1
|
Dong J, Xu Y, Yu D, Zhang X, Wang A, Lv L, Li Z. Gq/G11 oncogenic mutations promote PD-L1 expression and suppress tumor immunity. Eur J Cell Biol 2024; 103:151467. [PMID: 39550833 DOI: 10.1016/j.ejcb.2024.151467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/24/2024] [Accepted: 11/08/2024] [Indexed: 11/19/2024] Open
Abstract
Uveal melanoma (UM) is the predominant form of eye cancer. The genes GNAQ and GNA11, encoding Gq and G11 respectively, are most frequently mutated in UM and are considered the major drivers of UM carcinogenesis by activating YAP. However, the mechanisms by which metastatic UM evades the immune system remain poorly understood. In this study, we found that oncogenic mutations of Gq/G11 promoted YAP and PD-L1 expression, modifying the tumor microenvironment and promoting immune evasion of UM. Consistently, the levels of GNAQ/GNA11 and YAP positively correlated to PD-L1 expression in UM patients. Furthermore, silencing YAP or treating with its inhibitor, Verteporfin, attenuated PD-L1 expression induced by Gq/G11 mutations, thereby enhancing T cell activation and T cell-mediated cytotoxicity. Collectively, this study reveals a potential role of Gq/G11 mutations on immune evasion of UM, a new mechanism of Gq/11 mutations-induced tumorigenesis, highlighting Gq/G11 and YAP as potential immunotherapeutic targets and suggesting Verteporfin as an adjuvant for immunotherapy of UM patients with GNAQ or GNA11 mutations.
Collapse
Affiliation(s)
- Jingyan Dong
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China; Shanxi Eye Hospital, Taiyuan 030001, China
| | - Yue Xu
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200032, China
| | - Dawei Yu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China
| | - Xiaoling Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China
| | - Anqi Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China
| | - Lei Lv
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Zhiqing Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China.
| |
Collapse
|
2
|
Dwyer MB, Aumiller JL, Wedegaertner PB. Going Rogue: Mechanisms, Regulation, and Roles of Mutationally Activated G α in Human Cancer. Mol Pharmacol 2024; 106:198-215. [PMID: 39187387 PMCID: PMC11493338 DOI: 10.1124/molpharm.124.000743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024] Open
Abstract
G protein-coupled receptors (GPCRs) couple to heterotrimeric G proteins, comprised of α and βγ subunits, to convert extracellular signals into activation of intracellular signaling pathways. Canonically, GPCR-mediated activation results in the exchange of GDP for GTP on G protein α subunits (Gα) and the dissociation of Gα-GTP and G protein βγ subunits (Gβγ), both of which can regulate a variety of signaling pathways. Hydrolysis of bound GTP by Gα returns the protein to Gα-GDP and allows reassociation with Gβγ to reform the inactive heterotrimer. Naturally occurring mutations in Gα have been found at conserved glutamine and arginine amino acids that disrupt the canonical G protein cycle by inhibiting GTP hydrolysis, rendering these mutants constitutively active. Interestingly, these dysregulated Gα mutants are found in many different cancers due to their ability to sustain aberrant signaling without a need for activation by GPCRs. This review will highlight an increased recognition of the prevalence of such constitutively activating Gα mutations in cancers and the signaling pathways activated. In addition, we will discuss new knowledge regarding how these constitutively active Gα are regulated, how different mutations are biochemically distinct, and how mutationally activated Gα are unique compared with GPCR-activated Gα Lastly, we will discuss recent progress in developing inhibitors directly targeting constitutively active Gα mutants. SIGNIFICANCE STATEMENT: Constitutively activating mutations in G protein α subunits (Gα) widely occur in and contribute to the development of many human cancers. To develop ways to inhibit dysregulated, oncogenic signaling by these mutant Gα, it is crucial to better understand mechanisms that lead to constitutive Gα activation and unique mechanisms that regulate mutationally activated Gα in cells. The prevalence of activating mutations in Gα in various cancers makes Gα proteins compelling targets for the development of therapeutics.
