1
|
Malikova I, Worth A, Aliyeva D, Khassenova M, Kriajevska MV, Tulchinsky E. Proteolysis of TAM receptors in autoimmune diseases and cancer: what does it say to us? Cell Death Dis 2025; 16:155. [PMID: 40044635 PMCID: PMC11883011 DOI: 10.1038/s41419-025-07480-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/09/2025]
Abstract
Proteolytic processing of Receptor Tyrosine Kinases (RTKs) leads to the release of ectodomains in the extracellular space. These soluble ectodomains often retain the ligand binding activity and dampen canonical pathways by acting as decoy receptors. On the other hand, shedding the ectodomains may initiate new molecular events and diversification of signalling. Members of the TAM (TYRO3, AXL, MER) family of RTKs undergo proteolytic cleavage, and their soluble forms are present in the extracellular space and biological fluids. TAM receptors are expressed in professional phagocytes, mediating apoptotic cell clearance, and suppressing innate immunity. Enhanced shedding of TAM ectodomains is documented in autoimmune and some inflammatory conditions. Also, soluble TAM receptors are present at high levels in the biological fluids of cancer patients and are associated with poor survival. We outline the biology of TAM receptors and discuss how their proteolytic processing impacts autoimmunity and tumorigenesis. In autoimmune diseases, proteolysis of TAM receptors likely reflects reduced canonical signalling in professional phagocytes. In cancer, TAM receptors are expressed in the immune cells of the tumour microenvironment, where they control pathways facilitating immune evasion. In tumour cells, ectodomain shedding activates non-canonical TAM pathways, leading to epithelial-mesenchymal transition, metastasis, and drug resistance.
Collapse
Affiliation(s)
- Ilona Malikova
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana, 020000, Kazakhstan
| | - Anastassiya Worth
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana, 020000, Kazakhstan
| | - Diana Aliyeva
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana, 020000, Kazakhstan
| | - Madina Khassenova
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana, 020000, Kazakhstan
| | - Marina V Kriajevska
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana, 020000, Kazakhstan
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Eugene Tulchinsky
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana, 020000, Kazakhstan.
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK.
| |
Collapse
|
2
|
Vázquez-Bellón N, Martínez-Bosch N, García de Frutos P, Navarro P. Hallmarks of pancreatic cancer: spotlight on TAM receptors. EBioMedicine 2024; 107:105278. [PMID: 39137571 PMCID: PMC11367522 DOI: 10.1016/j.ebiom.2024.105278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/14/2024] [Accepted: 07/29/2024] [Indexed: 08/15/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents the most prevalent type of pancreatic cancer and ranks among the most aggressive tumours, with a 5-year survival rate of less than 11%. Projections indicate that by 2030, it will become the second leading cause of cancer-related deaths. PDAC presents distinctive hallmarks contributing to its dismal prognosis: (i) late diagnosis, (ii) heterogenous and complex mutational landscape, (iii) high metastatic potential, (iv) dense fibrotic stroma, (v) immunosuppressive microenvironment, and (vi) high resistance to therapy. Mounting evidence has shown a role for TAM (Tyro3, AXL, MerTK) family of tyrosine kinase receptors in PDAC initiation and progression. This review aims to describe the impact of TAM receptors on the defining hallmarks of PDAC and discuss potential future directions using these proteins as novel biomarkers for early diagnosis and targets for precision therapy in PDAC, an urgent unmet clinical need.
Collapse
Affiliation(s)
- Núria Vázquez-Bellón
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB)-CSIC and Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain; PhD Program in Biomedicine, Facultat de Medicina (Campus Clínic), Universitat de Barcelona, Barcelona, Spain
| | - Neus Martínez-Bosch
- Cancer Research Program, Hospital del Mar Research Institute (HMRI), Unidad Asociada IIBB-CSIC, Barcelona, Spain
| | - Pablo García de Frutos
- Department of Cell Death and Proliferation, IIBB-CSIC, Unidad Asociada IMIM/IIBB-CSIC, Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), and IDIBAPS, Barcelona, Spain.
| | - Pilar Navarro
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB)-CSIC and Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain; Cancer Research Program, Hospital del Mar Research Institute (HMRI), Unidad Asociada IIBB-CSIC, Barcelona, Spain.
| |
Collapse
|
3
|
Sinha S, Guo R, Del Busso MD, Han W, Boysen J, Wellik LE, Ghosh AK, Kay NE. Aberrant activation of AXL may drive progression of squamous cell carcinoma in CLL patients: a mechanistic study with clinical implications. Br J Cancer 2024; 131:589-600. [PMID: 38886556 PMCID: PMC11300914 DOI: 10.1038/s41416-024-02752-1] [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: 10/19/2023] [Revised: 05/23/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Occurrence of squamous cell carcinoma (SCC) even in early-stage, untreated chronic lymphocytic leukemia (CLL) patients can be a significant morbidity issue with occasional transformation into metastatic skin lesions. METHODS CLL cells and extracellular vesicles (EVs) from CLL patients' blood/plasma were purified and used. Expression/activation of AXL and its functions in normal keratinocytes (HEKa) were assessed in vitro co-culture system and in SCC tissues. RESULTS We detected aberrant activation of AXL, AKT and ERK-1/2 in SCC cell lines compared to HEKa. We also detected increased expression of AXL in primary SCC tissues obtained from CLL patients. Increased activation of AXL, AKT, ERK-1/2 and Src was discernible in HEKa upon co-culturing with CLL cells. Further analysis suggests that Gas6, a ligand of AXL, regulates AXL activation in co-cultured HEKa. Interestingly, exposure of HEKa cells to CLL plasma-derived EVs induced expression of AXL, P-AKT, and EMT-associated markers leading to migration of the cells. Finally, pharmacologic inhibition of AXL induced cell death in SCC lines in a dose dependent manner. CONCLUSIONS Our findings that CLL cells likely are involved in driving SCC progression, at least in part, via activation of the AXL signaling axis, indicating that AXL inhibition may be beneficial for our CLL patients with SCC.
Collapse
Affiliation(s)
- Sutapa Sinha
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| | - Ray Guo
- Division of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Weiguo Han
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Justin Boysen
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Asish K Ghosh
- Stephenson Cancer Center and Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Neil E Kay
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
4
|
Tenstad O, Christakou E, Hodneland Nilsson L, Gausdal G, Micklem D, Kursula P, Lorens JB, Reed RK. In vivo turnover and biodistribution of soluble AXL: implications for biomarker development. Sci Rep 2024; 14:16141. [PMID: 38997436 PMCID: PMC11245488 DOI: 10.1038/s41598-024-66665-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: 03/13/2024] [Accepted: 07/03/2024] [Indexed: 07/14/2024] Open
Abstract
Soluble biomarkers are paramount to personalized medicine. However, the in vivo turnover and biodistribution of soluble proteins is seldom characterized. The cleaved extracellular domain of the AXL receptor (sAXL) is a prognostic biomarker in several diseases and a predictive marker of AXL targeting agents. Plasma sAXL reflects a balance between production in tissues with lymphatic transport into the circulation and removal from blood by degradation or excretion. It is unclear how this transport cycle affects plasma sAXL levels that are the metric for biomarker development. Radiolabeled mouse sAxl was monitored after intravenous injection to measure degradation and urinary excretion of sAxl, and after intradermal injection to mimic tissue or tumor production. sAxl was rapidly taken-up and degraded by the liver and kidney cortex. Surprisingly, intact sAxl was detectable in urine, indicating passage through the glomerular filter and a unique sampling opportunity. The structure of sAxl showed an elongated, flexible molecule with a length of 18 nm and a thickness of only 3 nm, allowing passage through the glomerulus and excretion into the urine. Intradermally injected sAxl passed through local and distant lymph nodes, followed by uptake in liver and kidney cortex. Low levels of sAxl were seen in the plasma, consistent with an extended transit time from local tissue to circulation. The rapid plasma clearance of sAxl suggests that steady-state levels in blood will sensitively and dynamically reflect the rate of production of sAxl in the tissues but will be influenced by perturbations of liver and kidney function.
