151
|
Wang M, Zhao F, Li Z, Li X, Dong L. Tectoridin and PLK1 inhibitor synergistically promote the apoptosis of lung adenocarcinoma cells: Bioinformatic analysis of TCGA and TCMSP. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2417-2426. [PMID: 37014402 DOI: 10.1007/s00210-023-02460-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/08/2023] [Indexed: 04/05/2023]
Abstract
Lung cancer is still the most common cancer in the world, especially lung adenocarcinoma (LUAD). Despite years of effort, including the application of immunotherapy and targeted therapy, the survival rate of LUAD has not improved significantly. Exploring effective targets and combination drugs is crucial for the treatment of LUAD. We characterized differentially expressed genes between LUAD and normal lung tissue based on The Cancer Genome Atlas (TCGA) database and identified polo-like kinase 1 (PLK1) as the hub gene. Through an analysis using the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP), we obtained a combination of Chinese medicine with PLK1 inhibitor, whose biological function we confirmed by western blot and TdT-UTP nick-end labelling (TUNEL) assays. After combined analysis of protein expression with clinical characteristics, GNPNAT1, CCT6A, SMOX, UCK2, PLK1, HMMR and ANLN expression were significantly correlated with age, sex and stage. Among them, the survival rate was lower in patients with high PLK1 expression than in those with low PLK1 expression, making PLK1 a promising therapeutic target for LUAD. Stage and PLK1 expression could be used as independent prognostic factors for LUAD. By TCMSP analysis, tectoridin had the strongest correlation with PLK1. Tectoridin synergized with PLK1 inhibitor to suppress autophagy and ferroptosis but promoted caspase-3-mediated apoptosis in A549 cells. Our findings highlight a potential drug target and the combination therapy strategy of PLK1 inhibitor and tectoridin for LUAD patients.
Collapse
Affiliation(s)
- Meng Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China
- Department of Respiratory, Binzhou Central Hospital, Binzhou, China
| | - Fei Zhao
- Department of Endocrinology, Binzhou Central Hospital, Binzhou, China
| | - Zhishu Li
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China
| | - Xin Li
- Department of Respiratory, Binzhou Central Hospital, Binzhou, China
| | - Lixia Dong
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China.
| |
Collapse
|
152
|
Kajita Y, Teranishi S, Sawazumi T, Watanabe H, Nagaoka S, Tanaka A, Suzukawa Y, Motobayashi Y, Hirose T, Maeda C, Seki K, Tashiro K, Kobayashi N, Yamamoto M, Kudo M, Inayama Y, Kaneko T. Comparison of the slow-pull and aspiration methods of endobronchial ultrasound-guided transbronchial needle aspiration for next-generation sequencing-compatible tissue collection in non-small cell lung cancer. Cancer Med 2023; 12:19512-19522. [PMID: 37732488 PMCID: PMC10587950 DOI: 10.1002/cam4.6561] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/07/2023] [Accepted: 09/09/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Personalized treatment for non-small cell lung cancer (NSCLC) has advanced rapidly, and elucidating the genetic changes that trigger this disease is crucial for appropriate treatment selection. Both slow-pull and aspiration methods of endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) are accepted methods for collecting samples suitable for next-generation sequencing (NGS) to examine driver gene mutations and translocations in NSCLC. Here, we aimed to determine which of these two methods is superior for obtaining higher-quality samples from patients with NSCLC. METHODS Seventy-one patients diagnosed with NSCLC via EBUS-TBNA using the slow-pull or aspiration (20-mL negative pressure) methods between July 2019 and September 2022 were included. A total of 203 tissue samples from the 71 patients were fixed in formalin, embedded in paraffin, and mounted on slides. The presence of tissue cores, degree of blood contamination, and number of tumor cells were compared between the groups. The success rate of NGS, using Oncomine Dx Target Test Multi-CDx, was also compared between the groups. RESULTS The slow-pull method was associated with a higher yield of tissue cores, lower degree of blood contamination, and higher number of tumor cells than the aspiration method. The success rate of the NGS was also significantly higher for the slow-pull group (95%) than for the aspiration group (68%). CONCLUSION Overall, these findings suggest that the slow-pull method is a superior technique for EBUS-TBNA to obtain high-quality tissue samples for NGS. The slow-pull method may contribute to the identification of driver gene mutations and translocations and facilitate personalized treatment of NSCLC.
Collapse
Affiliation(s)
- Yukihito Kajita
- Respiratory Disease CenterYokohama City University Medical CenterYokohamaJapan
| | - Shuhei Teranishi
- Respiratory Disease CenterYokohama City University Medical CenterYokohamaJapan
| | - Tomoe Sawazumi
- Division of PathologyYokohama City University Medical CenterYokohamaJapan
| | - Haruka Watanabe
- Respiratory Disease CenterYokohama City University Medical CenterYokohamaJapan
| | - Satoshi Nagaoka
- Respiratory Disease CenterYokohama City University Medical CenterYokohamaJapan
| | - Anna Tanaka
- Respiratory Disease CenterYokohama City University Medical CenterYokohamaJapan
| | - Yuichirou Suzukawa
- Respiratory Disease CenterYokohama City University Medical CenterYokohamaJapan
| | - Yuto Motobayashi
- Respiratory Disease CenterYokohama City University Medical CenterYokohamaJapan
| | - Tomofumi Hirose
- Respiratory Disease CenterYokohama City University Medical CenterYokohamaJapan
| | - Chihiro Maeda
- Respiratory Disease CenterYokohama City University Medical CenterYokohamaJapan
| | - Kenichi Seki
- Respiratory Disease CenterYokohama City University Medical CenterYokohamaJapan
| | - Ken Tashiro
- Respiratory Disease CenterYokohama City University Medical CenterYokohamaJapan
| | - Nobuaki Kobayashi
- Department of PulmonologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Masaki Yamamoto
- Respiratory Disease CenterYokohama City University Medical CenterYokohamaJapan
| | - Makoto Kudo
- Respiratory Disease CenterYokohama City University Medical CenterYokohamaJapan
| | - Yoshiaki Inayama
- Division of PathologyYokohama City University Medical CenterYokohamaJapan
| | - Takeshi Kaneko
- Department of PulmonologyYokohama City University Graduate School of MedicineYokohamaJapan
| |
Collapse
|
153
|
Lau SC, Perdrizet K, Fung AS, Mata DGM, Weiss J, Holzapfel N, Liu G, Bradbury PA, Shepherd FA, Sacher AG, Feilotter H, Sheffield B, Hwang D, Tsao MS, Cheng S, Cheema P, Leighl NB. Programmed Cell Death Protein 1 Inhibitors and MET Targeted Therapies in NSCLC With MET Exon 14 Skipping Mutations: Efficacy and Toxicity as Sequential Therapies. JTO Clin Res Rep 2023; 4:100562. [PMID: 37744308 PMCID: PMC10514105 DOI: 10.1016/j.jtocrr.2023.100562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/10/2023] [Accepted: 08/12/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction NSCLC with MET exon 14 skipping mutation (METex14) is associated with poor outcomes. Integration of novel targeted therapies is challenging because of barriers in testing and drug access. We, therefore, sought to characterize the treatment patterns, outcomes, and emerging issues of treatment sequencing in patients with METex14-mutant NSCLC. Methods We reviewed all NSCLC cases with METex14 alterations between 2014 and 2020 across four Canadian cancer centers. Demographics, disease characteristics, systemic therapy, overall response rates (ORRs), survival, and toxicity were summarized. Results Among 64 patients with METex14-mutant NSCLC, the median overall survival was 23.1 months: 127.0 months in stage 1, 27.3 months in resected stage 2 and 3, and 16.6 months in unresectable stage 3 or 4 disease, respectively. In patients with advanced disease, 22% were too unwell for systemic treatment. MET tyrosine kinase inhibitors (TKIs) were administered to 28 patients with an ORR of 33%, median progression-free survival of 2.7 months, and 3.8 months for selective TKIs. Programmed cell death protein-1 (PD-1) inhibitors were given to 25 patients-the ORR was 44% and progression-free survival was 10.6 months. No responses were seen with subsequent MET TKIs after initial TKI treatment. Grade 3 or higher toxicities occurred in 64% of patients who received MET TKI after PD-1 inhibitors versus 8% in those who did not receive PD-1 inhibitors. Conclusions Many patients with advanced METex14 NSCLC were too unwell to receive treatment. PD-1 inhibitors seem effective as an initial treatment, although greater toxicity was seen with subsequent MET TKIs. Thus, timely testing for METex14 skipping and initial therapy are imperative to improving patient survival.
Collapse
Affiliation(s)
- Sally C.M. Lau
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University (NYU) Langone Health, NYU Grossman School of Medicine, New York, New York
| | - Kirstin Perdrizet
- William Osler Health System, Brampton Civic Hospital, Brampton Ontario, Canada
| | - Andrea S. Fung
- Department of Medical Oncology, Cancer Centre of Southeastern Ontario, Kingston Health Sciences Centre, Queen’s University, Kingston, Ontario, Canada
| | - Danilo Giffoni M.M. Mata
- Department of Medical Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Jessica Weiss
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Nick Holzapfel
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Geoffrey Liu
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Penelope A. Bradbury
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Frances A. Shepherd
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Adrian G. Sacher
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Harriet Feilotter
- Department of Pathology, Cancer Centre of Southeastern Ontario, Kingston Health Sciences Centre, Queen’s University, Kingston, Ontario, Canada
| | - Brandon Sheffield
- William Osler Health System, Brampton Civic Hospital, Brampton Ontario, Canada
| | - David Hwang
- Department of Pathology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Ming Sound Tsao
- Department of Pathology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Susanna Cheng
- Department of Medical Oncology, Cancer Centre of Southeastern Ontario, Kingston Health Sciences Centre, Queen’s University, Kingston, Ontario, Canada
| | - Parneet Cheema
- William Osler Health System, Brampton Civic Hospital, Brampton Ontario, Canada
| | - Natasha B. Leighl
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
154
|
Balnis J, Lauria EJM, Yucel R, Singer HA, Alisch RS, Jaitovich A. Peripheral Blood Omics and Other Multiplex-based Systems in Pulmonary and Critical Care Medicine. Am J Respir Cell Mol Biol 2023; 69:383-390. [PMID: 37379507 PMCID: PMC10557924 DOI: 10.1165/rcmb.2023-0153ps] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/28/2023] [Indexed: 06/30/2023] Open
Abstract
Over the last years, the use of peripheral blood-derived big datasets in combination with machine learning technology has accelerated the understanding, prediction, and management of pulmonary and critical care conditions. The goal of this article is to provide readers with an introduction to the methods and applications of blood omics and other multiplex-based technologies in the pulmonary and critical care medicine setting to better appreciate the current literature in the field. To accomplish that, we provide essential concepts needed to rationalize this approach and introduce readers to the types of molecules that can be obtained from the circulating blood to generate big datasets; elaborate on the differences between bulk, sorted, and single-cell approaches; and the basic analytical pipelines required for clinical interpretation. Examples of peripheral blood-derived big datasets used in recent literature are presented, and limitations of that technology are highlighted to qualify both the current and future value of these methodologies.
Collapse
Affiliation(s)
- Joseph Balnis
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Eitel J. M. Lauria
- School of Computer Science and Mathematics, Marist College, Poughkeepsie, New York
| | - Recai Yucel
- Department of Epidemiology and Biostatistics, Temple University, Philadelphia, Pennsylvania; and
| | - Harold A. Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Reid S. Alisch
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| |
Collapse
|
155
|
Spitaleri G, Trillo Aliaga P, Attili I, Del Signore E, Corvaja C, Corti C, Uliano J, Passaro A, de Marinis F. MET in Non-Small-Cell Lung Cancer (NSCLC): Cross 'a Long and Winding Road' Looking for a Target. Cancers (Basel) 2023; 15:4779. [PMID: 37835473 PMCID: PMC10571577 DOI: 10.3390/cancers15194779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Non-Small-Cell Lung Cancer (NSCLC) can harbour different MET alterations, such as MET overexpression (MET OE), MET gene amplification (MET AMP), or MET gene mutations. Retrospective studies of surgical series of patients with MET-dysregulated NSCLC have shown worse clinical outcomes irrespective of the type of specific MET gene alteration. On the other hand, earlier attempts failed to identify the 'druggable' molecular gene driver until the discovery of MET exon 14 skipping mutations (METex14). METex14 are rare and amount to around 3% of all NSCLCs. Patients with METex14 NSCLC attain modest results when they are treated with immune checkpoint inhibitors (ICIs). New selective MET inhibitors (MET-Is) showed a long-lasting clinical benefit in patients with METex14 NSCLC and modest activity in patients with MET AMP NSCLC. Ongoing clinical trials are investigating new small molecule tyrosine kinase inhibitors, bispecific antibodies, or antibodies drug conjugate (ADCs). This review focuses on the prognostic role of MET, the summary of pivotal clinical trials of selective MET-Is with a focus on resistance mechanisms. The last section is addressed to future developments and challenges.
Collapse
Affiliation(s)
- Gianluca Spitaleri
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| | - Pamela Trillo Aliaga
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| | - Ilaria Attili
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| | - Ester Del Signore
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| | - Carla Corvaja
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| | - Chiara Corti
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (C.C.); (J.U.)
- Department of Oncology and Haematology (DIPO), University of Milan, 20122 Milan, Italy
| | - Jacopo Uliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (C.C.); (J.U.)
- Department of Oncology and Haematology (DIPO), University of Milan, 20122 Milan, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| | - Filippo de Marinis
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| |
Collapse
|
156
|
Hashimoto T, Owada Y, Katagiri H, Yakuwa K, Tyo K, Sugai M, Fuzimura I, Utsumi Y, Akiyama M, Nagashima H, Terasaki H, Yanagawa N, Saito H, Sugai T, Maemondo M. Characteristics and prognostic analysis of patients with detected KRAS mutations in resected lung adenocarcinomas by peptide nucleic acid-locked nucleic acid polymerase chain reaction (PNA-LNA PCR) clamp method. Transl Lung Cancer Res 2023; 12:1862-1875. [PMID: 37854155 PMCID: PMC10579836 DOI: 10.21037/tlcr-23-15] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 08/03/2023] [Indexed: 10/20/2023]
Abstract
Background Kirsten rat sarcoma virus (KRAS) gene mutations are a type of driver mutation discovered in the 1980s, but for a long time no molecular targeted drugs were available for them. Recently, sotorasib was developed as a molecular targeted drug for KRAS mutations. It is therefore necessary to identify the characteristics of patients with KRAS mutations. Methods This was the single-institution retrospective study. Surgically resected tumors from lung adenocarcinoma patients were collected at a single institution from June 2016 to September 2019. Peptide nucleic acid-locked nucleic acid polymerase chain reaction (PNA-LNA PCR) clamp analysis of KRAS G12X mutations was compared with analysis by therascreen KRAS RGQ kit. The association between KRAS mutation status and patient characteristics and prognosis was assessed. Results Among 499 lung adenocarcinomas, KRAS mutations were evaluated in 197 cases, excluding stage IV lung cancer and tumors with epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) mutations. KRAS G12X mutations were detected in 59 cases (29.9%). The highest frequency by gene mutation subtype was G12V in 23 cases (39.0%), followed by G12C in 16 cases (27.1%), G12D in 12 cases (20.3%), G12S in 4 cases (6.8%) and G12A in 2 cases. For the G12C mutation, the PNA-LNA PCR clamp and therascreen methods were consistent, but for the G12D and G12S mutations, the PNA-LNA PCR clamp method showed higher detection rates. In operable tumors, G12C mutations were more frequent in males, smokers, and patients with high expression of programmed death-ligand 1 (PD-L1), and had no correlation with prognosis. Conclusions By the PNA-LNA PCR clamp method, G12C mutation of surgical specimens was detected successfully. The PNA-LNA PCR clamp method is expected to be applied to the detection of druggable G12C mutations.
