1
|
Kounatidou NE, Vitkos E, Palioura S. Ocular surface squamous neoplasia: Update on genetics, epigenetics and opportunities for targeted therapy. Ocul Surf 2025; 35:1-14. [PMID: 39608452 DOI: 10.1016/j.jtos.2024.11.006] [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: 08/02/2024] [Revised: 11/09/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
PURPOSE The purpose of this review is to explore the molecular foundations of ocular surface squamous neoplasia (OSSN), focusing on the genetic and epigenetic aspects. While current management strategies include surgical excision and medical therapies, the understanding of OSSN's molecular basis remains limited, hindering the development of targeted treatments. METHODS A comprehensive MEDLINE search was conducted for literature published between January 1993 and October 2023. Only studies with original data on molecular, genetic, or epigenetic mechanisms, such as mutations, gene expression, and genetic predispositions were included. Articles were excluded if they focused solely on clinical management without addressing these factors, or if they were reviews, editorials, or opinion pieces. RESULTS The search yielded a total of 108 articles, out of which 39 articles met the criteria for further analysis. Investigations into OSSN have identified key DNA mutations in the TP53, HGF, EGFR, TERT, and CDKN2A genes, indicating common oncogenic pathways shared with other squamous cell carcinomas (SCCs). Significant epigenetic changes were identified, including DNA methylation, histone modifications, and altered miRNA expression patterns. Epigenetic dysregulation of critical tumor suppressors and oncoproteins, further highlight the complex genetic landscape of OSSN. CONCLUSION The molecular alterations identified in OSSN not only enhance our understanding of its biology but also have potential as novel biomarkers for early detection, prognostic evaluation, and as therapeutic targets. The identification of genetic and epigenetic markers in OSSN signifies progress towards personalized medicine approaches. Further studies and collaborative efforts are essential to validate these molecular markers and translate them into clinical practice, potentially revolutionizing OSSN management and improving patient outcomes.
Collapse
Affiliation(s)
| | - Evangelos Vitkos
- Department of Oral and Maxillofacial Surgery, Klinikum Dortmund, Dortmund, Germany
| | - Sotiria Palioura
- Department of Ophthalmology, University of Cyprus Medical School, Nicosia, Cyprus.
| |
Collapse
|
2
|
Elasbali AM, Anjum F, Al-Ghabban BA, Shafie A, Mohammad T, Hassan MI. Phytochemicals Neogitogenin and Samogenin Hold Potentials for Hepatocyte Growth Factor Receptor-Targeted Cancer Treatment. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:573-583. [PMID: 39388097 DOI: 10.1089/omi.2024.0169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Protein kinases are key targets for cancer therapies, with the c-Met receptor tyrosine kinase (MET) and its ligand, hepatocyte growth factor, playing a role in various cancers, including non-small cell lung cancer, gastric cancer, and hepatocellular carcinoma. Although small-molecule inhibitors have been designed to target MET, the development of drug resistance remains a significant challenge to advancing therapeutic strategies. In this study, we employed virtual screening of plant-based compounds sourced from the IMPPAT 2.0 databank to identify potent inhibitors of MET. Preliminary filtering based on the physicochemical parameters following Lipinski's rule of five and pan-assay interference compounds criteria were applied to prioritize hits. Subsequent molecular docking, pharmacokinetic evaluation, prediction of activity spectra for biologically active substances, and specificity assessments facilitated the identification of two promising phytochemicals, neogitogenin and samogenin. Both phytochemicals exhibited considerable drug-like properties with notable binding affinity and selectivity toward MET. Molecular dynamics simulation studies showed the conformational stability of MET with neogitogenin and samogenin. Taken together, these findings suggest that neogitogenin and samogenin hold potential as lead molecules for the development of MET-targeted therapeutics. We call for further evaluations of these phytochemicals in preclinical and experimental studies for anticancer drug discovery and development.
Collapse
Affiliation(s)
- Abdelbaset Mohamed Elasbali
- Department of Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Qurayyat, KSA, Saudi Arabia
| | - Farah Anjum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | | | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
3
|
Mer AH, Mirzaei Y, Misamogooe F, Bagheri N, Bazyari A, Keshtkaran Z, Meyfour A, Shahedi A, Amirkhani Z, Jafari A, Barpour N, Jahandideh S, Rezaei B, Nikmanesh Y, Abdollahpour-Alitappeh M. Progress of antibody-drug conjugates (ADCs) targeting c-Met in cancer therapy; insights from clinical and preclinical studies. Drug Deliv Transl Res 2024; 14:2963-2988. [PMID: 38597995 DOI: 10.1007/s13346-024-01564-3] [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] [Accepted: 02/29/2024] [Indexed: 04/11/2024]
Abstract
The cell-surface receptor tyrosine kinase c-mesenchymal-epithelial transition factor (c-Met) is overexpressed in a wide range of solid tumors, making it an appropriate target antigen for the development of anticancer therapeutics. Various antitumor c-Met-targeting therapies (including monoclonal antibodies [mAbs] and tyrosine kinases) have been developed for the treatment of c-Met-overexpressing tumors, most of which have so far failed to enter the clinic because of their efficacy and complications. Antibody-drug conjugates (ADCs), a new emerging class of cancer therapeutic agents that harness the target specificity of mAbs to deliver highly potent small molecules to the tumor with the minimal damage to normal cells, could be an attractive therapeutic approach to circumvent these limitations in patients with c-Met-overexpressing tumors. Of great note, there are currently nine c-Met-targeting ADCs being examined in different phases of clinical studies as well as eight preclinical studies for treating various solid tumors. The purpose of this study is to present a broad overview of clinical- and preclinical-stage c-Met-targeting ADCs.
