51
|
Liu Y, Smith MR, Wang Y, D'Agostino R, Ruiz J, Lycan T, Kucera GL, Miller LD, Li W, Chan MD, Farris M, Su J, Song Q, Zhao D, Chandrasekaran A, Xing F. c-Met Mediated Cytokine Network Promotes Brain Metastasis of Breast Cancer by Remodeling Neutrophil Activities. Cancers (Basel) 2023; 15:cancers15092626. [PMID: 37174093 PMCID: PMC10177081 DOI: 10.3390/cancers15092626] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/20/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
The brain is one of the most common metastatic sites among breast cancer patients, especially in those who have Her2-positive or triple-negative tumors. The brain microenvironment has been considered immune privileged, and the exact mechanisms of how immune cells in the brain microenvironment contribute to brain metastasis remain elusive. In this study, we found that neutrophils are recruited and influenced by c-Met high brain metastatic cells in the metastatic sites, and depletion of neutrophils significantly suppressed brain metastasis in animal models. Overexpression of c-Met in tumor cells enhances the secretion of a group of cytokines, including CXCL1/2, G-CSF, and GM-CSF, which play critical roles in neutrophil attraction, granulopoiesis, and homeostasis. Meanwhile, our transcriptomic analysis demonstrated that conditioned media from c-Met high cells significantly induced the secretion of lipocalin 2 (LCN2) from neutrophils, which in turn promotes the self-renewal of cancer stem cells. Our study unveiled the molecular and pathogenic mechanisms of how crosstalk between innate immune cells and tumor cells facilitates tumor progression in the brain, which provides novel therapeutic targets for treating brain metastasis.
Collapse
Affiliation(s)
- Yin Liu
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Margaret R Smith
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Yuezhu Wang
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Ralph D'Agostino
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Jimmy Ruiz
- Department of Hematology and Oncology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Thomas Lycan
- Department of Hematology and Oncology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Gregory L Kucera
- Department of Hematology and Oncology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Lance D Miller
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Wencheng Li
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Michael D Chan
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Michael Farris
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Jing Su
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN 47405, USA
| | - Qianqian Song
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Dawen Zhao
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Arvind Chandrasekaran
- Bioinspired Microengineering Laboratory (BIOME), Department of Chemical, Biological and Bioengineering, NC A&T State University, Greensboro, NC 27411, USA
| | - Fei Xing
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
52
|
Yang M, Vioix H, Sachdev R, Stargardter M, Tosh J, Pfeiffer BM, Paik PK. Cost-Effectiveness of Tepotinib Versus Capmatinib for the Treatment of Adult Patients With Metastatic Non-Small Cell Lung Cancer Harboring Mesenchymal-Epithelial Transition Exon 14 Skipping. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:487-497. [PMID: 36503033 PMCID: PMC10424058 DOI: 10.1016/j.jval.2022.11.018] [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: 06/20/2022] [Revised: 11/23/2022] [Accepted: 11/29/2022] [Indexed: 05/06/2023]
Abstract
OBJECTIVES From the US Medicare perspective, this study compared the cost-effectiveness of tepotinib and capmatinib for treating metastatic non-small cell lung cancer with tumors harboring mesenchymal-epithelial transition factor gene exon 14 skipping. METHODS A 3-state partitioned survival model assessed outcomes over a lifetime horizon. Parametric survival analysis of the phase 2 VISION trial informed clinical inputs for tepotinib. Capmatinib inputs were captured using hazard ratios derived from an unanchored matching-adjusted indirect comparison study and published literature. National cost databases, trial data, and literature furnished drug, treatment monitoring, and disease/adverse event management expenditures (2021 US dollars) and utility inputs. Outcomes were discounted at 3% annually. RESULTS In the base case, tepotinib dominated capmatinib in frontline settings (incremental discounted quality-adjusted life-years [QALYs] and costs of 0.2127 and -$47 756, respectively) while realizing an incremental cost-effectiveness ratio of $274 514/QALY in subsequent lines (incremental QALYs and costs of 0.3330 and $91 401, respectively). In a line agnostic context, tepotinib produced an incremental cost-effectiveness ratio of $105 383/QALY (incremental QALYs and costs of 0.2794 and $29 447, respectively). Sensitivity and scenarios analyses for individual lines typically supported the base case, whereas those for the line agnostic setting suggested sensitivity to drug acquisition costs and efficacy inputs. CONCLUSIONS Tepotinib could be cost-effective versus capmatinib in frontline and line agnostic contexts, considering the range of willingness-to-pay thresholds recommended by the Institute for Clinical and Economic Review ($100 000-$150 000/QALY). Tepotinib could be cost-effective in subsequent lines at higher willingness-to-pay levels. These results are to be interpreted cautiously, considering uncertainty in key model inputs.
