1
|
Rogmans C, Dittrich J, Hamm E, Weimer JP, Holthaus D, Arnold N, Flörkemeier I, Maass N, Jansen P, Dempfle A, Bauerschlag DO, Hedemann N. Inhibiting ADAM17 enhances the efficacy of olaparib in ovarian cancer spheroids. Sci Rep 2024; 14:26926. [PMID: 39506058 PMCID: PMC11541916 DOI: 10.1038/s41598-024-78442-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024] Open
Abstract
Acquired or de novo resistance to poly (ADP-ribose) polymerase inhibitors (PARPi) is a major challenge to ovarian cancer treatment. Therefore, strategies to overcome PARPi resistance are critical to improve prognosis. The purpose of this study is to evaluate whether inhibition of ADAM17 sensitizes ovarian cancer to treatment with olaparib, a PARPi, thereby bypassing resistance mechanisms and improving treatment response. Thus, we analyzed the effect of olaparib in combination with the ADAM17 inhibitor GW280264X in ovarian cancer using a 2D monolayer and a 3D spheroid model followed by a multicontent readout (viability, caspase activation and cytotoxicity). To emphasize the translational aspect of our work, we performed corresponding experiments on primary cells derived from ovarian cancer patients initially screened for their mutation status of the breast cancer gene (BRCA 1/2). In 2D, we observed a significant reduction in cell viability and a subsequent increase in apoptosis of the combined treatment (olaparib + GW280264X) compared with olaparib mono-treatment. The combined treatment allows a substantial dose reduction of olaparib rendering a strong synergistic effect. Using a 3D spheroid model from primary cells, we confirmed the 2D monoculture results and demonstrated not only increased caspase activity under the combined treatment but also a substantial gain in cytotoxicity compared to the mono-treatment. Our study proposes ADAM17 inhibition sensitizing ovarian cancer to olaparib treatment and improving treatment response.
Collapse
Affiliation(s)
- Christoph Rogmans
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany
| | - Jan Dittrich
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany
| | - Emily Hamm
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany
| | - Jörg Paul Weimer
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany
| | - David Holthaus
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany
- Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany
| | - Norbert Arnold
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany
| | - Inken Flörkemeier
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany
| | - Nicolai Maass
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany
| | - Peer Jansen
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany
| | - Astrid Dempfle
- Institute of Medical Informatics and Statistics, Kiel University and University Medical Center, Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany
| | - Dirk O Bauerschlag
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany
| | - Nina Hedemann
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany.
| |
Collapse
|
2
|
Saha N, Lee SG, Brockmann EC, de la Cruz MJ, Goldgur Y, Mendoza RP, Stanchina ED, Love TM, Marvald J, Xu Y, Xu K, Himanen JP, Lamminmäki U, Veach D, Nikolov DB. Fully human monoclonal antibody targeting the cysteine-rich substrate-interacting region of ADAM17 on cancer cells. Biomed Pharmacother 2024; 180:117605. [PMID: 39461016 DOI: 10.1016/j.biopha.2024.117605] [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: 08/08/2024] [Revised: 10/05/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
ADAM17 sheds EGFR/erbB ligands and triggers oncogenic pathways that lead to the progression of solid tumors. We targeted the ADAM17 disintegrin and cysteine rich domain region (D+C) to generate a panel of single-chain antibody fragments (scFvs) that selectively bind to the D or C domains of ADAM17, but not of ADAM10 or ADAM19. From the panel, we selected one scFv, referred to as C12, based on its high binding affinity towards the target, and re-formatted it to a full IgG for further studies. High-resolution cryo-electron microscopy studies documented that the mAb binds to the ADAM17 C-domain that in ADAM proteases, notably ADAM10 and ADAM17, is known to impart substrate-specificity. The C12 mAb significantly inhibited EGFR phosphorylation in cancer cell lines by hindering the cleavage of EGFR ligands tethered to the cell surface. This inhibition provides a mechanism for potential anti-tumor effects, and indeed C12 diminished the viability of a variety of EGFR-expressing cancer cell lines. Cell-based ELISA studies revealed that C12 preferentially bound to activated ADAM17 present on tumor cells, as compared to the autoinhibited ADAM17 that is the predominant form on HEK293 and other non-tumor cells. C12 also exhibited tumor growth inhibition in an ovarian cancer xenograft mouse model. Consistent with its selective tumor cell binding in vitro, radioimmuno PET (positron emission tomography) imaging with 89Zr-DFO-C12 in mouse xenograft models confirmed tumoral accumulation of the C12 mAb.
