7284, Team 3, Antoine Lacassagne Cancer Center, Nice, France.,Nice Hospital-Related Biobank (BB 0025-00033), Pasteur Hospital, Nice, France
| |
Collapse
|
111
|
Tomasello C, Baldessari C, Napolitano M, Orsi G, Grizzi G, Bertolini F, Barbieri F, Cascinu S. Resistance to EGFR inhibitors in non-small cell lung cancer: Clinical management and future perspectives. Crit Rev Oncol Hematol 2018; 123:149-161. [DOI: 10.1016/j.critrevonc.2018.01.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 11/09/2017] [Accepted: 01/31/2018] [Indexed: 12/18/2022] Open
|
112
|
Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, Colasacco C, Dacic S, Hirsch FR, Kerr K, Kwiatkowski DJ, Ladanyi M, Nowak JA, Sholl L, Temple-Smolkin R, Solomon B, Souter LH, Thunnissen E, Tsao MS, Ventura CB, Wynes MW, Yatabe Y. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. Arch Pathol Lab Med 2018; 142:321-346. [PMID: 29355391 DOI: 10.5858/arpa.2017-0388-cp] [Citation(s) in RCA: 573] [Impact Index Per Article: 81.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
CONTEXT - In 2013, an evidence-based guideline was published by the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology to set standards for the molecular analysis of lung cancers to guide treatment decisions with targeted inhibitors. New evidence has prompted an evaluation of additional laboratory technologies, targetable genes, patient populations, and tumor types for testing. OBJECTIVE - To systematically review and update the 2013 guideline to affirm its validity; to assess the evidence of new genetic discoveries, technologies, and therapies; and to issue an evidence-based update. DESIGN - The College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology convened an expert panel to develop an evidence-based guideline to help define the key questions and literature search terms, review abstracts and full articles, and draft recommendations. RESULTS - Eighteen new recommendations were drafted. The panel also updated 3 recommendations from the 2013 guideline. CONCLUSIONS - The 2013 guideline was largely reaffirmed with updated recommendations to allow testing of cytology samples, require improved assay sensitivity, and recommend against the use of immunohistochemistry for EGFR testing. Key new recommendations include ROS1 testing for all adenocarcinoma patients; the inclusion of additional genes ( ERBB2, MET, BRAF, KRAS, and RET) for laboratories that perform next-generation sequencing panels; immunohistochemistry as an alternative to fluorescence in situ hybridization for ALK and/or ROS1 testing; use of 5% sensitivity assays for EGFR T790M mutations in patients with secondary resistance to EGFR inhibitors; and the use of cell-free DNA to "rule in" targetable mutations when tissue is limited or hard to obtain.
Collapse
Affiliation(s)
- Neal I Lindeman
- From the Departments of Pathology (Drs Lindeman and Sholl) and Medicine (Dr Kwiatkowski), Brigham and Women's Hospital, Boston, Massachusetts; the Cancer Center (Dr Bernicker) and the Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas (Dr Cagle); the Department of Pathology, University of Colorado School of Medicine, Denver (Dr Aisner); the Diagnostic and Molecular Pathology Laboratory (Dr Arcila) and the Molecular Diagnostics Service (Dr Ladanyi), Memorial Sloan Kettering Cancer Center, New York, New York; the Department of Pathology & Medicine, Pulmonary, Critical Care and Sleep Medicine, New York, New York (Dr Beasley); the Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois (Mss Colasacco and Ventura); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Medicine and Pathology, University of Colorado, Denver (Dr Hirsch); the Department of Pathology, University of Aberdeen, Aberdeen, Scotland (Dr Kerr); the Department of Molecular Pathology, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Clinical and Scientific Affairs Division, Association for Molecular Pathology, Bethesda, Maryland (Dr Temple-Smolkin); the Molecular Therapeutics and Biomarkers Laboratory, Peter Maccallum Cancer Center, Melbourne, Australia (Dr Solomon); the Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands (Dr Thunnissen); the Department of Laboratory Medicine and Pathobiology, Princess Margaret Cancer Center, Toronto, Ontario, Canada (Dr Tsao); Scientific Affairs, International Association for the Study of Lung Cancer, Aurora, Colorado (Dr Wynes); and the Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan (Dr Yatabe). Dr Souter is in private practice in Wellanport, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, Colasacco C, Dacic S, Hirsch FR, Kerr K, Kwiatkowski DJ, Ladanyi M, Nowak JA, Sholl L, Temple-Smolkin R, Solomon B, Souter LH, Thunnissen E, Tsao MS, Ventura CB, Wynes MW, Yatabe Y. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J Thorac Oncol 2018; 13:323-358. [PMID: 29396253 DOI: 10.1016/j.jtho.2017.12.001] [Citation(s) in RCA: 357] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2017] [Indexed: 12/15/2022]
Abstract
CONTEXT In 2013, an evidence-based guideline was published by the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology to set standards for the molecular analysis of lung cancers to guide treatment decisions with targeted inhibitors. New evidence has prompted an evaluation of additional laboratory technologies, targetable genes, patient populations, and tumor types for testing. OBJECTIVE To systematically review and update the 2013 guideline to affirm its validity; to assess the evidence of new genetic discoveries, technologies, and therapies; and to issue an evidence-based update. DESIGN The College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology convened an expert panel to develop an evidence-based guideline to help define the key questions and literature search terms, review abstracts and full articles, and draft recommendations. RESULTS Eighteen new recommendations were drafted. The panel also updated 3 recommendations from the 2013 guideline. CONCLUSIONS The 2013 guideline was largely reaffirmed with updated recommendations to allow testing of cytology samples, require improved assay sensitivity, and recommend against the use of immunohistochemistry for EGFR testing. Key new recommendations include ROS1 testing for all adenocarcinoma patients; the inclusion of additional genes (ERBB2, MET, BRAF, KRAS, and RET) for laboratories that perform next-generation sequencing panels; immunohistochemistry as an alternative to fluorescence in situ hybridization for ALK and/or ROS1 testing; use of 5% sensitivity assays for EGFR T790M mutations in patients with secondary resistance to EGFR inhibitors; and the use of cell-free DNA to "rule in" targetable mutations when tissue is limited or hard to obtain.
Collapse
Affiliation(s)
- Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Philip T Cagle
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Dara L Aisner
- Department of Pathology, University of Colorado School of Medicine, Denver, New York
| | - Maria E Arcila
- Diagnostic and Molecular Pathology Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mary Beth Beasley
- Department of Pathology & Medicine, Pulmonary, Critical Care and Sleep Medicine, New York, New York
| | | | - Carol Colasacco
- Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois
| | - Sanja Dacic
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fred R Hirsch
- Department of Medicine and Pathology, University of Colorado, Denver, New York
| | - Keith Kerr
- Department of Pathology, University of Aberdeen, Aberdeen, Scotland
| | | | - Marc Ladanyi
- Molecular Diagnostics Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jan A Nowak
- Department of Molecular Pathology, Roswell Park Cancer Institute, Buffalo, New York
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Robyn Temple-Smolkin
- Clinical and Scientific Affairs Division, Association for Molecular Pathology, Bethesda, Maryland
| | - Benjamin Solomon
- Molecular Therapeutics and Biomarkers Laboratory, Peter Maccallum Cancer Center, Melbourne, Australia
| | | | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Ming S Tsao
- Department of Laboratory Medicine and Pathobiology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Christina B Ventura
- Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois
| | - Murry W Wynes
- Scientific Affairs, International Association for the Study of Lung Cancer, Aurora, Colorado
| | - Yasushi Yatabe
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| |
Collapse
|
114
|
Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, Colasacco C, Dacic S, Hirsch FR, Kerr K, Kwiatkowski DJ, Ladanyi M, Nowak JA, Sholl L, Temple-Smolkin R, Solomon B, Souter LH, Thunnissen E, Tsao MS, Ventura CB, Wynes MW, Yatabe Y. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J Mol Diagn 2018; 20:129-159. [PMID: 29398453 DOI: 10.1016/j.jmoldx.2017.11.004] [Citation(s) in RCA: 248] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
CONTEXT In 2013, an evidence-based guideline was published by the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology to set standards for the molecular analysis of lung cancers to guide treatment decisions with targeted inhibitors. New evidence has prompted an evaluation of additional laboratory technologies, targetable genes, patient populations, and tumor types for testing. OBJECTIVE To systematically review and update the 2013 guideline to affirm its validity; to assess the evidence of new genetic discoveries, technologies, and therapies; and to issue an evidence-based update. DESIGN The College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology convened an expert panel to develop an evidence-based guideline to help define the key questions and literature search terms, review abstracts and full articles, and draft recommendations. RESULTS Eighteen new recommendations were drafted. The panel also updated 3 recommendations from the 2013 guideline. CONCLUSIONS The 2013 guideline was largely reaffirmed with updated recommendations to allow testing of cytology samples, require improved assay sensitivity, and recommend against the use of immunohistochemistry for EGFR testing. Key new recommendations include ROS1 testing for all adenocarcinoma patients; the inclusion of additional genes (ERBB2, MET, BRAF, KRAS, and RET) for laboratories that perform next-generation sequencing panels; immunohistochemistry as an alternative to fluorescence in situ hybridization for ALK and/or ROS1 testing; use of 5% sensitivity assays for EGFR T790M mutations in patients with secondary resistance to EGFR inhibitors; and the use of cell-free DNA to "rule in" targetable mutations when tissue is limited or hard to obtain.