Collapse
Affiliation(s)
- Morgan B Dwyer
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jenna L Aumiller
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Philip B Wedegaertner
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
3
|
Tonelotto V, Costa-Garcia M, O'Reilly E, Smith KF, Slater K, Dillon ET, Pendino M, Higgins C, Sist P, Bosch R, Passamonti S, Piulats JM, Villanueva A, Tramer F, Vanella L, Carey M, Kennedy BN. 1,4-dihydroxy quininib activates ferroptosis pathways in metastatic uveal melanoma and reveals a novel prognostic biomarker signature. Cell Death Discov 2024; 10:70. [PMID: 38341410 DOI: 10.1038/s41420-023-01773-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 02/12/2024] Open
Abstract
Uveal melanoma (UM) is an ocular cancer, with propensity for lethal liver metastases. When metastatic UM (MUM) occurs, as few as 8% of patients survive beyond two years. Efficacious treatments for MUM are urgently needed. 1,4-dihydroxy quininib, a cysteinyl leukotriene receptor 1 (CysLT1) antagonist, alters UM cancer hallmarks in vitro, ex vivo and in vivo. Here, we investigated the 1,4-dihydroxy quininib mechanism of action and its translational potential in MUM. Proteomic profiling of OMM2.5 cells identified proteins differentially expressed after 1,4-dihydroxy quininib treatment. Glutathione peroxidase 4 (GPX4), glutamate-cysteine ligase modifier subunit (GCLM), heme oxygenase 1 (HO-1) and 4 hydroxynonenal (4-HNE) expression were assessed by immunoblots. Biliverdin, glutathione and lipid hydroperoxide were measured biochemically. Association between the expression of a specific ferroptosis signature and UM patient survival was performed using public databases. Our data revealed that 1,4-dihydroxy quininib modulates the expression of ferroptosis markers in OMM2.5 cells. Biochemical assays validated that GPX4, biliverdin, GCLM, glutathione and lipid hydroperoxide were significantly altered. HO-1 and 4-HNE levels were significantly increased in MUM tumor explants from orthotopic patient-derived xenografts (OPDX). Expression of genes inhibiting ferroptosis is significantly increased in UM patients with chromosome 3 monosomy. We identified IFerr, a novel ferroptosis signature correlating with UM patient survival. Altogether, we demontrated that in MUM cells and tissues, 1,4-dihydroxy quininib modulates key markers that induce ferroptosis, a relatively new type of cell death driven by iron-dependent peroxidation of phospholipids. Furthermore, we showed that high expression of specific genes inhibiting ferroptosis is associated with a worse UM prognosis, thus, the IFerr signature is a potential prognosticator for which patients develop MUM. All in all, ferroptosis has potential as a clinical biomarker and therapeutic target for MUM.
Collapse
Affiliation(s)
- Valentina Tonelotto
- UCD Conway Institute, University College Dublin, D04 V1W8, Dublin, Ireland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8, Dublin, Ireland
| | - Marcel Costa-Garcia
- Medical Oncology Department, Catalan Institute of Cancer (ICO), IDIBELL-OncoBell, Barcelona, Spain
| | - Eve O'Reilly
- UCD Conway Institute, University College Dublin, D04 V1W8, Dublin, Ireland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8, Dublin, Ireland
| | - Kaelin Francis Smith
- UCD Conway Institute, University College Dublin, D04 V1W8, Dublin, Ireland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8, Dublin, Ireland
| | - Kayleigh Slater
- UCD Conway Institute, University College Dublin, D04 V1W8, Dublin, Ireland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8, Dublin, Ireland
| | - Eugene T Dillon
- Mass Spectrometry Resource, Conway Institute of Biomolecular & Biomedical Research, University College Dublin, D04 V1W8, Dublin, Ireland
| | - Marzia Pendino
- UCD Conway Institute, University College Dublin, D04 V1W8, Dublin, Ireland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8, Dublin, Ireland
| | - Catherine Higgins
- UCD School of Mathematics & Statistics, University College Dublin, D04 V1W8, Dublin, Ireland
| | - Paola Sist
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Rosa Bosch
- Xenopat S.L., Business Bioincubator, Bellvitge Health Science Campus, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Sabina Passamonti
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Josep M Piulats
- Medical Oncology Department, Catalan Institute of Cancer (ICO), IDIBELL-OncoBell, Barcelona, Spain
| | - Alberto Villanueva
- Xenopat S.L., Business Bioincubator, Bellvitge Health Science Campus, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
- Program Against Cancer Therapeutic Resistance (ProCURE), ICO, IDIBELL, Barcelona, Spain
| | - Federica Tramer
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
- CERNUT-Research Centre on Nutraceuticals and Health Products, University of Catania, 95125, Catania, Italy
| | - Michelle Carey
- Mass Spectrometry Resource, Conway Institute of Biomolecular & Biomedical Research, University College Dublin, D04 V1W8, Dublin, Ireland
| | - Breandán N Kennedy
- UCD Conway Institute, University College Dublin, D04 V1W8, Dublin, Ireland.