Collapse
Affiliation(s)
- Ali Hussein Mer
- Department of Nursing, Mergasour Technical Institute, Erbil Polytechnic University, Erbil, Iraq
| | - Yousef Mirzaei
- Department of Medical Biochemical Analysis, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Fatemeh Misamogooe
- Student Research Committee, Larestan University of Medical Sciences, Larestan, Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, 8813733450, Iran
| | - Ahmadreza Bazyari
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Keshtkaran
- Department of Nursing, School of Nursing and Midwifery, Community Based Psychiatric Care Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Anna Meyfour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Shahedi
- Student Research Committee, Larestan University of Medical Sciences, Larestan, Iran
| | - Zahra Amirkhani
- Department of Nursing, School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Ameneh Jafari
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nesa Barpour
- Department of Genetics, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Saeed Jahandideh
- Department of Research and Development, Orchidgene Co, Tehran, 1387837584, Iran
| | - Behzad Rezaei
- Laparoscopy Research Center, Department of Surgery, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Fars Province, Iran
| | - Yousef Nikmanesh
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | | |
Collapse
|
4
|
Detchou D, Barrie U. Interleukin 6 and cancer resistance in glioblastoma multiforme. Neurosurg Rev 2024; 47:541. [PMID: 39231832 DOI: 10.1007/s10143-024-02783-5] [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: 08/16/2024] [Revised: 08/16/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
Despite unprecedented survival in patients with glioblastoma (GB), the aggressive primary brain cancer remains largely incurable and its mechanisms of treatment resistance have gained particular attention. The cytokine interleukin 6 (IL-6) and its receptor weave through the hallmarks of malignant gliomas and may represent a key vulnerability to GB. Known for activating the STAT3 pathway in autocrine fashion, IL-6 is amplified in GB and has been recognized as a negative biomarker for GB prognosis, rendering it a putative target of novel GB therapies. While it has been recognized as a biologically active component of GB for three decades only with concurrent advances in understanding of complementary immunotherapy has the concept of targeting IL-6 for a human clinical trial gained scientific footing.
Collapse
Affiliation(s)
- Donald Detchou
- School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| | - Umaru Barrie
- Department of Neurosurgery, New York University Grossman School of Medicine, New York City, NYC, USA
| |
Collapse
|
5
|
Contreras-Zentella ML, Alatriste-Contreras MG, Suárez-Cuenca JA, Hernández-Muñoz R. Gender effect of glucose, insulin/glucagon ratio, lipids, and nitrogen-metabolites on serum HGF and EGF levels in patients with diabetes type 2. Front Mol Biosci 2024; 11:1362305. [PMID: 38654922 PMCID: PMC11035728 DOI: 10.3389/fmolb.2024.1362305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Hepatocyte growth factor (HGF) exhibits potent growth-inducing properties across various tissues, while epidermal growth factor (EGF) acts as a molecular integration point for diverse stimuli. HGF plays a crucial role in hepatic metabolism, tissue repair, and offers protective effects on epithelial and non-epithelial organs, in addition to its involvement in reducing apoptosis and inflammation, underscoring its anti-inflammatory capabilities. The HGF-Met system is instrumental in hepatic metabolism and enhancing insulin sensitivity in animal diabetes models. Similarly, the EGF and its receptor tyrosine kinase family (EGFR) are critical in regulating cell growth, proliferation, migration, and differentiation in both healthy and diseased states, with EGF also contributing to insulin sensitivity. In this observational study, we aimed to identify correlations between serum levels of HGF and EGF, insulin, glucagon, glucose, and primary serum lipids in patients with type 2 diabetes mellitus (DM), taking into account the impact of gender. We noted differences in the management of glucose, insulin, and glucagon between healthy men and women, potentially due to the distinct influences of sexual hormones on the development of type 2 DM. Additionally, metabolites such as glucose, albumin, direct bilirubin, nitrites, and ammonia might influence serum levels of growth factors and hormones. In summary, our results highlight the regulatory role of insulin and glucagon in serum glucose and lipids, along with variations in HGF and EGF levels, which are affected by gender. This link is especially significant in DM, where impaired cell proliferation or repair mechanisms lead to metabolic changes. The gender-based differences in growth factors point to their involvement in the pathophysiology of the disease.
Collapse
Affiliation(s)
- Martha Lucinda Contreras-Zentella
- Departamento de Biología Celular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Martha Gabriela Alatriste-Contreras
- Departamento de Métodos Cuantitativos, División de Estudios Profesionales, Facultad de Economía, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Juan Antonio Suárez-Cuenca
- Departamento de Medicina Interna, Hospital General “Xoco”, Secretaría de Salud (SS), Mexico City, Mexico
| | - Rolando Hernández-Muñoz
- Departamento de Biología Celular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
6
|
Redmer T, Schumann E, Peters K, Weidemeier ME, Nowak S, Schroeder HWS, Vidal A, Radbruch H, Lehmann A, Kreuzer-Redmer S, Jürchott K, Radke J. MET receptor serves as a promising target in melanoma brain metastases. Acta Neuropathol 2024; 147:44. [PMID: 38386085 PMCID: PMC10884227 DOI: 10.1007/s00401-024-02694-1] [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: 10/16/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/23/2024]
Abstract
The development of brain metastases hallmarks disease progression in 20-40% of melanoma patients and is a serious obstacle to therapy. Understanding the processes involved in the development and maintenance of melanoma brain metastases (MBM) is critical for the discovery of novel therapeutic strategies. Here, we generated transcriptome and methylome profiles of MBM showing high or low abundance of infiltrated Iba1high tumor-associated microglia and macrophages (TAMs). Our survey identified potential prognostic markers of favorable disease course and response to immune checkpoint inhibitor (ICi) therapy, among them APBB1IP and the interferon-responsive gene ITGB7. In MBM with high ITGB7/APBB1IP levels, the accumulation of TAMs correlated significantly with the immune score. Signature-based deconvolution of MBM via single sample GSEA revealed enrichment of interferon-response and immune signatures and revealed inflammation, stress and MET receptor signaling. MET receptor phosphorylation/activation maybe elicited by inflammatory processes in brain metastatic melanoma cells via stroma cell-released HGF. We found phospho-METY1234/1235 in a subset of MBM and observed a marked response of brain metastasis-derived cell lines (BMCs) that lacked druggable BRAF mutations or developed resistance to BRAF inhibitors (BRAFi) in vivo to MET inhibitors PHA-665752 and ARQ197 (tivantinib). In summary, the activation of MET receptor in brain colonizing melanoma cells by stromal cell-released HGF may promote tumor self-maintenance and expansion and might counteract ICi therapy. Therefore, therapeutic targeting of MET possibly serves as a promising strategy to control intracranial progressive disease and improve patient survival.
Collapse
Affiliation(s)
- Torben Redmer
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria.