Collapse
Affiliation(s)
- Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Philip T Cagle
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Dara L Aisner
- Department of Pathology, University of Colorado School of Medicine, Denver, Colorado
| | - Maria E Arcila
- Diagnostic and Molecular Pathology Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mary Beth Beasley
- Department of Pathology & Medicine, Pulmonary, Critical Care and Sleep Medicine, New York, New York
| | - Eric H Bernicker
- Cancer Research Program, Houston Methodist Research Institute, Houston, Texas
| | - Carol Colasacco
- Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois
| | - Sanja Dacic
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fred R Hirsch
- Department of Medicine and Pathology, University of Colorado, Denver, Colorado
| | - Keith Kerr
- Department of Pathology, University of Aberdeen, Aberdeen, Scotland
| | | | - Marc Ladanyi
- Molecular Diagnostics Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jan A Nowak
- Department of Molecular Pathology, Roswell Park Cancer Institute, Buffalo, New York
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Robyn Temple-Smolkin
- Clinical and Scientific Affairs Division, Association for Molecular Pathology, Bethesda, Maryland
| | - Benjamin Solomon
- Molecular Therapeutics and Biomarkers Laboratory, Peter Maccallum Cancer Center, Melbourne, Australia
| | | | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Ming S Tsao
- Department of Laboratory Medicine and Pathobiology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Christina B Ventura
- Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois
| | - Murry W Wynes
- Scientific Affairs, International Association for the Study of Lung Cancer, Aurora, Colorado
| | - Yasushi Yatabe
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| |
Collapse
|
115
|
Liu Q, Yu S, Zhao W, Qin S, Chu Q, Wu K. EGFR-TKIs resistance via EGFR-independent signaling pathways. Mol Cancer 2018; 17:53. [PMID: 29455669 PMCID: PMC5817859 DOI: 10.1186/s12943-018-0793-1] [Citation(s) in RCA: 257] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 02/01/2018] [Indexed: 01/29/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs)-treatments bring significant benefit for patients harboring epidermal growth factor receptor (EGFR) mutations, especially for those with lung cancer. Unfortunately, the majority of these patients ultimately develop to the acquired resistance after a period of treatment. Two central mechanisms are involved in the resistant process: EGFR secondary mutations and bypass signaling activations. In an EGFR-dependent manner, acquired mutations, such as T790 M, interferes the interaction between TKIs and the kinase domain of EGFR. While in an EGFR-independent manner, dysregulation of other receptor tyrosine kinases (RTKs) or abnormal activation of downstream compounds both have compensatory functions against the inhibition of EGFR through triggering phosphatidylinositol 3-kinase (PI3K)/Akt and mitogen-activated protein kinase (MAPK) signaling axes. Nowadays, many clinical trials aiming to overcome and prevent TKIs resistance in various cancers are ongoing or completed. EGFR-TKIs in accompany with the targeted agents for resistance-related factors afford a promising first-line strategy to further clinical application.
Collapse
Affiliation(s)
- Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
116
|
Gallant JN, Lovly CM. Established, emerging and elusive molecular targets in the treatment of lung cancer. J Pathol 2018; 244:565-577. [PMID: 29344953 PMCID: PMC10182407 DOI: 10.1002/path.5038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/22/2017] [Accepted: 12/24/2017] [Indexed: 12/31/2022]
Abstract
Although histological subtype still underlies tumour classification and treatment, the recognition that lung cancer is, largely, a genetic disease has prompted a push to reconfigure cancer taxonomies according to molecular criteria. In this review, we discuss established (e.g. EGFR, ALK, ROS1, and programmed cell death 1/programmed death-ligand 1), emerging (e.g. MET, RET, and NTRK) and elusive (e.g. TP53, KRAS, and MYC) molecular targets in the treatment of lung cancer. We synthesize a large and rapidly growing body of literature regarding the discovery and therapeutic inhibition of these targets in lung cancer. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jean-Nicolas Gallant
- Vanderbilt University School of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christine M Lovly
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
117
|
Villaflor V, Won B, Nagy R, Banks K, Lanman RB, Talasaz A, Salgia R. Biopsy-free circulating tumor DNA assay identifies actionable mutations in lung cancer. Oncotarget 2018; 7:66880-66891. [PMID: 27602770 PMCID: PMC5341844 DOI: 10.18632/oncotarget.11801] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 08/13/2016] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION The potential of oncogene-driven targeted therapy is perhaps most fully realized in non-small cell lung cancer (NSCLC), given the number of genomic targets and approved matched therapies. However, invasive tissue biopsy at the time of each disease progression may not be possible and is associated with high morbidity and cost. Use of newly available "liquid biopsies" can circumvent these issues. RESULTS 83% of subjects had at least one genomic alteration identified in plasma. Most commonly mutated genes were TP53, KRAS and EGFR. Subjects with no detectable ctDNA were more likely to have small volume disease, lepidic growth pattern, mucinous tumors or isolated leptomeningeal disease. METHODS Subjects were individuals with NSCLC undergoing analysis of cell-free circulating tumor DNA using a validated, commercially-available next-generation sequencing assay at a single institution. Demographic, clinicopathologic information and results from tissue and plasma-based genomic testing were reviewed for each subject. CONCLUSIONS This is the first clinic-based series of NSCLC patients assessing outcomes of targeted therapies using a commercially available ctDNA assay. Over 80% of patients had detectable ctDNA, concordance between paired tissue and blood for truncal oncogenic drivers was high and patients with biomarkers identified in plasma had PFS in the expected range. These data suggest that biopsy-free ctDNA analysis is a viable first choice when the diagnostic tissue biopsy is insufficient for genotyping or at the time of progression when a repeated invasive tissue biopsy is not possible/preferred.
Collapse
Affiliation(s)
- Victoria Villaflor
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Brian Won
- University of Chicago School of Medicine, MC 2115, Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|
118
|
MET amplification assessed using optimized FISH reporting criteria predicts early distant metastasis in patients with non-small cell lung cancer. Oncotarget 2018; 9:12959-12970. [PMID: 29560123 PMCID: PMC5849187 DOI: 10.18632/oncotarget.24430] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 01/30/2018] [Indexed: 01/02/2023] Open
Abstract
To investigate the prognostic impact of MET copy number (MET-CN) in patients with non-small cell lung cancer (NSCLC), we retrospectively reviewed clinical and pathologic data of NSCLC patients whose tumors were assessed for MET-CN using fluorescence in situ hybridization (FISH). We correlated MET-CN status with patient overall survival (OS) and optimized MET-FISH reporting criteria. The study group included 384 patients with NSCLC of which 88% were adenocarcinoma and 55.7% of patients had distant metastases. There were 170 patients with stages I-III and 214 patients with stage IV disease. Based on the MET-CN and MET/CEP7 ratio the patients were classified into 3 categories: MET-amplification (METamp): MET/CEP7 ≥ 2 or MET-CN ≥ 5; MET-CN-gain (METcng): MET-CN ≥ 4 to < 5; and MET-negative (METneg): MET-CN < 4. METamp was associated with high fatality (P=.036) and stage IV tumors (P=.038). In patients with stages I-III NSCLC, patients in the METamp category had the shortest OS (P=.015) and more often developed distant metastases within 1 year (P=.004). In patients with stage IV tumors, METamp did not further impact the OS. Patients in the METcng category had the longest OS (P=.053). Multivariate analysis confirmed METamp to be an independent high-risk factor (HR 3.26; P=.026) and predicted earlier progression to distant metastasis (HR 4.86; P=.001). In conclusion, we suggest that the MET-FISH criteria presented optimizes risk stratification by defining 3 categories of NSCLC patients. METamp is an independent risk factor predicting early distant metastasis and patients with METcng could represent a lower-risk group.