- UCD School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8, Dublin, Ireland.
| |
Collapse
|
4
|
Khan SA, Almalki WH, Arora S, Kesharwani P. Recent approaches for the treatment of uveal melanoma: Opportunities and challenges. Crit Rev Oncol Hematol 2024; 193:104218. [PMID: 38040071 DOI: 10.1016/j.critrevonc.2023.104218] [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: 10/11/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/03/2023] Open
Abstract
Uveal melanoma (UM) is the most prevalent primary intraocular cancer in adult population. Primary methods for treatment of UM involves surgery Proton Beam Therapy (PBT), Plaque Brachytherapy, phototherapy, and Charged Particle Radiation Therapy (CPT). It has been found that approximately 50 % of patients diagnosed with UM ultimately experience development of metastatic disease. Furthermore, it has been identified that majority of the patient experience metastasis in liver with a prevalence of 95 %. Management of metastatic UM (MUM) involves various therapeutic modalities, including systemic chemotherapy, molecular targeted therapy, immunotherapy and liver directed interventions. We outline gene mutation in UM and addresses various treatment modalities, including molecular targeted therapy, miRNA-based therapy, and immunotherapy. Additionally, inclusion of ongoing clinical trials aimed at developing novel therapeutic options for management of UM are also mentioned.
Collapse
Affiliation(s)
- Sauban Ahmed Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Swaranjeet Arora
- Department of Finance and Management, Lal Bahadur Shastri Institute of Management, 11/07 Dwarka Sector 11, Near Metro Station, New Delhi, Delhi 110075, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
5
|
Taskaeva I, Shatruk A, Bgatova N, Yeremina A, Trunov A, Kononova N, Chernykh V. Autophagy and vesicular trafficking in human uveal melanoma: A histopathological study. Microsc Res Tech 2024; 87:122-132. [PMID: 37698482 DOI: 10.1002/jemt.24417] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 08/04/2023] [Accepted: 09/03/2023] [Indexed: 09/13/2023]
Abstract
Uveal melanoma is an ocular tumor with a high risk of developing metastases. The endo-lysosomal system can affect the melanoma progression by accelerating and facilitating invasion or metastasis. This study aims to conduct comparative analysis of normal choroidal melanocytes and uveal melanoma cells ultrastructure with a focus on intracellular transport system, and to examine the patterns of autophagy- and vesicular trafficking-related proteins expression in a case series of uveal melanomas. Transmission electron microscopy was used to assess the ultrastructure of normal choroidal melanocytes and uveal melanoma cells. The expression levels of autophagy- and vesicular trafficking-related proteins in three histological types of uveal melanoma were analyzed by immunofluorescence staining. Electron microscopy results showed that the autophagic vacuoles were more abundant in normal choroidal melanocytes, than in uveal melanoma cells. The normal choroidal melanocytes were characterized by active intracellular vesicular trafficking; however, the proportion of caveolae was higher in uveal melanoma cells. The spindle type of tumor was characterized by a high expression levels of LC3 beta, while Rab7 and Rab11 proteins expression was significantly up-regulated in the mixed-type tumor cells. The results indicate that uveal melanoma cells probably have lower basal levels of autophagy and higher receptor-mediated endocytic trafficking-associated with caveolae than normal choroidal melanocytes. RESEARCH HIGHLIGHTS: The autophagic vacuoles are abundant in normal choroidal melanocytes. Uveal melanoma cells are characterized by a high proportion of caveolae. The high expression levels of LC3 beta were revealed in a spindle type of tumor, while Rab7 and Rab11 proteins expression was up-regulated in the mixed-type tumor cells.