- Institute of Pathology, Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria.
| | - Elisa Schumann
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, CCCC (Campus Mitte), Berlin, Germany
| | - Kristin Peters
- Institute of Pathology, University Medicine Greifswald, Greifswald, Germany
| | - Martin E Weidemeier
- Department of Neurosurgery, University Medicine Greifswald, Greifswald, Germany
| | - Stephan Nowak
- Department of Neurosurgery, University Medicine Greifswald, Greifswald, Germany
| | - Henry W S Schroeder
- Department of Neurosurgery, University Medicine Greifswald, Greifswald, Germany
| | - Anna Vidal
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Helena Radbruch
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Annika Lehmann
- Institute of Pathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Susanne Kreuzer-Redmer
- Nutrigenomics Unit, Institute of Animal Nutrition and Functional Plant Compounds, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Karsten Jürchott
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Josefine Radke
- Institute of Pathology, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
7
|
Kermorgant S. Rationale for Cotargeting Hepatocyte Growth Factor and Epidermal Growth Factor Receptor in Recurrent/Metastatic Head and Neck Cancer. J Clin Oncol 2023:JCO2300460. [PMID: 37319388 DOI: 10.1200/jco.23.00460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/03/2023] [Accepted: 05/10/2023] [Indexed: 06/17/2023] Open
Affiliation(s)
- Stéphanie Kermorgant
- Spatial Signalling Group, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| |
Collapse
|
8
|
Choi SW, Oh H, Park SY, Cho W, Abd El-Aty AM, Hacimuftuoglu A, Jeong JH, Jung TW. Adipokine gremlin-1 promotes hepatic steatosis via upregulation of ER stress by suppressing autophagy-mediated signaling. J Cell Physiol 2023; 238:966-975. [PMID: 36890751 DOI: 10.1002/jcp.30982] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/01/2023] [Accepted: 02/10/2023] [Indexed: 03/10/2023]
Abstract
Gremlin-1 (GR1) is a novel adipokine that is highly expressed in human adipocytes and has been shown to inhibit the BMP2/4-TGFb signaling pathway. It has an effect on insulin sensitivity. Elevated levels of Gremlin have been shown to lead to insulin resistance in skeletal muscle, adipocytes, and hepatocytes. In this study, we investigated the effect of GR1 on hepatic lipid metabolism under hyperlipidemic conditions and explored the molecular mechanisms associated with GR1 by in vitro and in vivo studies. We found that palmitate increased GR1 expression in visceral adipocytes. Recombinant GR1 increased lipid accumulation, lipogenesis, and ER stress markers in cultured primary hepatocytes. Treatment with GR1 increased EGFR expression and mTOR phosphorylation and reduced autophagy markers. EGFR or rapamycin siRNA reduced the effects of GR1 on lipogenic lipid deposition and ER stress in cultured hepatocytes. Administration of GR1 via the tail vein induced lipogenic proteins and ER stress while suppressing autophagy in the livers of experimental mice. Suppression of GR1 by in vivo transfection reduced the effects of a high-fat diet on hepatic lipid metabolism, ER stress, and autophagy in mice. These results suggest that the adipokine GR1 promotes hepatic ER stress due to the impairment of autophagy, ultimately causing hepatic steatosis in the obese state. The current study demonstrated that targeting GR1 may be a potential therapeutic approach for treating metabolic diseases, including metabolic-associated fatty liver disease (MAFLD).
Collapse
Affiliation(s)
- Sung Woo Choi
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Heeseung Oh
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Seung Yeon Park
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.,Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Wonjun Cho
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.,Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, Turkey
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, Turkey.,Vaccine Development Application and Research Center, Ataturk University, Erzurum, Turkey
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.,Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
9
|
Sun H, Li X, Dai S, Shen X, Qiu M. A case report of response to crizotinib in chemotherapy-refractory metastatic gallbladder cancer with met amplification and acquired resistance resulting from the loss of MET amplification. PRECISION CLINICAL MEDICINE 2021; 4:209-214. [PMID: 35693217 PMCID: PMC8982584 DOI: 10.1093/pcmedi/pbab017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/17/2021] [Accepted: 07/21/2021] [Indexed: 02/05/2023] Open
Abstract
Gallbladder cancer (GBC) is a highly invasive disease and the most prevalent malignancy of the biliary system. Patients with GBC are commonly diagnosed at a late stage and have an unfavorable prognosis. Palliative chemotherapy has been the standard care for recurrent or metastatic disease in the past decades. Recently, several targeted therapies have been investigated in advanced biliary tract cancer (BTC) including inhibitors of genes or pathways such as FGFR2 fusions or rearrangements, IDH1 mutations, and NTRK gene fusions. Also, several clinical studies involving molecular stratification have been performed in defined patient groups, for example, BRAF V600E and HER2. Mesenchymal epithelial transition(MET)encodes a tyrosine kinase receptor and its ligand hepatocyte growth factor is a proto-oncogene. Targeting the MET signaling pathway is an effective strategy in numerous cancer types. However, the poor efficacy of MET inhibitors has been demonstrated in several phase II studies, but currently no reports have explained the potential mechanisms of resistance to MET inhibitors in BTC. In this article, we report a case of metastatic GBC with MET amplification that exhibited a rapid response to crizotinib after the failure of two lines of chemotherapy. After the patient had progressed and discontinued crizotinib, cabozantinib was introduced. Analysis of circulating tumor DNA (ctDNA) by next-generation sequencing (NGS) indicated a loss of MET amplification status. To our knowledge, this is the first case study demonstrating the use of NGS in ctDNA to monitor the development of acquired resistance during anti-MET treatment in GBC.