Collapse
Affiliation(s)
- Mo Yang
- EMD Serono, Rockland, MA, USA.
| | - Helene Vioix
- the healthcare business of Merck KGaA, Darmstadt, Germany
| | | | | | | | | | - Paul K Paik
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| |
Collapse
|
53
|
Hendriks LE, Kerr KM, Menis J, Mok TS, Nestle U, Passaro A, Peters S, Planchard D, Smit EF, Solomon BJ, Veronesi G, Reck M. Oncogene-addicted metastatic non-small-cell lung cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2023; 34:339-357. [PMID: 36872130 DOI: 10.1016/j.annonc.2022.12.009] [Citation(s) in RCA: 323] [Impact Index Per Article: 161.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 01/24/2023] Open
Affiliation(s)
- L E Hendriks
- Department of Pulmonology, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - K M Kerr
- Aberdeen Royal Infirmary, Aberdeen University Medical School, Aberdeen, UK
| | - J Menis
- Medical Oncology Department, University and Hospital Trust of Verona, Verona, Italy
| | - T S Mok
- Department of Clinical Oncology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - U Nestle
- Department of Radiation Oncology, University Hospital Freiburg, Freiburg, Germany; Department of Radiation Oncology, Kliniken Maria Hilf, Moenchengladbach, Germany
| | - A Passaro
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - S Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland
| | - D Planchard
- Department of Medical Oncology, Thoracic Group, Gustave-Roussy Villejuif, France
| | - E F Smit
- Thoracic Oncology Service, Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Pulmonary Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - B J Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - G Veronesi
- Faculty of Medicine and Surgery-Vita-Salute San Raffaele University, Milan, Italy; Division of Thoracic Surgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M Reck
- Department of Thoracic Oncology, Airway Research Center North, German Center for Lung Research, Lung Clinic, Grosshansdorf, Germany
| |
Collapse
|
54
|
Silginer M, Papa E, Szabó E, Vasella F, Pruschy M, Stroh C, Roth P, Weiss T, Weller M. Immunological and tumor-intrinsic mechanisms mediate the synergistic growth suppression of experimental glioblastoma by radiotherapy and MET inhibition. Acta Neuropathol Commun 2023; 11:41. [PMID: 36915128 PMCID: PMC10009975 DOI: 10.1186/s40478-023-01527-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/05/2023] [Indexed: 03/14/2023] Open
Abstract
The hepatocyte growth factor (HGF)/MET signaling pathway has been proposed to be involved in the resistance to radiotherapy of glioblastoma via proinvasive and DNA damage response pathways.Here we assessed the role of the MET pathway in the response to radiotherapy in vitro and in vivo in syngeneic mouse glioma models. We find that the murine glioma cell lines GL-261, SMA-497, SMA-540 and SMA-560 express HGF and its receptor MET and respond to exogenous HGF with MET phosphorylation. Glioma cell viability or proliferation are unaffected by genetic or pharmacological MET inhibition using tepotinib or CRISPR/Cas9-engineered Met gene knockout and MET inhibition fails to sensitize glioma cells to irradiation in vitro. In contrast, the combination of tepotinib with radiotherapy prolongs survival of orthotopic SMA-560 or GL-261 glioma-bearing mice compared with radiotherapy or tepotinib treatment alone. Synergy is lost when such experiments are conducted in immunodeficient Rag1-/- mice, and, importantly, also when Met gene expression is disrupted in the tumor cells. Combination therapy suppresses a set of pro-inflammatory mediators including matrix metalloproteases that are upregulated by radiotherapy alone and that have been linked to poor outcome in glioblastoma. Several of these mediators are positively regulated by transforming growth factor (TGF)-β, and pSMAD2 levels as a surrogate marker of TGF-β pathway activity are suppressed by combination treatment. We conclude that synergistic suppression of experimental syngeneic glioma growth by irradiation and MET inhibition requires MET expression in the tumor as well as an intact immune system. Clinical evaluation of this combined strategy in newly diagnosed glioblastoma is warranted.
Collapse
Affiliation(s)
- Manuela Silginer
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital of Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland.
| | - Eleanna Papa
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University of Zurich, Zurich, Switzerland
| | - Emese Szabó
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University of Zurich, Zurich, Switzerland
| | - Flavio Vasella
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital of Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | - Martin Pruschy
- Laboratory for Molecular Radiobiology, Department of Radiation Oncology, University Hospital and University of Zurich, Zurich, Switzerland
| | | | - Patrick Roth
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital of Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland.,Laboratory of Molecular Neuro-Oncology, Department of Neurology, University of Zurich, Zurich, Switzerland
| | - Tobias Weiss
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital of Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | - Michael Weller
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital of Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland.,Laboratory of Molecular Neuro-Oncology, Department of Neurology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
55
|
Kraus S. Dramatic response to tepotinib in a highly symptomatic patient with metastatic MET exon 14 skipping non-small cell lung cancer and poor performance status: A case report. CURRENT PROBLEMS IN CANCER: CASE REPORTS 2023. [DOI: 10.1016/j.cpccr.2023.100230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2023] Open
|
56
|
D’Aiello A, Miao E, Cheng H. Advances in the Management of Central Nervous System Metastases in Non-Small Cell Lung Cancer. Cancers (Basel) 2023; 15:cancers15030844. [PMID: 36765802 PMCID: PMC9913558 DOI: 10.3390/cancers15030844] [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: 12/31/2022] [Revised: 01/24/2023] [Accepted: 01/28/2023] [Indexed: 01/31/2023] Open
Abstract
Central nervous system (CNS) metastases are common among patients with non-small cell lung cancer (NSCLC). While the presence of brain metastases has historically portended poor prognosis, recent advances in local and systemic therapies have greatly improved outcomes for NSCLC patients with CNS involvement. Stereotactic radiology surgery (SRS) has emerged as an effective radiotherapy technique with fewer toxicities compared to whole brain radiotherapy (WBRT). Furthermore, multi-generation tyrosine kinase inhibitors (TKIs) with CNS overall response rates (ORR) of up to 70-80% are now an accepted first-line approach for a subset of advanced NSCLC patients with targetable molecular alterations. In addition, while the CNS was once considered an immunologic sanctuary site, growing evidence shows that immune checkpoint inhibitors (ICIs) can induce durable responses in brain metastases as well. Ongoing efforts to optimize CNS metastases management are necessary to refine multimodal treatment approaches and develop new therapeutics with better CNS penetrance.