Collapse
|
119
|
The coexistence of MET over-expression and an EGFR T790M mutation is related to acquired resistance to EGFR tyrosine kinase inhibitors in advanced non-small cell lung cancer. Oncotarget 2018; 7:51311-51319. [PMID: 27259997 PMCID: PMC5239477 DOI: 10.18632/oncotarget.9697] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 05/13/2016] [Indexed: 12/16/2022] Open
Abstract
MET overexpression and the EGFR T790M mutation are both associated with acquired resistance (AR) to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) in advanced non-small cell lung cancer (NSCLC). We characterized the frequency, underlying molecular mechanisms, and subsequent treatment for AR in MET overexpressing NSCLC patients with or without the T790M mutation. The study participants were 207 patients with advanced NSCLC and AR to EGFR-TKIs. The percentages of MET-, T790M- and MET/T790M-positive patients were 20.3% (42/207), 34.8% (72/207) and 6.8% (14/207), respectively. The disease control rate was 100% (5/5) for five patients with MET overexpression who received EGFR-TKIs plus a MET inhibitor. Among the MET/T790M-positive patients, seven received EGFR-TKIs plus a MET inhibitor and four received a T790M inhibitor, but no response was observed. The median post-progression survival (PPS) was 14.1, 24.5, and 10.7 months for MET-overexpressing, T790M-positive and MET/T790M-positive patients, respectively (P=0.044). c-Met, p-Met, ERBB3, and p-ERBB3 were highly expressed in MET-positive and MET/T790M-positive patients, but were poorly expressed in T790M-positive patients. EGFR, p-EGFR, AKT, p-AKT, MAPK, and p-MAPK were highly expressed in all three groups. These results suggest that MET/T790M-positive patients are at higher risk of AR to EGFR-TKIs, and have a worse PPS than patients with only MET overexpression or the T790M mutation alone. Clinical trials are needed to determine the best treatment for patients with both MET overexpression and the EGFR T790M mutation.
Collapse
|
120
|
Duan J, Yang X, Zhao J, Zhuo M, Wang Z, An T, Bai H, Wang J. Correlation among genetic variations of c-MET in Chinese patients with non-small cell lung cancer. Oncotarget 2017; 9:2660-2667. [PMID: 29416799 PMCID: PMC5788667 DOI: 10.18632/oncotarget.23474] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/15/2017] [Indexed: 12/26/2022] Open
Abstract
Background The purpose of our research was to determine the correlation of amplification, protein expression and somatic mutation of c-MET in IIIb-IV stage NSCLC (Non-small cell lung cancer). We also explored correlation of c-MET variation with clinical outcome. Results c-MET expression was observed in 28.6% (56/196) cases, and among those 13.8% (27/196) were shown to be FISH positive. Only 2.67% patients in this study carried the c-MET mutation. Cases with c-MET FISH positive were all IHC positive ,but in IHC positive cases, only half were FISH positive. Among patients with IHC2+ staining, 35.5% was FISH positive, while cases with IHC3+ staining,64% was FISH positive. Both protein expression and copy number of c-MET did not significantly correlate with clinical prognosis in these patients treated with EGFR-TKIs. Conclusions IHC could be used as a preliminary screening method for c-MET copy number amplification and should be confirmed by FISH only in IHC positive case which facilitate selection of ALK or MET inhibitor therapy. Methods c-MET gene copy number, protein expression and somatic mutation for exon 14 were detected by fluorescent- In-Situ-Hybridization (FISH), Immunohistochemistry (IHC), and Denaturing-High-Performance-Liquid-Chromatography (DHPLC), respectively, in 196 NSCLC patients. The relationship between c-MET abnormalities and clinical outcome of targeted therapy was analyzed by McNemar's test.
Collapse
Affiliation(s)
- Jianchun Duan
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaodan Yang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Thoracic Medical Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Thoracic Medical Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Minglei Zhuo
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Thoracic Medical Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Zhijie Wang
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tongtong An
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Thoracic Medical Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Hua Bai
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Wang
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
121
|
Abstract
Lung cancer is the number one cause of cancer-related death in both men and women. However, over the last few years, we have witnessed improved outcomes that are largely attributable to early detection, increased efforts in tobacco control, improved surgical approaches, and the development of novel targeted therapies. Currently, there are several novel therapies in clinical practice, including those targeting actionable mutations and more recently immunotherapeutic agents. Immunotherapy represents the most significant step forward in eradicating this deadly disease. Given the ever-changing landscape of lung cancer management, here we present an overview of the most recent advances in the management of non-small cell lung cancer.
Collapse
Affiliation(s)
- Samira Shojaee
- Division of Pulmonary and Critical Care Medicine, Virginia Commonwealth University, Richmond, USA
| | - Patrick Nana-Sinkam
- Division of Pulmonary and Critical Care Medicine, Virginia Commonwealth University, Richmond, USA
| |
Collapse
|
122
|
Batra U, Jain A, Sharma M, Bajaj R, Suryavanshis M. Role of crizotinib in c-mesenchymal-epidermal transition-positive nonsmall cell lung cancer patients. Indian J Cancer 2017; 54:178-181. [PMID: 29199685 DOI: 10.4103/0019-509x.219592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The increasing cases of NSCLC and improved understanding of its molecular biology has lead to identification of its varied driver mutations. cMET amplification has an important role as resistance mechanism for EGFR TKIs. Crizotinib is a drug which shows its anti-tumoral effect in cMET positive cases. Here we present a case series of three such patients who achieved were cMET amplified and showed partial response on Crizotinib.
Collapse
Affiliation(s)
- U Batra
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - A Jain
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - M Sharma
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - R Bajaj
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - M Suryavanshis
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| |
Collapse
|
123
|
Parsons BM, Meier DR, Gurda GT, Lofgren KA, Kenny PA. Exceptional Response to Crizotinib in an MET-Amplified Triple-Negative Breast Tumor. JCO Precis Oncol 2017; 1:1-6. [DOI: 10.1200/po.17.00070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Benjamin M. Parsons
- Benjamin M. Parsons and Grzegorz T. Gurda, Gundersen Health System; David R. Meier, Kristopher A. Lofgren, and Paraic A. Kenny, Kabara Cancer Research Institute, Gundersen Medical Foundation, La Crosse; and Paraic A. Kenny, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - David R. Meier
- Benjamin M. Parsons and Grzegorz T. Gurda, Gundersen Health System; David R. Meier, Kristopher A. Lofgren, and Paraic A. Kenny, Kabara Cancer Research Institute, Gundersen Medical Foundation, La Crosse; and Paraic A. Kenny, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Grzegorz T. Gurda
- Benjamin M. Parsons and Grzegorz T. Gurda, Gundersen Health System; David R. Meier, Kristopher A. Lofgren, and Paraic A. Kenny, Kabara Cancer Research Institute, Gundersen Medical Foundation, La Crosse; and Paraic A. Kenny, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Kristopher A. Lofgren
- Benjamin M. Parsons and Grzegorz T. Gurda, Gundersen Health System; David R. Meier, Kristopher A. Lofgren, and Paraic A. Kenny, Kabara Cancer Research Institute, Gundersen Medical Foundation, La Crosse; and Paraic A. Kenny, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Paraic A. Kenny
- Benjamin M. Parsons and Grzegorz T. Gurda, Gundersen Health System; David R. Meier, Kristopher A. Lofgren, and Paraic A. Kenny, Kabara Cancer Research Institute, Gundersen Medical Foundation, La Crosse; and Paraic A. Kenny, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
124
|
Zhang YC, Zhou Q, Wu YL. The emerging roles of NGS-based liquid biopsy in non-small cell lung cancer. J Hematol Oncol 2017; 10:167. [PMID: 29061113 PMCID: PMC5654124 DOI: 10.1186/s13045-017-0536-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/17/2017] [Indexed: 12/25/2022] Open
Abstract
The treatment paradigm of non-small cell lung cancer (NSCLC) has evolved into oncogene-directed precision medicine. Identifying actionable genomic alterations is the initial step towards precision medicine. An important scientific progress in molecular profiling of NSCLC over the past decade is the shift from the traditional piecemeal fashion to massively parallel sequencing with the use of next-generation sequencing (NGS). Another technical advance is the development of liquid biopsy with great potential in providing a dynamic and comprehensive genomic profiling of NSCLC in a minimally invasive manner. The integration of NGS with liquid biopsy has been demonstrated to play emerging roles in genomic profiling of NSCLC by increasing evidences. This review summarized the potential applications of NGS-based liquid biopsy in the diagnosis and treatment of NSCLC including identifying actionable genomic alterations, tracking spatiotemporal tumor evolution, dynamically monitoring response and resistance to targeted therapies, and diagnostic value in early-stage NSCLC, and discussed emerging challenges to overcome in order to facilitate clinical translation in future.