Collapse
Affiliation(s)
- Nayanendu Saha
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| | - Sang Gyu Lee
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | | | - M Jason de la Cruz
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Yehuda Goldgur
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Rachelle P Mendoza
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Elisa de Stanchina
- Antitumor Assessment Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Tanzy M Love
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - Josh Marvald
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - Yan Xu
- Texas Therapeutic Institute, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Kai Xu
- Texas Therapeutic Institute, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Juha P Himanen
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Urpo Lamminmäki
- Department of Life Technologies, University of Turku, Turku, Finland
| | - Darren Veach
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Dimitar B Nikolov
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| |
Collapse
|
3
|
Rossi V, Hochkoeppler A, Govoni M, Di Stefano G. Lactate-Induced HBEGF Shedding and EGFR Activation: Paving the Way to a New Anticancer Therapeutic Opportunity. Cells 2024; 13:1533. [PMID: 39329717 PMCID: PMC11430493 DOI: 10.3390/cells13181533] [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: 07/17/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Cancer cells can release EGF-like peptides, acquiring the capacity of autocrine stimulation via EGFR-mediated signaling. One of these peptides (HBEGF) was found to be released from a membrane-bound precursor protein and is critically implicated in the proliferative potential of cancer cells. We observed that the increased lactate levels characterizing neoplastic tissues can induce the release of uPA, a protease promoting HBEGF shedding. This effect led to EGFR activation and increased ERK1/2 phosphorylation. Since EGFR-mediated signaling potentiates glycolytic metabolism, this phenomenon can induce a self-sustaining deleterious loop, favoring tumor growth. A well characterized HBEGF inhibitor is CRM197, a single-site variant of diphtheria toxin. We observed that, when administered individually, CRM197 did not trigger evident antineoplastic effects. However, its association with a uPA inhibitor caused dampening of EGFR-mediated signaling and apoptosis induction. Overall, our study highlights that the increased glycolytic metabolism and lactate production can foster the activated state of EGFR receptor and suggests that the inhibition of EGFR-mediated signaling can be attempted by means of CRM197 administered with an appropriate protease inhibitor. This attempt could help in overcoming the problem of the acquired resistance to the conventionally used EGFR inhibitors.
Collapse
Affiliation(s)
- Valentina Rossi
- Department of Medical and Surgical Sciences (DIMEC), Section of General Pathology, University of Bologna, 40126 Bologna, Italy; (V.R.); (M.G.)
| | - Alejandro Hochkoeppler
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy;
| | - Marzia Govoni
- Department of Medical and Surgical Sciences (DIMEC), Section of General Pathology, University of Bologna, 40126 Bologna, Italy; (V.R.); (M.G.)
| | - Giuseppina Di Stefano
- Department of Medical and Surgical Sciences (DIMEC), Section of General Pathology, University of Bologna, 40126 Bologna, Italy; (V.R.); (M.G.)
| |
Collapse
|
4
|
Holthaus D, Rogmans C, Gursinski I, Quevedo-Olmos A, Ehsani M, Mangler M, Flörkemeier I, Weimer JP, Meyer TF, Maass N, Bauerschlag DO, Hedemann N. Inhibition of ADAM17 increases the cytotoxic effect of cisplatin in cervical spheroids and organoids. Front Oncol 2024; 14:1432239. [PMID: 39286024 PMCID: PMC11402614 DOI: 10.3389/fonc.2024.1432239] [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: 05/13/2024] [Accepted: 08/07/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction Cervical cancer represents one of the main causes of female, cancer-related mortality worldwide. The majority of cancers are caused by human papillomaviruses such as HPV16 and HPV18. As chemotherapeutic resistance to first-line platinum treatment is still a predominant clinical challenge in advanced cervical cancer, novel treatment options including combinatorial therapies are urgently required to overcome chemotherapeutic resistance. Inhibition of A Disintegrin And Metalloproteinase (ADAM)-family members, heavily involved in tumour progression of a vast range of solid tumours, strongly improved response to chemotherapeutic treatment in other tumour entities including ovarian cancer. Methods We established two- and three-dimensional models derived from three traditional cervical cancer cell lines and ectocervical cancer-derived organoids. Following characterisation, these models were used to investigate their response to cisplatin treatment in the absence and presence of ADAM inhibitors using viability assays and automated live cell imaging. Results The pivotal role of the metalloprotease ADAM17 driving chemotherapy resistance was detectable in all ectocervical cultures irrespective of the model system used, whereas ADAM10 inhibition was predominantly effective only in loosely aggregated spheroids. We showed prominent differences regarding treatment responses between 2D monolayers compared to 3D spheroid and 3D organoid model systems. Particularly, the organoid system, regarded as the closest representation of primary tumours, exhibited reliably the combinatorial effect of ADAM17 inhibition and cisplatin in all three individual donors. Discussion As two- and three-dimensional models of the same cell lines differ in their responses to chemotherapy it is essential to validate treatment strategies in more advanced model systems representing the patient situation more realistically. Ectocervical organoids showed reliable results regarding treatment responses closely mimicking the primary tumours and could therefore serve as an important tool for personalized medicine in cervical cancer. These findings strengthen the role of ADAM17 as a potential novel target for combinatorial treatments to overcome chemoresistance in cervical cancer.