Collapse
Affiliation(s)
- Olav Tenstad
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009, Bergen, Norway
| | - Eleni Christakou
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009, Bergen, Norway
- BerGenBio ASA, 5009, Bergen, Norway
| | | | | | | | - Petri Kursula
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009, Bergen, Norway
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, 90014, Oulu, Finland
| | - James B Lorens
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009, Bergen, Norway.
- Centre for Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, 5021, Bergen, Norway.
| | - Rolf K Reed
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009, Bergen, Norway
| |
Collapse
|
5
|
Raymond D, Fukui M, Zwernik S, Kassam A, Rovin R, Akhtar P. Evaluating soluble Axl as a biomarker for glioblastoma: A pilot study. PLoS One 2024; 19:e0301739. [PMID: 38968207 PMCID: PMC11226040 DOI: 10.1371/journal.pone.0301739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/11/2024] [Indexed: 07/07/2024] Open
Abstract
With current imaging, discriminating tumor progression from treatment effect following immunotherapy or oncolytic virotherapy of glioblastoma (GBM) is challenging. A blood based diagnostic biomarker would therefore be helpful. Axl is a receptor tyrosine kinase that is highly expressed by many cancers including GBM. Axl expression is regulated through enzymatic cleavage of its extracellular domain. The resulting fragment can be detected in serum as soluble Axl (sAxl). sAxl levels can distinguish patients with melanoma, hepatocellular carcinoma, and pancreatic ductal adenocarcinoma from healthy controls. This is a pilot study to determine if sAxl is a candidate biomarker for GBM. The sAxl levels in the serum of 40 healthy volunteers and 20 GBM patients were determined using an enzyme-linked immunosorbent assay (ELISA). Pre- and post- operative sAxl levels were obtained. Volumetric MRI evaluation provided GBM tumor volume metrics. There was no significant difference in the sAxl levels of the volunteers (30.16±1.88 ng/ml) and GBM patients (30.74±1.96 ng/ml) p = 0.27. The postoperative sAxl levels were significantly higher than preoperative levels (32.32±2.26 ng/ml vs 30.74±1.96 ng/ml, p = 0.03). We found no correlation between tumor volume and sAxl levels. Axl expression was low or absent in 6 of 11 (55%) patient derived GBM cell lines. Given the wide range of Axl expression by GBM tumors, sAxl may not be a reliable indicator of GBM. However, given the small sample size in this study, a larger study may be considered.
Collapse
Affiliation(s)
- Daniel Raymond
- Department of Biology, Northern Michigan University, Marquette, Michigan, United States of America
| | - Melanie Fukui
- Aurora Neuroscience Innovation Institute, Milwaukee, Wisconsin, United States of America
| | - Samuel Zwernik
- Advocate Aurora Research Institute, Milwaukee, Wisconsin, United States of America
| | - Amin Kassam
- Aurora Neuroscience Innovation Institute, Milwaukee, Wisconsin, United States of America
| | - Richard Rovin
- Aurora Neuroscience Innovation Institute, Milwaukee, Wisconsin, United States of America
| | - Parvez Akhtar
- Advocate Aurora Research Institute, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
6
|
Khan H, Abu-Raisi M, Feasson M, Shaikh F, Saposnik G, Mamdani M, Qadura M. Current Prognostic Biomarkers for Abdominal Aortic Aneurysm: A Comprehensive Scoping Review of the Literature. Biomolecules 2024; 14:661. [PMID: 38927064 PMCID: PMC11201473 DOI: 10.3390/biom14060661] [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: 05/02/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a progressive dilatation of the aorta that can lead to aortic rupture. The pathophysiology of the disease is not well characterized but is known to be caused by the general breakdown of the extracellular matrix within the aortic wall. In this comprehensive literature review, all current research on proteins that have been investigated for their potential prognostic capabilities in patients with AAA was included. A total of 45 proteins were found to be potential prognostic biomarkers for AAA, predicting incidence of AAA, AAA rupture, AAA growth, endoleak, and post-surgical mortality. The 45 proteins fell into the following seven general categories based on their primary function: (1) cardiovascular health, (2) hemostasis, (3) transport proteins, (4) inflammation and immunity, (5) kidney function, (6) cellular structure, (7) and hormones and growth factors. This is the most up-to-date literature review on current prognostic markers for AAA and their functions. This review outlines the wide pathophysiological processes that are implicated in AAA disease progression.
Collapse
Affiliation(s)
- Hamzah Khan
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Mohamed Abu-Raisi
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Manon Feasson
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Farah Shaikh
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Gustavo Saposnik
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Muhammad Mamdani
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Mohammad Qadura
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
7
|
Iwase T, Wang X, Thi Hanh Phi L, Sridhar N, Ueno NT, Lee J. Advances in targets in inflammatory breast cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 384:125-152. [PMID: 38637096 DOI: 10.1016/bs.ircmb.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Affiliation(s)
- Toshiaki Iwase
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; University of Hawaii Cancer Center, Honolulu, HI, United States.
| | - Xiaoping Wang
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lan Thi Hanh Phi
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nithya Sridhar
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Naoto T Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; University of Hawaii Cancer Center, Honolulu, HI, United States
| | - Jangsoon Lee
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
8
|
Tutusaus A, Morales A, García de Frutos P, Marí M. GAS6/TAM Axis as Therapeutic Target in Liver Diseases. Semin Liver Dis 2024; 44:99-114. [PMID: 38395061 PMCID: PMC11027478 DOI: 10.1055/a-2275-0408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
TAM (TYRO3, AXL, and MERTK) protein tyrosine kinase membrane receptors and their vitamin K-dependent ligands GAS6 and protein S (PROS) are well-known players in tumor biology and autoimmune diseases. In contrast, TAM regulation of fibrogenesis and the inflammation mechanisms underlying metabolic dysfunction-associated steatohepatitis (MASH), cirrhosis, and, ultimately, liver cancer has recently been revealed. GAS6 and PROS binding to phosphatidylserine exposed in outer membranes of apoptotic cells links TAMs, particularly MERTK, with hepatocellular damage. In addition, AXL and MERTK regulate the development of liver fibrosis and inflammation in chronic liver diseases. Acute hepatic injury is also mediated by the TAM system, as recent data regarding acetaminophen toxicity and acute-on-chronic liver failure have uncovered. Soluble TAM-related proteins, mainly released from activated macrophages and hepatic stellate cells after hepatic deterioration, are proposed as early serum markers for disease progression. In conclusion, the TAM system is becoming an interesting pharmacological target in liver pathology and a focus of future biomedical research in this field.
Collapse
Affiliation(s)
- Anna Tutusaus
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| | - Albert Morales
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| | - Pablo García de Frutos
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Cardiovasculares (CIBERCV), Barcelona, Comunidad de Madrid, Spain
| | - Montserrat Marí
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| |
Collapse
|
9
|
Apostolo D, Ferreira LL, Vincenzi F, Vercellino N, Minisini R, Latini F, Ferrari B, Burlone ME, Pirisi M, Bellan M. From MASH to HCC: the role of Gas6/TAM receptors. Front Immunol 2024; 15:1332818. [PMID: 38298195 PMCID: PMC10827955 DOI: 10.3389/fimmu.2024.1332818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is the replacement term for what used to be called nonalcoholic steatohepatitis (NASH). It is characterized by inflammation and injury of the liver in the presence of cardiometabolic risk factors and may eventually result in the development of hepatocellular carcinoma (HCC), the most common form of primary liver cancer. Several pathogenic mechanisms are involved in the transition from MASH to HCC, encompassing metabolic injury, inflammation, immune dysregulation and fibrosis. In this context, Gas6 (Growth Arrest-Specific 6) and TAM (Tyro3, Axl, and MerTK) receptors may play important roles. The Gas6/TAM family is involved in the modulation of inflammation, lipid metabolism, fibrosis, tumor progression and metastasis, processes which play an important role in the pathophysiology of acute and chronic liver diseases. In this review, we discuss MASH-associated HCC and the potential involvement of the Gas6/TAM system in disease development and progression. In addition, since therapeutic strategies for MASH and HCC are limited, we also speculate regarding possible future treatments involving the targeting of Gas6 or TAM receptors.