Collapse
Affiliation(s)
- Yi-Chen Zhang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, People's Republic of China.
| |
Collapse
|
125
|
Bahrami A, Shahidsales S, Khazaei M, Ghayour-Mobarhan M, Maftouh M, Hassanian SM, Avan A. C-Met as a potential target for the treatment of gastrointestinal cancer: Current status and future perspectives. J Cell Physiol 2017; 232:2657-2673. [PMID: 28075018 DOI: 10.1002/jcp.25794] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 01/05/2025]
Abstract
Aberrant activation of the HGF/c-Met signalling pathways is shown to be related with cell proliferation, progression, metastasis, and worse prognosis in several tumor types, including gastrointestinal cancers, suggesting its value as a stimulating-target for cancer-therapy. Several approaches have been developed for targeting HGF and/or c-Met, and one of them, crizotinib (dual c-Met/ALK inhibitor), is recently been approved by FDA for lung-cancers with ALK-rearrangement. The main aim of current review is to give an overview on the role of c-Met/HGF pathway in gastrointestinal cancer, in preclinical and clinical trials. Although several important matters is still remained to be elucidated on the molecular pathways underlying the antitumor effects of this therapy in gastrointestinal-cancers. Further investigations are warranted to recognize the main determinants of the activity of c-Met inhibitors, for parallel targeting signalling pathway associated/activated via MET/HGF pathway or in response to the cell resistance to anti-c-Met agents. Additionally, identification of patients that might benefit from therapy could help to increase the selectivity and efficacy of the therapy.
Collapse
Affiliation(s)
- Afsane Bahrami
- Molecular Medicine Group, Department of Modern Sciences and Technology, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soodabeh Shahidsales
- Cancer Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Neurogenic Inflammatory Research Center and Department of Physiology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Metabolic syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Maftouh
- Metabolic syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
126
|
Li Q, Wu J, Yan LX, Huang JW, Zhang Z, Zhang JE, Gao XL, Luo ZR, Liu J, Yang SF, Liu YH. ALK and ROS1 Double-Rearranged Lung Squamous Cell Carcinoma Responding to Crizotinib Treatment: A Case Report. J Thorac Oncol 2017; 12:e193-e197. [PMID: 28911956 DOI: 10.1016/j.jtho.2017.08.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 08/29/2017] [Accepted: 08/29/2017] [Indexed: 11/30/2022]
Affiliation(s)
- Qiong Li
- Graduate Shool, Southern Medical University, Guangzhou, People's Republic of China
| | - Jian Wu
- Department of Respiratory Medicine, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, People's Republic of China.
| | - Li-Xu Yan
- Department of Pathology, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, People's Republic of China
| | - Jun-Wei Huang
- Department of Respiratory Medicine, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, People's Republic of China
| | - Zhou Zhang
- Burning Rock Biotech, Guangzhou, People's Republic of China
| | - Jin-E Zhang
- Department of Radiology, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, People's Republic of China
| | - Xing-Lin Gao
- Department of Respiratory Medicine, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, People's Republic of China
| | - Ze-Ru Luo
- Department of Respiratory Medicine, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, People's Republic of China
| | - Jing Liu
- Burning Rock Biotech, Guangzhou, People's Republic of China
| | - Shi-Fang Yang
- Department of Respiratory Medicine, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, People's Republic of China
| | - Yan-Hui Liu
- Department of Pathology, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, People's Republic of China
| |
Collapse
|
127
|
Lau KW, Seng C, Lim TKH, Tan DSW. Expanded molecular interrogation for potential actionable targets in non-squamous non-small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:372. [PMID: 29057232 PMCID: PMC5635263 DOI: 10.21037/atm.2017.08.42] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/03/2017] [Indexed: 12/15/2022]
Abstract
The advent of targeted therapies has established new standards of care for defined molecular subsets of non-small cell lung cancer (NSCLC). Not only has this led to significant changes in the routine clinical management of lung cancer e.g., multiplexed genomic testing, but it has provided important principles and benchmarks for determining "actionability". At present, the clinical paradigms are most evolved for EGFR mutations and ALK rearrangements, where multiple randomized phase III trials have determined optimal treatment strategies in both treatment naïve and resistant settings. However, this may not always be feasible with low prevalence alterations e.g., ROS1 and BRAF mutations. Another emerging observation is that not all targets are equally "actionable", necessitating a rigorous preclinical, clinical and translational framework to prosecute new targets and drug candidates. In this review, we will cover the role of targeted therapies for NSCLC harbouring BRAF, MET, HER2 and RET alterations, all of which have shown promise in non-squamous non-small cell lung cancer (ns-NSCLC). We further review some early epigenetic targets in NSCLC, an area of emerging interest. With increased molecular segmentation of lung cancer, we discuss the upcoming challenges in drug development and implementation of precision oncology approaches, especially in light of the complex and rapidly evolving therapeutic landscape.
Collapse
Affiliation(s)
- Kah Weng Lau
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
- Institute of Molecular and Cell Biology, ASTAR, Singapore
| | - Claudia Seng
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Tony K H Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
- Cancer Therapeutics Research Laboratory, Singapore
- Genome Institute of Singapore, ASTAR, Singapore
| |
Collapse
|
128
|
Xu C, Wang W, Wu M, Zhu Y, Zhuang W, Lin G, Du K, Huang Y, Chen Y, Chen G, Fang M. Comparison of the c-MET gene amplification between primary tumor and metastatic lymph nodes in non-small cell lung cancer. Thorac Cancer 2017; 8:417-422. [PMID: 28590585 PMCID: PMC5582467 DOI: 10.1111/1759-7714.12455] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/14/2017] [Accepted: 04/17/2017] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND c-MET has recently been identified as a promising novel target in non-small cell lung cancer (NSCLC). We detected the consistency of c-MET gene amplification in metastatic lymph nodes and tumor tissues of NSCLC patients and discuss the clinical application value of c-MET gene amplification in metastatic lymph nodes. METHODS Real-time fluorescent quantitative PCR was used to test tumor tissues in 368 NSCLC patients and 178 paired metastatic lymph node samples. The amplification consistency in metastatic lymph nodes and tissue samples were compared and the correlation between c-MET gene amplification and the clinical characteristics of patients was analyzed. RESULTS The c-MET gene amplification rate was 8.97% (33/368) in tumor tissues. Of the 178 paired cases, c-MET gene amplification was positive in 7.95% (15/178) of cancerous tissues and 18.54% (33/178) of metastatic lymph nodes. c-MET gene amplification was detected more frequently in metastatic lymph nodes than in primary cancerous tissue. When metastatic lymph nodes were used as surrogate samples of primary cancerous tissues, the sensitivity was 86.67% (13/15) and the specificity was 87.69% (143/163). CONCLUSIONS Screening for c-MET gene amplification in lymph node metastases could determine which patients are eligible for tyrosine kinase inhibitor therapy. Lymph node metastasis can predict c-MET gene amplification in a primary tumor and guide the clinical use of c-MET gene targeted drugs.