Collapse
Affiliation(s)
- David Holthaus
- Department of Gynaecology and Obstetrics, University Hospital Schleswig-Holstein, Kiel, Germany
- Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christoph Rogmans
- Department of Gynaecology and Obstetrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ina Gursinski
- Department of Gynaecology and Obstetrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Alvaro Quevedo-Olmos
- Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Marzieh Ehsani
- Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Mandy Mangler
- Department of Gynaecology and Obstetrics, Vivantes Auguste Viktoria-Klinikum, Berlin, Germany
- Department of Gynaecology, Charité University Medicine, Berlin, Germany
| | - Inken Flörkemeier
- Department of Gynaecology and Obstetrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jörg P Weimer
- Department of Gynaecology and Obstetrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thomas F Meyer
- Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Nicolai Maass
- Department of Gynaecology and Obstetrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Dirk O Bauerschlag
- Department of Gynaecology and Obstetrics, University Hospital Schleswig-Holstein, Kiel, Germany
- Clinic and Polyclinic for Gynaecology and Reproductive Medicine, University Hospital Jena, Jena, Germany
| | - Nina Hedemann
- Department of Gynaecology and Obstetrics, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
5
|
Wang K, Xuan Z, Liu X, Zheng M, Yang C, Wang H. Immunomodulatory role of metalloproteinase ADAM17 in tumor development. Front Immunol 2022; 13:1059376. [PMID: 36466812 PMCID: PMC9715963 DOI: 10.3389/fimmu.2022.1059376] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/03/2022] [Indexed: 12/25/2023] Open
Abstract
ADAM17 is a member of the a disintegrin and metalloproteinase (ADAM) family of transmembrane proteases involved in the shedding of some cell membrane proteins and regulating various signaling pathways. More than 90 substrates are regulated by ADAM17, some of which are closely relevant to tumor formation and development. Besides, ADAM17 is also responsible for immune regulation and its substrate-mediated signal transduction. Recently, ADAM17 has been considered as a major target for the treatment of tumors and yet its immunomodulatory roles and mechanisms remain unclear. In this paper, we summarized the recent understanding of structure and several regulatory roles of ADAM17. Importantly, we highlighted the immunomodulatory roles of ADAM17 in tumor development, as well as small molecule inhibitors and monoclonal antibodies targeting ADAM17.
Collapse
Affiliation(s)
- Kai Wang
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Zixue Xuan
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xiaoyan Liu
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Meiling Zheng
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Chao Yang
- National Engineering Research Center for Marine Aquaculture, Institute of Innovation & Application, Zhejiang Ocean University, Zhoushan, China
| | - Haiyong Wang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
6
|
Fabbi M, Costa D, Russo D, Arenare L, Gaggero G, Signoriello S, Scambia G, Pisano C, Colombo N, Losito NS, Filaci G, Spina A, Califano D, Scognamiglio G, Gadducci A, Mezzanzanica D, Bagnoli M, Ferrini S, Canzonieri V, Chiodini P, Perrone F, Pignata S. Analysis of A Disintegrin and Metalloprotease 17 (ADAM17) Expression as a Prognostic Marker in Ovarian Cancer Patients Undergoing First-Line Treatment Plus Bevacizumab. Diagnostics (Basel) 2022; 12:diagnostics12092118. [PMID: 36140519 PMCID: PMC9498026 DOI: 10.3390/diagnostics12092118] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/20/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
To find prognostic factors for advanced ovarian cancer patients undergoing first-line therapy with carboplatin, paclitaxel and bevacizumab, we investigated the expression of a disintegrin and metalloprotease 17 (ADAM17) in cancer tissues. ADAM17 has been involved in ovarian cancer development, progression and cell resistance to cisplatin. Tissue microarrays from 309 ovarian cancer patients enrolled in the MITO16A/MANGO-OV2 clinical trial were analyzed by immunohistochemistry for ADAM17 protein expression. Intensity and extent of staining were combined into a semi-quantitative visual grading system (H score) which was related to clinicopathological characteristics of cases and the clinical outcome of patients by univariate and multivariate Cox regression models. ADAM17 immunostaining was detected in most samples, mainly localized in the tumor cells, with variable intensity across the cohort. Kaplan–Meier survival curves, generated according to the best cut-off value for the ADAM17 H score, showed that high ADAM17 expression was associated with worse prognosis for PFS and OS. However, after the application of a shrinkage procedure to adjust for overfitting hazard ratio estimates, the ADAM17 value as prognostic factor was lost. As subgroup analysis suggested that ADAM17 expression could be prognostically relevant in cases with no residual disease at baseline, further studies in this patient category may be worth planning.