Collapse
Affiliation(s)
- Hongna Sun
- Department of Abdominal Cancer, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaofen Li
- Department of Abdominal Cancer, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuang Dai
- Department of Medical Oncology, Lung cancer center, West China Hospital, Sichuan University, Chengdu 611135, China
| | - Xudong Shen
- The Medical Department, 3D Medicines Inc., Shanghai 201202, China
| | - Meng Qiu
- Department of Abdominal Cancer, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
10
|
Moosavi F, Giovannetti E, Peters GJ, Firuzi O. Combination of HGF/MET-targeting agents and other therapeutic strategies in cancer. Crit Rev Oncol Hematol 2021; 160:103234. [PMID: 33497758 DOI: 10.1016/j.critrevonc.2021.103234] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/29/2020] [Accepted: 01/16/2021] [Indexed: 02/06/2023] Open
Abstract
MET receptor has emerged as a druggable target across several human cancers. Agents targeting MET and its ligand hepatocyte growth factor (HGF) including small molecules such as crizotinib, tivantinib and cabozantinib or antibodies including rilotumumab and onartuzumab have proven their values in different tumors. Recently, capmatinib was approved for treatment of metastatic lung cancer with MET exon 14 skipping. In this review, we critically examine the current evidence on how HGF/MET combination therapies may take advantage of synergistic effects, overcome primary or acquired drug resistance, target tumor microenvironment, modulate drug metabolism or tackle pharmacokinetic issues. Preclinical and clinical studies on the combination of HGF/MET-targeted agents with conventional chemotherapeutics or molecularly targeted treatments (including EGFR, VEGFR, HER2, RAF/MEK, and PI3K/Akt targeting agents) and also the value of biomarkers are examined. Our deeper understanding of molecular mechanisms underlying successful pharmacological combinations is crucial to find the best personalized treatment regimens for cancer patients.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, the Netherlands; Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, the Netherlands; Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
11
|
Garon EB, Brodrick P. Targeted Therapy Approaches for MET Abnormalities in Non-Small Cell Lung Cancer. Drugs 2021; 81:547-554. [PMID: 33638808 DOI: 10.1007/s40265-021-01477-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2021] [Indexed: 12/18/2022]
Abstract
The tyrosine kinase receptor mesenchymal epithelial transition (MET) is a proto-oncogene that, through the activation of the MET-hepatocyte growth factor (HGF) pathway, encodes a variety of biological processes, including cell development, proliferation, invasion, and migration. Abnormal activation of the MET pathway, occurring through MET protein overexpression, and gene amplification or mutation, can contribute to oncogenesis and has been implicated in non-small cell lung cancer (NSCLC). Though it is associated with poor clinical outcome in NSCLCs, MET overexpression and its role as a therapeutic target remains somewhat elusive due to discrepancies in its occurrence. Unlike MET overexpression, MET amplification has demonstrated a stronger potential as a biomarker for therapeutic treatment, with clinical data indicating a compelling connection between a high MET gene copy number and a high response rate to targeted therapies. However, MET exon 14 skipping mutations, occurring in 3%-4 % of lung adenocarcinomas, are of particular interest, as tumors harboring these mutations have shown a significant response to MET inhibitors. Following the discovery of MET as a potential therapeutic target, extensive clinical studies have proposed three approaches to targeting MET: (1) MET tyrosine kinase inhibitors (TKIs), including crizotinib, capmatinib, tepotinib, savolinitib, and cabozantinib; (2) MET or HGF monoclonal antibodies, including emibetuzumab and ficlatuzumab; and (3) MET or HGF antibody drug conjugates, including telisotuzumab. Herein, we discuss the relevant clinical trials, particularly focusing on the efficacy as well as the safety and tolerability of the treatment options, in the promising field of targeting MET in NSCLC.
Collapse
Affiliation(s)
- Edward B Garon
- David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Paige Brodrick
- David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Fang P, Zhou L, Lim LY, Fu H, Yuan ZX, Lin J. Targeting Strategies for Renal Cancer Stem Cell Therapy. Curr Pharm Des 2020; 26:1964-1978. [PMID: 32188377 DOI: 10.2174/1381612826666200318153106] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/27/2020] [Indexed: 12/24/2022]
Abstract
Renal cell carcinoma (RCC) is an intractable genitourinary malignancy that accounts for approximately 4% of adult malignancies. Currently, there is no approved targeted therapy for RCC that has yielded durable remissions, and they remain palliative in intent. Emerging evidence has indicated that renal tumorigenesis and RCC treatment-resistance may originate from renal cancer stem cells (CSCs) with tumor-initiating capacity (CSC hypothesis). A better understanding of the mechanism underlying renal CSCs will help to dissect RCC heterogeneity and drug treatment efficiency, to promote more personalized and targeted therapies. In this review, we summarized the stem cell characteristics of renal CSCs. We outlined the targeting strategies and challenges associated with developing therapies that target renal CSCs angiogenesis, immunosuppression, signaling pathways, surface biomarkers, microRNAs and nanomedicine. In conclusion, CSCs are an important role in renal carcinogenesis and represent a valid target for treatment of RCC patients.
Collapse
Affiliation(s)
- Pengchao Fang
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Liuting Zhou
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lee Y Lim
- Department of Pharmacy, School of Medicine and Pharmacology, The University of Western Australia, Crawley WA 6009, Perth, Australia
| | - Hualin Fu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhi-Xiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Juchun Lin
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
13
|
Xie B, Wang Y, He J, Ni Z, Chai D. Aberrant Cyclin E and Hepatocyte Growth Factor Expression, Microvascular Density, and Micro-Lymphatic Vessel Density in Esophageal Squamous Cell Carcinoma. Cancer Control 2020; 26:1073274819875736. [PMID: 31533461 PMCID: PMC6753519 DOI: 10.1177/1073274819875736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cyclin E and hepatocyte growth factor (HGF) have been observed as a multifaceted
factor in many cancers, and the assessment of microvascular density (MVD) and
micro-lymphatic vessel density (MLVD) has been used to quantify tumor
angiogenesis and lymphangiogenesis. The aim of this study was to explore the
association between expression of cyclin E, HGF, MVD, and MLVD, and
clinicopathologic parameters in esophageal squamous cell carcinoma (ESCC). The
expression of cyclin E, HGF, MVD, and MLVD were detected using
immunohistochemically anticyclin E, HGF, CD34, and lymphatic vessel endothelial
hyaluronan receptor 1 in 168 surgically resected ESCC cases and 30 normal
esophageal mucosal samples. The expression levels of cyclin E, HGF, MVD, and
MLVD were higher compared to controls. High cyclin E and HGF expression was
found more frequently in the tumors larger than 5 cm (P <
.001), with poorer differentiation (P = .034) and higher tumor
node metastasis (TNM) staging (P = .009) compared to their
counterparts. Both MVD and MLVD values were found to be higher in the tumors
larger than 5 cm (P < .001), with poorer differentiation
(P < .001) and higher TNM staging (P
< .001) compared to their counterparts. Furthermore, the expression of MVD
and MLVD in both the high cyclin E and high HGF expression groups was
significantly higher compared to the low cyclin E and HGF expression groups
(P < .001). This study demonstrated that high cyclin E
and HGF expression is closely correlated with tumor size, tumor differentiation
degree, and TNM stage in patients with ESCC. These findings proposed that cyclin
E and HGF could serve as novel molecular markers for preoperational evaluation
of ESCC.