Collapse
Affiliation(s)
- Chun‐wei Xu
- Department of Pathology, Fujian Provincial Cancer HospitalFujian Medical University Cancer HospitalFuzhouChina
| | - Wen‐xian Wang
- Department of ChemotherapyZhejiang Cancer HospitalHangzhouChina
| | - Mei‐juan Wu
- Department of PathologyZhejiang Cancer HospitalHangzhouChina
| | - You‐cai Zhu
- Department of Thoracic Disease CenterZhejiang Rongjun HospitalJiaxingChina
| | - Wu Zhuang
- Department of Medical Thoracic Oncology, Fujian Provincial Cancer HospitalFujian Medical University Cancer HospitalFuzhouChina
| | - Gen Lin
- Department of Medical Thoracic Oncology, Fujian Provincial Cancer HospitalFujian Medical University Cancer HospitalFuzhouChina
| | - Kai‐qi Du
- Department of Thoracic Disease CenterZhejiang Rongjun HospitalJiaxingChina
| | - Yun‐jian Huang
- Department of Medical Thoracic Oncology, Fujian Provincial Cancer HospitalFujian Medical University Cancer HospitalFuzhouChina
| | - Yan‐ping Chen
- Department of Pathology, Fujian Provincial Cancer HospitalFujian Medical University Cancer HospitalFuzhouChina
| | - Gang Chen
- Department of Pathology, Fujian Provincial Cancer HospitalFujian Medical University Cancer HospitalFuzhouChina
| | - Mei‐yu Fang
- Department of Comprehensive Medical OncologyZhejiang Cancer HospitalHangzhouChina
| |
Collapse
|
129
|
Smith MA, Licata T, Lakhani A, Garcia MV, Schildhaus HU, Vuaroqueaux V, Halmos B, Borczuk AC, Chen YA, Creelan BC, Boyle TA, Haura EB. MET-GRB2 Signaling-Associated Complexes Correlate with Oncogenic MET Signaling and Sensitivity to MET Kinase Inhibitors. Clin Cancer Res 2017; 23:7084-7096. [PMID: 28855353 DOI: 10.1158/1078-0432.ccr-16-3006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 05/01/2017] [Accepted: 08/23/2017] [Indexed: 12/31/2022]
Abstract
Purpose: Targeting MET in cancer is hampered by lack of diagnostics that accurately reflect high MET signaling and dependence. We hypothesized that assays reflecting MET signaling associated protein complexes could redefine tumors dependent on MET and could add additional precision beyond genomic assessments.Experimental Design: We used biochemical approaches, cellular viability studies, and proximity ligation assays to assess MET dependence. We examined MET signaling complexes in lung cancer patient specimens (N = 406) and patient-derived xenograft (PDX) models of solid tumors (N = 308). We evaluated response to crizotinib in a MET-amplified cohort of PDX models of lung cancer (N = 6) and provide a case report of a lung cancer patient harboring a Δexon14 MET splice variant.Results: We found the interaction of MET with the adaptor protein GRB2 is necessary for oncogenic survival signaling by MET. MET-GRB2 complexes were identified only within MET-amplified PDX models and patient specimens but exhibit substantial variability. Lack of MET-GRB2 complexes was associated with lack of response to MET TKI in cell lines and PDX models. Presence of MET-GRB2 complexes can further subtype tumors with Δexon14 MET splice variants. Presence of these complexes correlated with response to crizotinib in one patient with Δexon14 MET lacking MET gene amplification.Conclusions: Proximity assays measuring MET-GRB2 signaling complexes provide novel insights into MET-mediated signaling and could complement current clinical genomics-based assay platforms. Clin Cancer Res; 23(22); 7084-96. ©2017 AACR.
Collapse
Affiliation(s)
- Matthew A Smith
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Thomas Licata
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Aliya Lakhani
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | | | | | | | - Balazs Halmos
- Department of Oncology, Montefiore/Albert Einstein Cancer Center, Bronx, New York
| | - Alain C Borczuk
- Department of Pathology, Weill-Cornell Medicine, New York, New York
| | - Y Ann Chen
- Department of Biostatistics, Moffitt Cancer Center, Tampa, Florida
| | | | - Theresa A Boyle
- Department of Molecular Pathology, Moffitt Cancer Center, Tampa, Florida
| | - Eric B Haura
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida.
| |
Collapse
|
130
|
Davies KD, Ng TL, Estrada-Bernal A, Le AT, Ennever PR, Camidge DR, Doebele RC, Aisner DL. Dramatic Response to Crizotinib in a Patient with Lung Cancer Positive for an HLA-DRB1-MET Gene Fusion. JCO Precis Oncol 2017. [PMID: 29527595 DOI: 10.1200/po.17.00117] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Kurtis D Davies
- Department of Pathology, University of Colorado - Anschutz Medical Campus, Aurora CO
| | - Terry L Ng
- Division of Medical Oncology - University of Colorado - Anschutz Medical Campus, Aurora CO
| | - Adriana Estrada-Bernal
- Division of Medical Oncology - University of Colorado - Anschutz Medical Campus, Aurora CO
| | - Anh T Le
- Division of Medical Oncology - University of Colorado - Anschutz Medical Campus, Aurora CO
| | | | - D Ross Camidge
- Division of Medical Oncology - University of Colorado - Anschutz Medical Campus, Aurora CO
| | - Robert C Doebele
- Division of Medical Oncology - University of Colorado - Anschutz Medical Campus, Aurora CO
| | - Dara L Aisner
- Department of Pathology, University of Colorado - Anschutz Medical Campus, Aurora CO
| |
Collapse
|
131
|
Pilotto S, Carbognin L, Karachaliou N, Ma PC, Rosell R, Tortora G, Bria E. Tracking MET de-addiction in lung cancer: A road towards the oncogenic target. Cancer Treat Rev 2017; 60:1-11. [PMID: 28843992 DOI: 10.1016/j.ctrv.2017.08.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 08/05/2017] [Accepted: 08/09/2017] [Indexed: 02/06/2023]
Abstract
The discovery of druggable oncogenic drivers (i.e. EGFR and ALK), along with the introduction of comprehensive tumor genotyping techniques into the daily clinical practice define non-small-cell lung cancer (NSCLC) asa group of heterogeneous diseases, requiring a context-personalized clinico-therapeutical approach. Among the most investigated biomarkers, the MET proto-oncogene has been extensively demonstrated to play a crucial role throughout the lung oncogenesis, unbalancing the proliferation/apoptosis signaling and influencing the epithelial-mesenchymal transition and the invasive phenotype. Nevertheless, although different mechanisms eliciting the aberrant MET-associated oncogenic stimulus have been detected in lung cancer (such as gene amplification, increased gene copy number, mutations and MET/HGF overexpression), to date no clinically impactful results have been achieved with anti-MET tyrosine kinase inhibitors and monoclonal antibodies in the context of an unselected or MET enriched population. Recently, MET exon 14 splicing abnormalities have been identified asa potential oncogenic target in lung cancer, able to drive the activity of MET inhibitors in molecularly selected patients. In this paper, the major advancement and drawbacks of MET history in lung cancer are reviewed, underlying the renewed scientific euphoria related to the recent identification of MET exon 14 splicing variants asan actionable oncogenic target.
Collapse
Affiliation(s)
- S Pilotto
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy.
| | - L Carbognin
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy.
| | | | - P C Ma
- WVU Cancer Institute, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV, United States; WV Clinical and Translational Science Institute, Morgantown, WV, United States.
| | - R Rosell
- Pangaea Biotech, Barcelona, Spain; Instituto Oncológico Dr Rosell, Quiron-Dexeus University Hospital, Barcelona, Spain; Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona, Spain; Molecular Oncology Research (MORe) Foundation, Barcelona, Spain; Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Spain.
| | - G Tortora
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy.
| | - E Bria
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy.
| |
Collapse
|
132
|
Oh DY, Jung K, Song JY, Kim S, Shin S, Kwon YJ, Oh E, Park WY, Song SY, Choi YL. Precision medicine approaches to lung adenocarcinoma with concomitant MET and HER2 amplification. BMC Cancer 2017; 17:535. [PMID: 28806950 PMCID: PMC5557466 DOI: 10.1186/s12885-017-3525-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 08/01/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Patient-derived xenograft (PDX) models are important tools in precision medicine and for the development of targeted therapies to treat cancer patients. This study aimed to evaluate our precision medicine strategy that integrates genomic profiling and preclinical drug-screening platforms, in order to personalize cancer treatments using PDX models. METHODS We performed array-comparative genomic hybridization, microarray, and targeted next-generation sequencing analyses, in order to determine the oncogenic driver mutations. PDX cells were obtained from PDXs and subsequently screened in vitro with 17 targeted agents. RESULTS PDX tumors recapitulated the histopathologic and genetic features of the patient tumors. Among the samples from lung cancer patients that were molecularly-profiled, copy number analysis identified unique focal MET amplification in one sample, 033 T, without RTK/RAS/RAF oncogene mutations. Although HER2 amplification in 033 T was not detected in the cancer panel, the selection of HER2-amplified clones was found in PDXs and PDX cells. Additionally, MET and HER2 overexpression were found in patient tumors, PDXs, and PDX cells. Crizotinib or EGFR tyrosine kinase inhibitor treatments significantly inhibited cell growth and impaired tumor sphere formation in 033 T PDX cells. CONCLUSIONS We established PDX cell models using surgical samples from lung cancer patients, and investigated their preclinical and clinical implications for personalized targeted therapy. Additionally, we suggest that MET and EGFR inhibitor-based therapy can be used to treat MET and HER2-overexpressing lung cancers, without receptor tyrosine kinase /RAS/RAF pathway alterations.