Collapse
Affiliation(s)
- Daria Apostolo
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Luciana L. Ferreira
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Federica Vincenzi
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Nicole Vercellino
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Rosalba Minisini
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Federico Latini
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Barbara Ferrari
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Michela E. Burlone
- Department of Internal Medicine, Azienda Ospedaliero-Universitaria Maggiore Della Carità, Novara, Italy
| | - Mario Pirisi
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- Department of Internal Medicine, Azienda Ospedaliero-Universitaria Maggiore Della Carità, Novara, Italy
- Center on Autoimmune and Allergic Diseases, Università del Piemonte Orientale, Novara, Italy
| | - Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- Department of Internal Medicine, Azienda Ospedaliero-Universitaria Maggiore Della Carità, Novara, Italy
- Center on Autoimmune and Allergic Diseases, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
10
|
Brown BA, Lazzara MJ. Single-cell RNA sequencing reveals microenvironment context-specific routes for epithelial-mesenchymal transition in pancreas cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542969. [PMID: 37398348 PMCID: PMC10312528 DOI: 10.1101/2023.05.30.542969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
In the PDAC tumor microenvironment, multiple factors initiate the epithelial-mesenchymal transition (EMT) that occurs heterogeneously among transformed ductal cells, but it is unclear if different drivers promote EMT through common or distinct signaling pathways. Here, we use single-cell RNA sequencing (scRNA-seq) to identify the transcriptional basis for EMT in pancreas cancer cells in response to hypoxia or EMT-inducing growth factors. Using clustering and gene set enrichment analysis, we find EMT gene expression patterns that are unique to the hypoxia or growth factor conditions or that are common between them. Among the inferences from the analysis, we find that the FAT1 cell adhesion protein is enriched in epithelial cells and suppresses EMT. Further, the receptor tyrosine kinase AXL is preferentially expressed in hypoxic mesenchymal cells in a manner correlating with YAP nuclear localization, which is suppressed by FAT1 expression. AXL inhibition prevents EMT in response to hypoxia but not growth factors. Relationships between FAT1 or AXL expression with EMT were confirmed through analysis of patient tumor scRNA-seq data. Further exploration of inferences from this unique dataset will reveal additional microenvironment context-specific signaling pathways for EMT that may represent novel drug targets for PDAC combination therapies.
Collapse
|
11
|
Bhadresha K, Mirza S, Penny C, Mughal MJ. Targeting AXL in Mesothelioma: from functional characterization to clinical implication. Crit Rev Oncol Hematol 2023:104043. [PMID: 37268175 DOI: 10.1016/j.critrevonc.2023.104043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/04/2023] Open
Abstract
Malignant pleural mesothelioma (MM) is a highly aggressive and lethal cancer with a poor survival rate. Current treatment approaches primarily rely on chemotherapy and radiation, but their effectiveness is limited. Consequently, there is an urgent need for alternative treatment strategies, a comprehensive understanding of the molecular mechanisms underlying MM, and the identification of potential therapeutic targets. Extensive studies over the past decade have emphasized the role of Axl in driving tumor development and metastasis, while high levels of Axl expression have been associated with immune evasion, drug resistance, and reduced patient survival in various cancer types. Ongoing clinical trials are investigating the efficacy of Axl inhibitors for different cancers. However, the precise role of Axl in MM progression, development, and metastasis, as well as its regulatory mechanisms within MM, remain inadequately understood. This review aims to comprehensively investigate the involvement of Axl in MM. We discuss Axl role in MM progression, development, and metastasis, along with its specific regulatory mechanisms. Additionally, we examined the Axl associated signaling pathways, the relationship between Axl and immune evasion, and the clinical implications of Axl for MM treatment. Furthermore, we discussed the potential utility of liquid biopsy as a non-invasive diagnostic technique for early detection of Axl in MM. Lastly, we evaluated the potential of a microRNA signature that targets Axl. By consolidating existing knowledge and identifying research gaps, this review contributes to a better understanding of Axl's role in MM and sets the stage for future investigations and the development of effective therapeutic interventions.
Collapse
Affiliation(s)
- Kinjal Bhadresha
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sheefa Mirza
- Department of Internal Medicine, Common Epithelial Cancer Research Center, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Clement Penny
- Department of Internal Medicine, Common Epithelial Cancer Research Center, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Muhammed Jameel Mughal
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Science, The George Washington University, Washington DC, United States of America.
| |
Collapse
|
12
|
Vago JP, Valdrighi N, Blaney-Davidson EN, Hornikx DLAH, Neefjes M, Barba-Sarasua ME, Thielen NGM, van den Bosch MHJ, van der Kraan PM, Koenders MI, Amaral FA, van de Loo FAJ. Gas6/Axl Axis Activation Dampens the Inflammatory Response in Osteoarthritic Fibroblast-like Synoviocytes and Synovial Explants. Pharmaceuticals (Basel) 2023; 16:ph16050703. [PMID: 37242486 DOI: 10.3390/ph16050703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent joint disease, and it is characterized by cartilage degeneration, synovitis, and bone sclerosis, resulting in swelling, stiffness, and joint pain. TAM receptors (Tyro3, Axl, and Mer) play an important role in regulating immune responses, clearing apoptotic cells, and promoting tissue repair. Here, we investigated the anti-inflammatory effects of a TAM receptor ligand, i.e., growth arrest-specific gene 6 (Gas6), in synovial fibroblasts from OA patients. TAM receptor expression was determined in synovial tissue. Soluble Axl (sAxl), a decoy receptor for the ligand Gas6, showed concentrations 4.6 times higher than Gas6 in synovial fluid of OA patients. In OA fibroblast-like synoviocytes (OAFLS) exposed to inflammatory stimuli, the levels of sAxl in the supernatants were increased, while the expression of Gas6 was downregulated. In OAFLS under TLR4 stimulation by LPS (Escherichia coli lipopolysaccharide), the addition of exogenous Gas6 by Gas6-conditioned medium (Gas6-CM) reduced pro-inflammatory markers including IL-6, TNF-α, IL-1β, CCL2, and CXCL8. Moreover, Gas6-CM downregulated IL-6, CCL2, and IL-1β in LPS-stimulated OA synovial explants. Pharmacological inhibition of TAM receptors by a pan inhibitor (RU301) or by a selective Axl inhibitor (RU428) similarly abrogated Gas6-CM anti-inflammatory effects. Mechanistically, Gas6 effects were dependent on Axl activation, determined by Axl, STAT1, and STAT3 phosphorylation, and by the downstream induction of the suppressors of the cytokine signaling family (SOCS1 and SOCS3). Taken together, our results showed that Gas6 treatment dampens inflammatory markers of OAFLS and synovial explants derived from OA patients associated with SOCS1/3 production.
Collapse
Affiliation(s)
- Juliana P Vago
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Natália Valdrighi
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Esmeralda N Blaney-Davidson
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Daniel L A H Hornikx
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Margot Neefjes
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - María E Barba-Sarasua
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Nathalie G M Thielen
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Martijn H J van den Bosch
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Peter M van der Kraan
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Marije I Koenders
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Flávio A Amaral
- Departament of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Fons A J van de Loo
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
13
|
Yong J, Elisabeth Groeger S, Ruf S, Ruiz-Heiland G. Influence of leptin and compression in GAS-6 mediated homeostasis of periodontal ligament cell. Oral Dis 2023; 29:1172-1183. [PMID: 34861742 DOI: 10.1111/odi.14092] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 11/21/2021] [Accepted: 12/01/2021] [Indexed: 11/29/2022]
Abstract
Growth arrest-specific protein 6 (GAS-6) regulates immunomodulatory and inflammatory mechanisms in periodontium and may participate in obesity predisposition. This study aimed to determine whether GAS-6 is associated with the homeostasis of periodontal ligament (SV-PDL) cells in the presence of adipokines or compressive forces. The SV-PDL cell line was used. Western blots were employed for TAM receptors detection. Cells were stimulated using different concentrations of GAS-6. The migration, viability, and proliferation were measured by a standard scratch test, MTS assay, and immunofluorescent staining. The mRNA expression was analyzed by RT-PCR. Release of TGF-β1, GAS-6, and Axl were verified by ELISA. Western blot shows that TAM receptors are expressed in SV-PDL cells. GAS-6 has a promoting effect on cell migration and proliferation. RT-PCR analysis showed that GAS-6 induces Collagen-1, Collagen-3, Periostin, and TGF-β1 mRNA expression whereas it reduces Caspase-3, Caspase-8, Caspase-9, and IL-6 mRNA expression. Further, secreted GAS-6 in SV-PDL is reduced in response to both compressive forces and leptin and upregulated by IL-6. Additionally, ADAM-10 inhibition reduces GAS-6 and Axl release on SV-PDL cells. TAM receptors especially Axl are identified as the receptors of GAS-6. GAS-6/TAM interactions contribute to periodontal ligament cells homeostasis. Leptin inhibits the GAS-6 release independently of ADAM-10 metalloprotease.