Collapse
Affiliation(s)
- Angelica D’Aiello
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Emily Miao
- Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Haiying Cheng
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Correspondence: ; Tel.: +1-718-430-2430
| |
Collapse
|
57
|
Souza VGP, de Araújo RP, Santesso MR, Seneda AL, Minutentag IW, Felix TF, Hamamoto Filho PT, Pewarchuk ME, Brockley LJ, Marchi FA, Lam WL, Drigo SA, Reis PP. Advances in the Molecular Landscape of Lung Cancer Brain Metastasis. Cancers (Basel) 2023; 15:722. [PMID: 36765679 PMCID: PMC9913505 DOI: 10.3390/cancers15030722] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/16/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023] Open
Abstract
Lung cancer is one of the most frequent tumors that metastasize to the brain. Brain metastasis (BM) is common in advanced cases, being the major cause of patient morbidity and mortality. BMs are thought to arise via the seeding of circulating tumor cells into the brain microvasculature. In brain tissue, the interaction with immune cells promotes a microenvironment favorable to the growth of cancer cells. Despite multimodal treatments and advances in systemic therapies, lung cancer patients still have poor prognoses. Therefore, there is an urgent need to identify the molecular drivers of BM and clinically applicable biomarkers in order to improve disease outcomes and patient survival. The goal of this review is to summarize the current state of knowledge on the mechanisms of the metastatic spread of lung cancer to the brain and how the metastatic spread is influenced by the brain microenvironment, and to elucidate the molecular determinants of brain metastasis regarding the role of genomic and transcriptomic changes, including coding and non-coding RNAs. We also present an overview of the current therapeutics and novel treatment strategies for patients diagnosed with BM from NSCLC.
Collapse
Affiliation(s)
- Vanessa G. P. Souza
- Molecular Oncology Laboratory, Experimental Research Unit, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Rachel Paes de Araújo
- Molecular Oncology Laboratory, Experimental Research Unit, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Mariana R. Santesso
- Molecular Oncology Laboratory, Experimental Research Unit, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Ana Laura Seneda
- Molecular Oncology Laboratory, Experimental Research Unit, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Iael W. Minutentag
- Molecular Oncology Laboratory, Experimental Research Unit, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Tainara Francini Felix
- Molecular Oncology Laboratory, Experimental Research Unit, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Pedro Tadao Hamamoto Filho
- Department of Neurology, Psychology and Psychiatry, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | | | - Liam J. Brockley
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Fábio A. Marchi
- Faculty of Medicine, University of São Paulo, São Paulo 01246-903, Brazil
| | - Wan L. Lam
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Sandra A. Drigo
- Molecular Oncology Laboratory, Experimental Research Unit, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Patricia P. Reis
- Molecular Oncology Laboratory, Experimental Research Unit, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| |
Collapse
|
58
|
[Research Progresses in the Treatment of NSCLC with MET Gene Variants: A Riview]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:877-887. [PMID: 36617474 PMCID: PMC9845091 DOI: 10.3779/j.issn.1009-3419.2022.101.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mesenchymal-epithelial transition factor (MET) has long been considered as the most crucial and promising driver gene in the occurrence and development of non-small cell lung cancer (NSCLC), except for epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), and c-ROS oncogene 1 receptor tyrosine kinase (ROS1). In recent years, therapeutic drugs targeting MET have been continuously developed and applied in clinical practice. First, the curative effect of NSCLC patients with MET exon 14 skipping mutations has been further improved. In addition, when MET amplification occurs after resistance to EGFR tyrosine kinase inhibitors (EGFR-TKIs) in patients with advanced EGFR-mutant NSCLC, the combination of MET-TKIs and EGFR-TKIs has brought significant survival benefits and many other advances. This article reviews the treatment progress of NSCLC patients with different types of MET variants under different circumstances, which provides reference for the selection of clinical treatment strategies.
.