Collapse
Affiliation(s)
- Doo-Yi Oh
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Kyungsoo Jung
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Ji-Young Song
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Seokhwi Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Sang Shin
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Yong-Jun Kwon
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Ensel Oh
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Woong-Yang Park
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.,Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea
| | - Sang Yong Song
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.,Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, South Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.
| |
Collapse
|
133
|
Ying X, Wang M, Verma V, Wang M, Ye S, Bi J, Zhou X, Han G, Zhen W. Metastatic spread of solid subtype lung adenocarcinoma to the small intestine with anemia and melena: A case report. Medicine (Baltimore) 2017; 96:e7768. [PMID: 28834880 PMCID: PMC5572002 DOI: 10.1097/md.0000000000007768] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
RATIONALE Metastasis to the small intestine from a primary lung cancer is rare, and is associated with a poor prognosis. Early diagnosis of small intestine metastasis is difficult because of the low incidence of clinically apparent symptoms. PATIENT CONCERNS Clinical data and treatment of a 59-year-old man with small intestine metastasis from primary solid subtype lung adenocarcinoma are summarized. DIAGNOSES A man who was previously diagnosed with stage IIIA (T3N2M0) lung adenocarcinoma (solid subtype) came to our hospital for postoperative radiotherapy. Laboratory tests indicated anemia and melena. The patient was initially believed to have digestive ulcer and was treated with omeprazole, which proved to be ineffective. We conducted an abdominal computed tomography (CT) contrast scan and discovered a mass in the small intestine mass. Further positron emission tomography-computed tomography (PET-CT) imaging indicated the small intestine mass with fluorodeoxyglucose uptake. INTERVENTIONS The patient underwent an enterectomy and anastomosis. Pathological analysis confirmed the diagnosis of small intestinal metastasis from lung cancer with concomitant mesenteric lymph node metastasis. OUTCOMES One month after the operation, hemoglobin levels became normal, and the patient had good quality of life. However, 3 months after the operation, the patient suffered from anemia again. An abdominal CT scan indicated a new small intestine mass. Progression continued rapidly, and the patient died of hemorrhagic shock 5.5 months after the resection of the small intestine mass. LESSONS Although uncommon, if lung cancer patients present with anemia and melena, enteric metastasis should be part of the differential diagnosis. Abdominal CT scans and PET-CT are effective for early diagnosis. The prognosis of metastatic spread of solid subtype lung adenocarcinoma to the small intestine with mesenteric lymph node metastasis is poor. Subgroups of patients benefitting from metastasectomy and more effective systemic therapy need to be further investigated.
Collapse
Affiliation(s)
| | | | - Vivek Verma
- Department of Radiation Oncology, University of Nebraska Medical Centre, Omaha, NE
| | | | - Shengwei Ye
- Department of Gastrointestinal Surgery, Hubei Cancer Hospital
| | | | | | - Guang Han
- Department of Radiation Oncology
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Weining Zhen
- Department of Radiation Oncology, University of Nebraska Medical Centre, Omaha, NE
| |
Collapse
|
134
|
Engstrom LD, Aranda R, Lee M, Tovar EA, Essenburg CJ, Madaj Z, Chiang H, Briere D, Hallin J, Lopez-Casas PP, Baños N, Menendez C, Hidalgo M, Tassell V, Chao R, Chudova DI, Lanman RB, Olson P, Bazhenova L, Patel SP, Graveel C, Nishino M, Shapiro GI, Peled N, Awad MM, Jänne PA, Christensen JG. Glesatinib Exhibits Antitumor Activity in Lung Cancer Models and Patients Harboring MET Exon 14 Mutations and Overcomes Mutation-mediated Resistance to Type I MET Inhibitors in Nonclinical Models. Clin Cancer Res 2017; 23:6661-6672. [DOI: 10.1158/1078-0432.ccr-17-1192] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/13/2017] [Accepted: 07/24/2017] [Indexed: 11/16/2022]
|
135
|
Al-Saad S, Richardsen E, Kilvaer TK, Donnem T, Andersen S, Khanehkenari M, Bremnes RM, Busund LT. The impact of MET, IGF-1, IGF1R expression and EGFR mutations on survival of patients with non-small-cell lung cancer. PLoS One 2017; 12:e0181527. [PMID: 28742836 PMCID: PMC5526580 DOI: 10.1371/journal.pone.0181527] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 07/03/2017] [Indexed: 01/10/2023] Open
Abstract
Introduction To compare the efficacy of silver in situ hybridization (SISH) and immunohistochemistry (IHC) in detecting MET and IGF1R alterations and to investigate their prevalence and prognostic significance. A possible correlation between MET receptor expression, MET gene alterations and the two most frequent occurring EGFR gene mutations was also investigated. Materials and methods Stage I to IIIA tumors from 326 patients with NSCLC were immunohistochemically tested for protein expression of MET and IGF-1. Their cytoplasmic expression was compared with the gene copy number of the MET and IGF1Rgenes by SISH in paraffin-embedded, formalin-fixed material. Correlations were made with the immunohistochemical expression of two frequent EGFR mutations and clinicopathological variables. Univariate and multivariate survival analyses was used to evaluate the prognostic efficacy of the tested markers. Results In univariate analyses, high cytoplasmic MET expression showed a significant negative prognostic effect in adenocarcinoma patients (p = 0.026). MET gene to chromosome 7 ratio was a significant positive prognostic marker (p = 0.005), probably only due to the highly negative prognostic significance of chromosome 7 polysomy (p = 0.002). High IGF1R gene copy number was a negative prognostic marker for all NSCLC patients (p = 0.037). In the multivariate analysis, polysomy of chromosome 7 in tumor cells correlated significantly and independently with a poor prognosis (p = 0.011). In patients with adenocarcinoma, a high cytoplasmic MET expression was an independent negative prognostic marker (p = 0.013). In males a high IGF1R gene copy number to chromosome 15 count ratio was significantly and independently correlated to a poor prognosis (p = 0.018). Conclusion MET protein expression provides superior prognostic information compared with SISH. Polysomy of chromosome 7 is an independent negative prognostic factor in NSCLC patients. This finding has an important implication while examining genes located on chromosome 7 by means of SISH. High IGF1R gene copy number to chromosome 15 count ratio is an independent predictor of inferior survival in male patients with primary NSCLC.
Collapse
Affiliation(s)
- Samer Al-Saad
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
- Department of Clinical Pathology, University Hospital of Northern Norway, Tromso, Norway
- * E-mail:
| | - Elin Richardsen
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
- Department of Clinical Pathology, University Hospital of Northern Norway, Tromso, Norway
| | - Thomas K. Kilvaer
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- Department of Oncology, University Hospital of Northern Norway, Tromso, Norway
| | - Tom Donnem
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- Department of Oncology, University Hospital of Northern Norway, Tromso, Norway
| | - Sigve Andersen
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- Department of Oncology, University Hospital of Northern Norway, Tromso, Norway
| | - Mehrdad Khanehkenari
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Roy M. Bremnes
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- Department of Oncology, University Hospital of Northern Norway, Tromso, Norway
| | - Lill-Tove Busund
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
- Department of Clinical Pathology, University Hospital of Northern Norway, Tromso, Norway
| |
Collapse
|
136
|
Losanno T, Gridelli C. Recent advances in targeted advanced lung cancer therapy in the elderly. Expert Rev Anticancer Ther 2017; 17:787-797. [DOI: 10.1080/14737140.2017.1348232] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Tania Losanno
- Medical Oncology, Azienda Ospedaliera San Camillo Forlanini, Roma, Italy
| | | |
Collapse
|
137
|
First macrocyclic 3 rd -generation ALK inhibitor for treatment of ALK/ROS1 cancer: Clinical and designing strategy update of lorlatinib. Eur J Med Chem 2017; 134:348-356. [DOI: 10.1016/j.ejmech.2017.04.032] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/26/2017] [Accepted: 04/12/2017] [Indexed: 11/21/2022]
|
138
|
Miao Y, Ling L, Zhang X, Huang J. [A Case Report of Pulmonary Sarcomatoid Carcinoma with ALK Rearrangement]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2017; 20:366-370. [PMID: 28532546 PMCID: PMC5973067 DOI: 10.3779/j.issn.1009-3419.2017.05.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Yehong Miao
- Department of Respiratory Medicine, the First Affiliated Hospital of Suzhou University, Suzhou 215006, China
| | - Lin Ling
- Department of Respiratory Medicine, the First Affiliated Hospital of Suzhou University, Suzhou 215006, China
| | - Xiuqin Zhang
- Department of Respiratory Medicine, the First Affiliated Hospital of Suzhou University, Suzhou 215006, China
| | - Jian'an Huang
- Department of Respiratory Medicine, the First Affiliated Hospital of Suzhou University, Suzhou 215006, China
| |
Collapse
|
139
|
Martin P, Leighl NB. Review of the use of pretest probability for molecular testing in non-small cell lung cancer and overview of new mutations that may affect clinical practice. Ther Adv Med Oncol 2017; 9:405-414. [PMID: 28607579 PMCID: PMC5455881 DOI: 10.1177/1758834017704329] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 03/14/2017] [Indexed: 11/17/2022] Open
Abstract
This article considers the use of pretest probability in non-small cell lung cancer (NSCLC) and how its use in EGFR testing has helped establish clinical guidelines on selecting patients for EGFR testing. With an ever-increasing number of molecular abnormalities being identified and often limited tissue available for testing, the use of pretest probability will need to be increasingly considered in the future for selecting investigations and treatments in patients. In addition we review new mutations that have the potential to affect clinical practice.