Collapse
Affiliation(s)
- Marina Fabbi
- UO Bioterapie, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence:
| | - Delfina Costa
- UO Oncologia Molecolare e Angiogenesi, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Daniela Russo
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | - Laura Arenare
- Clinical Trials Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | - Gabriele Gaggero
- UO Anatomia Patologica Ospedaliera, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Simona Signoriello
- Department of Mental Health and Public Medicine, Section of Statistics, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Giovanni Scambia
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Life Science and Public Health, Catholic University of Sacred Heart, Largo Agostino Gemelli, 00168 Rome, Italy
| | - Carmela Pisano
- Urogynecological Medical Oncology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | - Nicoletta Colombo
- European Institute of Oncology IRCCS, University of Milan-Bicocca, 20126 Milan, Italy
| | - Nunzia Simona Losito
- Pathology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | - Gilberto Filaci
- UO Bioterapie, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
| | - Anna Spina
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | - Daniela Califano
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | - Giosuè Scognamiglio
- Pathology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | - Angiolo Gadducci
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, 56127 Pisa, Italy
| | - Delia Mezzanzanica
- Molecular Therapies Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Marina Bagnoli
- Molecular Therapies Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Silvano Ferrini
- UO Bioterapie, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Paolo Chiodini
- Department of Mental Health and Public Medicine, Section of Statistics, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Francesco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | - Sandro Pignata
- Urogynecological Medical Oncology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| |
Collapse
|
7
|
Garrido MP, Vallejos C, Girardi S, Gabler F, Selman A, López F, Vega M, Romero C. NGF/TRKA Promotes ADAM17-Dependent Cleavage of P75 in Ovarian Cells: Elucidating a Pro-Tumoral Mechanism. Int J Mol Sci 2022; 23:ijms23042124. [PMID: 35216240 PMCID: PMC8877415 DOI: 10.3390/ijms23042124] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/03/2022] [Accepted: 01/13/2022] [Indexed: 02/08/2023] Open
Abstract
Nerve growth factor (NGF) and its high-affinity receptor TRKA are overexpressed in epithelial ovarian cancer (EOC) displaying a crucial role in the disease progression. Otherwise, NGF interacts with its low-affinity receptor P75, activating pro-apoptotic pathways. In neurons, P75 could be cleaved by metalloproteinases (α and γ-secretases), leading to a decrease in P75 signaling. Therefore, this study aimed to evaluate whether the shedding of P75 occurs in EOC cells and whether NGF/TRKA could promote the cleavage of the P75 receptor. The immunodetection of the α-secretase, ADAM17, TRKA, P75, and P75 fragments was assessed by immunohisto/cytochemistry and Western blot in biopsies and ovarian cell lines. The TRKA and secretases' inhibition was performed using specific inhibitors. The results show that P75 immunodetection decreased during EOC progression and was negatively correlated with the presence of TRKA in EOC biopsies. NGF/TRKA increases ADAM17 levels and the fragments of P75 in ovarian cells. This effect is abolished when cells are previously treated with ADAM17, γ-secretase, and TRKA inhibitors. These results indicate that NGF/TRKA promotes the shedding of P75, involving the activation of secretases such as ADAM17. Since ADAM17 has been proposed as a screening marker for early detection of EOC, our results contribute to understanding better the role of ADAM17 and NGF/TRKA in EOC pathogenesis, which includes the NGF/TRKA-mediated cleavage of P75.
Collapse
Affiliation(s)
- Maritza P. Garrido
- Laboratorio de Endocrinología y Biología de la Reproducción, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile; (M.P.G.); (C.V.); (S.G.); (F.L.); (M.V.)
- Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
| | - Christopher Vallejos
- Laboratorio de Endocrinología y Biología de la Reproducción, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile; (M.P.G.); (C.V.); (S.G.); (F.L.); (M.V.)
| | - Silvanna Girardi
- Laboratorio de Endocrinología y Biología de la Reproducción, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile; (M.P.G.); (C.V.); (S.G.); (F.L.); (M.V.)
| | - Fernando Gabler
- Departamento de Patología, Escuela de Medicina, Hospital San Borja Arriarán, Universidad de Chile, Santiago 8360160, Chile;
| | - Alberto Selman
- Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
- Instituto Nacional del Cáncer, Santiago 8380455, Chile
| | - Fernanda López
- Laboratorio de Endocrinología y Biología de la Reproducción, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile; (M.P.G.); (C.V.); (S.G.); (F.L.); (M.V.)
| | - Margarita Vega
- Laboratorio de Endocrinología y Biología de la Reproducción, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile; (M.P.G.); (C.V.); (S.G.); (F.L.); (M.V.)
- Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
| | - Carmen Romero
- Laboratorio de Endocrinología y Biología de la Reproducción, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile; (M.P.G.); (C.V.); (S.G.); (F.L.); (M.V.)
- Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
- Correspondence:
| |
Collapse
|
8
|
ADAM17-A Potential Blood-Based Biomarker for Detection of Early-Stage Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13215563. [PMID: 34771725 PMCID: PMC8583642 DOI: 10.3390/cancers13215563] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 01/11/2023] Open
Abstract
Simple Summary Ovarian cancer has the highest lethality among gynecological tumors. Therefore, it is essential to find reliable biomarkers to improve early detection. This is the first report describing ADAM17 detection in serum and ascites fluid of ovarian cancer patients. A high ADAM17 concentration in serum at primary diagnosis is associated with early FIGO stages and predicts complete resection of the tumor mass. In addition, ADAM17 and CA-125 complement each other, especially in the diagnosis of early stages. In summary, ADAM17 appears to be a promising screening marker for detecting early-stage ovarian cancer. Abstract Ovarian cancer has the highest mortality rate among gynecological tumors. This is based on late diagnosis and the lack of early symptoms. To improve early detection, it is essential to find reliable biomarkers. The metalloprotease ADAM17 could be a potential marker, as it is highly expressed in many solid tumors, including ovarian and breast cancer. The aim of this work is to evaluate the relevance of ADAM17 as a potential diagnostic blood-based biomarker in ovarian cancer. Ovarian cancer cell lines IGROV-1 and A2780, as well as primary patient-derived tumor cells obtained from tumor tissue and ascitic fluid, were cultured to analyze ADAM17 abundance in the culture supernatant. In a translational approach, a cohort of 117 well-characterized ovarian cancer patients was assembled and ADAM17 levels in serum and corresponding ascitic fluid were determined at primary diagnosis. ADAM17 was quantified by enzyme-linked immunosorbent assay (ELISA). In the present study, ADAM17 was detected in the culture supernatant of ovarian cancer cell lines and primary cells. In addition, ADAM17 was found in serum and ascites of ovarian cancer patients. ADAM17 level was significantly increased in ovarian cancer patients compared to an age-matched control group (p < 0.0001). Importantly early FIGO I/II stages, which would not have been detected by CA-125, were associated with higher ADAM17 concentrations (p = 0.007). This is the first study proposing ADAM17 as a serum tumor marker in the setting of a gynecological tumor disease. Usage of ADAM17 in combination with CA-125 and other markers could help detect early stages of ovarian cancer.
Collapse
|
9
|
Implications of ADAM17 activation for hyperglycaemia, obesity and type 2 diabetes. Biosci Rep 2021; 41:228464. [PMID: 33904577 PMCID: PMC8128101 DOI: 10.1042/bsr20210029] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
In this review, we focus specifically on the role that the metalloproteinase, A Disintegrin and Metalloproteinase 17 [ADAM17] plays in the development and progression of the metabolic syndrome. There is a well-recognised link between the ADAM17 substrate tumour necrosis factor α (TNF-α) and obesity, inflammation and diabetes. In addition, knocking out ADAM17 in mice leads to an extremely lean phenotype. Importantly, ADAM17-deficient mice exhibit one of the most pronounced examples of hypermetabolism in rodents to date. It is vital to further understand the mechanistic role that ADAM17 plays in the metabolic syndrome. Such studies will demonstrate that ADAM17 is a valuable therapeutic target to treat obesity and diabetes.
Collapse
|
10
|
ADAM17 Inhibition Increases the Impact of Cisplatin Treatment in Ovarian Cancer Spheroids. Cancers (Basel) 2021; 13:cancers13092039. [PMID: 33922533 PMCID: PMC8122950 DOI: 10.3390/cancers13092039] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/15/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Ovarian cancer (OvCa) treatment is still a challenge, mainly due to acquired resistance mechanisms during the course of chemotherapy. Here, we show the enhanced cytotoxicity of the combined treatment with the ADAM17 inhibitor GW280264X and cisplatin in comparison with cisplatin monotherapy. This effect was visible in five of five ovarian cancer cell lines grown as a monolayer and two of three tested cell lines in three-dimensional tumor spheroids. Tumor spheroids derived from primary tumor and ascites cells were sensitized to cisplatin treatment by GW280264X. In summary, the combination of ADAM17 inhibition with conventional chemotherapy seems to be a promising strategy to overcome chemotherapy resistance in OvCa. Abstract Chemotherapy resistance is a major challenge in ovarian cancer (OvCa). Thus, novel treatment combinations are highly warranted. However, many promising drug candidates tested in two-dimensional (2D) cell culture have not proved successful in the clinic. For this reason, we analyzed our drug combination not only in monolayers but also in three-dimensional (3D) tumor spheroids. One potential therapeutic target for OvCa is A disintegrin and metalloprotease 17 (ADAM17). ADAM17 can be activated by chemotherapeutics, which leads to enhanced tumor growth due to concomitant substrate cleavage. Therefore, blocking ADAM17 during chemotherapy may overcome resistance. Here, we tested the effect of the ADAM17 inhibitor GW280264X in combination with cisplatin on ovarian cancer cells in 2D and 3D. In 2D, the effect on five cell lines was analyzed with two readouts. Three of these cell lines formed dense aggregates or spheroids (HEY, SKOV-3, and OVCAR-8) in 3D and the treatment effect was analyzed with a multicontent readout (cytotoxicity, viability, and caspase3/7 activation). We tested the combined therapy on tumor spheroids derived from primary patient cells. In 2D, we found a significant reduction in the half minimal (50%) inhibitory concentration (IC50) value of the combined treatment (GW280264X plus cisplatin) in comparison with cisplatin monotherapy in all five cell lines with both 2D readout assays (viability and caspase activation). In contrast, the combined treatment only showed an IC50 reduction in HEY and OVCAR-8 3D tumor spheroid models using caspase3/7 activity or CelltoxTM Green as the readout. Finally, we found an improved effect of GW280264X with cisplatin in tumor spheroids derived from patient samples. In summary, we demonstrate that ADAM17 inhibition is a promising treatment strategy in ovarian cancer.