Collapse
|
59
|
Ma X, Zheng D, Zhang J, Dong Y, Li L, Jie B, Jiang S. Clinical outcomes of vinorelbine loading CalliSpheres beads in the treatment of previously treated advanced lung cancer with progressive refractory obstructive atelectasis. Front Bioeng Biotechnol 2022; 10:1088274. [PMID: 36605253 PMCID: PMC9810263 DOI: 10.3389/fbioe.2022.1088274] [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: 11/03/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Drug-eluting beads bronchial arterial chemoembolization (DEB-BACE) has been used in the treatment of locally advanced lung cancer and has the potential to improve outcomes and reduce recurrence. However, DEB-BACE shows a poor therapeutic effect in advanced lung cancer after failure of multiple therapies. This study assessed the effect of DEB-BACE in the treatment of progressive lung cancer with refractory obstructive atelectasis. Methods: Progressive advanced lung cancer patients with refractory obstructive atelectasis were voluntarily enrolled in this study after failure of multiple conventional therapies. Baseline information, DEB-BACE treatment process, and changes in clinical symptoms were recorded. The primary endpoints were the objective response rate (ORR) and improvement rate of dyspnea. The secondary endpoints were time-to-progression (TTP), overall survival (OS), and rate of pulmonary re-expansion. Treatment-related adverse events and serious adverse events were analyzed to assess the safety of DEB-BACE. The Cox regression model was performed to analyze the possible factors impacting prognosis of DEB-BACE. Results: DEB-BACE was successfully performed with CalliSpheres beads loaded with vinorelbine in the 20 enrolled patients. ORR and disease control rate were 80% and 85%, respectively, at the first follow-up (43.4 ± 15.26 days). The improvement rate of dyspnea was 85% and 80% at 1 week and 1 month (p < 0.0001, p < 0.0001), respectively. TTP was 41.25 ± 14.43 days and 89.55 ± 61.7 days before and after DEB-BACE, respectively; DEB-BACE delayed the progression of advanced lung cancer (p < 0.0001). OS was 238.03 ± 33.74 days (95% confidence interval: 171.9-304.16). The rate of pulmonary re-expansion was 80% at the first follow-up. The reasons for poor prognosis were tumor necrosis, longer disease duration, and pulmonary atelectasis duration (p = 0.012, p = 0.038, p = 0.029). Massive hemoptysis was observed in two cases, and one patient died of asphyxia caused by hemoptysis. Moderate hemoptysis occurred in one case. All three adverse events were considered as the result of the tumor cavity after DEB-BACE. Conclusion: DEB-BACE loaded with vinorelbine is a feasible option for progressive advanced lung cancer with obstructive atelectasis after failure of other treatments.
Collapse
Affiliation(s)
- Xu Ma
- Department of Radiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Di Zheng
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jie Zhang
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu Dong
- Department of Radiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lingling Li
- Department of Radiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bing Jie
- Department of Radiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China,*Correspondence: Bing Jie, ; Sen Jiang,
| | - Sen Jiang
- Department of Radiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China,*Correspondence: Bing Jie, ; Sen Jiang,
| |
Collapse
|
60
|
Zhu X, Lu Y, Lu S. Landscape of Savolitinib Development for the Treatment of Non-Small Cell Lung Cancer with MET Alteration-A Narrative Review. Cancers (Basel) 2022; 14:cancers14246122. [PMID: 36551608 PMCID: PMC9776447 DOI: 10.3390/cancers14246122] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is increasingly being treated with targeted therapies. Savolitinib (Orpathys®) is highly selective mesenchymal epithelial transition (MET)-tyrosine kinase inhibitor (TKI), which is conditionally approved in China for advanced NSCLC with MET exon 14 skipping mutations (METex14). This article summarizes the clinical development of savolitinib, as a monotherapy in NSCLC with METex14 mutation and in combination with epidermal growth factor receptor (EGFR) inhibitor in post EGFR-TKI resistance NSCLC due to MET-based acquired resistance. Preclinical models demonstrated anti-tumor activities in MET-driven cancer cell line and xenograft tumor models. The Phase Ia/Ib study established an optimized, recommended phase II dose in Chinese NSCLC patients, while TATTON study of savolitinib plus osimertinib in patients with EGFR mutant, MET-amplified and TKI-progressed NSCLC showed beneficial efficacy with acceptable safety profile. In a pivotal phase II study, Chinese patients with pulmonary sarcomatoid carcinoma, brain metastasis and other NSCLC subtype positive for METex14 mutation showed notable responses and acceptable safety profile with savolitinib. Currently, results from ongoing clinical trials are eagerly anticipated to confirm the efficacious and safety benefits of savolitinib as monotherapy and in combination with EGFR-TKI in acquired resistance setting in advanced NSCLC and its subtypes with MET alterations.
Collapse
Affiliation(s)
- Xiaokuan Zhu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yao Lu
- AstraZeneca China, Shanghai 201200, China
| | - Shun Lu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence:
| |
Collapse
|
61
|
Xu L, Wang F, Luo F. MET-targeted therapies for the treatment of non-small-cell lung cancer: A systematic review and meta-analysis. Front Oncol 2022; 12:1013299. [PMID: 36387098 PMCID: PMC9646943 DOI: 10.3389/fonc.2022.1013299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/17/2022] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Dysregulation of the mesenchymal epithelial transition (MET) pathway contributes to poor clinical outcomes in patients with non-small cell lung cancer (NSCLC). Numerous clinical trials are currently investigating several therapies based on modulation of the MET pathway. OBJECTIVES This study aimed to systematically evaluate the activity and safety of MET inhibitors in patients with NSCLC. METHODS We searched PubMed, Embase, and the Cochrane Library from inception to June 02, 2022. The objective response rate (ORR) and disease control rate (DCR) were extracted as the main outcomes and pooled using the weighted mean proportion with fixed- or random-effects models in cases of significant heterogeneity (I 2>50%). Safety analysis was performed based on adverse events reported in all studies. RESULTS Eleven studies (882 patients) were included in the meta-analysis. The pooled ORR was 28.1% (95% confidence interval [CI], 0.223-0.354), while the pooled DCR was 69.1% (95% CI, 0.631-0.756). ORRs were higher for tepotinib (44.7% [95% CI, 0.365-0.530]) and savolitinib (42.9% [95% CI, 0.311-0.553]) than for other types of MET inhibitors. Patients with NSCLC with exon 14 skipping exhibited higher ORRs (39.3% (95% CI, 0.296-0.522)) and DCRs (77.8% (95% CI, 0.714-0.847)) than those with MET protein overexpression or amplification. Intracranial response rate and intracranial disease control rates were 40.1% (95% CI, 0.289-0.556) and 95.4% (95% CI, 0.892-0.100), respectively. Adverse events were mild (grade 1 to 2) in 87.2% of patients. Common adverse events above grade 3 included lower extremity edema (3.5% [95% CI, 0.027-0.044]), alanine aminotransferase (ALT) elevation (2.4% [95% CI, 0.014-0.033]), and lipase elevation (2.2% [95% CI, 0.016-0.031]). CONCLUSION MET inhibitors, which exhibited a satisfactory safety profile in the current study, may become a new standard of care for addressing MET dysregulation in patients with advanced or metastatic NSCLC, and even in those with brain metastases, particularly tepotinib, savolitinib and capmatinib. Further randomized trials are required to establish standard predictive biomarkers for MET therapies and to compare the effects of different MET inhibitors in NSCLC with MET dysregulation.