Collapse
Affiliation(s)
- Tatsuya Hashimoto
- Division of Pulmonary Medicine, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Yoshihisa Owada
- Division of Pulmonary Medicine, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Hiroshi Katagiri
- Division of Pulmonary Medicine, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Kazuhiro Yakuwa
- Division of Pulmonary Medicine, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Katuya Tyo
- Division of Pulmonary Medicine, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Mayu Sugai
- Division of Pulmonary Medicine, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Itaru Fuzimura
- Division of Pulmonary Medicine, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Yu Utsumi
- Division of Pulmonary Medicine, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Masachika Akiyama
- Division of Pulmonary Medicine, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Hiromi Nagashima
- Division of Pulmonary Medicine, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Hiroshi Terasaki
- Medical Solution Segment, Advanced Technology Center, Genome Analysis Department, LSI Medience Corporation, Tokyo, Japan
| | - Naoki Yanagawa
- Department of Molecular Diagnostic Pathology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Hajime Saito
- Division of Thoracic Surgery, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Tamotsu Sugai
- Department of Molecular Diagnostic Pathology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Makoto Maemondo
- Division of Pulmonary Medicine, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| |
Collapse
|
157
|
Sasaki A, Fujimoto Y, Inada T, Ishizuka A, Ehara J, Ogita S, Norisue Y. Efficacy of tyrosine kinase inhibitors in patients with non-small-cell lung cancer with performance status 4: a case series and review of the literature. J Med Case Rep 2023; 17:410. [PMID: 37759318 PMCID: PMC10538240 DOI: 10.1186/s13256-023-04145-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Current guidelines for non-small-cell lung cancer (NSCLC) recommend that each tyrosine kinase inhibitor (TKI) is indicated even for driver mutation-positive patients with a poor performance status (PS). In previous studies, most patients had a PS of 2-3, but those with a PS of 4 were very few. Therefore, the efficacy of TKIs in patients with NSCLC with a PS of 4 remains unclear. CASE PRESENTATION We retrospectively reviewed the clinical records of four patients with NSCLC with PS 4 treated with TKIs: an 89-year-old Japanese woman (Case 1), a 80-year-old Japanese woman (Case 2), an 50-year-old Japanese man (Case 3), and a 81-year-old Japanese woman (Case 4). Genetic alterations were epidermal growth factor receptor (EGFR), MET exon 14 skipping, BRAFV600E, and ROS1 proto-oncogene receptor tyrosine kinase (ROS1). One case with ROS1 fusion showed a significant response with the recovery of PS. However, in the remaining three cases (i.e., EGFR, MET exon 14 skipping, and BRAFV600E mutations), patients died despite the administration of TKIs. These three patients had to be hospitalized at the end of their life to receive treatment. CONCLUSIONS This is the first case series to summarize the efficacy of TKIs in patients with NSCLC with a PS of 4. Additionally, this case series poses a question concerning the indication of TKIs for older patients with a PS of 4.
Collapse
Affiliation(s)
- Akinori Sasaki
- Department of Oncology, Tokyo Bay Urayasu Ichikawa Medical Center, 3-4-32 Toudaijima, Urayasu, Chiba, 279-0001, Japan.
| | - Yutaro Fujimoto
- Department of Pulmonology, Tokyo Bay Urayasu Ichikawa Medical Center, 3-4-32 Toudaijima, Urayasu, Chiba, 279-0001, Japan
| | - Takashi Inada
- Department of Pulmonology, Tokyo Bay Urayasu Ichikawa Medical Center, 3-4-32 Toudaijima, Urayasu, Chiba, 279-0001, Japan
| | - Azusa Ishizuka
- Department of Pulmonology, Tokyo Bay Urayasu Ichikawa Medical Center, 3-4-32 Toudaijima, Urayasu, Chiba, 279-0001, Japan
| | - Jun Ehara
- Department of Pulmonology, Tokyo Bay Urayasu Ichikawa Medical Center, 3-4-32 Toudaijima, Urayasu, Chiba, 279-0001, Japan
| | - Shin Ogita
- Department of Oncology, St. Luke's International Hospital, 9-1 Akashi-Cho, Chuo, Tokyo, 104-8560, Japan
| | - Yasuhiro Norisue
- Department of Pulmonology, Tokyo Bay Urayasu Ichikawa Medical Center, 3-4-32 Toudaijima, Urayasu, Chiba, 279-0001, Japan
| |
Collapse
|
158
|
Altintas DM, Comoglio PM. An Observatory for the MET Oncogene: A Guide for Targeted Therapies. Cancers (Basel) 2023; 15:4672. [PMID: 37760640 PMCID: PMC10526818 DOI: 10.3390/cancers15184672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
The MET proto-oncogene encodes a pivotal tyrosine kinase receptor, binding the hepatocyte growth factor (HGF, also known as scatter factor, SF) and governing essential biological processes such as organogenesis, tissue repair, and angiogenesis. The pleiotropic physiological functions of MET explain its diverse role in cancer progression in a broad range of tumors; genetic/epigenetic alterations of MET drive tumor cell dissemination, metastasis, and acquired resistance to conventional and targeted therapies. Therefore, targeting MET emerged as a promising strategy, and many efforts were devoted to identifying the optimal way of hampering MET signaling. Despite encouraging results, however, the complexity of MET's functions in oncogenesis yields intriguing observations, fostering a humbler stance on our comprehension. This review explores recent discoveries concerning MET alterations in cancer, elucidating their biological repercussions, discussing therapeutic avenues, and outlining future directions. By contextualizing the research question and articulating the study's purpose, this work navigates MET biology's intricacies in cancer, offering a comprehensive perspective.
Collapse
Affiliation(s)
| | - Paolo M. Comoglio
- IFOM ETS—The AIRC Institute of Molecular Oncology, 20139 Milano, Italy;
| |
Collapse
|
159
|
LI S, ZHANG X. [Drug Resistance Mechanism and Therapeutic Strategy of Targeted Therapy of
Non-small Cell Lung Cancer with MET Alterations]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2023; 26:684-691. [PMID: 37985154 PMCID: PMC10600752 DOI: 10.3779/j.issn.1009-3419.2023.102.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Indexed: 11/22/2023]
Abstract
Mesenchymal to epithelial transition factor (MET) gene alterations involve in the proliferation, invasion, and metastasis of non-small cell lung cancer. MET-tyrosine kinase inhibitors (TKIs) have been approved to treat non-small cell lung cancer with MET alterations, and resistance to these TKIs is inevitable. Molecular mechanisms of resistance to MET-TKIs are completely unclear. The review focused on potential mechanisms of MET-TKIs resistance and therapeutics strategies to delay and prevent resistance.
.
Collapse
|
160
|
Kumaki Y, Oda G, Ikeda S. Targeting MET Amplification: Opportunities and Obstacles in Therapeutic Approaches. Cancers (Basel) 2023; 15:4552. [PMID: 37760522 PMCID: PMC10526812 DOI: 10.3390/cancers15184552] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/01/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
The MET gene plays a vital role in cellular proliferation, earning it recognition as a principal oncogene. Therapies that target MET amplification have demonstrated promising results both in preclinical models and in specific clinical cases. A significant obstacle to these therapies is the ability to distinguish between focal amplification and polysomy, a task for which simple MET copy number measurement proves insufficient. To effectively differentiate between the two, it is crucial to utilize comparative measures, including in situ hybridization (ISH) with the centromere or next generation sequencing (NGS) with adjacent genes. Despite the promising potential of MET amplification treatment, the judicious selection of patients is paramount to maximize therapeutic efficacy. The effectiveness of MET inhibitors can fluctuate depending on the extent of MET amplification. Future research must seek to establish the ideal threshold value for MET amplification, identify the most efficacious combination therapies, and innovate new targeted treatments for patients exhibiting MET amplification.
Collapse
Affiliation(s)
- Yuichi Kumaki
- Department of Specialized Surgery, Tokyo Medical and Dental University, Tokyo 113-8519, Japan;
| | - Goshi Oda
- Department of Specialized Surgery, Tokyo Medical and Dental University, Tokyo 113-8519, Japan;
| | - Sadakatsu Ikeda
- Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
161
|
McLaughlin J, Berkman J, Nana-Sinkam P. Targeted therapies in non-small cell lung cancer: present and future. Fac Rev 2023; 12:22. [PMID: 37675274 PMCID: PMC10477963 DOI: 10.12703/r/12-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023] Open
Abstract
Lung cancer is the leading cause of malignancy-related death in the United States and the second most common cancer diagnosis worldwide. In the last two decades, lung cancer treatment has evolved to include advances in the development of mutation-based targeting, immunotherapy, radiation therapy, and minimally invasive surgical techniques. The discovery of lung cancer as a molecularly heterogeneous disease has driven investigation into the development of targeted therapies resulting in improved patient outcomes. Despite these advances, there remain opportunities, through further investigation of mechanisms of resistance, to develop novel therapeutics that better direct the personalization of lung cancer therapy. In this review, we highlight developments in the evolution of targeted therapies in non-small cell lung cancer, as well as future directions shaped by emerging patterns of resistance.
Collapse
Affiliation(s)
- Jessica McLaughlin
- Department of Internal Medicine, Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University Health System, Richmond, VA 23298
| | - Jonathan Berkman
- Department of Internal Medicine, Division of Hematology, Oncology and Palliative Care, Virginia Commonwealth University Health System, Richmond, VA 23298
| | - Patrick Nana-Sinkam
- Department of Internal Medicine, Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University Health System, Richmond, VA 23298
| |
Collapse
|
162
|
Choi HY, Chang JE. Targeted Therapy for Cancers: From Ongoing Clinical Trials to FDA-Approved Drugs. Int J Mol Sci 2023; 24:13618. [PMID: 37686423 PMCID: PMC10487969 DOI: 10.3390/ijms241713618] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/23/2023] [Accepted: 09/02/2023] [Indexed: 09/10/2023] Open
Abstract
The development of targeted therapies has revolutionized cancer treatment, offering improved efficacy with reduced side effects compared with traditional chemotherapy. This review highlights the current landscape of targeted therapy in lung cancer, colorectal cancer, and prostate cancer, focusing on key molecular targets. Moreover, it aligns with US Food and Drug Administration (FDA)-approved drugs and drug candidates. In lung cancer, mutations in the epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) gene rearrangements have emerged as significant targets. FDA-approved drugs like osimertinib and crizotinib specifically inhibit these aberrant pathways, providing remarkable benefits in patients with EGFR-mutated or ALK-positive lung cancer. Colorectal cancer treatment has been shaped by targeting the vascular endothelial growth factor (VEGF) and EGFR. Bevacizumab and cetuximab are prominent FDA-approved agents that hinder VEGF and EGFR signaling, significantly enhancing outcomes in metastatic colorectal cancer patients. In prostate cancer, androgen receptor (AR) targeting is pivotal. Drugs like enzalutamide, apalutamide, and darolutamide effectively inhibit AR signaling, demonstrating efficacy in castration-resistant prostate cancer. This review further highlights promising targets like mesenchymal-epithelial transition (MET), ROS1, BRAF, and poly(ADP-ribose) polymeras (PARP) in specific cancer subsets, along with ongoing clinical trials that continue to shape the future of targeted therapy.
Collapse
Affiliation(s)
| | - Ji-Eun Chang
- College of Pharmacy, Dongduk Women’s University, Seoul 02748, Republic of Korea
| |
Collapse
|
163
|
Tamura T, Hata S, Baba T, Koyanagi T, Umeno T, Nishii K, Kuyama S. A case of successful desensitization treatment with tepotinib after tepotinib-induced rash. Respir Med Case Rep 2023; 45:101911. [PMID: 37706029 PMCID: PMC10495622 DOI: 10.1016/j.rmcr.2023.101911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/06/2023] [Accepted: 08/21/2023] [Indexed: 09/15/2023] Open
Abstract
Tepotinib is one of the key drugs for MET exon 14-skipping mutation-positive non-small cell lung cancer (NSCLC). The main adverse event of tepotinib treatment is edema. Rash is a rare adverse event, affecting only 0.7% of patients. We report a case of successful desensitization after skin rash caused by tepotinib. A 61-year-old male was treated with tepotinib 500 mg as second-line therapy for NSCLC with MET exon 14-skipping mutation. Treatment was discontinued on day 12 due to grade 3 erythema throughout the body. After improvement of the skin rash, he was started on 250 mg tepotinib with an oral antihistamine and topical steroid. Treatment was discontinued on day 11 due to skin rash exacerbation. One month of treatment-free follow-up showed skin rash improvement but lung carcinoma growth. Tepotinib desensitization therapy was started at a dose of 12.5 mg and gradually increased to 250 mg/day. The patient has since continued tepotinib treatment without skin rashes. Desensitization therapy may be effective for managing skin rash due to tepotinib.