Collapse
|
11
|
Expression of ADAM Proteases in Bladder Cancer Patients with BCG Failure: A Pilot Study. J Clin Med 2021; 10:jcm10040764. [PMID: 33672843 PMCID: PMC7917772 DOI: 10.3390/jcm10040764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 01/02/2023] Open
Abstract
Although Bacillus Calmette Guérin (BCG) remains a mainstay of adjuvant treatment in high-risk, non-muscle-invasive bladder cancer, BCG failure occurs in up to 40% of patients, with radical cystectomy (RC) as the inevitable therapeutic consequence. Current data suggest that PD-L1 immunosuppressive signaling is responsible for BCG failure, supporting the therapeutic rationale of combining checkpoint inhibitors with BCG. To address the immune cascade in 19 RC specimens obtained after BCG failure, we applied a small immunohistochemical (IHC) panel consisting of selected markers (PD-L1, GATA-3, a disintegrin and metalloproteinase (ADAM) proteases, IL-10/IL-10R). A modified quick score was used for IHC semi-quantification of these markers in tumor cells (TC) and immune cells (IC) within two different regions: muscle-invasive bladder cancer (MIBC) and primary/concurrent carcinoma in situ (CIS). Contrary to expectation, PD-L1 was consistently low, irrespective of tumor region and cell type. Intriguingly, expression of ADAM17, which has been reported to release membrane-bound PD-L1, was high in both tumor regions and cell types. Moreover, expression of GATA3, IL-10, and IL-10R was also increased, indicative of a generally immunosuppressive tumor microenvironment in BCG failure. ADAM10 expression was associated with advanced tumor disease at RC. Our findings raise the possibility that ADAM proteases may cleave PD-L1 from the surface of bladder TC and possibly also from IC. Therefore, IHC assessment of PD-L1 expression seems to be insufficient and should be supplemented by ADAM10/17 in patients with BCG failure.
Collapse
|
12
|
Borlak F, Reutzel-Selke A, Schirmeier A, Gogolok J, von Hoerschelmann E, Sauer IM, Pratschke J, Bahra M, Schmuck RB. Notch Signaling Pathway in Pancreatobiliary Tumors. ACTA ACUST UNITED AC 2021; 57:medicina57020105. [PMID: 33498866 PMCID: PMC7911049 DOI: 10.3390/medicina57020105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/13/2021] [Accepted: 01/16/2021] [Indexed: 11/30/2022]
Abstract
Background and Objectives: The Notch signaling pathway plays an important role both in the development of the ductal systems of the pancreas and the bile ducts as well as in cancer development and progression. The aim of this study was to examine the expression of central proteins of the Notch signaling pathway in pancreatobiliary tumors and its influence on patient survival. Materials and Methods: We compared the receptors (Notch1, Notch4), activating splicing factors (ADAM17), and target genes (HES1) of the Notch pathway and progenitor cell markers with relevance for the Notch signaling pathway (CD44, MSI1) between pancreatic adenocarcinomas (PDAC, n = 14), intrahepatic cholangiocarcinoma (iCC, n = 24), and extrahepatic cholangiocarcinoma (eCC, n = 22) cholangiocarcinomas via immunohistochemistry and ImageJ software-assisted analysis. An Immunohistochemistry (IHC)-score was determined by the percentage and intensity of stained (positive) cells (scale 0–7) and normal and malignant tissue was compared. In the IHC results, patients’ (gender, age) and tumor (TNM Classification of Malignant Tumors, Union Internationale contre le Cancer (UICC) stages, grading, and lymphangitic carcinomatosa) characteristics were correlated to patient survival. Results: For eCC, the expression of CD44 (p = 0.043, IHC-score 3.94 vs. 3.54) and for iCC, the expression of CD44 (p = 0.026, IHC-score 4.04 vs. 3.48) and Notch1 (p < 0.001, IHC-score 2.87 vs. 1.78) was significantly higher in the tumor compared to non-malignant tissue. For PDAC, the expression of ADAM17 (p = 0.008, IHC-score 3.43 vs. 1.73), CD44 (p = 0.012, IHC-score 3.64 vs. 2.27), Notch1 (p = 0.012, IHC-score 2.21 vs. 0.64), and Notch4 (p = 0.008, IHC-score 2.86 vs. 0.91) was significantly higher in the tumor tissue. However, none of the analyzed Notch-signaling related components showed an association to patient survival. Conclusion: A significant overexpression of almost all studied components of the Notch signaling pathway can be found in the tumor tissue, however, without a significant influence on patient survival. Therefore, further studies are warranted to draw conclusions on Notch pathway’s relevance for patient survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Rosa B. Schmuck
- Correspondence: ; Tel.: +49-30450652184; Fax: +49-304507652184
| |
Collapse
|
13
|
Wullweber A, Strick R, Lange F, Sikic D, Taubert H, Wach S, Wullich B, Bertz S, Weyerer V, Stoehr R, Breyer J, Burger M, Hartmann A, Strissel PL, Eckstein M. Bladder Tumor Subtype Commitment Occurs in Carcinoma In Situ Driven by Key Signaling Pathways Including ECM Remodeling. Cancer Res 2021; 81:1552-1566. [PMID: 33472889 DOI: 10.1158/0008-5472.can-20-2336] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/24/2020] [Accepted: 01/13/2021] [Indexed: 11/16/2022]
Abstract