Collapse
Affiliation(s)
- Linrui Xu
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Pulmonary Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Faping Wang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Pulmonary Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fengming Luo
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Pulmonary Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
62
|
Tseng LW, Chang JWC, Wu CE. Safety of Tepotinib Challenge after Capmatinib-Induced Pneumonitis in a Patient with Non-Small Cell Lung Cancer Harboring MET Exon 14 Skipping Mutation: A Case Report. Int J Mol Sci 2022; 23:ijms231911809. [PMID: 36233109 PMCID: PMC9570266 DOI: 10.3390/ijms231911809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
The targeted agents capmatinib and tepotinib provide a new treatment for patients with non-small cell lung cancer (NSCLC) with MET exon 14 skipping mutation (METex14). However, drug-induced pneumonitis is an uncommon but threatening adverse effect found in patients treated with both capmatinib and tepotinib. The safety of treating a patient with a MET inhibitor after drug-induced pneumonitis by another MET inhibitor remains unclear. Here, we present a case of a patient with NSCLC harboring a METex14 who was treated with a standard dose of tepotinib after advanced capmatinib-induced pneumonitis and did not present pneumonitis relapse. Tepotinib may be a safe option when medical professionals consider switching MET inhibitors after patients experience pneumonitis.
Collapse
Affiliation(s)
- Liang-Wei Tseng
- Division of Chinese Internal Medicine, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan
| | - John Wen-Cheng Chang
- Division of Haematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, 5, Fu-Hsing Street, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Chiao-En Wu
- Division of Haematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, 5, Fu-Hsing Street, Kwei-Shan, Taoyuan 33302, Taiwan
- Correspondence: ; Tel.: +886-3-3281200; Fax: +886-3-3278211
| |
Collapse
|
63
|
Remon J, Hendriks LE, Mountzios G, García-Campelo R, Saw SP, Uprety D, Recondo G, Villacampa G, Reck M. MET alterations in NSCLC—Current Perspectives and Future Challenges. J Thorac Oncol 2022; 18:419-435. [PMID: 36441095 DOI: 10.1016/j.jtho.2022.10.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/17/2022] [Accepted: 10/19/2022] [Indexed: 11/24/2022]
Abstract
Targeted therapies have revolutionized the treatment and improved the outcome for oncogene-driven NSCLC and an increasing number of oncogenic driver therapies have become available. For MET-dysregulated NSCLC (especially MET exon 14 skipping mutations and MET-amplifications, which is one of the most common bypass mechanisms of resistance in oncogene-addicted NSCLC), several anti-MET-targeted therapies have been approved recently (MET exon 14 skipping mutation) and multiple others are in development. In this narrative review, we summarize the role of MET as an oncogenic driver in NSCLC, discuss the different testing methods for exon 14 skipping mutations, gene amplification, and protein overexpression, and review the existing data and ongoing clinical trials regarding targeted therapies in MET-altered NSCLC. As immunotherapy with or without chemotherapy has become the standard of care for advanced NSCLC, immunotherapy data for MET-dysregulated NSCLC are put into perspective. Finally, we discuss future challenges in this rapidly evolving landscape.
Collapse
|
64
|
Ahn L, Alexander T, Vlassak S, Berghoff K, Lemmens L. Tepotinib: Management of Adverse Events in Patients With MET Exon 14 Skipping Non-Small Cell Lung Cancer. Clin J Oncol Nurs 2022; 26:543-551. [PMID: 36108212 PMCID: PMC10034867 DOI: 10.1188/22.cjon.543-551] [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] [Indexed: 11/17/2022]
Abstract
BACKGROUND Tepotinib, a highly selective, oral, once-daily MET inhibitor, has been approved for treatment of metastatic MET exon 14 skipping non-small cell lung cancer. OBJECTIVES This article provides nurse-specific recommendations for identification and management of tepotinib adverse events (AEs). METHODS Guidance on monitoring and proactive/reactive AE management was developed based on published literature and real-world nursing experience. Case studies of VISION trial participants were summarized to illustrate key principles. FINDINGS Tepotinib AEs are generally mild to moderate and manageable, and can include peripheral edema, hypoalbuminemia, nausea, diarrhea, and creatinine increase. Alongside supportive care, tepotinib interruption and dose reduction is recommended for grade 3 AEs. For peripheral edema, proactive monitoring is crucial, and treatment interruption (including frequent, short treatment holidays) should be considered early. Nursing management of tepotinib AEs includes proactive monitoring, patient education, and interprofessional team coordination.