Collapse
Affiliation(s)
- Tomoki Tamura
- Department of Respiratory Medicine, National Hospital Organization, Iwakuni Clinical Center, Iwakuni, Japan
| | - Sakura Hata
- Department of Respiratory Medicine, National Hospital Organization, Iwakuni Clinical Center, Iwakuni, Japan
| | - Takahiro Baba
- Department of Respiratory Medicine, National Hospital Organization, Iwakuni Clinical Center, Iwakuni, Japan
- Department of Internal Medicine, National Hospital Organization, Otake Medical Center, Otake, Japan
| | - Taisaku Koyanagi
- Department of Respiratory Medicine, National Hospital Organization, Iwakuni Clinical Center, Iwakuni, Japan
| | - Takahiro Umeno
- Department of Respiratory Medicine, National Hospital Organization, Iwakuni Clinical Center, Iwakuni, Japan
| | - Kazuya Nishii
- Department of Respiratory Medicine, National Hospital Organization, Iwakuni Clinical Center, Iwakuni, Japan
| | - Shoichi Kuyama
- Department of Respiratory Medicine, National Hospital Organization, Iwakuni Clinical Center, Iwakuni, Japan
| |
Collapse
|
164
|
Lam M, Olivier T, Haslam A, Tuia J, Prasad V. Cost of Drug Wastage From Dose Modification and Discontinuation of Oral Anticancer Drugs. JAMA Oncol 2023; 9:1238-1244. [PMID: 37471095 PMCID: PMC10360002 DOI: 10.1001/jamaoncol.2023.2306] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/03/2023] [Indexed: 07/21/2023]
Abstract
Importance Oral chemotherapy is often dispensed to patients as a 1-month supply, with pill dose and package size predetermined by the drug manufacturer; thus, changing the patient dosage may waste the remaining initial drug supply. The cost of pills wasted due to dose modification and discontinuation is often unreported. Objective To estimate the cost of pill wastage due to dose modification and discontinuation for oral anticancer drugs that were recently approved by the US Food and Drug Administration (FDA) or that are commonly prescribed. Design, Setting, and Participants This retrospective cross-sectional economic evaluation initially identified 26 oral anticancer drugs newly approved between January 1, 2020, and August 31, 2022, from the FDA website and the top 50 best-selling pharmaceuticals in 2021 abstracted from the Drug Discovery Trends website managed by Drug Discovery and Development. The monthly costs of each agent were extracted from the Micromedex RED BOOK database. The FDA package insert, and in some cases PubMed, of each identified drug and indication was searched (matching on trial registration number) for information on registration trials. Information extracted for each drug included the name of the drug approved, drug target, cost of the drug, number of pills per bottle, available strengths, indication, name of the trial, number of patients exposed to treatment drug, number of dose level reductions, median duration of treatment, percentage of patients who received dose reduction, and percentage of dose discontinuation. All variables included in calculations were derived from the package insert or original trial publication. Main Outcomes and Measures The cost of wastage for selected oral anticancer drugs due to dose reduction or discontinuation and the percentage of wastage in comparison with the total cost of treatment. Results After removing duplicates, 22 oral anticancer medications were included in the study. Because some drugs had more than 1 indication, data from 35 clinical trials were analyzed. Eight of the medications (covering 9 indications) had pill strengths divisible at each dose-reduction level; thus the cost of reduction for these pills was assumed to be zero. Two medications did not allow for dose reduction. The median cost of wastage from dose reduction and discontinuation was $1750 (range, $43-$27 200), with a mean cost of $4290 (SD, $5720) per patient. The median percentage of wastage from the total cost of treatment was 1.04% (range, 0.04%-10.80%) with a mean of 1.78% (SD, 2.21%). Conclusions and Relevance This economic evaluation found that due to both the high cost per pill and limited pill strength availability, the mean cost of wastage associated with dose reduction or discontinuation was $4290 per patient. These results suggest that to reduce the financial burden for patients with cancer, regulatory bodies should enforce availability of pill strengths that will limit pill wastage during dose modification or recommend that drug manufacturers issue credit for unused pills.
Collapse
Affiliation(s)
- Michael Lam
- School of Pharmacy, University of California, San Diego, La Jolla
| | - Timothée Olivier
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Alyson Haslam
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Jordan Tuia
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Vinay Prasad
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| |
Collapse
|
165
|
Sisi M, Vitale G, Fusaroli M, Riefolo M, Giunchi V, D’Errico A, Ardizzoni A, Raschi E, Gelsomino F. Capmatinib-Induced Liver Injury as Emerging Toxicity of MET Inhibitors in Patients With NSCLC Pretreated With Immune Checkpoint Inhibitors. JTO Clin Res Rep 2023; 4:100563. [PMID: 37745898 PMCID: PMC10511802 DOI: 10.1016/j.jtocrr.2023.100563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/31/2023] [Accepted: 08/12/2023] [Indexed: 09/26/2023] Open
Abstract
Safety data on MET inhibitors in patients with advanced NSCLC harboring MET exon 14 mutation and treated with frontline immune checkpoint inhibitors (ICIs) are still limited. Here, we describe clinical characteristics, liver biopsy features, and management of liver injury of two patients with a diagnosis of MET exon 14-mutant NSCLC receiving capmatinib after ICI failure. On the basis of histologic findings and exclusion of other potential causes, a diagnosis of drug-induced liver injury (DILI) associated with portal fibrosis was made in both cases. The use of hepatoprotective drugs, in addition to oral ursodeoxycholic acid, resulted in liver blood tests normalization. To provide a global safety perspective, we queried the Food and Drug Administration Adverse Event Reporting System and detected a robust disproportionality signal. Out of the 918 total reports with capmatinib from the Food and Drug Administration Adverse Event Reporting System database, DILI was recorded in 43 cases (4.7%), mostly serious (93.0%) with hospitalization and death recorded in 25.6% and 16.3% of the cases, respectively. The median time to onset was 42 days, with discontinuation and positive dechallenge documented in 41.9% and 39.5% of the cases, respectively. Anti-programmed cell death protein-1 agents were coreported in 11 DILI cases. Only two cases of DILI out of 105 reports were found for tepotinib. Our data support a potential association between capmatinib and DILI in patients who have also been previously exposed to immunotherapy. Considering the potential implications for sequence strategy and timing of ICI and MET inhibitor, further investigation is warranted.
Collapse
Affiliation(s)
- Monia Sisi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giovanni Vitale
- Internal Medicine Unit for the Treatment of Severe Organ Failure, Istituto di Ricovero e Cura a Carattere (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Michele Fusaroli
- Pharmacology Unit, DIMEC, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Mattia Riefolo
- Department of Pathology, Istituto di Ricovero e Cura a Carattere (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Valentina Giunchi
- Pharmacology Unit, DIMEC, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Antonietta D’Errico
- Department of Pathology, Istituto di Ricovero e Cura a Carattere (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea Ardizzoni
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Emanuel Raschi
- Pharmacology Unit, DIMEC, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Francesco Gelsomino
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
166
|
Hatswell AJ. Incorporating Prior Beliefs Into Meta-Analyses of Health-State Utility Values Using the Bayesian Power Prior. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:1389-1397. [PMID: 37187235 DOI: 10.1016/j.jval.2023.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/17/2023] [Accepted: 04/28/2023] [Indexed: 05/17/2023]
Abstract
OBJECTIVES Health-state utility values (HSUVs) directly affect estimates of Quality-Adjusted Life-Years and thus the cost-utility estimates. In practice a single preferred value (SPV) is often selected for HSUVs, despite meta-analysis being an option when multiple (credible) HSUVs are available. Nevertheless, the SPV approach is often reasonable because meta-analysis implicitly considers all HSUVs as equally relevant. This article presents a method for the incorporation of weights to HSUV synthesis, allowing more relevant studies to have greater influence. METHODS Using 4 case studies in lung cancer, hemodialysis, compensated liver cirrhosis, and diabetic retinopathy blindness, a Bayesian Power Prior (BPP) approach is used to incorporate beliefs on study applicability, reflecting the authors' perceived suitability for UK decision making. Older studies, non-UK value sets, and vignette studies are thus downweighted (but not disregarded). BPP HSUV estimates were compared with a SPV, random effects meta-analysis, and fixed effects meta-analysis. Sensitivity analyses were conducted iteratively updating the case studies, using alternative weighting methods, and simulated data. RESULTS Across all case studies, SPVs did not accord with meta-analyzed values, and fixed effects meta-analysis produced unrealistically narrow CIs. Point estimates from random effects meta-analysis and BPP models were similar in the final models, although BPP reflected additional uncertainty as wider credible intervals, particularly when fewer studies were available. Differences in point estimates were seen in iterative updating, weighting approaches, and simulated data. CONCLUSIONS The concept of the BPP can be adapted for synthesizing HSUVs, incorporating expert opinion on relevance. Because of the downweighting of studies, the BPP reflected structural uncertainty as wider credible intervals, with all forms of synthesis showing meaningful differences compared with SPVs. These differences would have implications for both cost-utility point estimates and probabilistic analyses.
Collapse
Affiliation(s)
- Anthony J Hatswell
- Delta Hat Limited, Nottingham, England, UK; Department of Statistical Science, University College London, London, England, UK.
| |
Collapse
|
167
|
Mazieres J, Vioix H, Pfeiffer BM, Campden RI, Chen Z, Heeg B, Cortot AB. MET Exon 14 Skipping in NSCLC: A Systematic Literature Review of Epidemiology, Clinical Characteristics, and Outcomes. Clin Lung Cancer 2023; 24:483-497. [PMID: 37451931 DOI: 10.1016/j.cllc.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/30/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION MET exon 14 (METex14) skipping is a rare oncogenic driver in non-small-cell lung cancer (NSCLC) for which targeted therapy with MET tyrosine kinase inhibitors (TKIs) was recently approved. Given the heterogeneity in published data of METex14 skipping NSCLC, we conducted a systematic literature review to evaluate its frequency, patient characteristics, and outcomes. METHODS On June 13, 2022 we conducted a systematic literature review of publications and conference abstracts reporting frequency, patient characteristics, or outcomes of patients with METex14 skipping NSCLC. RESULTS We included 139 studies reporting frequency or patient characteristics (350,997 patients), and 39 studies reporting clinical outcomes (3989 patients). Median METex14 skipping frequency was 2.0% in unselected patients with NSCLC, with minimal geographic variation. Median frequency was 2.4% in adenocarcinoma or nonsquamous subgroups, 12.0% in sarcomatoid, and 1.3% in squamous histology. Patients with METex14 skipping NSCLC were more likely to be elderly, have adenocarcinoma histology; there was no marked sex or smoking status distribution. In first line of treatment, median objective response rate ranged from 50.7% to 68.8% with targeted therapies (both values correspond to MET TKIs), was 33.3% with immunotherapy, and ranged from 23.1% to 27.0% with chemotherapy. CONCLUSIONS Patients with METex14 skipping are more likely to have certain characteristics, but no patient subgroup can be ruled out; thus, it is crucial to test all patients with NSCLC to identify suitable candidates for MET inhibitor therapy. MET TKIs appeared to result in higher efficacy outcomes, although no direct comparison with chemotherapy or immunotherapy regimens was found.
Collapse
Affiliation(s)
- Julien Mazieres
- CHU de Toulouse, Université Paul Sabatier, Toulouse, France.
| | - Helene Vioix
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | | | | - Zhiyuan Chen
- Ingress Health, A Cytel Company, Rotterdam, The Netherlands
| | - Bart Heeg
- Ingress Health, A Cytel Company, Rotterdam, The Netherlands
| | - Alexis B Cortot
- Université Lille, Centre Hospitalier Universitaire de Lille, Centre national de la recherche scientifique, Inserm, Institute Pasteur de Lille, Lille, France
| |
Collapse
|
168
|
Mazieres J, Paik PK, Garassino MC, Le X, Sakai H, Veillon R, Smit EF, Cortot AB, Raskin J, Viteri S, Wu YL, Yang JCH, Ahn MJ, Ma R, Zhao J, O’Brate A, Berghoff K, Bruns R, Otto G, Johne A, Felip E, Thomas M. Tepotinib Treatment in Patients With MET Exon 14-Skipping Non-Small Cell Lung Cancer: Long-term Follow-up of the VISION Phase 2 Nonrandomized Clinical Trial. JAMA Oncol 2023; 9:1260-1266. [PMID: 37270698 PMCID: PMC10240398 DOI: 10.1001/jamaoncol.2023.1962] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 04/10/2023] [Indexed: 06/05/2023]
Abstract
Importance MET inhibitors have recently demonstrated clinical activity in patients with MET exon 14 (METex14)-skipping non-small cell lung cancer (NSCLC); however, data with longer follow-up and in larger populations are needed to further optimize therapeutic approaches. Objective To assess the long-term efficacy and safety of tepotinib, a potent and highly selective MET inhibitor, in patients with METex14-skipping NSCLC in the VISION study. Design, Setting, and Participants The VISION phase 2 nonrandomized clinical trial was a multicohort, open-label, multicenter study that enrolled patients with METex14-skipping advanced/metastatic NSCLC (cohorts A and C) from September 2016 to May 2021. Cohort C (>18 months' follow-up) was an independent cohort, designed to confirm findings from cohort A (>35 months' follow-up). Data cutoff was November 20, 2022. Intervention Patients received tepotinib, 500 mg (450 mg active moiety), once daily. Main Outcomes and Measures The primary end point was objective response by independent review committee (RECIST v1.1). Secondary end points included duration of response (DOR), progression-free survival (PFS), overall survival (OS), and safety. Results Cohorts A and C included 313 patients (50.8% female, 33.9% Asian; median [range] age, 72 [41-94] years). The objective response rate (ORR) was 51.4% (95% CI, 45.8%-57.1%) with a median (m)DOR of 18.0 (95% CI, 12.4-46.4) months. In cohort C (n = 161), an ORR of 55.9% (95% CI, 47.9%-63.7%) with an mDOR of 20.8 (95% CI, 12.6-not estimable [NE]) months was reported across treatment lines, comparable to cohort A (n = 152). In treatment-naive patients (cohorts A and C; n = 164), ORR was 57.3% (95% CI, 49.4%-65.0%) and mDOR was 46.4 (95% CI, 13.8-NE) months. In previously treated patients (n = 149), ORR was 45.0% (95% CI, 36.8%-53.3%) and mDOR was 12.6 (95% CI, 9.5-18.5) months. Peripheral edema, the most common treatment-related adverse event, occurred in 210 patients (67.1%) (35 [11.2%] experienced grade ≥3 events). Conclusions and Relevance The findings from cohort C in this nonrandomized clinical trial supported the results from original cohort A. Overall, the long-term outcomes of VISION demonstrated robust and durable clinical activity following treatment with tepotinib, particularly in the treatment-naive setting, in the largest known clinical trial of patients with METex14-skipping NSCLC, supporting the global approvals of tepotinib and enabling clinicians to implement this therapeutic approach for such patients. Trial Registration ClinicalTrials.gov Identifier: NCT02864992.