Basal and luminal subtypes of invasive bladder tumors have significant prognostic and predictive impacts for patients. However, it remains unclear whether tumor subtype commitment occurs in noninvasive urothelial lesions or in carcinoma in situ (CIS) and which gene pathways are important for bladder tumor progression. To understand the timing of this commitment, we used gene expression and protein analysis to create a global overview of 36 separate tissues excised from a whole bladder encompassing urothelium, noninvasive urothelial lesions, CIS, and invasive carcinomas. Additionally investigated were matched CIS, noninvasive urothelial lesions, and muscle-invasive bladder cancers (MIBC) from 22 patients. The final stage of subtype commitment to either a luminal or basal MIBC occurred at the CIS transition. For all tissues combined, hierarchical clustering of subtype gene expression revealed three subtypes: "luminal," "basal," and a "luminal p53-/extracellular matrix (ECM)-like" phenotype of ECM-related genes enriched in tumor-associated urothelium, noninvasive urothelial lesions, and CIS, but rarely invasive, carcinomas. A separate cohort of normal urothelium from noncancer patients showed significantly lower expression of ECM-related genes compared with tumor-associated urothelium, noninvasive urothelial lesions, and CIS. A PanCancer Progression Panel of 681 genes unveiled pathways specific for the luminal p53-/ECM-like cluster, for example, ECM remodeling, angiogenesis, epithelial-to-mesenchymal transition, cellular discohesion, cell motility involved in tumor progression, and cell proliferation and oncogenic ERBB2/ERBB3 signaling for invasive carcinomas. In conclusion, this study provides insights into bladder cancer subtype commitment and associated signaling pathways, which could help predict therapy response and enhance our understanding of therapy resistance. SIGNIFICANCE: This study demonstrates that CIS is the stage of commitment for determining MIBC tumor subtype, which is relevant for patient prognosis and therapy response.
Collapse
Affiliation(s)
- Adrian Wullweber
- Department of Internal Medicine, Evangelisches Krankenhaus Düsseldorf, Düsseldorf, Germany.,Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Reiner Strick
- Translational Research Centre (TRC), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Fabienne Lange
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Danijel Sikic
- Department of Urology and Pediatric Urology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Helge Taubert
- Department of Urology and Pediatric Urology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sven Wach
- Department of Urology and Pediatric Urology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Bernd Wullich
- Department of Urology and Pediatric Urology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Simone Bertz
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Veronika Weyerer
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Robert Stoehr
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Johannes Breyer
- Department of Urology, Caritas Hospital St. Josef, University of Regensburg, Regensburg, Germany
| | - Maximilian Burger
- Department of Urology, Caritas Hospital St. Josef, University of Regensburg, Regensburg, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Pamela L Strissel
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.,Translational Research Centre (TRC), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| |
Collapse
|
14
|
Orzechowska M, Anusewicz D, Bednarek AK. Functional Gene Expression Differentiation of the Notch Signaling Pathway in Female Reproductive Tract Tissues-A Comprehensive Review With Analysis. Front Cell Dev Biol 2021; 8:592616. [PMID: 33384996 PMCID: PMC7770115 DOI: 10.3389/fcell.2020.592616] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
The Notch pathway involves evolutionarily conserved signaling regulating the development of the female tract organs such as breast, ovary, cervix, and uterine endometrium. A great number of studies revealed Notch aberrancies in association with their carcinogenesis and disease progression, the management of which is still challenging. The present study is a comprehensive review of the available literature on Notch signaling during the normal development and carcinogenesis of the female tract organs. The review has been enriched with our analyses of the TCGA data including breast, cervical, ovarian, and endometrial carcinomas concerning the effects of Notch signaling at two levels: the core components and downstream effectors, hence filling the lack of global overview of Notch-driven carcinogenesis and disease progression. Phenotype heterogeneity regarding Notch signaling was projected in two uniform manifold approximation and projection algorithm dimensions, preceded by the principal component analysis step reducing the data burden. Additionally, overall and disease-free survival analyses were performed with the optimal cutpoint determination by Evaluate Cutpoints software to establish the character of particular Notch components in tumorigenesis. In addition to the review, we demonstrated separate models of the examined cancers of the Notch pathway and its targets, although expression profiles of all normal tissues were much more similar to each other than to its cancerous compartments. Such Notch-driven cancerous differentiation resulted in a case of opposite association with DFS and OS. As a consequence, target genes also show very distinct profiles including genes associated with cell proliferation and differentiation, energy metabolism, or the EMT. In conclusion, the observed Notch associations with the female tract malignancies resulted from differential expression of target genes. This may influence a future analysis to search for new therapeutic targets based on specific Notch pathway profiles.