Collapse
Affiliation(s)
- Linda Ahn
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
- Corresponding author. Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA. Linda Ahn
| | - Terri Alexander
- MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Soetkin Vlassak
- the healthcare business of Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Karin Berghoff
- the healthcare business of Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | | |
Collapse
|
65
|
Management of Peripheral Edema in Patients with MET Exon 14-Mutated Non-small Cell Lung Cancer Treated with Small Molecule MET Inhibitors. Target Oncol 2022; 17:597-604. [PMID: 36087188 PMCID: PMC9512730 DOI: 10.1007/s11523-022-00912-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2022] [Indexed: 11/03/2022]
Abstract
Small molecule mesenchymal-epithelial transition (MET) inhibitors, such as crizotinib, capmatinib, and tepotinib, are treatment options for metastatic non-small cell lung cancer (NSCLC) in adult patients whose tumors have a mutation that leads to MET exon 14 skipping. In clinical trials, these MET inhibitors were associated with a high incidence of peripheral edema, although this was generally mild-to-moderate in severity. There is limited information about the mechanism involved in MET inhibitor-induced peripheral edema. Perturbation of hepatocyte growth factor (HGF)/MET signaling may disrupt the permeability balance in the vascular endothelium and thus promote edema development. Another potential mechanism is through effects on renal function, although this is unlikely to be the primary mechanism. Because edema is common in cancer patients and may not necessarily be caused by the cancer treatment, or other conditions that have similar symptoms to peripheral edema, a thorough assessment is required to ascertain the underlying cause. Before starting MET-inhibitor therapy, patients should be educated about the possibility of developing peripheral edema. Patient limb volume should be measured before initiating treatment, to aid assessment if symptoms develop. Since the exact mechanism of MET inhibitor-induced edema is unknown, management is empiric, with common approaches including compression stockings, specific exercises, massage, limb elevation, and/or diuretic treatment. Although not usually required, discontinuation of MET inhibitor treatment generally resolves peripheral edema. Early diagnosis and management, as well as patient information and education, are vital to decrease the clinical burden associated with edema, and to reinforce capmatinib treatment adherence.
Collapse
|
66
|
Ai X, Yu Y, Zhao J, Sheng W, Bai J, Fan Z, Liu X, Ji W, Chen R, Lu S. Comprehensive analysis of MET mutations in NSCLC patients in a real-world setting. Ther Adv Med Oncol 2022; 14:17588359221112474. [PMID: 35860830 PMCID: PMC9290171 DOI: 10.1177/17588359221112474] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Aberrant mesenchymal–epithelial transition/hepatocyte growth factor (MET/HGF) regulation presented in a wide variety of human cancers. MET exon 14 skipping, copy number gain (CNG), and kinase domain mutations/arrangements were associated with increased MET activity, and considered to be oncogenic drivers of non-small cell lung cancers (NSCLCs). Methods: We retrospectively analyzed 564 patients with MET alterations. MET alterations were classified into structural mutations or small mutations. MET CNG, exon 14 skipping, gain of function (GOF) mutations, and kinase domain rearrangement were defined as actionable mutations. Results: Six hundred thirty-two MET mutations were identified including 199 CNG, 117 exon 14 skipping, 12 GOF mutations, and 2 actionable fusions. Higher percentage of MET structural alterations (CNG + fusion) were detected in advanced NSCLC patients. Moreover, MET CNG was enriched while exon 14 skipping was rare in epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKI)-treated advanced NSCLC patients. Ten of the 12 MET GOF mutations were also in EGFR-TKI-treated patients. Fifteen (68.1%) of the 22 patients treated with crizotinib or savolitinib had a partial response. Interestingly, one patient had a great response to savolitinib with a novel MET exon 14 skipping mutation identified after failure of immune-checkpoint inhibitor. Conclusions: Half of the MET alterations were actionable mutations. MET CNG, exon 14 skipping and GOF mutations had different distribution in different clinical scenario but all defined a molecular subgroup of NSCLCs for which MET inhibition was active.