Collapse
Affiliation(s)
- Iuliia Taskaeva
- Laboratory of Ultrastructural Research, Research Institute of Clinical and Experimental Lymphology - Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Anastasia Shatruk
- Laboratory of Ultrastructural Research, Research Institute of Clinical and Experimental Lymphology - Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Nataliya Bgatova
- Laboratory of Ultrastructural Research, Research Institute of Clinical and Experimental Lymphology - Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alena Yeremina
- S. N. Fyodorov Federal State Institution National Medical Research Center Intersectoral Research and Technology Complex "Eye Microsurgery" Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Aleksander Trunov
- S. N. Fyodorov Federal State Institution National Medical Research Center Intersectoral Research and Technology Complex "Eye Microsurgery" Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Natalya Kononova
- S. N. Fyodorov Federal State Institution National Medical Research Center Intersectoral Research and Technology Complex "Eye Microsurgery" Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Valeriy Chernykh
- S. N. Fyodorov Federal State Institution National Medical Research Center Intersectoral Research and Technology Complex "Eye Microsurgery" Ministry of Health of the Russian Federation, Novosibirsk, Russia
| |
Collapse
|
6
|
Falzone ME, MacKinnon R. The mechanism of Gαq regulation of PLCβ3-catalyzed PIP2 hydrolysis. Proc Natl Acad Sci U S A 2023; 120:e2315011120. [PMID: 37991948 PMCID: PMC10691244 DOI: 10.1073/pnas.2315011120] [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: 08/30/2023] [Accepted: 10/13/2023] [Indexed: 11/24/2023] Open
Abstract
PLCβ (Phospholipase Cβ) enzymes cleave phosphatidylinositol 4,5-bisphosphate (PIP2) producing IP3 and DAG (diacylglycerol). PIP2 modulates the function of many ion channels, while IP3 and DAG regulate intracellular Ca2+ levels and protein phosphorylation by protein kinase C, respectively. PLCβ enzymes are under the control of G protein coupled receptor signaling through direct interactions with G proteins Gβγ and Gαq and have been shown to be coincidence detectors for dual stimulation of Gαq and Gαi-coupled receptors. PLCβs are aqueous-soluble cytoplasmic enzymes but partition onto the membrane surface to access their lipid substrate, complicating their functional and structural characterization. Using newly developed methods, we recently showed that Gβγ activates PLCβ3 by recruiting it to the membrane. Using these same methods, here we show that Gαq increases the catalytic rate constant, kcat, of PLCβ3. Since stimulation of PLCβ3 by Gαq depends on an autoinhibitory element (the X-Y linker), we propose that Gαq produces partial relief of the X-Y linker autoinhibition through an allosteric mechanism. We also determined membrane-bound structures of the PLCβ3·Gαq and PLCβ3·Gβγ(2)·Gαq complexes, which show that these G proteins can bind simultaneously and independently of each other to regulate PLCβ3 activity. The structures rationalize a finding in the enzyme assay, that costimulation by both G proteins follows a product rule of each independent stimulus. We conclude that baseline activity of PLCβ3 is strongly suppressed, but the effect of G proteins, especially acting together, provides a robust stimulus upon G protein stimulation.