Collapse
Affiliation(s)
- Bo Xie
- Department of Thoracic surgery, the First Affiliated Hospital of Suchow University, Suzhou, Jiangsu, China.,Department of Oncology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Anhui, China
| | - Yuanyuan Wang
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, Anhui, China
| | - Jingkang He
- Department of Thoracic surgery, the First Affiliated Hospital of Suchow University, Suzhou, Jiangsu, China
| | - Zhengzheng Ni
- Department of Thoracic surgery, the First Affiliated Hospital of Suchow University, Suzhou, Jiangsu, China
| | - Damin Chai
- Department of Oncology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Anhui, China
| |
Collapse
|
14
|
Jabbarpour Z, Kiani J, Keshtkar S, Saidijam M, Ghahremani MH, Ahmadbeigi N. Effects of human placenta-derived mesenchymal stem cells with NK4 gene expression on glioblastoma multiforme cell lines. J Cell Biochem 2020; 121:1362-1373. [PMID: 31595570 DOI: 10.1002/jcb.29371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/13/2019] [Indexed: 12/11/2022]
Abstract
Poor prognosis and low survival are commonly seen in patients with glioblastoma multiforme (GBM). Due to the specific nature of solid tumors such as GBM, delivery of therapeutic agents to the tumor sites is difficult. So, one of the major challenges in the treatment of these tumors is a selection of appropriate method for drug delivery. Mesenchymal stem cells (MSCs) have a unique characteristic in migration toward the tumor tissue. In this regard, the present study examined the antitumor effects of manipulating human placenta-derived mesenchymal stem cells (PDMSCs) with NK4 expression (PDMSC-NK4) on GBM cells. After separation and characterization of PDMSCs, these cells were transduced with NK4 which was known as the antagonist of hepatocyte growth factor (HGF). The results indicated that engineered PDMSCs preferably migrate into GBM cells by transwell coculture system. In addition, the proliferation of the GBM cells significantly reduced after coculture with these cells. In fact, manipulated PDMSCs inhibited growth of tumor cells by induction of apoptosis. Our findings suggested that besides having antitumor effects, PDMSCs can also be applied as an ideal cellular vehicle to target the glioblastoma multiforme.
Collapse
Affiliation(s)
- Zahra Jabbarpour
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Somayeh Keshtkar
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Massoud Saidijam
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad H Ghahremani
- Department of Pharmacology-Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Naser Ahmadbeigi
- Cell-Based Therapies Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Kupsamy K, Moodley J, Naicker T. Hepatocyte growth factor and epidermal growth factor in HIV infected women with preeclampsia. Eur J Obstet Gynecol Reprod Biol 2019; 240:9-14. [PMID: 31202974 DOI: 10.1016/j.ejogrb.2019.05.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/15/2019] [Accepted: 05/25/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Epidermal growth factor (EGF) and Hepatocyte growth factor (HGF) both have tyrosine kinase receptors (EGFR and c-Met) which upon binding, activates and regulates many important cellular processes such as cell survival, growth, proliferation, differentiation, invasion, repair and so forth via the RAS/MAPK/ERK1/2, PI3K/AKT and JAK STAT3 pathways. These processes are crucial for the development of a placenta and other functions in order for a normal pregnancy to occur. Hence, this study determined the concentrations of HGF and EGF to find the correlation between HIV and preeclampsia (PE). STUDY DESIGN A total sample size of n = 80 was used, n = 40 preeclamptic women and n = 40 normotensive women these were further stratified into HIV-positive and HIV-negative women. Analysis of the growth factors were done by using the multiplex Bio-Plex immunoassay method. RESULTS Irrespective of HIV status, based on pregnancy type, EGF in PE women displayed an upregulation compared to normotensive women. However, for HGF no variance was found between pregnancy type. Based on HIV status, regardless of pregnancy type, both HGF and EGF levels were significantly increased in HIV-positive women compared to HIV-negative women. Across all groups for HGF, significant difference was found between HIV-negative normotensive women (lower) vs HIV-positive normotensive women (higher). Nevertheless, for EGF across all groups, a statistically significant decrease was found in HIV-negative normotensive women compared to HIV-positive normotensive women, HIV-positive PE women and HIV-negative PE women. CONCLUSION The study demonstrates that there is a strong association between HIV and PE and that HGF and EGF are promising biomarkers to use as a diagnostic tool for PE.
Collapse
Affiliation(s)
- Kyle Kupsamy
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa.
| | - Jagidesa Moodley
- Women's Health and HIV Research Group, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| |
Collapse
|
16
|
Geng F, Wang Z, Yin H, Yu J, Cao B. Molecular Targeted Drugs and Treatment of Colorectal Cancer: Recent Progress and Future Perspectives. Cancer Biother Radiopharm 2018. [PMID: 28622036 DOI: 10.1089/cbr.2017.2210] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nowadays, colorectal cancer is the fourth most common type of tumor all over the world. When diagnosed, ∼50%-60% of tumors have metastasized, thus resulting in a grim prognosis. Chemotherapy is regarded as standard treatment for patients with colorectal cancer, however, limitations of chemotherapy cannot be ignored, such as low selectivity, insufficient concentrations in tumor tissues, and systemic toxicity. Recently, six targeted drugs have been approved by the U.S. Food and Drug Administration (FDA) for treatment of metastatic colorectal cancer (mCRC), including bevacizumab, aflibercept, regorafenib, cetuximab, and panitumumab. The development of these drugs marked significant advancement in the field of mCRC therapy. The addition of biologic agents to chemotherapy has prolonged the median overall survival. Now, many investigational drugs are under clinical trials, of which programmed death (PD)-1/L1 has drawn much attention. In this review, new biologic agents under clinical trials such as MEK/MET/RAS/RAF/PD-1 inhibitors with potentials for mCRC treatment are concluded by describing targeted drugs approved by FDA, to offer new insights into global trends and future development.