Collapse
Affiliation(s)
- Xinghao Ai
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yongfeng Yu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Wang Sheng
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jing Bai
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zaiwen Fan
- Department of Medical Oncology, Air Force Medical Center, PLA, Beijing, China
| | - Xuemei Liu
- Department of Radiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenxiang Ji
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Rongrong Chen
- Geneplus-Beijing, Floor 9, Building 6, Medical Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, No.241, Huaihai West Road, Shanghai 200032, China
| |
Collapse
|
67
|
Uehara Y, Watanabe K, Hakozaki T, Yomota M, Hosomi Y. Efficacy of first-line immune checkpoint inhibitors in patients with advanced NSCLC with KRAS, MET, FGFR, RET, BRAF, and HER2 alterations. Thorac Cancer 2022; 13:1703-1711. [PMID: 35491960 PMCID: PMC9161348 DOI: 10.1111/1759-7714.14448] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/14/2022] [Accepted: 04/16/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND In patients with non-small cell lung cancer (NSCLC) harboring driver alterations, the efficacy of immune checkpoint inhibitors (ICIs) remains uncertain. Our study aimed to examine the first-line ICI efficacy in patients with NSCLC harboring KRAS, MET, FGFR, RET, BRAF, and HER2 alterations in a real-world setting. METHODS This single-center, retrospective cohort study included patients with advanced NSCLC harboring KRAS, MET, FGFR, RET, BRAF, HER2 alterations or driver-negative, and were treated with first-line ICI therapy. Best overall response, progression-free survival (PFS), and overall survival (OS) were evaluated. RESULTS Seventy-eight patients with NSCLC were included (median age, 72 years): 67% were men, 15% were never-smokers, and 83% had adenocarcinoma. The driver alterations involved KRAS (n = 21), MET (n = 6), FGFR (n = 3), RET (n = 2), BRAF (n = 2), HER2 (n = 1), and driver-negative (n = 43). The partial responses for KRAS, MET, FGFR, RET, BRAF, HER2, and driver-negative were 57%, 50%, 100%, 50%, 100%, 0%, and 47%, respectively. The median PFS (months) was 16.2 (95% confidence interval [CI]: 6.3- not reached [NR]) for KRAS, 2.8 (95% CI: 2.7-NR) for MET, 11.7 (95% CI: 5.9-NR) for other alterations (FGFR, RET, BRAF, and HER2), and 10.0 (95% CI: 3.7-14.3) for driver-negative, respectively. The median OS (months) was 31.3 (95% CI: 9.0-NR) for KRAS, not reached for MET, 23.5 (95% CI: 18.3-NR) for other alterations, and 21.1 (95% CI: 15.2-NR) for driver-negative, respectively. CONCLUSIONS The benefit of the first-line ICI was similar in advanced NSCLC regardless of the driver alterations, except for MET alterations.
Collapse
Affiliation(s)
- Yuji Uehara
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases CenterKomagome HospitalBunkyo‐kuTokyoJapan
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityBunkyo‐kuTokyoJapan
| | - Kageaki Watanabe
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases CenterKomagome HospitalBunkyo‐kuTokyoJapan
| | - Taiki Hakozaki
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases CenterKomagome HospitalBunkyo‐kuTokyoJapan
- Department of Life Science and Medical BioscienceWaseda UniversityShinjukuTokyoJapan
| | - Makiko Yomota
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases CenterKomagome HospitalBunkyo‐kuTokyoJapan
| | - Yukio Hosomi
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases CenterKomagome HospitalBunkyo‐kuTokyoJapan
| |
Collapse
|
68
|
Alvarez-Breckenridge C, Remon J, Piña Y, Nieblas-Bedolla E, Forsyth P, Hendriks L, Brastianos PK. Emerging Systemic Treatment Perspectives on Brain Metastases: Moving Toward a Better Outlook for Patients. Am Soc Clin Oncol Educ Book 2022; 42:1-19. [PMID: 35522917 DOI: 10.1200/edbk_352320] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The diagnosis of brain metastases has historically been a dreaded, end-stage complication of systemic disease. Additionally, with the increasing effectiveness of systemic therapies that prolong life expectancy and improved imaging tools, the incidence of intracranial progression is becoming more common. Within this context, there has been increasing attention directed at understanding the molecular underpinnings of intracranial progression. Exploring the unique features of brain metastases compared with their extracranial counterparts to identify aberrant signaling pathways, which can be targeted pharmacologically, may help lead to new treatments for this patient population. Additionally, critical discoveries outside the sphere of the central nervous system are increasingly being applied to brain metastases with the emergence of immune checkpoint inhibition, becoming a prevalent treatment option for patients with brain metastases across multiple histologies. As novel treatment strategies are considered, they require thoughtful incorporation of agents that can cross the blood-brain barrier and can synergize with pre-existing agents through rational combinations. Lastly, as clinicians and scientists continue to understand key molecular features of these tumors, they will continue to influence the treatment algorithms that are developing for the management of these patients. Due to the complexity of treatment decisions for patients with brain metastases, an emerging tool is the utilization of multidisciplinary brain metastasis tumor boards to ensure optimal treatment decisions are made and that patients are provided access to applicable clinical trials. Looking to the future, the collective effort to understand the various tumor-intrinsic and tumor-extrinsic factors that promote central nervous system seeding and propagation will have the potential to change the clinical trajectory for these patients.