Collapse
Affiliation(s)
- Maria E. Falzone
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, New York, NY10065
- HHMI, The Rockefeller University, New York, NY10065
| | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, New York, NY10065
- HHMI, The Rockefeller University, New York, NY10065
| |
Collapse
|
7
|
Luo Y, Ni R, Jin X, Feng P, Dai C, Jiang L, Chen P, Yang L, Zhu Y. FOXD1 expression-based prognostic model for uveal melanoma. Heliyon 2023; 9:e21333. [PMID: 38027647 PMCID: PMC10651470 DOI: 10.1016/j.heliyon.2023.e21333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
FOXD1, a new member of the FOX transcription factor family, serves as a mediator and biomarker for cell reprogramming. But its contribution to prognosis of uveal melanoma (UVM) is unclear. This study demonstrated that FOXD1 might promote tumor growth and invasion, because FOXD1 expression was negatively correlated with overall survival, progression-free survival, and disease-specific survival in UVM patients. This conjecture was verified in cell culture with human uveal melanoma cell line (MUM2B) as model cells. Additionally, the biological mechanisms of FOXD1 based on FOXD1-related genomic spectrum, molecular pathways, tumor microenvironment, and drug treatment sensitivity were examined using The Cancer Genome Atlas (TCGA) database, aiming to reasonably explain why FOXD1 leads to poor prognosis of UVM. On these bases, a novel tumor prognostic model was established using the FOXD1-related immunomodulators TMEM173, TNFRSF4, TNFSF13, and ULBP1, which will enable the stratification of disease seriousness and clinical treatment for patients.
Collapse
Affiliation(s)
- Yang Luo
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Renhao Ni
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Xiaojun Jin
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Peipei Feng
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Centre Lihuili Hospital, Ningbo, 315000, China
| | - Chenyi Dai
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Lingjing Jiang
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | | | - Lu Yang
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo, 315211, China
| |
Collapse
|
8
|
García-Mulero S, Fornelino R, Punta M, Lise S, Varela M, del Carpio LP, Moreno R, Costa-García M, Rieder D, Trajanoski Z, Gros A, Alemany R, Piulats JM, Sanz-Pamplona R. Driver mutations in GNAQ and GNA11 genes as potential targets for precision immunotherapy in uveal melanoma patients. Oncoimmunology 2023; 12:2261278. [PMID: 38126027 PMCID: PMC10732647 DOI: 10.1080/2162402x.2023.2261278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/17/2023] [Indexed: 12/23/2023] Open
Abstract
Uveal melanoma (UM) is the most common ocular malignancy in adults. Nearly 95% of UM patients carry the mutually exclusive mutations in the homologous genes GNAQ (amino acid change Q209L/Q209P) and GNA11 (aminoacid change Q209L). UM is located in an immunosuppressed organ and does not suffer immunoediting. Therefore, we hypothesize that driver mutations in GNAQ/11 genes could be recognized by the immune system. Genomic and transcriptomic data from primary uveal tumors were collected from the TCGA-UM dataset (n = 80) and used to assess the immunogenic potential for GNAQ/GNA11 Q209L/Q209P mutations using a variety of tools and HLA type information. All prediction tools showed stronger GNAQ/11 Q209L binding to HLA than GNAQ/11 Q209P. The immunogenicity analysis revealed that Q209L is likely to be presented by more than 73% of individuals in 1000 G databases whereas Q209P is only predicted to be presented in 24% of individuals. GNAQ/11 Q209L showed a higher likelihood to be presented by HLA-I molecules than almost all driver mutations analyzed. Finally, samples carrying Q209L had a higher immune-reactive phenotype. Regarding cancer risk, seven HLA genotypes with low Q209L affinity show higher frequency in uveal melanoma patients than in the general population. However, no clear association was found between any HLA genotype and survival. Results suggest a high potential immunogenicity of the GNAQ/11 Q209L variant that could allow the generation of novel therapeutic tools to treat UM like neoantigen vaccinations.