Collapse
Affiliation(s)
- Jiawen Yong
- Department of Orthodontics, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Sabine Elisabeth Groeger
- Department of Orthodontics, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Sabine Ruf
- Department of Orthodontics, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Gisela Ruiz-Heiland
- Department of Orthodontics, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
14
|
Zhou S, Li Y, Zhang Z, Yuan Y. An insight into the TAM system in Alzheimer's disease. Int Immunopharmacol 2023; 116:109791. [PMID: 36738678 DOI: 10.1016/j.intimp.2023.109791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/13/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
The TAM receptors may help delay the progression of Alzheimer's disease (AD). AD is the most common neurodegenerative disease associated with human aging. The TAM receptors, derived from the first letter of its three constituents -Tyro3, Axl, and Mertk, are associated with immune responses, cellular differentiation and migration, and clearance of apoptotic cells and debris, with the two canonical ligands, Growth Arrest Specific 6 (Gas6) and ProS1. Several kinds of research have indicated the participation of the TAM system in AD pathology. Also, the TAMs regulate multiple features of microglia, the significant sensors of disorder in the central nervous system (CNS). In this review, we describe the biology of the TAM receptors and ligands in the CNS. Then, we discuss the relationship between the TAM system and AD, specially focusing on its functional expression in the microglia. Finally, we also summarize some agents that could interfere with the TAM signaling pathways and discuss potential difficulties and strategies for drug development.
Collapse
Affiliation(s)
- Shiqi Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yanyan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yuhe Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
15
|
Phase I Study Evaluating Glesatinib (MGCD265), An Inhibitor of MET and AXL, in Patients with Non-small Cell Lung Cancer and Other Advanced Solid Tumors. Target Oncol 2023; 18:105-118. [PMID: 36459255 DOI: 10.1007/s11523-022-00931-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Heightened signaling by mesenchymal epithelial transition factor (MET) is implicated in tumorigenesis. Glesatinib is an investigational, oral inhibitor of MET and AXL. OBJECTIVE This phase I study determined the maximum tolerated dose (MTD), recommended phase II dose (RP2D), and safety profile of glesatinib in patients with advanced or unresectable solid tumors. Antitumor activity and pharmacokinetics (PK) were secondary objectives. PATIENTS AND METHODS Four formulations of glesatinib glycolate salt (capsule, unmicronized, micronized, and micronized version 2 [V2] tablets) and two free-base formulations (free-base suspension [FBS] capsule and spray-dried dispersion [SDD] tablet), developed to enhance drug exposure and optimize manufacturing processes, were evaluated in patients with genetically unselected advanced/unresectable solid tumors. MTD, based on dose-limiting toxicities (DLTs) observed during the first 21-day treatment cycle, was further evaluated in dose-expansion cohorts comprising patients with overexpression of MET and/or AXL, MET/AXL amplification, MET-activating mutations, or MET/AXL rearrangements for confirmation as the RP2D. RESULTS Glesatinib was evaluated across 27 dose-escalation cohorts (n = 108). Due to suboptimal exposure with glesatinib glycolate salt formulations in the initial cohorts, investigations subsequently focused on the FBS capsule and SDD tablet; for these formulations, MTD was identified as 1050 mg twice daily and 750 mg twice daily, respectively. An additional 71 patients received glesatinib in the FBS and SDD dose-expansion cohorts. At MTDs, the most frequent treatment-related adverse events were diarrhea (FBS, 83.3%; SDD, 75.0%), nausea (57.1%, 30.6%), vomiting (45.2%, 25.0%), increased alanine aminotransferase (45.2%, 30.6%), and increased aspartate aminotransferase (47.6%, 27.8%). Exploratory pharmacodynamic analyses indicated target engagement and inhibition of MET by glesatinib. Antitumor activity was observed with glesatinib FBS 1050 mg twice daily and SDD 750 mg twice daily in tumors harboring MET/AXL alteration or aberrant protein expression, particularly in patients with non--small cell lung cancer (NSCLC). In patients with NSCLC, the objective response rate was 25.9% in those with MET/AXL mutation or amplification and 30.0% in a subset with MET-activating mutations. All six partial responses occurred in patients with tumors carrying MET exon 14 deletion mutations. CONCLUSIONS The safety profile of single-agent glesatinib was acceptable. SDD 750 mg twice daily was selected as the preferred glesatinib formulation and dose based on clinical activity, safety, and PK data. Observations from this study led to initiation of a phase II study of glesatinib in patients with NSCLC stratified by type of MET alteration (NCT02544633). CLINICAL TRIALS REGISTRATION ClinicalTrials.gov NCT00697632; June 2008.
Collapse
|
16
|
van Aalen EA, Wouters SFA, Verzijl D, Merkx M. Bioluminescent RAPPID Sensors for the Single-Step Detection of Soluble Axl and Multiplex Analysis of Cell Surface Cancer Biomarkers. Anal Chem 2022; 94:6548-6556. [PMID: 35438976 PMCID: PMC9069438 DOI: 10.1021/acs.analchem.2c00297] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Early diagnosis of
cancer is essential for the efficacy of treatment.
Our group recently developed RAPPID, a bioluminescent immunoassay
platform capable of measuring a wide panel of biomarkers directly
in solution. Here, we developed and systematically screened different
RAPPID sensors for sensitive detection of the soluble fraction of
Axl (sAxl), a cell surface receptor that is overexpressed in several
types of cancer. The best-performing RAPPID sensor, with a limit of
detection of 8 pM and a >9-fold maximal change in
emission
ratio, was applied to measure Axl in three different contexts: clinically
relevant sAxl levels (∼0.5 and ∼1 nM) in diluted blood
plasma, proteolytically cleaved Axl in the cell culture medium of
A431 and HeLa cancer cells, and Axl on the membrane of A431 cells.
We further extended the sensor toolbox by developing dual-color RAPPID
for simultaneous detection of Axl and EGFR on A431 and HeLa cells,
as well as an AND-gate RAPPID that measures the concurrent presence
of these two cell surface receptors on the same cell. These new RAPPID
sensors provide attractive alternatives for more laborious protein
detection and quantification methods such as FACS and immunostainings,
due to their simple practical implantation and low intrinsic background
signal.