Collapse
Affiliation(s)
| | - Jordi Remon
- Department of Medical Oncology, HM CIOCC Barcelona (Centro Integral Oncológico Clara Campal), Hospital HM Delfos, HM Hospitales, Barcelona, Spain
| | - Yolanda Piña
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL
| | | | - Peter Forsyth
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL
| | - Lizza Hendriks
- Department of Pulmonary Diseases - GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, Netherlands
| | | |
Collapse
|
69
|
Veillon R, Sakai H, Le X, Felip E, Cortot AB, Egbert S, Park K, Griesinger F, Britschgi C, Wu YL, Melosky B, Baijal S, Jr GDC, Sedova M, Berghoff K, Otto G, Paik PK. Safety of Tepotinib in Patients with MET Exon 14 Skipping NSCLC and Recommendations for Management. Clin Lung Cancer 2022; 23:320-332. [PMID: 35466070 PMCID: PMC10068910 DOI: 10.1016/j.cllc.2022.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 11/03/2022]
Abstract
INTRODUCTION The MET inhibitor tepotinib demonstrated durable clinical activity in patients with advanced MET exon 14 (METex14) skipping NSCLC. We report detailed analyses of adverse events of clinical interest (AECIs) in VISION, including edema, a class effect of MET inhibitors. PATIENTS AND METHODS Incidence, management, and time to first onset/resolution were analyzed for all-cause AECIs, according to composite categories (edema, hypoalbuminemia, creatinine increase, and ALT/AST increase) or individual preferred terms (pleural effusion, nausea, diarrhea, and vomiting), for patients with METex14 skipping NSCLC in the phase II VISION trial. RESULTS Of 255 patients analyzed (median age: 72 years), edema, the most common AECI, was reported in 69.8% (grade 3, 9.4%; grade 4, 0%). Median time to first edema onset was 7.9 weeks (range: 0.1-58.3). Edema was manageable with supportive measures, dose reduction (18.8%), and/or treatment interruption (23.1%), and rarely prompted discontinuation (4.3%). Other AECIs were also manageable and predominantly mild/moderate: hypoalbuminemia, 23.9% (grade 3, 5.5%); pleural effusion, 13.3% (grade ≥ 3, 5.1%); creatinine increase, 25.9% (grade 3, 0.4%); nausea, 26.7% (grade 3, 0.8%), diarrhea, 26.3% (grade 3, 0.4%), vomiting 12.9% (grade 3, 1.2%), and ALT/AST increase, 12.2% (grade ≥ 3, 3.1%). GI AEs typically occurred early and resolved in the first weeks. CONCLUSION Tepotinib was well tolerated in the largest trial of a MET inhibitor in METex14 skipping NSCLC. The most frequent AEs were largely mild/moderate and manageable with supportive measures and/or dose reduction/interruption, and caused few withdrawals in this elderly population.
Collapse
|
70
|
Rosner S, Spira AI. Has the Enemy "MET" Its Match? Subgroup Analysis Results from VISION Study. Clin Cancer Res 2022; 28:1055-1057. [PMID: 34983790 DOI: 10.1158/1078-0432.ccr-21-4109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/09/2021] [Accepted: 12/28/2021] [Indexed: 11/16/2022]
Abstract
Emerging therapies have recently received approval for the treatment of MET exon 14 skipping non-small cell lung cancer (NSCLC). Further characterization of these therapies, such as tepotinib, is needed to identify patient subgroups most likely to derive benefit from these novel agents.See related article by Le et al., p. XXXX.
Collapse
Affiliation(s)
- Samuel Rosner
- Medical Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Alexander I Spira
- Virginia Cancer Specialists (VCS) Research Institute, Fairfax, Virginia. .,NEXT Oncology-Virginia, Fairfax, Virginia.,US Oncology Research, The Woodlands, Texas
| |
Collapse
|
71
|
Batra U, Nathany S. MET: A narrative review of exon 14 skipping mutation in non-small-cell lung carcinoma. CANCER RESEARCH, STATISTICS, AND TREATMENT 2022. [DOI: 10.4103/crst.crst_158_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
72
|
Brain penetration and efficacy of tepotinib in orthotopic patient-derived xenograft models of MET-driven non-small cell lung cancer brain metastases. Lung Cancer 2021; 163:77-86. [PMID: 34942492 DOI: 10.1016/j.lungcan.2021.11.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/25/2021] [Accepted: 11/28/2021] [Indexed: 11/20/2022]
Abstract
Central nervous system-penetrant therapies with intracranial efficacy against non-small cell lung cancer (NSCLC) brain metastases are urgently needed. We report preclinical studies investigating brain penetration and intracranial activity of the MET inhibitor tepotinib. After intravenous infusion of tepotinib in Wistar rats (n = 3), mean (±standard deviation) total tepotinib concentration was 2.87-fold higher in brain (505 ± 22 ng/g) than plasma (177 ± 20 ng/mL). In equilibrium dialysis experiments performed in triplicate, mean tepotinib unbound fraction was 0.35% at 0.3 and 3.0 µM tepotinib in rat brain tissue, and 4.0% at 0.3 and 1.0 µM tepotinib in rat plasma. The calculated unbound brain-to-plasma ratio was 0.25, indicating brain penetration sufficient for intracranial target inhibition. Of 20 screened subcutaneous patient-derived xenograft (PDX) models from lung cancer brain metastases (n = 1), two NSCLC brain metastases models (LU5349 and LU5406) were sensitive to the suboptimal dose of tepotinib of 30 mg/kg/qd (tumor volume change [%TV]: -12% and -88%, respectively). Molecular profiling (nCounter®; NanoString) revealed high-level MET amplification in both tumors (mean MET gene copy number: 11.2 and 24.2, respectively). Tepotinib sensitivity was confirmed for both subcutaneous models at a clinically relevant dose (125 mg/kg/qd; n = 5). LU5349 and LU5406 were orthotopically implanted into brains of mice and monitored by magnetic resonance imaging (MRI). Tepotinib 125 mg/kg/qd induced pronounced tumor regression, including complete or near-complete regressions, compared with vehicle in both orthotopic models (n = 10; median %TV: LU5349, -84%; LU5406, -63%). Intracranial antitumor activity of tepotinib did not appear to correlate with blood-brain barrier leakiness assessed in T1-weighted gadolinium contrast-enhanced MRI.
Collapse
|