Collapse
Affiliation(s)
- Fang Geng
- 1 Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University , Beijing, China .,2 Cancer Center, Beijing Friendship Hospital, Capital Medical University , Beijing, China
| | - Zheng Wang
- 1 Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University , Beijing, China
| | - Hang Yin
- 1 Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University , Beijing, China
| | - Junxian Yu
- 1 Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University , Beijing, China
| | - Bangwei Cao
- 2 Cancer Center, Beijing Friendship Hospital, Capital Medical University , Beijing, China
| |
Collapse
|
17
|
Doi T, Yamaguchi K, Komatsu Y, Muro K, Nishina T, Nakajima TE, Tang R, Yang H, Zhang Y, Jung AS, Ang A, Yasui H. A Phase 1/1b tolerability study of rilotumumab alone or in combination with cisplatin and capecitabine in Japanese patients with gastric cancer. Jpn J Clin Oncol 2018; 47:1002-1009. [PMID: 28973403 DOI: 10.1093/jjco/hyx114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/09/2017] [Indexed: 12/11/2022] Open
Abstract
Objective To evaluate the safety (including adverse events and dose-limiting toxicities [DLTs]), tolerability, pharmacokinetics and antitumor activity of the investigational MET inhibitor rilotumumab alone in patients with advanced solid tumors (Part 1) or in combination with cisplatin plus capecitabine (CX) in patients with MET-positive advanced gastric or gastroesophageal junction cancer (Part 2). Methods Adult patients received 10 or 20 mg/kg intravenous (IV) rilotumumab every 2 weeks (Part 1) or 15 mg/kg IV rilotumumab every 3 weeks plus 80 mg/m2 cisplatin on Day 1 and 1000 mg/m2 capecitabine twice daily on Days 1-14 of every 21-day cycle (Part 2). Results Nine patients enrolled in Part 1; 12 patients enrolled in Part 2. One DLT occurred (Grade 3 decreased appetite and stomatitis [Part 2]). Adverse events related to any treatment occurred in 17 patients (81%) and were Grade ≥3 in nine patients (43%). Rilotumumab pharmacokinetics appeared linear, and exposure was unaffected by CX. No patient who received rilotumumab monotherapy in Part 1 had a response. In Part 2, five of eight patients (63%) with measureable disease at baseline had a partial response and two patients (25%) had stable disease; median (95% CI) progression-free survival was 7.0 (2.4-15.4) months; overall survival was 18.2 (5.6-20.4) months. Conclusions In combination with CX, rilotumumab appeared tolerable and showed antitumor activity in Japanese patients with MET-positive gastric/gastroesophageal junction cancer. However, owing to the results of recent Phase 3 trials of MET inhibitors (including rilotumumab), further development of rilotumumab in this setting is not being pursued. ClinicalTrials.gov Identifier: NCT01791374.
Collapse
Affiliation(s)
- Toshihiko Doi
- National Cancer Center Hospital East, Kashiwa, Chiba
| | - Kensei Yamaguchi
- Saitama Cancer Center, Kita Adachi-gun, Saitama.,Cancer Institute Hospital of Japanese Foundation for Cancer Research, Ariake, Tokyo
| | | | - Kei Muro
- Aichi Cancer Center Hospital, Nagoya
| | | | | | - Rui Tang
- Amgen Inc., Thousand Oaks, CA, USA
| | - Hui Yang
- Amgen Inc., Thousand Oaks, CA, USA
| | | | | | | | | |
Collapse
|
18
|
Glisson B, Besse B, Dols MC, Dubey S, Schupp M, Jain R, Jiang Y, Menon H, Nackaerts K, Orlov S, Paz-Ares L, Ramlau R, Tang R, Zhang Y, Zhu M. A Randomized, Placebo-Controlled, Phase 1b/2 Study of Rilotumumab or Ganitumab in Combination With Platinum-Based Chemotherapy as First-Line Treatment for Extensive-Stage Small-Cell Lung Cancer. Clin Lung Cancer 2017; 18:615-625.e8. [DOI: 10.1016/j.cllc.2017.05.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/26/2017] [Accepted: 05/02/2017] [Indexed: 11/28/2022]
|
19
|
Role and Therapeutic Targeting of the HGF/MET Pathway in Glioblastoma. Cancers (Basel) 2017; 9:cancers9070087. [PMID: 28696366 PMCID: PMC5532623 DOI: 10.3390/cancers9070087] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 06/29/2017] [Accepted: 07/06/2017] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma (GBM) is a lethal brain tumor with dismal prognosis. Current therapeutic options, consisting of surgery, chemotherapy and radiation, have only served to marginally increase patient survival. Receptor tyrosine kinases (RTKs) are dysregulated in approximately 90% of GBM; attributed to this, research has focused on inhibiting RTKs as a novel and effective therapy for GBM. Overexpression of RTK mesenchymal epithelial transition (MET), and its ligand, hepatocyte growth factor (HGF), in GBM highlights a promising new therapeutic target. This review will discuss the role of MET in cell cycle regulation, cell proliferation, evasion of apoptosis, cell migration and invasion, angiogenesis and therapeutic resistance in GBM. It will also discuss the modes of deregulation of HGF/MET and their regulation by microRNAs. As the HGF/MET pathway is a vital regulator of multiple pro-survival pathways, efforts and strategies for its exploitation for GBM therapy are also described.
Collapse
|
20
|
Progress of antibody-based inhibitors of the HGF-cMET axis in cancer therapy. Exp Mol Med 2017; 49:e307. [PMID: 28336955 PMCID: PMC5382561 DOI: 10.1038/emm.2017.17] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 12/22/2016] [Indexed: 12/27/2022] Open
Abstract
Dysregulated receptor tyrosine kinase signaling in human cancer cells leads to tumor progression, invasion and metastasis. The receptor tyrosine kinase cMET is frequently overexpressed in cancer tissue, and activation of cMET signaling is related to drug resistance and the processes of carcinogenesis, invasion and metastasis. For that reason, cMET and its ligand, hepatocyte growth factor (HGF), are considered prime targets for the development of anticancer drugs. At least eight anti-cMET and four anti-HGF antibodies have been tested or are being tested in clinical trials. However, to date none of these HGF/cMET inhibitors have shown significant efficacy in clinical trials. Furthermore, no receptor tyrosine kinase inhibitors primarily targeting cMET have been approved. Given that neutralization of HGF or cMET does not cause significant adverse effects, inhibition of the HGF/cMET signaling pathway appears to be safe. In this review, we summarized the completed and ongoing clinical trials testing antibody- or protein-based anticancer drugs targeting cMET and HGF.