Collapse
Affiliation(s)
- Petra Martin
- Division of Medical Oncology, Princess Margaret Cancer Centre, 5-105 610 University Avenue, Toronto, ON, M5G 2M9, Canada
| | - Natasha B Leighl
- Division of Medical Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
140
|
Shim HS, Choi YL, Kim L, Chang S, Kim WS, Roh MS, Kim TJ, Ha SY, Chung JH, Jang SJ, Lee GK. Molecular Testing of Lung Cancers. J Pathol Transl Med 2017; 51:242-254. [PMID: 28427247 PMCID: PMC5445209 DOI: 10.4132/jptm.2017.04.10] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 04/09/2017] [Indexed: 12/25/2022] Open
Abstract
Targeted therapies guided by molecular diagnostics have become a standard treatment of lung cancer. Epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) rearrangements are currently used as the best predictive biomarkers for EGFR tyrosine kinase inhibitors and ALK inhibitors, respectively. Besides EGFR and ALK, the list of druggable genetic alterations has been growing, including ROS1 rearrangements, RET rearrangements, and MET alterations. In this situation, pathologists should carefully manage clinical samples for molecular testing and should do their best to quickly and accurately identify patients who will benefit from precision therapeutics. Here, we grouped molecular biomarkers of lung cancers into three categories—mutations, gene rearrangements, and amplifications—and propose expanded guidelines on molecular testing of lung cancers.
Collapse
Affiliation(s)
- Hyo Sup Shim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Lucia Kim
- Department of Pathology, Inha University School of Medicine, Incheon, Korea
| | - Sunhee Chang
- Department of Pathology, Inje University Ilsan Paik Hospital, Inje University, Goyang, Korea
| | - Wan-Seop Kim
- Department of Pathology, Konkuk University School of Medicine, Seoul, Korea
| | - Mee Sook Roh
- Department of Pathology, Dong-A University College of Medicine, Busan, Korea
| | - Tae-Jung Kim
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung Yeon Ha
- Department of Pathology, Gachon University Gil Medical Center, Incheon, Korea
| | - Jin-Haeng Chung
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Se Jin Jang
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Geon Kook Lee
- Department of Pathology, National Cancer Center, Goyang, Korea
| | | | | |
Collapse
|
141
|
Yu W, Wang Y, Jiang Y, Zhang W, Li Y. Genetic analysis and clinicopathological features of ALK-rearranged renal cell carcinoma in a large series of resected Chinese renal cell carcinoma patients and literature review. Histopathology 2017; 71:53-62. [PMID: 28199742 DOI: 10.1111/his.13185] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 02/08/2017] [Indexed: 12/27/2022]
Abstract
AIMS Anaplastic lymphoma kinase (ALK)-rearranged renal cell carcinoma (RCC) is a rare subtype of RCC reported in recent years, with eight cases so far. The aims of the present study were to screen ALK-rearranged cases from a large cohort of RCCs in China to determine the frequency of ALK rearrangement and investigate the clinicopathological features and outcomes. METHODS AND RESULTS Tissues from a total of 477 RCC patients in China were embedded into tissue microarrays for immunostaining. Fluorescence in-situ hybridization (FISH) and fluorescence quantitative reverse transcription polymerase chain reaction (RT-PCR) were applied to identify and confirm the rearrangement of ALK, and to identify the genes fused with ALK. ALK expression was identified in two of 477 RCCs. By FISH analysis, the two tumours showed either a 1R1G1F or a 2R2G signal pattern, indicating rearrangement involving ALK. Fluorescence quantitative RT-PCR detected the TPM3-ALK fusion and EML4-ALK fusion transcripts in the two tumours, respectively. Follow-up data were analyzed for the two cases and eight other ALK-rearranged RCCs reported in the literature. Two patients died from RCCs, on the 16th month and 48th month after surgery, respectively. The 5-year cancer-specific survival rate of patients with the 10 ALK-rearranged RCCs was lower than that of patients with International Society of Urological Pathology (ISUP) G1, G2 and G3 clear cell RCC (CCRCC) and papillary RCC (PRCC), but higher than that of patients with G4 CCRCC and PRCC. CONCLUSIONS ALK-rearranged RCC is a rare subtype of adult RCC and is associated with distinct histological features and a poor prognosis. Identification of ALK-rearranged RCC has important clinical significance, because patients might benefit from ALK inhibitor therapy as used in lung adenocarcinoma.
Collapse
Affiliation(s)
- Wenjuan Yu
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuewei Wang
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanxia Jiang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wei Zhang
- Department of Pathology, 401 Hospital of People's Liberation Army, Qingdao, China
| | - Yujun Li
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
142
|
Li A, Yang JJ, Zhang XC, Zhang Z, Su J, Gou LY, Bai Y, Zhou Q, Yang Z, Han-Zhang H, Zhong WZ, Chuai S, Zhang Q, Xie Z, Gao H, Chen H, Wang Z, Wang Z, Yang XN, Wang BC, Gan B, Chen ZH, Jiang BY, Wu SP, Liu SY, Xu CR, Wu YL. Acquired MET Y1248H and D1246N Mutations Mediate Resistance to MET Inhibitors in Non-Small Cell Lung Cancer. Clin Cancer Res 2017; 23:4929-4937. [PMID: 28396313 DOI: 10.1158/1078-0432.ccr-16-3273] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 12/20/2016] [Accepted: 04/04/2017] [Indexed: 11/16/2022]
Abstract
Purpose:MET amplification, responsible for 20% of acquired resistance to EGFR tyrosine kinase inhibitor (TKI) in patients with advanced non-small cell lung cancer (NSCLC), presents an attractive target. Numerous studies have conferred susceptibility of MET mutations and focal amplification to targeted MET-TKIs. However, the mechanism underlying MET-TKIs-induced resistance remains elusive.Experimental Design: We conducted a cohort of 12 patients with advanced NSCLC who developed resistance to a combinatorial therapy consisting of gefitinib and a type I MET-TKI. We performed capture-based targeted ultra-deep sequencing on serial tumor biopsies and plasmas ctDNA samples to detect and quantify genetic alterations.Results: We identified 2 newly acquired MET mutations, Y1248H and D1246N, in 2 patients and further confirmed their resistance against type I MET-TKIs in silco, in vitro, and in vivo Interestingly, NIH3T3 cells harboring either mutation exhibited responses to type II MET-TKIs, suggesting sequential use of MET-TKIs may offer a more durable response. In addition, we also discovered that EGFR amplification may act as an alternative MET-TKI resistance mechanism.Conclusions: Our study provides insight into the diversity of mechanisms underlying MET-TKI-induced resistance and highlights the potential of sequential use of MET-TKIs. Clin Cancer Res; 23(16); 4929-37. ©2017 AACR.
Collapse
Affiliation(s)
- Anna Li
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Xu-Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Zhou Zhang
- Burning Rock Biotech, Guangzhou, Guangdong Province, P.R. China
| | - Jian Su
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Lan-Ying Gou
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Yu Bai
- Asia Innovative Medicines and Early Development, AstraZeneca, Shanghai, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Zhenfan Yang
- Asia Innovative Medicines and Early Development, AstraZeneca, Shanghai, China
| | - Han Han-Zhang
- Burning Rock Biotech, Guangzhou, Guangdong Province, P.R. China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Shannon Chuai
- Burning Rock Biotech, Guangzhou, Guangdong Province, P.R. China
| | - Qi Zhang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Zhi Xie
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Hongfei Gao
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Huajun Chen
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Zhen Wang
- Asia Innovative Medicines and Early Development, AstraZeneca, Shanghai, China
| | - Zheng Wang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Xue-Ning Yang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Bin-Chao Wang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Bin Gan
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Zhi-Hong Chen
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Ben-Yuan Jiang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Si-Pei Wu
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Si-Yang Liu
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Chong-Rui Xu
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China.
| |
Collapse
|
143
|
Calvayrac O, Pradines A, Pons E, Mazières J, Guibert N. Molecular biomarkers for lung adenocarcinoma. Eur Respir J 2017; 49:49/4/1601734. [PMID: 28381431 DOI: 10.1183/13993003.01734-2016] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 12/28/2016] [Indexed: 12/28/2022]
Abstract
The identification of oncogenic driver alterations that underlie sensitivity to small inhibitors has led to growing interest in identifying additional targetable oncogenes in nonsmall cell lung cancer. Although the therapeutic impact of the discovery of these alterations has now been widely demonstrated, the epidemiological data associated with each of these biomarkers remain insufficiently studied. In this review, we discuss the techniques used to discover each of these candidate oncogenes, their prevalence in nonsmall cell lung cancer, and briefly outline the epidemiological features of the major oncogenes and ways in which their identification can determine therapeutic strategies.