Collapse
Affiliation(s)
| | - Paul K. Paik
- Thoracic Oncology Service, Memorial Sloan-Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Marina C. Garassino
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Now with Department of Medicine, Section of Hematology/Oncology, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, Illinois
| | - Xiuning Le
- Department of Thoracic Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Hiroshi Sakai
- Department of Thoracic Oncology, Saitama Cancer Center, Kitaadachi-gun, Japan
- Now with Department of Thoracic Oncology, Ageo Central General Hospital, Saitama, Japan
| | - Remi Veillon
- CHU Bordeaux, service des maladies respiratoires, Bordeaux, France
| | - Egbert F. Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Now with Department of Pulmonary Diseases, Leiden University Medical Centre, Leiden, the Netherlands
| | - Alexis B. Cortot
- Univ. Lille, CHU Lille, CNRS, Inserm, Institut Pasteur de Lille, UMR9020 – UMR-S 1277 - Canther, Lille, France
| | - Jo Raskin
- Department of Pulmonology and Thoracic Oncology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Santiago Viteri
- Instituto Oncológico Dr. Rosell, Hospital Universitario Dexeus, Grupo Quiron Salud, Barcelona, Spain
- Now with UOMI cancer center, Clínica Mi NovAliança, Lleida, Spain
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - James C. H. Yang
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Myung-Ju Ahn
- Division of Hematology Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Rui Ma
- Medical Oncology Department of Thoracic Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Jun Zhao
- Department of Thoracic Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Aurora O’Brate
- Global Medical Affairs, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Karin Berghoff
- Global Patient Safety, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Rolf Bruns
- Department of Biostatistics, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Gordon Otto
- Global Clinical Development, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Andreas Johne
- Global Clinical Development, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Enriqueta Felip
- Department of Oncology, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Michael Thomas
- Thoraxklinik and National Center for Tumor Diseases at Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
169
|
Elkrief A, Odintsov I, Markov V, Caeser R, Sobczuk P, Tischfield SE, Bhanot U, Vanderbilt CM, Cheng EH, Drilon A, Riely GJ, Lockwood WW, de Stanchina E, Tirunagaru VG, Doebele RC, Quintanal-Villalonga Á, Rudin CM, Somwar R, Ladanyi M. Combination Therapy With MDM2 and MEK Inhibitors Is Effective in Patient-Derived Models of Lung Adenocarcinoma With Concurrent Oncogenic Drivers and MDM2 Amplification. J Thorac Oncol 2023; 18:1165-1183. [PMID: 37182602 PMCID: PMC10524759 DOI: 10.1016/j.jtho.2023.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/01/2023] [Accepted: 05/08/2023] [Indexed: 05/16/2023]
Abstract
INTRODUCTION Although targeted therapies have revolutionized the therapeutic landscape of lung adenocarcinomas (LUADs), disease progression on single-agent targeted therapy against known oncogenic drivers is common, and therapeutic options after disease progression are limited. In patients with MDM2 amplification (MDM2amp) and a concurrent oncogenic driver alteration, we hypothesized that targeting of the tumor-suppressor pathway (by means of restoration of p53 using MDM2 inhibition) and simultaneous targeting of co-occurring MAPK oncogenic pathway might represent a more durably effective therapeutic strategy. METHODS We evaluated genomic next-generation sequencing data using the Memorial Sloan Kettering Cancer Center-Integrated Mutation Profiling of Actionable Cancer Targets platform to nominate potential targets for combination therapy in LUAD. We investigated the small molecule MDM2 inhibitor milademetan in cell lines and patient-derived xenografts of LUAD with a known driver alteration and MDM2amp. RESULTS Of 10,587 patient samples from 7121 patients with LUAD profiled by next-generation sequencing, 6% (410 of 7121) harbored MDM2amp. MDM2amp was significantly enriched among tumors with driver alterations in METex14 (36%, p < 0.001), EGFR (8%, p < 0.001), RET (12%, p < 0.01), and ALK (10%, p < 0.01). The combination of milademetan and the MEK inhibitor trametinib was synergistic in growth inhibition of ECLC5-GLx (TRIM33-RET/MDM2amp), LUAD12c (METex14/KRASG12S/MDM2amp), SW1573 (KRASG12C, TP53 wild type), and A549 (KRASG12S) cells and in increasing expression of proapoptotic proteins PUMA and BIM. Treatment of ECLC5-GLx and LUAD12c with single-agent milademetan increased ERK phosphorylation, consistent with previous data on ERK activation with MDM2 inhibition. This ERK activation was effectively suppressed by concomitant administration of trametinib. In contrast, ERK phosphorylation induced by milademetan was not suppressed by concurrent RET inhibition using selpercatinib (in ECLC5-GLx) or MET inhibition using capmatinib (in LUAD12c). In vivo, combination milademetan and trametinib was more effective than either agent alone in ECLC5-GLx, LX-285 (EGFRex19del/MDM2amp), L13BS1 (METex14/MDM2amp), and A549 (KRASG12S, TP53 wild type). CONCLUSIONS Combined MDM2/MEK inhibition was found to have efficacy across multiple patient-derived LUAD models harboring MDM2amp and concurrent oncogenic drivers. This combination, potentially applicable to LUADs with a wide variety of oncogenic driver mutations and kinase fusions activating the MAPK pathway, has evident clinical implications and will be investigated as part of a planned phase 1/2 clinical trial.
Collapse
Affiliation(s)
- Arielle Elkrief
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Igor Odintsov
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vladimir Markov
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rebecca Caeser
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pawel Sobczuk
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sam E Tischfield
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Umesh Bhanot
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chad M Vanderbilt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Emily H Cheng
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Gregory J Riely
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - William W Lockwood
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | | | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Romel Somwar
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
170
|
Gu L, Zhao Y, Wen F, Zhang D, Cai J, Chen Z. A durable response to savolitinib in a patient with lung adenocarcinoma harboring two novel MET exon 14 skipping sites. Anticancer Drugs 2023; 34:949-953. [PMID: 36846984 DOI: 10.1097/cad.0000000000001495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
MET exon 14 ( METex14 ) skipping variants are oncogenic drivers in non-small-cell lung cancer. Several METex14 skipping alterations have been identified, but different mesenchymal-epithelial transition (MET) exon splicing variants tend to present different clinical outcomes. Here, we reported that a patient with lung adenocarcinoma harbored two novel METex14 skipping mutations (c.2888-35_2888-16del and c.2888-4T>G) identified by the tissue-based next-generation sequencing (NGS) and received savolitinib treatment after chemotherapy failed with brain metastasis. The patient responded well to savolitinib until disease progression in brain lesions and achieved a progress-free survival (PFS) of over 19.7 months. Considering the durable response for extracranial lesions and the same METex14 skipping sites identified by circulating tumor DNA-based NGS, the patient was still given savolitinib combined with stereotactic body radiation therapy for brain lesions. An extracranial PFS of 28 months was achieved. This is the first report of a patient with lung adenocarcinoma harboring two novel METex14 skipping mutations that responded to the MET inhibitor savolitinib. Our case may provide evidence for the treatment of patients with two novel METex14 skipping variants and offer a therapy regimen for those with intracranial progression.
Collapse
Affiliation(s)
- Linping Gu
- Department of Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai
| | - Yi Zhao
- Department of Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai
| | - Fengcai Wen
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Ding Zhang
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Jinping Cai
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Zhiwei Chen
- Department of Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai
| |
Collapse
|
171
|
Li MSC, Mok KKS, Mok TSK. Developments in targeted therapy & immunotherapy-how non-small cell lung cancer management will change in the next decade: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:358. [PMID: 37675321 PMCID: PMC10477626 DOI: 10.21037/atm-22-4444] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 03/08/2023] [Indexed: 09/08/2023]
Abstract
Background and Objective The adoption of targeted therapy and immunotherapy has revolutionised the treatment landscape of non-small cell lung cancer. For early staged disease, incorporation of targeted therapy and immunotherapy has recently been demonstrated to reduce recurrence. Development of targeted therapies in advanced lung cancer is driven by advanced genomic sequencing techniques, better understanding of drug resistance mechanisms, and improved drug designs. The list of targetable molecular alteration is continuously expanding, and next generation molecular therapies have shown promise in circumventing drug resistance. Lung cancer patients may achieve durable disease control with immune checkpoint inhibitors however most patients develop immunotherapy resistance. A wide spectrum of resistance mechanisms, ranging from impaired T-cell activation, presence of coinhibitory immune checkpoints, to immunosuppressive tumour microenvironment, have been proposed. A multitude of novel immunotherapy strategies are under development to target such resistance mechanisms. This review aims to provide a succinct overview in the latest development in targeted therapy and immunotherapy for NSCLC management. Methods We searched all original papers and reviews on targeted therapy and immunotherapy in non-small cell lung cancer (NSCLC) using PubMed in June 2022. Search terms included "non-small cell lung cancer", "targeted therapy", "immunotherapy", "EGFR", "ALK", "ROS1", "BRAF V600E", "MET", "RET", "KRAS", "HER2", "ERBB2", "NRG1", "immune checkpoint", "PD-1", "PD-L1", "CTLA4", "TIGIT", "VEGF", "cancer vaccine", "cellular therapy", "tumour microenvironment", "cytokine", and "gut microbiota". Key Content and Findings We first discuss the incorporation of targeted therapy and immunotherapy in early staged NSCLC. This includes the latest clinical data that led to the approval of neoadjuvant immunotherapy, adjuvant immunotherapy and adjuvant targeted therapy for early staged NSCLC. The second section focuses on targeted therapy in metastatic NSCLC. The list of targetable alteration now includes but is not limited to EGFR, ALK, ROS1, BRAF V600E, MET exon 14 skipping, RET, KRAS G12C, HER2 and NRG1. Potential drug resistance mechanisms and novel therapeutics under development are also discussed. The third section on immunotherapy in metastatic NSCLC, covers immunotherapy that are currently approved [anti-PD-(L)1 and anti-CTLA4], and agents that are under active research (e.g., anti-TIGIT, cancer vaccine, cellular therapy, cytokine and other TME modulating agents). Conclusions This review encompasses the latest updates in targeted therapy and immunotherapy in lung cancer management and discusses the future direction in the field.
Collapse
Affiliation(s)
- Molly S. C. Li
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Kevin K. S. Mok
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, China
| | - Tony S. K. Mok
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
172
|
Kujtan L, Subramanian J. Telisotuzumab vedotin with erlotinib in the treatment of non-small cell lung cancer: a well MET combination? Transl Lung Cancer Res 2023; 12:1826-1829. [PMID: 37691875 PMCID: PMC10483073 DOI: 10.21037/tlcr-23-288] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/07/2023] [Indexed: 09/12/2023]
Affiliation(s)
- Lara Kujtan
- Richard and Annette Bloch Cancer Center, University of Missouri at Kansas City, Kansas City, MO, USA
| | | |
Collapse
|
173
|
Gilson P, Pouget C, Belmonte R, Fadil S, Demange J, Rouyer M, Lacour J, Betz M, Dardare J, Witz A, Merlin JL, Harlé A. Validation of the Idylla GeneFusion assay to detect fusions and MET exon-skipping in non-small cell lung cancers. Sci Rep 2023; 13:12909. [PMID: 37558711 PMCID: PMC10412571 DOI: 10.1038/s41598-023-39749-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/30/2023] [Indexed: 08/11/2023] Open
Abstract
Gene fusions and MET exon skipping drive oncogenesis in 8-9% and 3% of non-small cell lung cancers (NSCLC) respectively. Their detection are essential for the management of patients since they confer sensitivity to specific targeted therapies with significant clinical benefit over conventional chemotherapy. Immunohistochemistry (IHC) and fluorescent in situ hybridization (FISH) account for historical reference techniques however molecular-based technologies (RNA-based sequencing and RT-PCR) are emerging as alternative or complementary methods. Here, we evaluated the analytical performance of the fully-automated RT-PCR Idylla GeneFusion assay compared to reference methods using 35 fixed NSCLC samples. Idylla demonstrated overall agreement, sensitivity and specificity of 100% compared to RNASeq. Interestingly, it succeeded in retrieving 10 out of 11 samples with inconclusive results due to insufficient RNA quality for sequencing. Idylla showed an overall agreement, sensitivity and specificity of 90.32%, 91.67% and 89.47% compared to IHC/FISH respectively. Using commercial standards, the limit of detection of the Idylla system for the most frequent fusions and exon skipping ranges between 5 and 10 ng RNA input. These results support that the Idylla assay is a reliable and rapid option for the detection of these alterations, however a particular attention is needed for the interpretation of the expression imbalance.
Collapse
Affiliation(s)
- Pauline Gilson
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France.
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France.
| | - Celso Pouget
- Service d'Anatomocytopathologie, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Richard Belmonte
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Smahane Fadil
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Jessica Demange
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Marie Rouyer
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Julien Lacour
- Service d'Anatomocytopathologie, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Margaux Betz
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Julie Dardare
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Andréa Witz
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Jean-Louis Merlin
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| | - Alexandre Harlé
- Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN, Université de Lorraine, 6 Avenue de Bourgogne, CS 30519, 54519, Vandœuvre-lès-Nancy Cedex, France
- Service de Biologie Moléculaire des Tumeurs, Département de Biopathologie, Institut de Cancérologie de Lorraine/CHRU Nancy, Rue du Morvan, 54511, Vandœuvre-lès-Nancy Cedex, France
| |
Collapse
|
174
|
Iwama E, Yamamoto H, Okubo F, Ijichi K, Ibusuki R, Shiaraishi Y, Yoneshima Y, Tanaka K, Oda Y, Okamoto I. Evaluation of appropriate conditions for Oncomine DxTT testing of FFPE specimens for driver gene alterations in non-small cell lung cancer. Thorac Cancer 2023; 14:2288-2296. [PMID: 37345344 PMCID: PMC10423657 DOI: 10.1111/1759-7714.15014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND The Oncomine Dx Target Test Multi-CDx System (ODxTT) is a next-generation sequencing panel approved as a companion diagnostic for drugs targeted to corresponding gene alterations in non-small cell lung cancer. However, appropriate slide conditions for ODxTT have remained unclear. METHODS We focused on the production of the number of tumor cells on a formalin-fixed paraffin-embedded (FFPE) section and the number of prepared slides, designated the TS value, and determined a TS value of ≥4000 as a target slide condition for ODxTT. We evaluated the impact of this condition on ODxTT testing with tumor specimens found to have a TS of <4000 (n = 23) or a TS of ≥4000 (n = 142). RESULTS A positive correlation was apparent between the TS value and the concentrations of both DNA and RNA. Among the 142 samples with a TS of ≥4000, a sufficient concentration of DNA or RNA for ODxTT analysis was achieved in 100% and 98% samples, respectively. Among samples explored for driver gene alterations after determination of the target slide condition (TS ≥4000), most (84.9%) had a TS of ≥4000 and were submitted for ODxTT analysis. CONCLUSION Our findings indicate that a TS of ≥4000 is a feasible and relevant criterion for ODxTT testing, and its adoption should help to improve the success rate of such testing in clinical practice.