Collapse
Affiliation(s)
- Eva A van Aalen
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, P.O Box 513, 5600 MB Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O Box 513, 5600 MB Eindhoven, The Netherlands
| | - Simone F A Wouters
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, P.O Box 513, 5600 MB Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O Box 513, 5600 MB Eindhoven, The Netherlands
| | | | - Maarten Merkx
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, P.O Box 513, 5600 MB Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O Box 513, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
17
|
Tajbakhsh A, Gheibihayat SM, Taheri RA, Fasihi-Ramandi M, Bajestani AN, Taheri A. Potential diagnostic and prognostic of efferocytosis-related unwanted soluble receptors/ligands as new non-invasive biomarkers in disorders: a review. Mol Biol Rep 2022; 49:5133-5152. [PMID: 35419645 DOI: 10.1007/s11033-022-07224-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/02/2022] [Indexed: 11/25/2022]
Abstract
Efferocytosis is the process by which apoptotic cells are removed without inflammation to maintain tissue homeostasis, prevent unwanted inflammatory responses, and inhibit autoimmune responses. Coordination of efferocytosis occurs via many surfaces and chemotactic molecules and adaptors. Recently, soluble positive or negative mediators of efferocytosis, have been more noticeable as non-invasive valuable biomarkers in prognosis and targeted therapy. These soluble factors can be detected in different bodily fluids, such as serum, plasma, and urine as a non-invasive method. There are lots of studies that have tried to show the importance of receptors and ligands in disorders; while a few studies tried to indicate the importance of soluble forms of receptors/ligands and their clinical aspects as a systemic compound and shedding of targets related to efferocytosis. Some of these soluble forms also can be as sensitive as specific biomarkers for certain diseases compared with routine biomarkers, such as soluble circulatory Lectin-like oxidized low-density lipoprotein receptor-1 vs. troponin T in the acute coronary syndrome. Thus, this review tried to gain more understanding about efferocytosis-related unwanted soluble receptors/ligands, their roles, the clinical significance, and potential for diagnosis, and prognosis related to different diseases.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Molecular Biology Research Center, Systems Biology and Poisoning Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ramezan Ali Taheri
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahdi Fasihi-Ramandi
- Molecular Biology Research Center, Systems Biology and Poisoning Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abolfazl Nesaei Bajestani
- Department of Medical Genetics, Ayatollah Madani Hospital, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Abolfazl Taheri
- School of Medicine, New Hearing Technologies Research Center, Baghiyyatollah Al-Azam Hospital, Baqiyatallah University of Medical Sciences, Tehran, Iran.
- Department of ENT, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Patnaik A, Gadgeel S, Papadopoulos KP, Rasco DW, Haas NB, Der-Torossian H, Faltaos D, Potvin D, Tassell V, Tawashi M, Chao R, O'Dwyer PJ. Phase I Study of Glesatinib (MGCD256) in Combination with Erlotinib or Docetaxel in Patients with Advanced Solid Tumors. Target Oncol 2022; 17:125-138. [PMID: 35347559 DOI: 10.1007/s11523-022-00875-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Oncogenic drivers in solid tumors include aberrant activation of mesenchymal epithelial transition factor (MET) and AXL. OBJECTIVE This study investigated the safety and antitumor activity of glesatinib, a multitargeted receptor tyrosine kinase inhibitor that inhibits MET and AXL at clinically relevant doses, in combination with erlotinib or docetaxel. PATIENTS AND METHODS The phase I portion of this open-label, multicenter study included two parallel arms in which ascending doses of oral glesatinib (starting dose 96 mg/m2) were administered with erlotinib or docetaxel (starting doses 100 mg once daily and 50 mg/m2, respectively) using a modified 3 + 3 design. Maximum tolerated dose (MTD) was based on dose-limiting toxicities (DLTs) during the first 21-day treatment cycle. Enrollment focused on patients with solid tumor types typically associated with MET aberration and/or AXL overexpression. The primary objective was to determine the safety profile of the treatment combinations. Antitumor activity and pharmacokinetics (PK) were also assessed. RESULTS Ten dose levels of glesatinib across three glycolate formulations (unmicronized, micronized, or micronized version 2 [V2] tablets) available during the course of the study were investigated in 14 dose-escalation cohorts (n = 126). MTDs of unmicronized glesatinib plus erlotinib or docetaxel, and micronized glesatinib plus erlotinib were not reached. Micronized glesatinib 96 mg/m2 plus docetaxel exceeded the MTD. Further dosing focused on glesatinib micronized V2: maximum administered dose (MAD) was 700 mg twice daily with erlotinib 150 mg once daily or docetaxel 75 mg/m2 every 3 weeks. DLTs, acceptable at lower glesatinib (micronized V2) dose levels, occurred in two of five and two of six patients at the MADs of glesatinib + erlotinib and glesatinib + docetaxel, respectively. Across all cohorts, the most frequent treatment-related adverse events were diarrhea (glesatinib + erlotinib: 84.1%; glesatinib + docetaxel: 45.6%), fatigue (46.4%, 70.4%), and nausea (30.4%, 35.1%). The objective response rate was 1.8% and 12.0% in all glesatinib + erlotinib and glesatinib + docetaxel cohorts, respectively. CONCLUSIONS The safety profile of glesatinib plus erlotinib or docetaxel was acceptable and there were no PK interactions. MADs of glesatinib 700 mg twice daily (micronized V2) with erlotinib 150 mg once daily or docetaxel 75 mg/m2 every 3 weeks exceeded the MTD by a small margin. Modest signals of efficacy were observed with these treatment combinations in non-genetically selected patients with advanced solid tumors. CLINICAL TRIALS REGISTRATION ClinicalTrials.gov NCT00975767; 11 September 2009.
Collapse
Affiliation(s)
- Amita Patnaik
- START, 4383 Medical Drive, Suite 4026, San Antonio, TX, 78229, USA.
| | - Shirish Gadgeel
- Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA.,Henry Ford Health System, Detroit, MI, USA
| | | | - Drew W Rasco
- START, 4383 Medical Drive, Suite 4026, San Antonio, TX, 78229, USA
| | - Naomi B Haas
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Demiana Faltaos
- Mirati Therapeutics Inc., San Diego, CA, USA.,Olema Therapeutics, San Francisco, CA, USA
| | | | | | - Manal Tawashi
- Mirati Therapeutics Inc., San Diego, CA, USA.,HUYABIO International, San Diego, CA, USA
| | | | - Peter J O'Dwyer
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
19
|
Scherschinski L, Prem M, Kremenetskaia I, Tinhofer I, Vajkoczy P, Karbe AG, Onken JS. Regulation of the Receptor Tyrosine Kinase AXL in Response to Therapy and Its Role in Therapy Resistance in Glioblastoma. Int J Mol Sci 2022; 23:ijms23020982. [PMID: 35055167 PMCID: PMC8781963 DOI: 10.3390/ijms23020982] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/04/2022] [Accepted: 01/13/2022] [Indexed: 01/08/2023] Open
Abstract
The receptor tyrosine kinase AXL (RTK-AXL) is implicated in therapy resistance and tumor progression in glioblastoma multiforme (GBM). Here, we investigated therapy-induced receptor modifications and how endogenous RTK-AXL expression and RTK-AXL inhibition contribute to therapy resistance in GBM. GBM cell lines U118MG and SF126 were exposed to temozolomide (TMZ) and radiation (RTX). Receptor modifications in response to therapy were investigated on protein and mRNA levels. TMZ-resistant and RTK-AXL overexpressing cell lines were exposed to increasing doses of TMZ and RTX, with and without RTK-AXL tyrosine kinase inhibitor (TKI). Colorimetric microtiter (MTT) assay and colony formation assay (CFA) were used to assess cell viability. Results showed that the RTK-AXL shedding product, C-terminal AXL (CT-AXL), rises in response to repeated TMZ doses and under hypoxia, acts as a surrogate marker for radio-resistance. Endogenous RTX-AXL overexpression leads to therapy resistance, whereas combination therapy of TZM and RTX with TKI R428 significantly increases therapeutic effects. This data proves the role of RTK-AXL in acquired and intrinsic therapy resistance. By demonstrating that therapy resistance may be overcome by combining AXL TKI with standard treatments, we have provided a rationale for future study designs investigating AXL TKIs in GBM.