Collapse
|
21
|
Price EW, Carnazza KE, Carlin SD, Cho A, Edwards KJ, Sevak KK, Glaser JM, de Stanchina E, Janjigian YY, Lewis JS. 89Zr-DFO-AMG102 Immuno-PET to Determine Local Hepatocyte Growth Factor Protein Levels in Tumors for Enhanced Patient Selection. J Nucl Med 2017; 58:1386-1394. [PMID: 28280216 DOI: 10.2967/jnumed.116.187310] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/15/2017] [Indexed: 11/16/2022] Open
Abstract
The hepatocyte growth factor (HGF) binding antibody rilotumumab (AMG102) was modified for use as a 89Zr-based immuno-PET imaging agent to noninvasively determine the local levels of HGF protein in tumors. Because recent clinical trials of HGF-targeting therapies have been largely unsuccessful in several different cancers (e.g., gastric, brain, lung), we have synthesized and validated 89Zr-DFO-AMG102 as a companion diagnostic for improved identification and selection of patients having high local levels of HGF in tumors. To date, patient selection has not been performed using the local levels of HGF protein in tumors. Methods: The chelator p-SCN-Bn-DFO was conjugated to AMG102, radiolabeling with 89Zr was performed in high radiochemical yields and purity (>99%), and binding affinity of the modified antibody was confirmed using an enzyme-linked immunosorbent assay (ELISA)-type binding assay. PET imaging, biodistribution, autoradiography and immunohistochemistry, and ex vivo HGF ELISA experiments were performed on murine xenografts of U87MG (HGF-positive, MET-positive) and MKN45 (HGF-negative, MET-positive) and 4 patient-derived xenografts (MET-positive, HGF unknown). Results: Tumor uptake of 89Zr-DFO-AMG102 at 120 h after injection in U87MG xenografts (HGF-positive) was high (36.8 ± 7.8 percentage injected dose per gram [%ID/g]), whereas uptake in MKN45 xenografts (HGF-negative) was 5.0 ± 1.3 %ID/g and a control of nonspecific human IgG 89Zr-DFO-IgG in U87MG tumors was 11.5 ± 3.3 %ID/g, demonstrating selective uptake in HGF-positive tumors. Similar experiments performed in 4 different gastric cancer patient-derived xenograft models showed low uptake of 89Zr-DFO-AMG102 (∼4-7 %ID/g), which corresponded with low HGF levels in these tumors (ex vivo ELISA). Autoradiography, immunohistochemical staining, and HGF ELISA assays confirmed that elevated levels of HGF protein were present only in U87MG tumors and that 89Zr-DFO-AMG102 uptake was closely correlated with HGF protein levels in tumors. Conclusion: The new immuno-PET imaging agent 89Zr-DFO-AMG102 was successfully synthesized, radiolabeled, and validated in vitro and in vivo to selectively accumulate in tumors with high local levels of HGF protein. These results suggest that 89Zr-DFO-AMG102 would be a valuable companion diagnostic tool for the noninvasive selection of patients with elevated local concentrations of HGF in tumors for planning any HGF-targeted therapy, with the potential to improve clinical outcomes.
Collapse
Affiliation(s)
- Eric W Price
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kathryn E Carnazza
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sean D Carlin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew Cho
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kimberly J Edwards
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kuntal K Sevak
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jonathan M Glaser
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yelena Y Janjigian
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York .,Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
22
|
Szturz P, Raymond E, Abitbol C, Albert S, de Gramont A, Faivre S. Understanding c-MET signalling in squamous cell carcinoma of the head & neck. Crit Rev Oncol Hematol 2017; 111:39-51. [DOI: 10.1016/j.critrevonc.2017.01.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 10/28/2016] [Accepted: 01/09/2017] [Indexed: 12/21/2022] Open
|
23
|
Zhang Y, Kuchimanchi M, Zhu M, Doshi S, Hoang T, Kasichayanula S. Assessment of pharmacokinetic interaction between rilotumumab and epirubicin, cisplatin and capecitabine (ECX) in a Phase 3 study in gastric cancer. Br J Clin Pharmacol 2016; 83:1048-1055. [PMID: 27966237 DOI: 10.1111/bcp.13179] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 10/20/2016] [Accepted: 10/27/2016] [Indexed: 02/06/2023] Open
Abstract
AIMS Rilotumumab is a fully human monoclonal antibody investigated for the treatment of MET-positive gastric cancer. The aim of this study was to evaluate the potential pharmacokinetic (PK)-based drug-drug interaction (DDI) between rilotumumab and epirubicin (E), cisplatin(C) and capecitabine (X). METHODS This was a Phase 3 double-blind, placebo-controlled study, in which rilotumumab, epirubicin and cisplatin were administered intravenously at 15 mg kg-1 , 50 mg m-2 , and 60 mg m-2 Q3W, respectively, while capecitabine was given orally at 625 mg m-2 twice daily. Rilotumumab PK samples were taken at pre-dose and at the end-of-infusion from all patients in cycles 1, 3, 5 and 7. ECX PK samples were taken in cycle 3 from patients who participated in the intensive PK assessment. ECX PK was assessed by non-compartmental (NCA) analyses and PK parameters were compared between two arms. Rilotumumab PK was assessed by comparing the observed rilotumumab serum concentrations with model-predicted concentrations using a population PK model developed from previous Phase 1 and Phase 2 studies. RESULTS The study enrolled 609 patients. ECX plasma concentrations in the presence and absence of rilotumumab were similar, as demonstrated by the geometric mean ratios for Cmax and AUC, which were close to 1.0, suggesting ECX PK was not affected by co-administration of rilotumumab. The observed rilotumumab serum concentrations were similar to the values predicted by population PK modelling on the basis of a prediction-corrected visual predictive check, indicating rilotumumab exposure was not affected by co-administration of ECX. CONCLUSIONS The results suggest lack of PK-based DDI between rilotumumab and ECX.