Collapse
Affiliation(s)
- Olivier Calvayrac
- Inserm, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France
| | - Anne Pradines
- Inserm, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France.,Institut Universitaire du Cancer, Toulouse, France.,Institut Claudius Regaud, IUCT-Oncopole, Laboratoire de Biologie Médicale Oncologique, Toulouse, France
| | - Elvire Pons
- Inserm, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France.,Institut Claudius Regaud, IUCT-Oncopole, Laboratoire de Biologie Médicale Oncologique, Toulouse, France
| | - Julien Mazières
- Inserm, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France .,Institut Universitaire du Cancer, Toulouse, France.,Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier, Toulouse, France
| | - Nicolas Guibert
- Inserm, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France.,Institut Universitaire du Cancer, Toulouse, France.,Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
144
|
Qian M, Zhu B, Wang X, Liebman M. Drug resistance in ALK-positiveNon-small cell lungcancer patients. Semin Cell Dev Biol 2017; 64:150-157. [DOI: 10.1016/j.semcdb.2016.09.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 09/28/2016] [Indexed: 02/07/2023]
|
145
|
MET exon 14 skipping mutation in triple-negative pulmonary adenocarcinomas and pleomorphic carcinomas: An analysis of intratumoral MET status heterogeneity and clinicopathological characteristics. Lung Cancer 2017; 106:131-137. [DOI: 10.1016/j.lungcan.2017.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/13/2016] [Accepted: 02/10/2017] [Indexed: 11/15/2022]
|
146
|
Soldera SV, Leighl NB. Update on the Treatment of Metastatic Squamous Non-Small Cell Lung Cancer in New Era of Personalized Medicine. Front Oncol 2017; 7:50. [PMID: 28396848 PMCID: PMC5366319 DOI: 10.3389/fonc.2017.00050] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/09/2017] [Indexed: 12/26/2022] Open
Abstract
Despite advances in molecular characterization and lung cancer treatment in recent years, treatment options for patients diagnosed with squamous cell carcinoma of the lung (SCC) remain limited as actionable mutations are rarely detected in this subtype. This article reviews potential molecular targets and associated novel agents for the treatment of advanced SCC in the era of personalized medicine. Elements of various pathways including epidermal growth factor receptor, PI3KCA, fibroblast growth factor receptor, retinoblastoma, cyclin-dependent kinases, discoidin domain receptor tyrosine kinase 2, and mesenchymal-to-epithelial transition may play pivotal roles in the development of SCC and are under investigation for drug development.
Collapse
Affiliation(s)
| | - Natasha B. Leighl
- Division of Medical Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
147
|
MET Gene Amplification and Overexpression in Chinese Non–Small-Cell Lung Cancer Patients Without EGFR Mutations. Clin Lung Cancer 2017; 18:213-219.e2. [DOI: 10.1016/j.cllc.2016.09.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/26/2016] [Accepted: 09/06/2016] [Indexed: 11/18/2022]
|
148
|
Rashdan S, Gerber DE. A crowded, but still varied, space: brigatinib in anaplastic lymphoma kinase-rearranged non-small cell lung cancer. Transl Cancer Res 2017; 6:S78-S82. [PMID: 28680831 DOI: 10.21037/tcr.2017.02.12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Sawsan Rashdan
- Department of Internal Medicine (Division of Hematology-Oncology), Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center. Dallas, Texas. USA
| | - David E Gerber
- Department of Internal Medicine (Division of Hematology-Oncology), Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center. Dallas, Texas. USA.,Department of Clinical Sciences, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center. Dallas, Texas. USA
| |
Collapse
|
149
|
Ivanov M, Laktionov K, Breder V, Chernenko P, Novikova E, Telysheva E, Musienko S, Baranova A, Mileyko V. Towards standardization of next-generation sequencing of FFPE samples for clinical oncology: intrinsic obstacles and possible solutions. J Transl Med 2017; 15:22. [PMID: 28137276 PMCID: PMC5282851 DOI: 10.1186/s12967-017-1125-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 01/19/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Next generation sequencing has a potential to revolutionize the management of cancer patients within the framework of precision oncology. Nevertheless, lack of standardization decelerated entering of the technology into the clinical testing space. Here we dissected a number of common problems of NGS diagnostics in oncology and introduced ways they can be resolved. METHODS DNA was extracted from 26 formalin fixed paraffin embedded (FFPE) specimens and processed with the TrueSeq Amplicon Cancer Panel (Illumina Inc, San Diego, California) targeting 48 cancer-related genes and sequenced in single run. Sequencing data were comparatively analyzed by several bioinformatics pipelines. RESULTS Libraries yielded sufficient coverage to detect even low prevalent mutations. We found that the number of FFPE sequence artifacts significantly correlates with pre-normalization concentration of libraries (rank correlation -0.81; p < 1e-10), thus, contributing to sample-specific variant detection cut-offs. Surprisingly, extensive validation of EGFR mutation calls by a combination of aligners and variant callers resulted in identification of two false negatives and one false positive that were due to complexity of underlying genomic change, confirmed by Sanger sequencing. Additionally, the study of the non-EGFR amplicons revealed 33 confirmed unique mutations in 17 genes, with TP53 being the most frequently mutated. Clinical relevance of these finding is discussed. CONCLUSIONS Reporting of entire mutational spectrum revealed by targeted sequencing is questionable, at least until the clinically-driven guidelines on reporting of somatic mutations are established. The standardization of sequencing protocols, especially their data analysis components, requires assay-, disease-, and, in many cases, even sample-specific customization that could be performed only in cooperation with clinicians.
Collapse
Affiliation(s)
- Maxim Ivanov
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Moscow Region, 141700 Russia
- Atlas Biomed Group, Moscow, 121069 Russia
- Institute of Chemical Biology and Fundamental Medicine of SB RAS, Novosibirsk, 630090 Russia
| | - Konstantin Laktionov
- N.N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow, 115478 Russia
| | - Valery Breder
- N.N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow, 115478 Russia
| | - Polina Chernenko
- N.N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow, 115478 Russia
| | - Ekaterina Novikova
- Federal State Budgetary Institution Russian Scientific Center of Roentgenoradiology (RSCRR) of the Ministry of Healthcare of the Russian Federation (Russian Scientific Center of Roentgenoradiology), Moscow, 117485 Russia
| | - Ekaterina Telysheva
- Federal State Budgetary Institution Russian Scientific Center of Roentgenoradiology (RSCRR) of the Ministry of Healthcare of the Russian Federation (Russian Scientific Center of Roentgenoradiology), Moscow, 117485 Russia
| | | | - Ancha Baranova
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Moscow Region, 141700 Russia
- Atlas Biomed Group, Moscow, 121069 Russia
- Research Centre for Medical Genetics, Moscow, 115478 Russia
- Center for the Study of Chronic Metabolic and Rare Diseases, School of System Biology, George Mason University, Fairfax, VA USA
| | - Vladislav Mileyko
- Atlas Biomed Group, Moscow, 121069 Russia
- Institute of Chemical Biology and Fundamental Medicine of SB RAS, Novosibirsk, 630090 Russia
| |
Collapse
|
150
|
张 权, 张 树. [Research Progress of Targeted Therapy for Anaplastic Lymphoma Kinase and Other Rare Driver Genes in Advanced Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2017; 20:66-72. [PMID: 28103976 PMCID: PMC5973289 DOI: 10.3779/j.issn.1009-3419.2017.01.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 11/11/2016] [Accepted: 11/12/2016] [Indexed: 11/05/2022]
Abstract
Targeted therapy was one of the major treatments in advanced non-small cell lung cancer (NSCLC) with positive driver genes. This area of research progresses day by day, with novel target discoveries, novel drug development, and use of novel combination treatments. Researchers have also undergone deep investigation about the molecular mechanisms underlying inherent or acquired resistance to these targeted therapies. This review aimed to summarize the advanced developments of targeted therapy for anaplastic lymphoma kinase (ALK) and other rare driver genes in NSCLC.
Collapse
Affiliation(s)
- 权 张
- />101149 北京,首都医科大学附属北京胸科医院肿瘤内科Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - 树才 张
- />101149 北京,首都医科大学附属北京胸科医院肿瘤内科Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| |
Collapse
|