Collapse
Affiliation(s)
- Lea Scherschinski
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
| | - Markus Prem
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
- Department of Neurosurgery, Technische Universität Dresden, 01069 Dresden, Germany
| | - Irina Kremenetskaia
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
| | - Ingeborg Tinhofer
- Department of Radiooncology and Radiotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany;
- German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung–DKTK), Partner Site Berlin, 10115 Berlin, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
| | - Anna-Gila Karbe
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
| | - Julia Sophie Onken
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
- German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung–DKTK), Partner Site Berlin, 10115 Berlin, Germany
- Correspondence: ; Tel.: +49-(0)30-450-660253
| |
Collapse
|
20
|
Axl regulated survival/proliferation network and its therapeutic intervention in mouse models of glomerulonephritis. Arthritis Res Ther 2022; 24:284. [PMID: 36578056 PMCID: PMC9795606 DOI: 10.1186/s13075-022-02965-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/02/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Lupus nephritis (LN) is the most common and serious complication of systemic lupus erythematosus (SLE). LN pathogenesis is not fully understood. Axl receptor tyrosine kinase is upregulated and contributes to the pathogenic progress in LN. We have reported that Axl disruption attenuates nephritis development in mice. METHODS In this study, we analyzed the gene expression profiles with RNA-seq using renal cortical samples from nephritic mice. Axl-KO mice were bred onto a B6.lpr spontaneous lupus background, and renal disease development was followed and compared to the Axl-sufficient B6.lpr mice. Finally, anti-glomerular basement membrane (anti-GBM) Ab-induced nephritic mice were treated with Axl small molecule inhibitor, R428, at different stages of nephritis development. Blood urine nitrogen levels and renal pathologies were evaluated. RESULTS Transcriptome analysis revealed that renal Axl activation contributed to cell proliferation, survival, and motility through regulation of the Akt, c-Jun, and actin pathways. Spontaneous lupus-prone B6.lpr mice with Axl deficiency showed significantly reduced kidney damages and decreased T cell infiltration compared to the renal damage and T cell infiltration in Axl-sufficient B6.lpr mice. The improved kidney function was independent of autoAb production. Moreover, R428 significantly reduced anti-GBM glomerulonephritis at different stages of GN development compared to the untreated nephritic control mice. R428 administration reduced inflammatory cytokine (IL-6) production, T cell infiltration, and nephritis disease activity. CONCLUSIONS Results from this study emphasize the important role of Axl signaling in LN and highlight Axl as an attractive target in LN.
Collapse
|
21
|
Brosseron F, Maass A, Kleineidam L, Ravichandran KA, González PG, McManus RM, Ising C, Santarelli F, Kolbe CC, Häsler LM, Wolfsgruber S, Marquié M, Boada M, Orellana A, de Rojas I, Röske S, Peters O, Cosma NC, Cetindag A, Wang X, Priller J, Spruth EJ, Altenstein S, Schneider A, Fliessbach K, Wiltfang J, Schott BH, Bürger K, Janowitz D, Dichgans M, Perneczky R, Rauchmann BS, Teipel S, Kilimann I, Göerß D, Laske C, Munk MH, Düzel E, Yakupov R, Dobisch L, Metzger CD, Glanz W, Ewers M, Dechent P, Haynes JD, Scheffler K, Roy N, Rostamzadeh A, Teunissen CE, Marchant NL, Spottke A, Jucker M, Latz E, Wagner M, Mengel D, Synofzik M, Jessen F, Ramirez A, Ruiz A, Heneka MT. Soluble TAM receptors sAXL and sTyro3 predict structural and functional protection in Alzheimer's disease. Neuron 2021; 110:1009-1022.e4. [PMID: 34995486 DOI: 10.1016/j.neuron.2021.12.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 11/17/2021] [Accepted: 12/10/2021] [Indexed: 10/19/2022]
Abstract
There is an urgent need to improve the understanding of neuroinflammation in Alzheimer's disease (AD). We analyzed cerebrospinal fluid inflammatory biomarker correlations to brain structural volume and longitudinal cognitive outcomes in the DELCODE study and in a validation cohort of the F.ACE Alzheimer Center Barcelona. We investigated whether respective biomarker changes are evident before onset of cognitive impairment. YKL-40; sTREM2; sAXL; sTyro3; MIF; complement factors C1q, C4, and H; ferritin; and ApoE protein were elevated in pre-dementia subjects with pathological levels of tau or other neurodegeneration markers, demonstrating tight interactions between inflammation and accumulating neurodegeneration even before onset of symptoms. Intriguingly, higher levels of ApoE and soluble TAM receptors sAXL and sTyro3 were related to larger brain structure and stable cognitive outcome at follow-up. Our findings indicate a protective mechanism relevant for intervention strategies aiming to regulate neuroinflammation in subjects with no or subjective symptoms but underlying AD pathology profile.
Collapse
Affiliation(s)
- Frederic Brosseron
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, Magdeburg 39120, Germany
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Kishore Aravind Ravichandran
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Pablo García González
- Research Center and Memory Clinic, Fundacío ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Christina Ising
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Francesco Santarelli
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Carl-Christian Kolbe
- University of Bonn Medical Center, Institute of Innate Immune, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Lisa M Häsler
- Hertie Institute for Clinical Brain Research, Department of Cellular Neurology, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, Tübingen 72076, Germany
| | - Steffen Wolfsgruber
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Marta Marquié
- Research Center and Memory Clinic, Fundacío ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, Fundacío ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Adelina Orellana
- Research Center and Memory Clinic, Fundacío ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Itziar de Rojas
- Research Center and Memory Clinic, Fundacío ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Röske
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin 10117, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Nicoleta-Carmen Cosma
- German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin 10117, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Arda Cetindag
- German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin 10117, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Xiao Wang
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin 10117, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany; Department of Psychiatry and Psychotherapy, Technical University Munich, 81675 Munich, Germany
| | - Eike J Spruth
- German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin 10117, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin 10117, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Jens Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, Göttingen 37075, Germany; Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany; Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Björn H Schott
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, Göttingen 37075, Germany; Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany; Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Katharina Bürger
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, Munich 81377, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Martin Dichgans
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, Munich 81377, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, Munich 81377, Germany; Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany; Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Boris-Stephan Rauchmann
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Gehlsheimer Str. 20, Rostock 18147, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Gehlsheimer Str. 20, Rostock 18147, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Doreen Göerß
- Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, Tübingen 72076, Germany; Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Matthias H Munk
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, Tübingen 72076, Germany; Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, Magdeburg 39120, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Renat Yakupov
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, Magdeburg 39120, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, Magdeburg 39120, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Coraline D Metzger
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, Magdeburg 39120, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany; Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, Magdeburg, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, Magdeburg 39120, Germany
| | - Michael Ewers
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, Munich 81377, Germany
| | - Peter Dechent
- MR-Research in Neurosciences, Department of Cognitive Neurology, Georg-August-University Göttingen, Göttingen, Germany
| | - John Dylan Haynes
- Bernstein Center for Computational Neurosciences, Charité - Universitätsmedizin, Berlin, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, 72076 Tübingen, Germany
| | - Nina Roy
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany
| | - Ayda Rostamzadeh
- Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical centers, Vrije Universiteit, Amsterdam, the Netherlands
| | | | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, Department of Cellular Neurology, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, Tübingen 72076, Germany
| | - Eicke Latz
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; University of Bonn Medical Center, Institute of Innate Immune, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - David Mengel
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, Tübingen 72076, Germany; Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - Matthis Synofzik
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, Tübingen 72076, Germany; Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Köln, Germany
| | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Köln, Germany; Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, USA
| | - Agustín Ruiz
- Research Center and Memory Clinic, Fundacío ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, 4362 Esch-sur- Alzette, Luxembourg; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, 55 Lake Avenue, North Worcester, Massachusetts 01655, USA.
| | | |
Collapse
|
22
|
Exploring the Gamut of Receptor Tyrosine Kinases for Their Promise in the Management of Non-Alcoholic Fatty Liver Disease. Biomedicines 2021; 9:biomedicines9121776. [PMID: 34944593 PMCID: PMC8698495 DOI: 10.3390/biomedicines9121776] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 11/17/2022] Open
Abstract
Recently, non-alcoholic fatty liver disease (NAFLD) has emerged as a predominant health concern affecting approximately a quarter of the world’s population. NAFLD is a spectrum of liver ailments arising from nascent lipid accumulation and leading to inflammation, fibrosis or even carcinogenesis. Despite its prevalence and severity, no targeted pharmacological intervention is approved to date. Thus, it is imperative to identify suitable drug targets critical to the development and progression of NAFLD. In this quest, a ray of hope is nestled within a group of proteins, receptor tyrosine kinases (RTKs), as targets to contain or even reverse NAFLD. RTKs control numerous vital biological processes and their selective expression and activity in specific diseases have rendered them useful as drug targets. In this review, we discuss the recent advancements in characterizing the role of RTKs in NAFLD progression and qualify their suitability as pharmacological targets. Available data suggests inhibition of Epidermal Growth Factor Receptor, AXL, Fibroblast Growth Factor Receptor 4 and Vascular Endothelial Growth Factor Receptor, and activation of cellular mesenchymal-epithelial transition factor and Fibroblast Growth Factor Receptor 1 could pave the way for novel NAFLD therapeutics. Thus, it is important to characterize these RTKs for target validation and proof-of-concept through clinical trials.