Collapse
Affiliation(s)
- Eiji Iwama
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Diagnostic PathologyKyushu University HospitalFukuokaJapan
- Department of PathologyOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | - Fumihiko Okubo
- Division of Diagnostic PathologyKyushu University HospitalFukuokaJapan
| | - Kayo Ijichi
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Diagnostic PathologyKyushu University HospitalFukuokaJapan
| | - Ritsu Ibusuki
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yoshimasa Shiaraishi
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yasuto Yoneshima
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kentaro Tanaka
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Diagnostic PathologyKyushu University HospitalFukuokaJapan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
175
|
Yang M, Vioix H, Hook ES, Hatswell AJ, Batteson RL, Gaumond BR, O'Brate A, Popat S, Paik PK. Health Utility Analysis of Tepotinib in Patients With Non-Small Cell Lung Cancer Harboring MET Exon 14 Skipping. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:1155-1163. [PMID: 36805576 PMCID: PMC11145519 DOI: 10.1016/j.jval.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 06/18/2023]
Abstract
OBJECTIVES The VISION trial showed durable activity of tepotinib in MET exon 14 (METex14) skipping non-small cell lung cancer. We analyzed health state utilities using patient-reported outcomes from VISION. METHODS 5-level version of EQ-5D (EQ-5D-5L) and European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire-Core 30 responses were collected at baseline, every 6 to 12 weeks during treatment, and at the end of treatment and safety follow-up. EQ-5D-5L and European Organisation for Research and Treatment of Cancer Quality of Life Utility Measure-Core 10 Dimensions (QLU-C10D) utilities were derived using United States, Canada, United Kingdom, and Taiwan value sets, where available. Utilities were analyzed with linear mixed models including covariates for progression or time-to-death (TTD). RESULTS Utilities were derived for 273/291 patients (EQ-5D-5L, 1545 observations; QLU-C10D, 1546 observations). Mean (± SD) US EQ-5D-5L utilities increased after tepotinib initiation, from 0.687 ± 0.287 at baseline to 0.754 ± 0.250 before independently assessed progression, and decreased post progression (0.704 ± 0.288). US QLU-C10D utilities showed similar trends (0.705 ± 0.215, 0.753 ± 0.195, and 0.708 ± 0.209, respectively). Progression-based models demonstrated a statistically significant impact of progression on utilities and predicted higher utilities pre versus post progression. TTD-based models showed statistically significant associations of TTD with utilities and predicted declining utilities as TTD decreased. Prior treatment (yes/no) did not significantly predict utilities in progression- or TTD-based models. Utilities for Canada, United Kingdom, and Taiwan showed comparable trends. CONCLUSIONS In this first analysis of health state utilities in patients with METex14 skipping non-small cell lung cancer, who received tepotinib, utilities were significantly associated with progression and TTD, but not prior treatment.
Collapse
Affiliation(s)
- Mo Yang
- EMD Serono, Rockland, MA, USA.
| | - Helene Vioix
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | | | | | | | | - Aurora O'Brate
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | | - Paul K Paik
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
176
|
Sumiyoshi A, Fujii H, Okuma Y. Targeting microbiome, drug metabolism, and drug delivery in oncology. Adv Drug Deliv Rev 2023; 199:114902. [PMID: 37263544 DOI: 10.1016/j.addr.2023.114902] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 05/13/2023] [Accepted: 05/24/2023] [Indexed: 06/03/2023]
Abstract
Recent emerging scientific evidence shows a relationship between gut microbiota (GM) and immunomodulation. In the recently published "Hallmarks of Cancer", the microbiome has been reported to play a crucial role in cancer research, and perspectives for its clinical implementation to improve the effectiveness of pharmacotherapy were explored. Several studies have shown that GM can affect the outcomes of pharmacotherapy in cancer, suggesting that GM may affect anti-tumor immunity. Thus, studies on GM that analyze big data using computer-based analytical methods are required. In order to successfully deliver GM to an environment conducive to the proliferation of immune cells both within and outside the tumor microenvironment (TME), it is crucial to address a variety of challenges associated with distinct delivery methods, specifically those pertaining to oral, endoscopic, and intravenous delivery. Clinical trials are in progress to evaluate the effects of targeting GM and whether it can enhance immunity or act on the TME, thereby to improve the clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Ai Sumiyoshi
- Department of Pharmacy, National Cancer Center Hospital 5-1-1 Tsukiji Chuo, Tokyo 104-0045, Japan
| | - Hiroyuki Fujii
- Department of Thoracic Oncology, National Cancer Center Hospital 5-1-1 Tsukiji Chuo, Tokyo 104-0045, Japan; Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo, Kyoto 602-8566, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology, National Cancer Center Hospital 5-1-1 Tsukiji Chuo, Tokyo 104-0045, Japan.
| |
Collapse
|
177
|
Wang J, Yao W, Wang W, Fan M, Huang K, Liu Z, Zhu D. Complete pathological response and negative postoperative ctDNA were not predictive of discontinuation of adjuvant crizotinib therapy in a patient with locally advanced MET ex14 skipping mutation-positive non-small cell lung cancer: a case report. Front Oncol 2023; 13:1164543. [PMID: 37554169 PMCID: PMC10405726 DOI: 10.3389/fonc.2023.1164543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/05/2023] [Indexed: 08/10/2023] Open
Abstract
Neoadjuvant targeted therapy is an alternative treatment for locally advanced non-small cell lung cancer (NSCLC) patients with driver gene mutation. MET ex14 mutation is considered a driver gene, and crizotinib is the first oral tyrosine kinase inhibitor (TKI) for metastatic MET ex14 mutation-positive NSCLC patients. Here, we reported a case of a locally advanced NSCLC patient harboring MET ex14 mutation who achieved pathological complete response following neoadjuvant crizotinib therapy but developed rapid metastasis due to discontinuation of short-term postoperative adjuvant crizotinib therapy. Although no driver gene mutation was found via next-generation sequencing (NGS) with blood samples before discontinuation of adjuvant crizotinib, the patient was given crizotinib rechallenge. Fortunately, the patient achieved durable complete response. This suggested that neither pathological complete response nor negative circulating tumor DNA (ctDNA) could be an effective predictor for discontinuation of adjuvant targeted therapy. This case report demonstrated the potential of crizotinib as neoadjuvant therapy in MET ex14 mutation-positive NSCLC patients as well as the importance of long-term postoperative therapy even with negative ctDNA in blood.
Collapse
Affiliation(s)
- Jiantao Wang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenqing Yao
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weiya Wang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingyu Fan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kaili Huang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenkun Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Daxing Zhu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
178
|
Reischmann N, Schmelas C, Molina-Vila MÁ, Jordana-Ariza N, Kuntze D, García-Roman S, Simard MA, Musch D, Esdar C, Albers J, Karachaliou N. Overcoming MET-mediated resistance in oncogene-driven NSCLC. iScience 2023; 26:107006. [PMID: 37534190 PMCID: PMC10391663 DOI: 10.1016/j.isci.2023.107006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/20/2023] [Accepted: 05/26/2023] [Indexed: 08/04/2023] Open
Abstract
This study evaluates the efficacy of combining targeted therapies with MET or SHP2 inhibitors to overcome MET-mediated resistance in different NSCLC subtypes. A prevalence study was conducted for MET amplification and overexpression in samples from patients with NSCLC who relapsed on ALK, ROS1, or RET tyrosine kinase inhibitors. MET-mediated resistance was detected in 37.5% of tissue biopsies, which allow the detection of MET overexpression, compared to 7.4% of liquid biopsies. The development of drug resistance by MET overexpression was confirmed in EGFRex19del-, KRASG12C-, HER2ex20ins-, and TPM3-NTRK1-mutant cell lines. The combination of targeted therapy with MET or SHP2 inhibitors was found to overcome MET-mediated resistance in both in vitro and in vivo assays. This study highlights the importance of considering MET overexpression as a resistance driver to NSCLC targeted therapies to better identify patients who could potentially benefit from combination approaches with MET or SHP2 inhibitors.
Collapse
Affiliation(s)
| | | | | | | | - Daniel Kuntze
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | | | | - Doreen Musch
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | | - Joachim Albers
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | |
Collapse
|
179
|
Hsu R, Benjamin DJ, Nagasaka M. The Development and Role of Capmatinib in the Treatment of MET-Dysregulated Non-Small Cell Lung Cancer-A Narrative Review. Cancers (Basel) 2023; 15:3561. [PMID: 37509224 PMCID: PMC10377299 DOI: 10.3390/cancers15143561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of death, but over the past decade, there has been tremendous progress in the field with new targeted therapies. The mesenchymal-epithelial transition factor (MET) proto-oncogene has been implicated in multiple solid tumors, including NSCLC, and dysregulation in NSCLC from MET can present most notably as MET exon 14 skipping mutation and amplification. From this, MET tyrosine kinase inhibitors (TKIs) have been developed to treat this dysregulation despite challenges with efficacy and reliable biomarkers. Capmatinib is a Type Ib MET TKI first discovered in 2011 and was FDA approved in August 2022 for advanced NSCLC with MET exon 14 skipping mutation. In this narrative review, we discuss preclinical and early-phase studies that led to the GEOMETRY mono-1 study, which showed beneficial efficacy in MET exon 14 skipping mutations, leading to FDA approval of capmatinib along with Foundation One CDx assay as its companion diagnostic assay. Current and future directions of capmatinib are focused on improving the efficacy, overcoming the resistance of capmatinib, and finding approaches for new indications of capmatinib such as acquired MET amplification from epidermal growth factor receptor (EGFR) TKI resistance. Clinical trials now involve combination therapy with capmatinib, including amivantamab, trametinib, and immunotherapy. Furthermore, new drug agents, particularly antibody-drug conjugates, are being developed to help treat patients with acquired resistance from capmatinib and other TKIs.
Collapse
Affiliation(s)
- Robert Hsu
- Division of Medical Oncology, Department of Internal Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | | | - Misako Nagasaka
- Division of Hematology and Oncology, Department of Medicine, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA 92868, USA
| |
Collapse
|
180
|
Teh JH, Amgheib A, Fu R, Barnes C, Abrahams J, Ashek A, Wang N, Yang Z, Mansoorudeen M, Long NJ, Aboagye EO. Evaluation of [ 18F]AlF-EMP-105 for Molecular Imaging of C-Met. Pharmaceutics 2023; 15:1915. [PMID: 37514101 PMCID: PMC10383791 DOI: 10.3390/pharmaceutics15071915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
C-Met is a receptor tyrosine kinase that is overexpressed in a range of different cancer types, and has been identified as a potential biomarker for cancer imaging and therapy. Previously, a 68Ga-labelled peptide, [68Ga]Ga-EMP-100, has shown promise for imaging c-Met in renal cell carcinoma in humans. Herein, we report the synthesis and preliminary biological evaluation of an [18F]AlF-labelled analogue, [18F]AlF-EMP-105, for c-Met imaging by positron emission tomography. EMP-105 was radiolabelled using the aluminium-[18F]fluoride method with 46 ± 2% RCY and >95% RCP in 35-40 min. In vitro evaluation showed that [18F]AlF-EMP-105 has a high specificity for c-Met-expressing cells. Radioactive metabolite analysis at 5 and 30 min post-injection revealed that [18F]AlF-EMP-105 has good blood stability, but undergoes transformation-transchelation, defluorination or demetallation-in the liver and kidneys. PET imaging in non-tumour-bearing mice showed high radioactive accumulation in the kidneys, bladder and urine, demonstrating that the tracer is cleared predominantly as [18F]fluoride by the renal system. With its high specificity for c-Met expressing cells, [18F]AlF-EMP-105 shows promise as a potential diagnostic tool for imaging cancer.
Collapse
Affiliation(s)
- Jin Hui Teh
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
| | - Ala Amgheib
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Ruisi Fu
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Chris Barnes
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Joel Abrahams
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Ali Ashek
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Ning Wang
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Zixuan Yang
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Muneera Mansoorudeen
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Nicholas J Long
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
| | - Eric O Aboagye
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| |
Collapse
|
181
|
Wang C, Yuan X, Xue J. Targeted therapy for rare lung cancers: Status, challenges, and prospects. Mol Ther 2023; 31:1960-1978. [PMID: 37179456 PMCID: PMC10362419 DOI: 10.1016/j.ymthe.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/15/2023] Open
Abstract
Lung cancer causes the most cancer-related deaths worldwide. In recent years, molecular and immunohistochemical techniques have rapidly developed, further inaugurating an era of personalized medicine for lung cancer. The rare subset of lung cancers accounts for approximately 10%, each displaying distinct clinical characteristics. Treatments for rare lung cancers are mainly based on evidence from common counterparts, which may lead to unsolid clinical benefits considering intertumoral heterogeneity. The increasing knowledge of molecular profiling of rare lung cancers has made targeting genetic alterations and immune checkpoints a powerful strategy. Additionally, cellular therapy has emerged as a promising way to target tumor cells. In this review, we first discuss the current status of targeted therapy and preclinical models for rare lung cancers, as well as provide mutational profiles by integrating the results of existing cohorts. Finally, we point out the challenges and future directions for developing targeted agents for rare lung cancer.
Collapse
Affiliation(s)
- Chunsen Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, the National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiang Yuan
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, the National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, the National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
182
|
Albers J, Friese-Hamim M, Clark A, Schadt O, Walter-Bausch G, Stroh C, Johne A, Karachaliou N, Blaukat A. The Preclinical Pharmacology of Tepotinib-A Highly Selective MET Inhibitor with Activity in Tumors Harboring MET Alterations. Mol Cancer Ther 2023; 22:833-843. [PMID: 36999986 PMCID: PMC10320478 DOI: 10.1158/1535-7163.mct-22-0537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/16/2022] [Accepted: 03/29/2023] [Indexed: 04/01/2023]
Abstract
The mesenchymal-epithelial transition factor (MET) proto-oncogene encodes the MET receptor tyrosine kinase. MET aberrations drive tumorigenesis in several cancer types through a variety of molecular mechanisms, including MET mutations, gene amplification, rearrangement, and overexpression. Therefore, MET is a therapeutic target and the selective type Ib MET inhibitor, tepotinib, was designed to potently inhibit MET kinase activity. In vitro, tepotinib inhibits MET in a concentration-dependent manner irrespective of the mode of MET activation, and in vivo, tepotinib exhibits marked, dose-dependent antitumor activity in MET-dependent tumor models of various cancer indications. Tepotinib penetrates the blood-brain barrier and demonstrates strong antitumor activity in subcutaneous and orthotopic brain metastasis models, in-line with clinical activity observed in patients. MET amplification is an established mechanism of resistance to EGFR tyrosine kinase inhibitors (TKI), and preclinical studies show that tepotinib in combination with EGFR TKIs can overcome this resistance. Tepotinib is currently approved for the treatment of adult patients with advanced or metastatic non-small cell lung cancer harboring MET exon 14 skipping alterations. This review focuses on the pharmacology of tepotinib in preclinical cancer models harboring MET alterations and demonstrates that strong adherence to the principles of the Pharmacological Audit Trail may result in a successful discovery and development of a precision medicine.