Collapse
Affiliation(s)
- Yilong Zhang
- Clinical Pharmacology, Modeling and Simulation, Amgen Inc., Thousand Oaks, California, 91320
| | - Mita Kuchimanchi
- Clinical Pharmacology, Modeling and Simulation, Amgen Inc., Thousand Oaks, California, 91320
| | - Min Zhu
- Clinical Pharmacology, Modeling and Simulation, Amgen Inc., Thousand Oaks, California, 91320
| | - Sameer Doshi
- Clinical Pharmacology, Modeling and Simulation, Amgen Inc., Thousand Oaks, California, 91320
| | - Tien Hoang
- Clinical Development, Amgen Inc., Thousand Oaks, California, 91320
| | | |
Collapse
|
24
|
Szturz P, Raymond E, Faivre S. c-MET-mediated resistance to EGFR inhibitors in head and neck cancer: How to move from bench to bedside. Oral Oncol 2016; 59:e12-e14. [PMID: 27302885 DOI: 10.1016/j.oraloncology.2016.05.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 05/26/2016] [Indexed: 01/31/2023]
Affiliation(s)
- Petr Szturz
- Department of Oncology, Bichat-Beaujon University Hospital, Paris, France; Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and School of Medicine, Masaryk University, Brno, Czech Republic.
| | - Eric Raymond
- Department of Oncology, Bichat-Beaujon University Hospital, Paris, France
| | - Sandrine Faivre
- Department of Oncology, Bichat-Beaujon University Hospital, Paris, France
| |
Collapse
|
25
|
Garajová I, Giovannetti E, Biasco G, Peters GJ. c-Met as a Target for Personalized Therapy. TRANSLATIONAL ONCOGENOMICS 2015; 7:13-31. [PMID: 26628860 DOI: 10.4137/togog.s30534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/20/2015] [Accepted: 09/23/2015] [Indexed: 12/18/2022]
Abstract
MET and its ligand HGF are involved in many biological processes, both physiological and pathological, making this signaling pathway an attractive therapeutic target in oncology. Downstream signaling effects are transmitted via mitogen-activated protein kinase (MAPK), PI3K (phosphoinositide 3-kinase protein kinase B)/AKT, signal transducer and activator of transcription proteins (STAT), and nuclear factor-κB. The final output of the terminal effector components of these pathways is activation of cytoplasmic and nuclear processes leading to increases in cell proliferation, survival, mobilization and invasive capacity. In addition to its role as an oncogenic driver, increasing evidence implicates MET as a common mechanism of resistance to targeted therapies including EGFR and VEGFR inhibitors. In the present review, we summarize the current knowledge on the role of the HGF-MET signaling pathway in cancer and its therapeutic targeting (HGF activation inhibitors, HGF inhibitors, MET antagonists and selective/nonselective MET kinase inhibitors). Recent advances in understanding the role of this pathway in the resistance to current anticancer strategies used in lung, kidney and pancreatic cancer are discussed.
Collapse
Affiliation(s)
- Ingrid Garajová
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands. ; Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands. ; Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Guido Biasco
- Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Garajová I, Giovannetti E, Biasco G, Peters GJ. c-Met as a Target for Personalized Therapy. TRANSLATIONAL ONCOGENOMICS 2015; 7:13-31. [PMID: 26628860 PMCID: PMC4659440 DOI: 10.4137/tog.s30534] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/20/2015] [Accepted: 09/23/2015] [Indexed: 12/30/2022]
Abstract
MET and its ligand HGF are involved in many biological processes, both physiological and pathological, making this signaling pathway an attractive therapeutic target in oncology. Downstream signaling effects are transmitted via mitogen-activated protein kinase (MAPK), PI3K (phosphoinositide 3-kinase protein kinase B)/AKT, signal transducer and activator of transcription proteins (STAT), and nuclear factor-κB. The final output of the terminal effector components of these pathways is activation of cytoplasmic and nuclear processes leading to increases in cell proliferation, survival, mobilization and invasive capacity. In addition to its role as an oncogenic driver, increasing evidence implicates MET as a common mechanism of resistance to targeted therapies including EGFR and VEGFR inhibitors. In the present review, we summarize the current knowledge on the role of the HGF-MET signaling pathway in cancer and its therapeutic targeting (HGF activation inhibitors, HGF inhibitors, MET antagonists and selective/nonselective MET kinase inhibitors). Recent advances in understanding the role of this pathway in the resistance to current anticancer strategies used in lung, kidney and pancreatic cancer are discussed.
Collapse
Affiliation(s)
- Ingrid Garajová
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, Sant’Orsola-Malpighi Hospital, Bologna, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Guido Biasco
- Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, Sant’Orsola-Malpighi Hospital, Bologna, Italy
| | - Godefridus J. Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Zhang Y, Doshi S, Zhu M. Pharmacokinetics and pharmacodynamics of rilotumumab: a decade of experience in preclinical and clinical cancer research. Br J Clin Pharmacol 2015; 80:957-64. [PMID: 25912961 DOI: 10.1111/bcp.12663] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 04/06/2015] [Accepted: 04/20/2015] [Indexed: 12/11/2022] Open
Abstract
Rilotumumab is a fully human monoclonal antibody against hepatocyte growth factor, the only known ligand of the MET receptor. Over the last decade, rilotumumab has been extensively tested in preclinical studies and in clinical studies in a variety of cancer types. In this review, we examine the pharmacokinetic and pharmacodynamic data that have been collected in the rilotumumab programme to date, and discuss retrospectively how the knowledge acquired in this programme can be applied to a number of key issues in oncology drug development, including: (i) using preclinical data to inform first-in-human study design; (ii) the role of biomarkers in the identification of a target patient population; (iii) the potential for drug interactions between therapeutic proteins and other anticancer agents; and (iv) pharmacokinetic and pharmacodynamic considerations in phase 3 study design.
Collapse
Affiliation(s)
- Y Zhang
- Department of Clinical Pharmacology, Modeling, and Simulation, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - S Doshi
- Department of Clinical Pharmacology, Modeling, and Simulation, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - M Zhu
- Department of Clinical Pharmacology, Modeling, and Simulation, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| |
Collapse
|