Collapse
|
23
|
Nicolè L, Cappello F, Cappellesso R, Piccin L, Ventura L, Guzzardo V, Del Fiore P, Chiarion-Sileni V, Dei Tos AP, Mocellin S, Fassina A. RIPK3 and AXL Expression Study in Primary Cutaneous Melanoma Unmasks AXL as Predictor of Sentinel Node Metastasis: A Pilot Study. Front Oncol 2021; 11:728319. [PMID: 34745951 PMCID: PMC8566987 DOI: 10.3389/fonc.2021.728319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/30/2021] [Indexed: 11/13/2022] Open
Abstract
Malignant melanoma (MM) is the most lethal skin cancer. AXL is a tyrosine kinase receptor involved in several oncogenic processes and might play a role in blocking necroptosis (a regulated cell death mechanism) in MM through the downregulation of the necroptotic-related driver RIPK3. The aim of this study was to evaluate the clinical impact of the expression of AXL and RIPK3 in 108 primary cutaneous MMs. Association between AXL and RIPK3 immunoreactivity and clinical-pathological variables, sentinel lymph node status, and tumor-infiltrating lymphocytes (TILs) was assessed. Immunoreaction in tumor cells was detected in 30 cases (28%; range, 5%-80%) and in 17 cases (16%; range, 5%-50%) for AXL and RIPK3, respectively. Metastases in the sentinel lymph nodes were detected in 14 out of 61 patients, and these were associated with AXL-positive immunoreactivity in the primary tumor (p < 0.0001). No association between AXL and TILs was found. RIPK3 immunoreactivity was not associated with any variables. A final logistic regression analysis showed Breslow and AXL-positive immunoreactivity as the stronger predictor for positive sentinel node status [area under the receiver operating characteristic curve (AUC) of 0.96]. AXL could be a potential new biomarker for MM risk assessment, and it deserves to be further investigated in larger studies.
Collapse
Affiliation(s)
- Lorenzo Nicolè
- Department of Medicine (DIMED), University of Padova, Padova, Italy.,Unit of Surgical Pathology & Cytopathology, Ospedale dell'Angelo, Mestre, Italy
| | - Filippo Cappello
- Department of Medicine (DIMED), University of Padova, Padova, Italy.,Pathological Anatomy Unit, University Hospital of Padova, Padova, Italy
| | - Rocco Cappellesso
- Pathological Anatomy Unit, University Hospital of Padova, Padova, Italy
| | - Luisa Piccin
- Melanoma Oncology Unit, Istituto Oncologico Veneto (IOV-IRCCS), Padova, Italy
| | - Laura Ventura
- Department of Statistical Sciences, University of Padova, Padova, Italy
| | | | - Paolo Del Fiore
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, IOV- IRCCS, Padua, Italy
| | | | - Angelo Paolo Dei Tos
- Department of Medicine (DIMED), University of Padova, Padova, Italy.,Pathological Anatomy Unit, University Hospital of Padova, Padova, Italy
| | - Simone Mocellin
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, IOV- IRCCS, Padua, Italy
| | - Ambrogio Fassina
- Department of Medicine (DIMED), University of Padova, Padova, Italy
| |
Collapse
|
24
|
Emerging Importance of Tyrosine Kinase Inhibitors against Cancer: Quo Vadis to Cure? Int J Mol Sci 2021; 22:ijms222111659. [PMID: 34769090 PMCID: PMC8584061 DOI: 10.3390/ijms222111659] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/24/2021] [Accepted: 10/24/2021] [Indexed: 12/12/2022] Open
Abstract
GLOBOCAN 2020 estimated more than 19.3 million new cases, and about 10 million patients were deceased from cancer in 2020. Clinical manifestations showed that several growth factor receptors consisting of transmembrane and cytoplasmic tyrosine kinase (TK) domains play a vital role in cancer progression. Receptor tyrosine kinases (RTKs) are crucial intermediaries of the several cellular pathways and carcinogenesis that directly affect the prognosis and survival of higher tumor grade patients. Tyrosine kinase inhibitors (TKIs) are efficacious drugs for targeted therapy of various cancers. Therefore, RTKs have become a promising therapeutic target to cure cancer. A recent report shows that TKIs are vital mediators of signal transduction and cancer cell proliferation, angiogenesis, and apoptosis. In this review, we discuss the structure and function of RTKs to explore their prime role in cancer therapy. Various TKIs have been developed to date that contribute a lot to treating several types of cancer. These TKI based anticancer drug molecules are also discussed in detail, incorporating their therapeutic efficacy, mechanism of action, and side effects. Additionally, this article focuses on TKIs which are running in the clinical trial and pre-clinical studies. Further, to gain insight into the pathophysiological mechanism of TKIs, we also reviewed the impact of RTK resistance on TKI clinical drugs along with their mechanistic acquired resistance in different cancer types.
Collapse
|
25
|
Khera L, Lev S. Accelerating AXL targeting for TNBC therapy. Int J Biochem Cell Biol 2021; 139:106057. [PMID: 34403827 DOI: 10.1016/j.biocel.2021.106057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/02/2021] [Accepted: 08/10/2021] [Indexed: 12/11/2022]
Abstract
The tyrosine kinase receptor AXL of the TAM (TYRO3, AXL and MERTK) family is considered as a promising therapeutic target for different hematological cancers and solid tumors. AXL is involved in multiple pro-tumorigenic processes including cell migration, invasion, epithelial-mesenchymal transition (EMT), and stemness, and recent studies demonstrated its impact on cancer metastasis and drug resistance. Extensive studies on AXL have highlighted its unique characteristics and physiological functions and suggest that targeting of AXL could be beneficial in combination with chemotherapy, radiotherapy, immunotherapy, and targeted therapy. In this mini review, we discuss possible outcomes of AXL targeting either alone or together with other therapeutic agents and emphasize its impact on triple negative breast cancer (TNBC).
Collapse
Affiliation(s)
- Lohit Khera
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Sima Lev
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
26
|
AXL Receptor in Cancer Metastasis and Drug Resistance: When Normal Functions Go Askew. Cancers (Basel) 2021; 13:cancers13194864. [PMID: 34638349 PMCID: PMC8507788 DOI: 10.3390/cancers13194864] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/15/2021] [Accepted: 09/21/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary AXL is a member of the TAM (TYRO3, AXL, MER) family of receptor tyrosine kinases. In normal physiological conditions, AXL is involved in removing dead cells and their remains, and limiting the duration of immune responses. Both functions are utilized by cancers in the course of tumour progression. Cancer cells use the AXL pathway to detect toxic environments and to activate molecular mechanisms, thereby ensuring their survival or escape from the toxic zone. AXL is instrumental in controlling genetic programs of epithelial-mesenchymal and mesenchymal-epithelial transitions, enabling cancer cells to metastasize. Additionally, AXL signaling suppresses immune responses in tumour microenvironment and thereby helps cancer cells to evade immune surveillance. The broad role of AXL in tumour biology is the reason why its inhibition sensitizes tumours to a broad spectrum of anti-cancer drugs. In this review, we outline molecular mechanisms underlying AXL function in normal tissues, and discuss how these mechanisms are adopted by cancers to become metastatic and drug-resistant. Abstract The TAM proteins TYRO3, AXL, and MER are receptor tyrosine kinases implicated in the clearance of apoptotic debris and negative regulation of innate immune responses. AXL contributes to immunosuppression by terminating the Toll-like receptor signaling in dendritic cells, and suppressing natural killer cell activity. In recent years, AXL has been intensively studied in the context of cancer. Both molecules, the receptor, and its ligand GAS6, are commonly expressed in cancer cells, as well as stromal and infiltrating immune cells. In cancer cells, the activation of AXL signaling stimulates cell survival and increases migratory and invasive potential. In cells of the tumour microenvironment, AXL pathway potentiates immune evasion. AXL has been broadly implicated in the epithelial-mesenchymal plasticity of cancer cells, a key factor in drug resistance and metastasis. Several antibody-based and small molecule AXL inhibitors have been developed and used in preclinical studies. AXL inhibition in various mouse cancer models reduced metastatic spread and improved the survival of the animals. AXL inhibitors are currently being tested in several clinical trials as monotherapy or in combination with other drugs. Here, we give a brief overview of AXL structure and regulation and discuss the normal physiological functions of TAM receptors, focusing on AXL. We present a theory of how epithelial cancers exploit AXL signaling to resist cytotoxic insults, in order to disseminate and relapse.