Collapse
Affiliation(s)
- Joachim Albers
- Research Unit Oncology, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Manja Friese-Hamim
- Corporate Animal Using Vendor and Vivarium Governance (SQ-AV), Corporate Sustainability, Quality, Trade Compliance (SQ), Animal Affairs (SQ-A), the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Anderson Clark
- Research Unit Oncology, EMD Serono Research and Development Institute, Inc., Billerica, Massachusetts
| | - Oliver Schadt
- Global Medicinal Chemistry, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Gina Walter-Bausch
- Research Unit Oncology, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Christopher Stroh
- Clinical Biomarkers and Companion Diagnostics, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Andreas Johne
- Global Clinical Development Unit, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Niki Karachaliou
- Global Clinical Development Unit, the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Andree Blaukat
- Research Unit Oncology, the healthcare business of Merck KGaA, Darmstadt, Germany
| |
Collapse
|
183
|
Mahrous M, Omar Jebriel A, Allehebi A, Shafik A, El Karak F, Venturini F, Alhusaini H, Meergans M, Ali Nahit Sendur M, Ouda M, Al-Nassar M, Kilickap S, Al Turki S, Al-Fayea T, Abdel Kader Y. Consensus Recommendations for the Diagnosis, Biomarker Testing, and Clinical Management of Advanced or Metastatic Non-small Cell Lung Cancer With Mesenchymal-Epithelial Transition Exon 14 Skipping Mutations in the Middle East, Africa, and Russia. Cureus 2023; 15:e41992. [PMID: 37492039 PMCID: PMC10365828 DOI: 10.7759/cureus.41992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2023] [Indexed: 07/27/2023] Open
Abstract
Mesenchymal-epithelial transition exon 14 (METex14) skipping mutations occur in about 3%-4% of patients with non-small cell lung cancer (NSCLC). This is an aggressive subtype associated with poor prognosis. METex14 skipping is a potentially targetable mutation. Targeted therapy is a promising treatment modality for patients with advanced/metastatic METex14-mutant NSCLC. Performing systematic molecular testing to detect the driver mutation is essential for initiating targeted therapy. However, there is a lack of guidelines on molecular testing for assessing the eligibility of patients for targeted therapy. Therefore, a multidisciplinary panel consisting of experts from the Middle East, Africa, and Russia convened via a virtual advisory board meeting to provide their insights on various molecular testing techniques for the diagnosis of METex14 skipping mutation, management of patients with targeted therapies, and developing consensus recommendations for improving the processes. The expert panel emphasized performing molecular testing and liquid biopsy before treatment initiation and tissue re-biopsy for patients with failed molecular testing. Liquid biopsy was recommended as complementary to tissue biopsy for disease monitoring and prognosis. Selective MET inhibitors were recommended as the first and subsequent lines of therapy. These consensus recommendations will facilitate the management of METex14 skipping NSCLC in routine practice and warrant optimum outcomes for these patients.
Collapse
Affiliation(s)
- Mervat Mahrous
- Oncology, Minia University, Minia, EGY
- Oncology, Prince Sultan Military Medical City, Riyadh, SAU
| | | | - Ahmed Allehebi
- Oncology, King Faisal Specialist Hospital & Research Center, Jeddah, SAU
| | - Amr Shafik
- Oncology, Faculty of Medicine, Ain Shams University, Cairo, EGY
| | - Fadi El Karak
- Oncology, Saint Joseph University of Beirut, School of Medicine, Beirut, LBN
- Oncology, Hôtel-Dieu de France, Beirut, LBN
- Oncology, Clemenceau Medical Center, Dubai, ARE
| | | | - Hamed Alhusaini
- Oncology, King Faisal Specialist Hospital & Research Center, Riyadh, SAU
| | | | | | - Mohamed Ouda
- Oncology, Merck Serono Middle East FZ-Ltd., Dubai, ARE
| | | | | | | | - Turki Al-Fayea
- Oncology, King Fahad Medical City - Ministry of National Guard, Jeddah, SAU
- Oncology, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
| | | |
Collapse
|
184
|
Inno A, Bogina G, Settanni G, Foti G, Tessari R, Gori S. Mucoepidermoid Carcinoma of the Lung Harboring MET Exon 14 Skipping Mutation Treated With Tepotinib: A Case Report. JCO Precis Oncol 2023; 7:e2300136. [PMID: 37499190 DOI: 10.1200/po.23.00136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/27/2023] [Accepted: 06/30/2023] [Indexed: 07/29/2023] Open
Abstract
A case of pulmonary mucoepidermoid cancer harboring MET exon 14 skipping mutation and responding to tepotinib.
Collapse
Affiliation(s)
- Alessandro Inno
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella (VR), Italy
| | - Giuseppe Bogina
- Pathology, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella (VR), Italy
| | - Giulio Settanni
- Pathology, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella (VR), Italy
| | - Giovanni Foti
- Radiology, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella (VR), Italy
| | - Roberto Tessari
- Pharmacy, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella (VR), Italy
| | - Stefania Gori
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella (VR), Italy
| |
Collapse
|
185
|
Nagakubo Y, Hirotsu Y, Amemiya K, Mochizuki H, Tsutsui T, Kakizaki Y, Miyashita Y, Higuchi R, Nakagomi T, Goto T, Oyama T, Omata M. Nucleic Acid Quality Assessment is Critical to the Success of the Oncomine Dx Target Test for Lung Cancer. Mol Diagn Ther 2023; 27:513-523. [PMID: 37198423 DOI: 10.1007/s40291-023-00653-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND AND OBJECTIVE The Oncomine Dx Target Test (ODxTT) has been used as a companion diagnostic test for lung cancer. Here, we evaluated whether the amount of nucleic acid and the degree of RNA degradation are related to the success of the ODxTT. METHODS This study included 223 samples from 218 patients with lung cancer. For all samples, DNA and RNA concentrations were quantified using Qubit, and the degree of RNA degradation was evaluated using the Bioanalyzer. RESULTS Of the 223 samples, 219 samples were successfully analyzed in the ODxTT and four were not. DNA analysis failed in two samples, which were attributed to low DNA concentrations and both were cytology specimens. Meanwhile, RNA analysis failed in the other two samples. These samples had sufficient amounts of RNA, but it was highly degraded with DV200 (the percentage of RNA fragments > 200 base pairs) less than 30. Compared with RNA samples with DV200 ≥ 30, analysis of RNA with DV200 < 30 yielded significantly fewer reads for the internal control genes. This test showed actionable mutations were identified in 38% (83/218) of all patients and in 46.6% (76/163) of patients with lung adenocarcinoma. CONCLUSIONS DNA concentration and degree of RNA degradation are key factors determining the success of diagnostic testing by the ODxTT.
Collapse
Affiliation(s)
- Yuki Nagakubo
- Division of Genetics and Clinical Laboratory, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Yosuke Hirotsu
- Genome Analysis Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, 400-8506, Japan.
| | - Kenji Amemiya
- Division of Genetics and Clinical Laboratory, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Hitoshi Mochizuki
- Genome Analysis Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, 400-8506, Japan
- Central Clinical Laboratory, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
- Department of Gastroenterology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Toshiharu Tsutsui
- Lung Cancer and Respiratory Disease Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Yumiko Kakizaki
- Lung Cancer and Respiratory Disease Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Yoshihiro Miyashita
- Lung Cancer and Respiratory Disease Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Rumi Higuchi
- Lung Cancer and Respiratory Disease Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Takahiro Nakagomi
- Lung Cancer and Respiratory Disease Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Taichiro Goto
- Lung Cancer and Respiratory Disease Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Toshio Oyama
- Pathology Division, Laboratory Department, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Masao Omata
- Department of Gastroenterology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
- The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
186
|
Wang F, Cali Daylan AE, Deng L, Yang J, Sharma J, Su C, Li S, Zang X, Halmos B, Borczuk A, Cheng H. Heterogeneous Expression of PD-L1, B7x, B7-H3, and HHLA2 in Pulmonary Sarcomatoid Carcinoma and the Related Regulatory Signaling Pathways. Cancers (Basel) 2023; 15:3372. [PMID: 37444481 DOI: 10.3390/cancers15133372] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/15/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
Immunotherapy has transformed lung cancer management, but PSC remains an aggressive subtype with a poor prognosis. This study investigates the differential expression of PD-L1 and alternative immune checkpoints (ICs; B7x, B7-H3, and HHLA2), and genetic alterations in PSCs. Tumor specimens of 41 PSC patients were evaluated. PD-L1, B7x, B7-H3, and HHLA2 were positive in 75.0%, 67.6%, 73.0%, and 91.9% of tumors, respectively. PD-L1 expression was significantly higher in the epithelial compared to the sarcomatoid component (median TPS: 50% vs. 0%, p = 0.010). Expression of PD-L1 in both components was only seen in 32.1% of patients. However, at least one IC was expressed in 92.9% of epithelial and 100% of sarcomatoid components. Furthermore, METex14 was detected in 19.5% of patients and was associated with a higher sarcomatoid percentage. Our preclinical studies revealed that METex14 induced PD-L1 expression via MAPK or PI3K/Akt pathways, and MET inhibitors decreased PD-L1 expression. Our findings demonstrate distinct expressions of ICs in PSC subcomponents. Thus, combination IC inhibition as a therapeutic strategy in PSC warrants further exploration. A high percentage of METex14 in PSC and its role in regulating PD-L1 expression reveal different therapeutic targets in this aggressive NSCLC subtype.
Collapse
Affiliation(s)
- Feng Wang
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Lei Deng
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Jihua Yang
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Janaki Sharma
- Department of Medicine, University of Miami Health System, Miami, FL 33136, USA
| | - Christopher Su
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Shenduo Li
- Department of Medicine, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA
| | - Xingxing Zang
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Balazs Halmos
- Department of Oncology, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Alain Borczuk
- Department of Pathology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Haiying Cheng
- Department of Oncology, Montefiore Medical Center, Bronx, NY 10467, USA
| |
Collapse
|
187
|
Stephan-Falkenau S, Streubel A, Mairinger T, Blum TG, Kollmeier J, Mairinger FD, Bauer T, Pfannschmidt J, Hollmann M, Wessolly M. Integrated Clinical, Molecular and Immunological Characterization of Pulmonary Sarcomatoid Carcinomas Reveals an Immune Escape Mechanism That May Influence Therapeutic Strategies. Int J Mol Sci 2023; 24:10558. [PMID: 37445733 DOI: 10.3390/ijms241310558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Pulmonary sarcomatoid carcinoma (PSC) has highly aggressive biological behaviour and poor clinical outcomes, raising expectations for new therapeutic strategies. We characterized 179 PSC by immunohistochemistry, next-generation sequencing and in silico analysis using a deep learning algorithm with respect to clinical, immunological and molecular features. PSC was more common in men, older ages and smokers. Surgery was an independent factor (p < 0.01) of overall survival (OS). PD-L1 expression was detected in 82.1% of all patients. PSC patients displaying altered epitopes due to processing mutations showed another PD-L1-independent immune escape mechanism, which also significantly influenced OS (p < 0.02). The effect was also maintained when only advanced tumour stages were considered (p < 0.01). These patients also showed improved survival with a significant correlation for immunotherapy (p < 0.05) when few or no processing mutations were detected, although this should be interpreted with caution due to the small number of patients studied. Genomic alterations for which there are already approved drugs were present in 35.4% of patients. Met exon 14 skipping was found more frequently (13.7%) and EGFR mutations less frequently (1.7%) than in other NSCLC. In summary, in addition to the divergent genomic landscape of PSC, the specific immunological features of this prognostically poor subtype should be considered in therapy stratification.
Collapse
Affiliation(s)
- Susann Stephan-Falkenau
- Institute for Tissue Diagnostics, MVZ at Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Anna Streubel
- Institute for Tissue Diagnostics, MVZ at Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Thomas Mairinger
- Institute for Tissue Diagnostics, MVZ at Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Torsten-Gerriet Blum
- Department of Pneumology, Heckeshorn Lung Clinic, Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Jens Kollmeier
- Department of Pneumology, Heckeshorn Lung Clinic, Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Fabian D Mairinger
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| | - Torsten Bauer
- Department of Pneumology, Heckeshorn Lung Clinic, Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Joachim Pfannschmidt
- Department of Thoracic Surgery, Heckeshorn Lung Clinic, Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Manuel Hollmann
- Institute for Tissue Diagnostics, MVZ at Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Michael Wessolly
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| |
Collapse
|
188
|
Spagnolo CC, Ciappina G, Giovannetti E, Squeri A, Granata B, Lazzari C, Pretelli G, Pasello G, Santarpia M. Targeting MET in Non-Small Cell Lung Cancer (NSCLC): A New Old Story? Int J Mol Sci 2023; 24:10119. [PMID: 37373267 PMCID: PMC10299133 DOI: 10.3390/ijms241210119] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
In recent years, we have seen the development and approval for clinical use of an increasing number of therapeutic agents against actionable oncogenic drivers in metastatic non-small cell lung cancer (NSCLC). Among them, selective inhibitors, including tyrosine kinase inhibitors (TKIs) and monoclonal antibodies targeting the mesenchymal-epithelial transition (MET) receptor, have been studied in patients with advanced NSCLC with MET deregulation, primarily due to exon 14 skipping mutations or MET amplification. Some MET TKIs, including capmatinib and tepotinib, have proven to be highly effective in this molecularly defined subgroup of patients and are already approved for clinical use. Other similar agents are being tested in early-stage clinical trials with promising antitumor activity. The purpose of this review is to provide an overview of MET signaling pathways, MET oncogenic alterations primarily focusing on exon 14 skipping mutations, and the laboratory techniques used to detect MET alterations. Furthermore, we will summarize the currently available clinical data and ongoing studies on MET inhibitors, as well as the mechanisms of resistance to MET TKIs and new potential strategies, including combinatorial approaches, to improve the clinical outcomes of MET exon 14-altered NSCLC patients.
Collapse
Affiliation(s)
- Calogera Claudia Spagnolo
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98122 Messina, Italy; (C.C.S.); (G.C.); (A.S.); (B.G.)
| | - Giuliana Ciappina
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98122 Messina, Italy; (C.C.S.); (G.C.); (A.S.); (B.G.)