Collapse
Affiliation(s)
| | - Dorota Anusewicz
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
15
|
Xu R, Kang HF, Liu HB, Qian JB. Role of a disintegrin and metalloproteinase 17 in malignant tumors of the digestive system. Shijie Huaren Xiaohua Zazhi 2020; 28:986-991. [DOI: 10.11569/wcjd.v28.i19.986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A disintegrin and metalloproteinase 17 (ADAM17), also known as tumor necrosis factor-alpha converting enzyme, is widely distributed in mammalian cells and closely related to cell adhesion, migration, leukocyte recruitment, proteolysis, and other functions. ADAM17 plays an important role in the development of malignant tumors. On the one hand, it activates signaling pathways by mediating membrane protein shedding to participate in cell proliferation and angiogenesis. On the other hand, it plays an important role in tumor invasion and metastasis by degrading the cell basement membrane and extracellular matrix. Therefore, ADAM17 may be used as a potential target for tumor therapy. This article reviews the role of ADAM17 in malignant tumors of the digestive system.
Collapse
Affiliation(s)
- Rong Xu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital of Nantong University, Nantong 226000, Jiangsu Province, China
| | - Hai-Feng Kang
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital of Nantong University, Nantong 226000, Jiangsu Province, China
| | - Hong-Bin Liu
- Department of Pathology, The Second Affiliated Hospital of Nantong University, Nantong 226000, Jiangsu Province, China
| | - Jun-Bo Qian
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital of Nantong University, Nantong 226000, Jiangsu Province, China
| |
Collapse
|
16
|
Murumkar PR, Ghuge RB, Chauhan M, Barot RR, Sorathiya S, Choudhary KM, Joshi KD, Yadav MR. Recent developments and strategies for the discovery of TACE inhibitors. Expert Opin Drug Discov 2020; 15:779-801. [PMID: 32281878 DOI: 10.1080/17460441.2020.1744559] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION TNF-α plays a central role in certain autoimmune diseases as well as in inflammation. The current strategy for excluding TNF-α from circulation is to selectively inhibit TNF-α converting enzyme (TACE), an enzyme that cleaves mTNF-α to active TNF-α. Various TACE inhibitors have been discovered by using different strategies to control inflammatory diseases, cancer, and cardiac hypertrophy. AREAS COVERED The present article summarizes the design and discovery of novel TACE inhibitors that have been reported in the literature since 2012 onwards. It also includes some reports concerning the new role that TACE plays in cancer and cardiac hypertrophy. EXPERT OPINION So far, undertaken studies that have looked to design and develop small TACE inhibitors have been discouraging due to the failure of any TACE inhibitors to hit the market. However, some of the latest developments, such as with tartrate-based inhibitors, has given hope to the potentiality of a viable novel selective TACE inhibitor therapeutic in the future. Indeed, some of the novel peptidomimetics and monoclonal antibodies have great potential to pave the way for an effective and safe therapy by selectively inhibiting TACE enzyme.
Collapse
Affiliation(s)
- Prashant R Murumkar
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda , Vadodara, India
| | - Rahul B Ghuge
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda , Vadodara, India
| | - Monica Chauhan
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda , Vadodara, India
| | - Rahul R Barot
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda , Vadodara, India
| | - Sharmishtha Sorathiya
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda , Vadodara, India
| | - Kailash M Choudhary
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda , Vadodara, India
| | - Karan D Joshi
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda , Vadodara, India
| | - Mange Ram Yadav
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda , Vadodara, India
| |
Collapse
|
17
|
Chen JC, Huang C, Lee IN, Wu YP, Tang CH. Amphiregulin enhances cell migration and resistance to doxorubicin in chondrosarcoma cells through the MAPK pathway. Mol Carcinog 2018; 57:1816-1824. [DOI: 10.1002/mc.22899] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/24/2018] [Accepted: 08/31/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Jui-Chieh Chen
- Department of Biochemical Science and Technology; National Chiayi University; Chiayi City Taiwan
| | - Cheng Huang
- Department of Biotechnology and Laboratory Science in Medicine; National Yang-Ming University; Taipei Taiwan
- Department of Earth and Life Sciences; University of Taipei; Taipei Taiwan
| | - I-Neng Lee
- Department of Medical Research; Chang Gung Memorial Hospital; Chiayi Taiwan
| | - Yu-Ping Wu
- Department of Biochemical Science and Technology; National Chiayi University; Chiayi City Taiwan
- Department of Medical Research; Chang Gung Memorial Hospital; Chiayi Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology; School of Medicine; China Medical University; Taichung Taiwan
- Chinese Medicine Research Center; China Medical University; Taichung Taiwan
- Department of Biotechnology; College of Medical and Health Science; Asia University; Taichung Taiwan
| |
Collapse
|
18
|
Zhang Q, Wang C, Han X, Yang G, Ge Z, Zhang G. Knockdown of ADAM17 inhibits cell proliferation and increases oxaliplatin sensitivity in HCT-8 colorectal cancer through EGFR-PI3K-AKT activation. Biochem Biophys Res Commun 2018; 503:2333-2339. [DOI: 10.1016/j.bbrc.2018.06.158] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 06/27/2018] [Indexed: 11/25/2022]
|