Collapse
|
27
|
Li M, Xue W, Li X, Song Y, Liu X, Qin L. Axl is related to inflammation in hemodialysis patients. Mol Immunol 2021; 133:146-153. [PMID: 33667984 DOI: 10.1016/j.molimm.2021.02.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/10/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Hemodialysis (HD) patients often develop chronic inflammation, which is associated with an increased risk of cardiovascular complications and mortality. Axl and its ligand, growth arrest 6 (Gas6), have been reported to play key roles in regulating the immune response. However, the function of Axl in HD patients has not been clarified. METHODS In the present study, we enrolled 130 HD patients and 117 normal controls (NCs) and evaluated the levels of inflammatory markers, soluble Axl (sAxl), membrane Axl (mAxl), and Gas6 in all participants. The potential downstream cascades of Gas6-Axl signaling in HD patients were identified by quantitative real time polymerase chain reaction and western blotting. RESULTS The levels of inflammatory cytokines-tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ)-plasma sAxl, and Gas6, were significantly increased in HD patients compared to NCs. Additionally, sAxl was positively associated with the inflammatory factor, interleukin-6 (IL-6), in HD patients. Moreover, we found that mAxl in CD14+ mononuclear cells and CD19+ B cells was increased upon HD. Furthermore, we discovered that the metalloproteinase ADAM17, also called TACE, contributed to the cleavage of mAxl into sAxl, and not ADAM10, in the peripheral blood mononuclear cells (PBMCs) of HD patients. The upregulation of Gas6-mAxl signaling caused the activation of the STAT1-SOCS3 pathway in the PBMCs of HD patients. After two years follow-up, patients with lower sAxl levels had longer survival time than those with higher sAxl levels. CONCLUSION Our results suggest that Axl may play a significant role in systemic inflammation in HD patients.
Collapse
Affiliation(s)
- Mengyuan Li
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301, Middle Yanchang Road, Shanghai, 200072, China
| | - Wen Xue
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301, Middle Yanchang Road, Shanghai, 200072, China
| | - Xinhua Li
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301, Middle Yanchang Road, Shanghai, 200072, China
| | - Yaxiang Song
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301, Middle Yanchang Road, Shanghai, 200072, China
| | - Xinying Liu
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301, Middle Yanchang Road, Shanghai, 200072, China.
| | - Ling Qin
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301, Middle Yanchang Road, Shanghai, 200072, China.
| |
Collapse
|
28
|
AXL Receptor in Breast Cancer: Molecular Involvement and Therapeutic Limitations. Int J Mol Sci 2020; 21:ijms21228419. [PMID: 33182542 PMCID: PMC7696061 DOI: 10.3390/ijms21228419] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022] Open
Abstract
Breast cancer was one of the first malignancies to benefit from targeted therapy, i.e., treatments directed against specific markers. Inhibitors against HER2 are a significant example and they improved the life expectancy of a large cohort of patients. Research on new biomarkers, therefore, is always current and important. AXL, a member of the TYRO-3, AXL and MER (TAM) subfamily, is, today, considered a predictive and prognostic biomarker in many tumor contexts, primarily breast cancer. Its oncogenic implications make it an ideal target for the development of new pharmacological agents; moreover, its recent role as immune-modulator makes AXL particularly attractive to researchers involved in the study of interactions between cancer and the tumor microenvironment (TME). All these peculiarities characterize AXL as compared to other members of the TAM family. In this review, we will illustrate the biological role played by AXL in breast tumor cells, highlighting its molecular and biological features, its involvement in tumor progression and its implication as a target in ongoing clinical trials.
Collapse
|
29
|
Winge-Main AK, Wälchli S, Inderberg EM. T cell receptor therapy against melanoma-Immunotherapy for the future? Scand J Immunol 2020; 92:e12927. [PMID: 32640053 DOI: 10.1111/sji.12927] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022]
Abstract
Malignant melanoma has seen monumental changes in treatment options the last decade from the very poor results of dacarbazine treatment to the modern-day use of targeted therapies and immune checkpoint inhibitors. Melanoma has a high mutational burden making it more capable of evoking immune responses than many other tumours. Even when considering double immune checkpoint blockade with anti-CTLA-4 and anti-PD-1, we still have far to go in melanoma treatment as 50% of patients with metastatic disease do not respond to current treatment. Alternative immunotherapy should therefore be considered. Since melanoma has a high mutational burden, it is considered more immunogenic than many other tumours. T cell receptor (TCR) therapy could be a possible way forward, either alone or in combination, to improve the response rates of this deadly disease. Melanoma is one of the cancers where TCR therapy has been frequently applied. However, the number of antigens targeted remains fairly limited, although advanced personalized therapies aim at also targeting private mutations. In this review, we look at possible aspects of targeting TCR therapy towards melanoma and provide an implication of its use in the future.
Collapse
Affiliation(s)
- Anna K Winge-Main
- Department of Cellular Therapy, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Department of Cellular Therapy, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Else Marit Inderberg
- Department of Cellular Therapy, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
30
|
Pyle MP, Hoa M. Applications of single-cell sequencing for the field of otolaryngology: A contemporary review. Laryngoscope Investig Otolaryngol 2020; 5:404-431. [PMID: 32596483 PMCID: PMC7314468 DOI: 10.1002/lio2.388] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES Single-cell RNA sequencing (scRNA-Seq) is a new technique used to interrogate the transcriptome of individual cells within native tissues that have already resulted in key discoveries in auditory basic science research. Rapid advances in scRNA-Seq make it likely that it will soon be translated into clinical medicine. The goal of this review is to inspire the use of scRNA-Seq in otolaryngology by giving examples of how it can be applied to patient samples and how this information can be used clinically. METHODS Studies were selected based on the scientific quality and relevance to scRNA-Seq. In addition to mouse auditory system (inner ear including hair cells and supporting cells, spiral ganglion neurons, and inner ear organoids), recent studies using human primary cell samples are discussed. We also perform our own analysis on publicly available, published scRNA-Seq data from oral head and neck squamous cell carcinoma (HNSCC) samples to serve as an example of a clinically relevant application of scRNA-Seq. RESULTS Studies focusing on patient tissues show that scRNA-Seq reveals tissue heterogeneity and rare-cell types responsible for disease pathogenesis. The heterogeneity detected by scRNA-Seq can result in both the identification of known or novel disease biomarkers and drug targets. Our analysis of HNSCC data gives an example for how otolaryngologists can use scRNA-Seq for clinical use. CONCLUSIONS Although there are limitations to the translation of scRNA-Seq to the clinic, we show that its use in otolaryngology can give physicians insight into the tissue heterogeneity within their patient's diseased tissue giving them information on disease pathogenesis, novel disease biomarkers or druggable targets, and aid in selecting patient-specific drug cocktails.
Collapse
Affiliation(s)
- Madeline P. Pyle
- Division of Intramural Research, Section on Auditory Development and Restoration, National Institute on Deafness and Other Communication Disorders (NIDCD) Otolaryngology Surgeon‐Scientist ProgramNational Institutes of HealthBethesdaMarylandUSA
| | - Michael Hoa
- Division of Intramural Research, Section on Auditory Development and Restoration, National Institute on Deafness and Other Communication Disorders (NIDCD) Otolaryngology Surgeon‐Scientist ProgramNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|