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrje Universiteit, 1081HV Amsterdam, The Netherlands;
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, 56017 San Giuliano, Italy
| | - Andrea Squeri
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98122 Messina, Italy; (C.C.S.); (G.C.); (A.S.); (B.G.)
| | - Barbara Granata
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98122 Messina, Italy; (C.C.S.); (G.C.); (A.S.); (B.G.)
| | - Chiara Lazzari
- Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia (FPO)-IRCCS, 10060 Torino, Italy;
| | - Giulia Pretelli
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (G.P.); (G.P.)
| | - Giulia Pasello
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (G.P.); (G.P.)
- Oncologia Medica 2, Istituto Oncologico Veneto, IRCCS, 35128 Padova, Italy
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98122 Messina, Italy; (C.C.S.); (G.C.); (A.S.); (B.G.)
| |
Collapse
|
189
|
Babey H, Jamme P, Curcio H, Assié JB, Veillon R, Doubre H, Pérol M, Guisier F, Huchot E, Decroisette C, Falchero L, Corre R, Cortot A, Chouaïd C, Descourt R. Real-World Treatment Outcomes of MET Exon14 Skipping in Non-small Cell Lung Cancer: GFPC 03-18 Study. Target Oncol 2023:10.1007/s11523-023-00976-4. [PMID: 37310660 DOI: 10.1007/s11523-023-00976-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2023] [Indexed: 06/14/2023]
Abstract
BACKGROUND MET-targeted tyrosine kinase inhibitors (TKIs) demonstrated efficacy in advanced non-small cell lung cancer (aNSCLC) with MET exon14 skipping mutations (METexon14); yet, data on the management of these patients in clinical practice is sparse. OBJECTIVE The aim of this study was to describe the management of METexon14 aNSCLC patients. PATIENTS AND METHODS This real-life, retrospective study analyzed the management of METexon14 aNSCLC. The primary endpoint was the median overall survival (mOS). Secondary endpoints were to assess investigator-progression-free survival (PFS) and mOS in different subgroups: patients treated with (a) crizotinib, regardless of treatment line; (b) anti-MET TKIs (crizotinib, tepotinib, capmatinib); and (c) immunotherapy. RESULTS A total of 118 patients were included between December 2015 and January 1, 2020 in 13 centers. Median age was 73 years, 62.7% were female, 83.9% had adenocarcinoma, 92.4% at stage IV, and 27% had more than three metastatic sites. The majority of the patients (106, 89.8%) received at least one systemic treatment; 73% received at least one anti-MET TKI: crizotinib (68.6%), tepotinib (16%), capmatinib (10%). Only 10% received two anti-MET TKIs in their treatment sequences. With a median follow-up of 16 months (95% CI 13.6-29.7), mOS was 27.1 months (95% CI 18-31.4). There was no significant difference between mOS of patients treated and never treated with crizotinib, 19.7 (95% CI 13.6-29.7) and 28 (95% CI 16.4-NR) months, respectively (p = 0.16); mOS of the TKI cohort and of the TKI-naïve patient cohort were 27.1 (95% CI 18-29.7) and 35.6 (95% CI 8.6-NR) months respectively, with no significant difference (p = 0.7). CONCLUSIONS In this real-life study, there was no evidence of benefit in mOS with anti-MET TKIs.
Collapse
Affiliation(s)
- Hélène Babey
- Institut de Cancérologie, Hôpital Morvan, CHU de Brest, Brest, France
| | | | | | - Jean Baptiste Assié
- Service de Pneumologie, Pneumologie, Centre Hospitalier Intercommunal, CHI Créteil, 40, avenue de Verdun, 94010, Créteil Cedex, France
| | - Remi Veillon
- Oncologie thoracique, CHU de Bordeaux, Bordeaux, France
| | | | | | | | - Eric Huchot
- CHU Saint Pierre de La Réunion, Saint-Pierre, La Réunion, France
| | | | | | | | | | - Christos Chouaïd
- Service de Pneumologie, Pneumologie, Centre Hospitalier Intercommunal, CHI Créteil, 40, avenue de Verdun, 94010, Créteil Cedex, France.
| | - Renaud Descourt
- Institut de Cancérologie, Hôpital Morvan, CHU de Brest, Brest, France
| |
Collapse
|
190
|
Wang C, Lu X. Targeting MET: Discovery of Small Molecule Inhibitors as Non-Small Cell Lung Cancer Therapy. J Med Chem 2023. [PMID: 37262349 DOI: 10.1021/acs.jmedchem.3c00028] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
MET has been considered as a promising drug target for the treatment of MET-dependent diseases, particularly non-small cell lung cancer (NSCLC). Small molecule MET inhibitors with mainly three types of binding modes (Ia/Ib, II, and III) have been developed. In this Review, we provide an overview of the structural features, activation mechanism, and dysregulation pathway of MET and summarize progress on the development and discovery strategies utilized for MET inhibitors as well as mechanisms of acquired resistance to current approved inhibitors. The insights will accelerate discovery of new generation MET inhibitors to overcome clinical acquired resistance.
Collapse
Affiliation(s)
- Chaofan Wang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 510632, China
| | - Xiaoyun Lu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450001, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 510632, China
| |
Collapse
|
191
|
Ariyasu R, Kakuto S, Miyadera K, Akita T, Kiritani A, Tsugitomi R, Amino Y, Uchibori K, Kitazono S, Yanagitani N, Nishio M. Real-World Outcome Analysis of Patients With Stage IV NSCLC Treated With Tyrosine Kinase and Immune Checkpoint Inhibitors. JTO Clin Res Rep 2023; 4:100524. [PMID: 37426307 PMCID: PMC10329142 DOI: 10.1016/j.jtocrr.2023.100524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/31/2023] [Accepted: 04/27/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Only a few reports have determined whether recently advanced anticancer drugs, particularly next-generation tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs), prolong the survival of patients with NSCLC in the real world. Methods To evaluate the association between recently advanced drugs and patient survival, survival data of 2078 patients with stage IV NSCLC from 1995 to 2022 were analyzed in the present study. The patients were classified into the following six groups in terms of the date of diagnosis: period A, 1995 to 1999; period B, 2000 to 2004; period C, 2005 to 2009; period D, 2010 to 2014; period E, 2015 to 2019; and period F, 2000 to 2022. They were further grouped in terms of EGFR mutation and ALK fusion. Results The median overall survival (mOS) times were 8.9, 11.0, 13.6, 17.9, and 25.2 months in periods A to E, respectively, and the mOS time was not reached in period F. This time was significantly longer in period E than in period D (25.2 versus 17.9 mo, p < 0.005). Moreover, the mOS times of patients with EGFR mutation, those with ALK fusion, and those without both alterations were significantly longer in period E than in period D (46.0 versus 32.0 mo, p < 0.005; not reached versus 36.2 mo, p = 0.018; 14.6 versus 11.7 mo, p < 0.005). The history of treatment with next-generation TKIs and ICIs was found to be associated with overall survival. Conclusions The survival of patients with NSCLC was improved from period D to period E, regardless of the presence of driver gene alteration. We found that next-generation TKIs and ICIs may be associated with improvements in overall survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Makoto Nishio
- Corresponding author. Address for correspondence: Makoto Nishio, MD, PhD, Department of Thoracic Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo 135-8550, Japan.
| |
Collapse
|
192
|
Feldt SL, Bestvina CM. The Role of MET in Resistance to EGFR Inhibition in NSCLC: A Review of Mechanisms and Treatment Implications. Cancers (Basel) 2023; 15:cancers15112998. [PMID: 37296959 DOI: 10.3390/cancers15112998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Utilizing targeted therapy against activating mutations has opened a new era of treatment paradigms for patients with advanced non-small cell lung cancer (NSCLC). For patients with epidermal growth factor (EGFR)-mutated cancers, EGFR inhibitors, including the third-generation tyrosine kinase inhibitor (TKI) osimertinib, significantly prolong progression-free survival and overall survival, and are the current standard of care. However, progression after EGFR inhibition invariably occurs, and further study has helped elucidate mechanisms of resistance. Abnormalities in the mesenchymal-epithelial transition (MET) oncogenic pathway have been implicated as common alterations after progression, with MET amplification as one of the most frequent mechanisms. Multiple drugs with inhibitory activity against MET, including TKIs, antibodies, and antibody-drug conjugates, have been developed and studied in advanced NSCLC. Combining MET and EGFR is a promising treatment strategy for patients found to have a MET-driven resistance mechanism. Combination TKI therapy and EGFR-MET bispecific antibodies have shown promising anti-tumor activity in early clinical trials. Future study including ongoing large-scale trials of combination EGFR-MET inhibition will help clarify if targeting this mechanism behind EGFR resistance will have meaningful clinical benefit for patients with advanced EGFR-mutated NSCLC.
Collapse
Affiliation(s)
- Susan L Feldt
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
193
|
Hines JB, Bowar B, Levine E, Esposito A, Garassino MC, Bestvina CM. Targeted Toxicities: Protocols for Monitoring the Adverse Events of Targeted Therapies Used in the Treatment of Non-Small Cell Lung Cancer. Int J Mol Sci 2023; 24:9429. [PMID: 37298380 PMCID: PMC10253830 DOI: 10.3390/ijms24119429] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/12/2023] Open
Abstract
Targeted therapies have revolutionized the treatment for many patients with non-small cell lung cancer (NSCLC). Multiple new oral targeted therapies have been approved in the last decade; however, their overall efficacy may be reduced by poor adherence, treatment interruptions, or dose reductions due to adverse events. Most institutions lack standard monitoring protocols for toxicities from these targeted agents. This review describes important adverse events observed in clinical trials and reported by the U.S. Food and Drug Administration for both currently approved and upcoming promising therapies in the treatment of NSCLC. These agents cause a range of toxicities, including dermatologic, gastroenteric, pulmonary, and cardiac toxicities. This review proposes protocols for routine monitoring of these adverse events, both prior to initiation of therapy and while on treatment.
Collapse
Affiliation(s)
- Jacobi B. Hines
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL 60637, USA; (J.B.H.); (B.B.); (A.E.); (C.M.B.)
| | - Benjamin Bowar
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL 60637, USA; (J.B.H.); (B.B.); (A.E.); (C.M.B.)
| | - Emma Levine
- Booth School of Business, University of Chicago, Chicago, IL 60637, USA;
| | - Alessandra Esposito
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL 60637, USA; (J.B.H.); (B.B.); (A.E.); (C.M.B.)
| | - Marina C. Garassino
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL 60637, USA; (J.B.H.); (B.B.); (A.E.); (C.M.B.)
| | - Christine M. Bestvina
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL 60637, USA; (J.B.H.); (B.B.); (A.E.); (C.M.B.)
| |
Collapse
|
194
|
Heydt C, Ihle MA, Merkelbach-Bruse S. Overview of Molecular Detection Technologies for MET in Lung Cancer. Cancers (Basel) 2023; 15:cancers15112932. [PMID: 37296895 DOI: 10.3390/cancers15112932] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
MET tyrosine kinase receptor pathway activation has become an important actionable target in solid tumors. Aberrations in the MET proto-oncogene, including MET overexpression, the activation of MET mutations, MET mutations that lead to MET exon 14 skipping, MET gene amplifications, and MET fusions, are known to be primary and secondary oncogenic drivers in cancer; these aberrations have evolved as predictive biomarkers in clinical diagnostics. Thus, the detection of all known MET aberrations in daily clinical care is essential. In this review, current molecular technologies for the detection of the different MET aberrations are highlighted, including the benefits and drawbacks. In the future, another focus will be on the standardization of detection technologies for the delivery of reliable, quick, and affordable tests in clinical molecular diagnostics.
Collapse
Affiliation(s)
- Carina Heydt
- Faculty of Medicine, Institute of Pathology, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Michaela Angelika Ihle
- Faculty of Medicine, Institute of Pathology, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Sabine Merkelbach-Bruse
- Faculty of Medicine, Institute of Pathology, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| |
Collapse
|
195
|
Li S, de Camargo Correia GS, Wang J, Manochakian R, Zhao Y, Lou Y. Emerging Targeted Therapies in Advanced Non-Small-Cell Lung Cancer. Cancers (Basel) 2023; 15:2899. [PMID: 37296863 PMCID: PMC10251928 DOI: 10.3390/cancers15112899] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/08/2023] [Accepted: 05/13/2023] [Indexed: 06/12/2023] Open
Abstract
Lung cancer remains the leading cause of cancer-related mortality worldwide. Non-small-cell lung cancer (NSCLC) is the most common type and is still incurable for most patients at the advanced stage. Targeted therapy is an effective treatment that has significantly improved survival in NSCLC patients with actionable mutations. However, therapy resistance occurs widely among patients leading to disease progression. In addition, many oncogenic driver mutations in NSCLC still lack targeted agents. New drugs are being developed and tested in clinical trials to overcome these challenges. This review aims to summarize emerging targeted therapy that have been conducted or initiated through first-in-human clinical trials in the past year.
Collapse
Affiliation(s)
- Shenduo Li
- Division of Hematology and Medical Oncology, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL 32224, USA (G.S.d.C.C.)
| | | | - Jing Wang
- Department of Medicine, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL 32224, USA
| | - Rami Manochakian
- Division of Hematology and Medical Oncology, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL 32224, USA (G.S.d.C.C.)
| | - Yujie Zhao
- Division of Hematology and Medical Oncology, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL 32224, USA (G.S.d.C.C.)
| | - Yanyan Lou
- Division of Hematology and Medical Oncology, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL 32224, USA (G.S.d.C.C.)
| |
Collapse
|
196
|
de Jong D, Das JP, Ma H, Pailey Valiplackal J, Prendergast C, Roa T, Braumuller B, Deng A, Dercle L, Yeh R, Salvatore MM, Capaccione KM. Novel Targets, Novel Treatments: The Changing Landscape of Non-Small Cell Lung Cancer. Cancers (Basel) 2023; 15:2855. [PMID: 37345192 PMCID: PMC10216085 DOI: 10.3390/cancers15102855] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/11/2023] [Accepted: 05/19/2023] [Indexed: 06/23/2023] Open
Abstract
Treatment of non-small cell lung cancer (NSCLC) has undergone a paradigm shift. Once a disease with limited potential therapies, treatment options for patients have exploded with the availability of molecular testing to direct management and targeted therapies to treat tumors with specific driver mutations. New in vitro diagnostics allow for the early and non-invasive detection of disease, and emerging in vivo imaging techniques allow for better detection and monitoring. The development of checkpoint inhibitor immunotherapy has arguably been the biggest advance in lung cancer treatment, given that the vast majority of NSCLC tumors can be treated with these therapies. Specific targeted therapies, including those against KRAS, EGFR, RTK, and others have also improved the outcomes for those individuals bearing an actionable mutation. New and emerging therapies, such as bispecific antibodies, CAR T cell therapy, and molecular targeted radiotherapy, offer promise to patients for whom none of the existing therapies have proved effective. In this review, we provide the most up-to-date survey to our knowledge regarding emerging diagnostic and therapeutic strategies for lung cancer to provide clinicians with a comprehensive reference of the options for treatment available now and those which are soon to come.
Collapse
Affiliation(s)
- Dorine de Jong
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA;
| | - Jeeban P. Das
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; (J.P.D.); (R.Y.)
| | - Hong Ma
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (H.M.); (J.P.V.); (C.P.); (T.R.); (B.B.); (L.D.); (M.M.S.)
| | - Jacienta Pailey Valiplackal
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (H.M.); (J.P.V.); (C.P.); (T.R.); (B.B.); (L.D.); (M.M.S.)
| | - Conor Prendergast
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (H.M.); (J.P.V.); (C.P.); (T.R.); (B.B.); (L.D.); (M.M.S.)
| | - Tina Roa
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (H.M.); (J.P.V.); (C.P.); (T.R.); (B.B.); (L.D.); (M.M.S.)
| | - Brian Braumuller
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (H.M.); (J.P.V.); (C.P.); (T.R.); (B.B.); (L.D.); (M.M.S.)
| | - Aileen Deng
- Department of Hematology and Oncology, Novant Health, 170 Medical Park Road, Mooresville, NC 28117, USA;
| | - Laurent Dercle
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (H.M.); (J.P.V.); (C.P.); (T.R.); (B.B.); (L.D.); (M.M.S.)
| | - Randy Yeh
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; (J.P.D.); (R.Y.)
| | - Mary M. Salvatore
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (H.M.); (J.P.V.); (C.P.); (T.R.); (B.B.); (L.D.); (M.M.S.)
| | - Kathleen M. Capaccione
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (H.M.); (J.P.V.); (C.P.); (T.R.); (B.B.); (L.D.); (M.M.S.)
| |
Collapse
|
197
|
Zhou J, Zhang XC, Xue S, Dai M, Wang Y, Peng X, Chen J, Wang X, Shen Y, Qin H, Chen B, Zheng Y, Gao X, Xie Z, Ding J, Jiang H, Wu YL, Geng M, Ai J. SYK-mediated epithelial cell state is associated with response to c-Met inhibitors in c-Met-overexpressing lung cancer. Signal Transduct Target Ther 2023; 8:185. [PMID: 37183231 PMCID: PMC10183461 DOI: 10.1038/s41392-023-01403-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 02/19/2023] [Accepted: 03/05/2023] [Indexed: 05/16/2023] Open
Abstract
Genomic MET amplification and exon 14 skipping are currently clinically recognized biomarkers for stratifying subsets of non-small cell lung cancer (NSCLC) patients according to the predicted response to c-Met inhibitors (c-Metis), yet the overall clinical benefit of this strategy is quite limited. Notably, c-Met protein overexpression, which occurs in approximately 20-25% of NSCLC patients, has not yet been clearly defined as a clinically useful biomarker. An optimized strategy for accurately classifying patients with c-Met overexpression for decision-making regarding c-Meti treatment is lacking. Herein, we found that SYK regulates the plasticity of cells in an epithelial state and is associated with their sensitivity to c-Metis both in vitro and in vivo in PDX models with c-Met overexpression regardless of MET gene status. Furthermore, TGF-β1 treatment resulted in SYK transcriptional downregulation, increased Sp1-mediated transcription of FRA1, and restored the mesenchymal state, which conferred resistance to c-Metis. Clinically, a subpopulation of NSCLC patients with c-Met overexpression coupled with SYK overexpression exhibited a high response rate of 73.3% and longer progression-free survival with c-Meti treatment than other patients. SYK negativity coupled with TGF-β1 positivity conferred de novo and acquired resistance. In summary, SYK regulates cell plasticity toward a therapy-sensitive epithelial cell state. Furthermore, our findings showed that SYK overexpression can aid in precisely stratifying NSCLC patients with c-Met overexpression regardless of MET alterations and expand the population predicted to benefit from c-Met-targeted therapy.
Collapse
Affiliation(s)
- Ji Zhou
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Xu-Chao Zhang
- Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, and Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, 510080, China
| | - Shan Xue
- Department of Respiratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mengdi Dai
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Yueliang Wang
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xia Peng
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jianjiao Chen
- Department of Neurobiology, Brain Institute, University of Pittsburgh, Pittsburgh, 15213, USA
| | - Xinyi Wang
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yanyan Shen
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hui Qin
- Department of Respiratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Bi Chen
- Department of Respiratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Yu Zheng
- Department of Respiratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiwen Gao
- Department of Respiratory Medicine, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Zuoquan Xie
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Jian Ding
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
| | - Handong Jiang
- Department of Respiratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Yi-Long Wu
- Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, and Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, 510080, China.
| | - Meiyu Geng
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China.
| | - Jing Ai
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China.
| |
Collapse
|
198
|
Yu Y, Zhou J, Li X, Goto K, Min X, Nishino K, Cui J, Wu L, Sakakibara J, Shu Y, Dong X, Li L, Yoneshima Y, Zhou C, Li X, Zhang Y, Huang D, Zang A, Zhang W, Wang X, Zhang L, Bai C, Fang J, Cao L, Zhao Y, Yu Y, Shi M, Zhong D, Li F, Li M, Wu Q, Zhou J, Sun M, Lu S. Gumarontinib in patients with non-small-cell lung cancer harbouring MET exon 14 skipping mutations: a multicentre, single-arm, open-label, phase 1b/2 trial. EClinicalMedicine 2023; 59:101952. [PMID: 37096188 PMCID: PMC10121392 DOI: 10.1016/j.eclinm.2023.101952] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/21/2023] [Accepted: 03/21/2023] [Indexed: 04/26/2023] Open
Abstract
Background Approximately 3-4% of patients with non-small-cell lung cancer (NSCLC) have MET exon 14 (METex14) skipping mutations. We report primary results from the phase 2 stage of a phase 1b/2 study of gumarontinib, a selective, potent, oral MET inhibitor, in patients with METex14 skipping mutation-positive (METex14-positive) NSCLC. Methods The single-arm, multicentre, open-label, phase 2 stage of the GLORY study was conducted at 42 centres across China and Japan. Adults with locally advanced or metastatic METex14-positive NSCLC received oral gumarontinib 300 mg once daily in continuous 21-day cycles until disease progression, intolerable toxicity, or withdrawal of consent. Eligible patients had failed one or two prior lines of therapy (not including a MET inhibitor), were ineligible for/refused chemotherapy, and had no genetic alterations targetable with standard therapies. The primary endpoint was objective response rate in patients with a valid baseline tumour assessment, by blinded independent review. The study was registered at ClinicalTrials.gov (NCT04270591). Findings Between Aug 2, 2019 and Apr 28, 2021, 84 patients were enrolled and received gumarontinib (median follow-up 13.5 months [IQR 8.7-17.1]), at data cut-off (Apr 28, 2022) five patients whose METex14 status could not be confirmed by a central laboratory were excluded from the efficacy analysis. The objective response rate was 66% (95% CI 54-76) overall (n = 79), 71% (95% CI 55-83) in treatment-naïve patients (n = 44), and 60% (95% CI 42-76) in previously-treated patients (n = 35). The most common treatment-related adverse events (any grade) were oedema (67/84 patients, 80%) and hypoalbuminuria (32/84, 38%). Grade ≥3 treatment-emergent adverse events occurred in 45 (54%) patients. Treatment-related adverse events leading to permanent discontinuation occurred in 8% (7/84) of patients. Interpretation Gumarontinib monotherapy had durable antitumour activity with manageable toxicity in patients with locally advanced or metastatic METex14-positive NSCLC when used in first line or later. Funding Haihe Biopharma Co., Ltd. Supported in part by grants from the National Science and Technology Major Project of China for "Clinical Research of Gumarontinib, a highly selective MET inhibitor" (2018ZX09711002-011-003); the National Natural Science Foundation of China (82030045 to S.L. and 82172633 to YF.Y); Shanghai Municipal Science & Technology Commission Research Project (19411950500 to S.L.); Shanghai Shenkang Action Plan (16CR3005A to S.L.) and Shanghai Chest Hospital Project of Collaborative Innovation (YJXT20190105 to S.L.).
Collapse
Affiliation(s)
- Yongfeng Yu
- Department of Medical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianya Zhou
- Respiratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingya Li
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Koichi Goto
- Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Xuhong Min
- Radiology Intervention Department, Anhui Chest Hospital, Hefei, China
| | - Kazumi Nishino
- Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Jiuwei Cui
- Oncology Department, The First Bethune Hospital of Jilin University, Changchun, China
| | - Lin Wu
- Second Department of Thoracic Medicine, Hunan Cancer Hospital, Changsha, China
| | - Jun Sakakibara
- Respiratory Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Yongqian Shu
- Oncology Department, Jiangsu Province Hospital, Nanjing, China
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Li
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yasuto Yoneshima
- Respiratory Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Chengzhi Zhou
- Oncology, The First Affiliate Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoling Li
- Thoracic Medicine, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yiping Zhang
- Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Dingzhi Huang
- Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Aimin Zang
- Medical Oncology, Affiliated Hospital of Hebei University, Baoding, China
| | - Wei Zhang
- Pneumology Department/Institute Office, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiuwen Wang
- Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Li Zhang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Chong Bai
- Pneumology Department, Changhai Hospital of Shanghai, Shanghai, China
| | - Jian Fang
- Thoracic Oncology Second Department, Beijing Cancer Hospital, Beijing, China
| | - Lejie Cao
- Pneumology Department, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Yanqiu Zhao
- Respiratory Department of Internal Medicine, Henan Cancer Hospital, Zhengzhou, China
| | - Yan Yu
- The Third Ward of Respiratory Medicine Department, Harbin Medical University Cancer Hospital, Harbin, China
| | - Meiqi Shi
- Oncology Department, Jiangsu Cancer Hospital, Nanjing, China
| | - Diansheng Zhong
- Internal Medicine-Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Fugen Li
- Clinical Science Department, Haihe Biopharma Co., Ltd, Shanghai, China
| | - Meng Li
- Biostatistic and Data Science, Haihe Biopharma Co., Ltd, Shanghai, China
| | - Qiuxia Wu
- Clinical Science Department, Haihe Biopharma Co., Ltd, Shanghai, China
| | - Jun Zhou
- Clinical Science Department, Haihe Biopharma Co., Ltd, Shanghai, China
| | - Minghui Sun
- Clinical Science Department, Haihe Biopharma Co., Ltd, Shanghai, China
| | - Shun Lu
- Department of Medical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Corresponding author. Department of Medical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241, Huaihai West Road, Shanghai 200030, China.
| |
Collapse
|
199
|
Schuler M, Bölükbas S, Darwiche K, Theegarten D, Herrmann K, Stuschke M. Personalized Treatment for Patients With Lung Cancer. DEUTSCHES ARZTEBLATT INTERNATIONAL 2023; 120:300-310. [PMID: 36790172 PMCID: PMC10391522 DOI: 10.3238/arztebl.m2023.0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/05/2022] [Accepted: 01/16/2023] [Indexed: 07/06/2024]
Abstract
BACKGROUND Lung cancer is the most common cause of death among all types of cancer in Germany, with an annual death rate of 45 000 patients. Over the past 15 years, innovations in diagnosis and treatment have prolonged the survival of patients with non-small-cell lung cancer in all tumor stages. METHODS This review of the diagnosis and treatment of lung cancer is based on current national and international guidelines, and on prospective trials with the highest possible level of evidence that were retrieved by a selective search of the literature. RESULTS Improved outcomes in patients with non-small-cell lung cancer (85% of new diagnoses) were achieved with the aid of precise diagnostic techniques, including functional imaging and endobronchial procedures for localized disease stage. Contemporary surgical and radio-oncological technologies reduce the morbidity and expand the boundaries of local therapy. Molecular pathology, including the assessment of predictive biomarkers, is an integral part of the diagnostic evaluation of non-small-cell lung cancer in all tumor stages; it enables stratified cytotoxic/molecularly targeted treatments and immunotherapies and improves patient-reported outcomes. The percentage of long-term survivors in the metastatic stage has doubled by the introduction of immunotherapy. In contrast, there has been no major improvement in the survival of patients with small-cell lung cancer (15% of new diagnoses). CONCLUSION In addition to the implementation of lung cancer screening in high-risk populations, the further development and consistent implementation of personalized diagnosis and treatment in certified lung cancer centers can be expected to prolong survival and improve the patients' quality of life.
Collapse
Affiliation(s)
- Martin Schuler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen
- Division of Thoracic Oncology, West German Cancer Center, University Medicine Essen – Ruhrlandklinik
| | - Servet Bölükbas
- West German Cancer Center, University Medicine Essen – Ruhrlandklinik
| | - Kaid Darwiche
- Division of Interventional Bronchology, Department of Pneumology, West German Cancer Center, University Medicine Essen – Ruhrlandklinik
| | - Dirk Theegarten
- Institute of Pathology, West German Cancer Center, University Hospital Essen, Essen
| | - Ken Herrmann
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen
| | - Martin Stuschke
- Department of Radiation Therapy, West German Cancer Center, University Hospital Essen
| |
Collapse
|
200
|
Amir M, Javed S. Elucidation of binding dynamics of tyrosine kinase inhibitor tepotinib, to human serum albumin, using spectroscopic and computational approach. Int J Biol Macromol 2023; 241:124656. [PMID: 37119913 DOI: 10.1016/j.ijbiomac.2023.124656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/07/2023] [Accepted: 04/24/2023] [Indexed: 05/01/2023]
Abstract
Tepotinib (TPT), an anticancer drug, is a fibroblast growth factor receptor inhibitor approved by the FDA for the chemotherapy of urothelial carcinoma. The binding of anticancer medicines to HSA can affect their pharmacokinetics and pharmacodynamics. The absorption, fluorescence emission, circular dichroism, molecular docking, and simulation studies were used to evaluate the binding relationship between TPT and HSA. The absorption spectra exhibited a hyperchromic effect upon the interaction of TPT with HSA. The Stern-Volmer and binding constant of the HSA-TPT complex demonstrates that fluorescence quenching is triggered by a static rather than a dynamic process. Further, the displacement assays and molecular docking results revealed that TPT preferred binding to site III of HSA. Circular dichroism spectroscopy confirmed that TPT binding to HSA induces conformational changes and reduces α-helical content. The thermal CD spectra reveal that tepotinib enhances protein's stability in the temperature range of 20 to 90 °C. The findings of MDS studies provide further evidence for the stability of the HSA-TPT complex. Consequently, the findings of the present investigation provide a clear picture of the impacts of TPT on HSA interaction. These interactions are thought to make the microenvironment around HSA more hydrophobic than in its native state.
Collapse
Affiliation(s)
- Mohd Amir
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, India
| | - Saleem Javed
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|