Collapse
Affiliation(s)
- Sandra García-Mulero
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERESP, Barcelona, Spain
- Anatomy Unit, Department of Pathology and Experimental Therapy, and Department of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Roberto Fornelino
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERESP, Barcelona, Spain
| | - Marco Punta
- Bioinformatics Core, Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Stefano Lise
- Bioinformatics Core, Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Mar Varela
- Department of Pathology, Bellvitge University Hospital, Barcelona, Spain
| | - Luis P. del Carpio
- Procure Program, Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Rafael Moreno
- Procure Program, Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Marcel Costa-García
- Procure Program, Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Dietmar Rieder
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Zlatko Trajanoski
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Alena Gros
- Tumor Immunology and Immunotherapy, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ramón Alemany
- Procure Program, Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | | | - Rebeca Sanz-Pamplona
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERESP, Barcelona, Spain
- Institute for Health Research Aragon (IISA), ARAID Foundation, Aragon Government, University Hospital Lozano Blesa, Zaragoza, Spain
| |
Collapse
|
9
|
Falzone ME, MacKinnon R. The mechanism of Gα q regulation of PLCβ3 -catalyzed PIP2 hydrolysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.29.555394. [PMID: 37693483 PMCID: PMC10491199 DOI: 10.1101/2023.08.29.555394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
PLCβ enzymes cleave PIP2 producing IP3 and DAG. PIP2 modulates the function of many ion channels, while IP3 and DAG regulate intracellular Ca 2+ levels and protein phosphorylation by protein kinase C, respectively. PLCβ enzymes are under the control of GPCR signaling through direct interactions with G proteins Gβγ and Gα q and have been shown to be coincidence detectors for dual stimulation of Gα q and G α i coupled receptors. PLCβs are aqueous-soluble cytoplasmic enzymes, but partition onto the membrane surface to access their lipid substrate, complicating their functional and structural characterization. Using newly developed methods, we recently showed that Gβγ activates PLCβ3 by recruiting it to the membrane. Using these same methods, here we show that Gα q increases the catalytic rate constant, k cat , of PLCβ3 . Since stimulation of PLCβ3 by Gα q depends on an autoinhibitory element (the X-Y linker), we propose that Gα q produces partial relief of the X-Y linker autoinhibition through an allosteric mechanism. We also determined membrane-bound structures of the PLCβ3-Gα q , and PLCβ3-Gβγ(2)-Gα q complexes, which show that these G proteins can bind simultaneously and independently of each other to regulate PLCβ3 activity. The structures rationalize a finding in the enzyme assay, that co-stimulation by both G proteins follows a product rule of each independent stimulus. We conclude that baseline activity of PLCβ3 is strongly suppressed, but the effect of G proteins, especially acting together, provides a robust stimulus upon G protein stimulation. Significance Statement For certain cellular signaling processes, the background activity of signaling enzymes must be minimal and stimulus-dependent activation robust. Nowhere is this truer than in signaling by PLCβ3 , whose activity regulates intracellular Ca 2+ , phosphorylation by Protein Kinase C, and the activity of numerous ion channels and membrane receptors. In this study we show how PLCβ3 enzymes are regulated by two kinds of G proteins, Gβγ and Gα q . Enzyme activity studies and structures on membranes show how these G proteins act by separate, independent mechanisms, leading to a product rule of co-stimulation when they act together. The findings explain how cells achieve robust stimulation of PLCβ3 in the setting of very low background activity, properties essential to cell health and survival.
Collapse
|
10
|
Dewaele S, Delhaye L, De Paepe B, Bogaert B, Martinez R, Anckaert J, Yigit N, Nuytens J, Van Coster R, Eyckerman S, Raemdonck K, Mestdagh P. mTOR Inhibition Enhances Delivery and Activity of Antisense Oligonucleotides in Uveal Melanoma Cells. Nucleic Acid Ther 2023; 33:248-264. [PMID: 37389884 DOI: 10.1089/nat.2023.0008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023] Open
Abstract
Uveal melanoma (UM) is the most common primary intraocular malignancy in adults. Owing to a lack of effective treatments, patients with metastatic disease have a median survival time of 6-12 months. We recently demonstrated that the Survival Associated Mitochondrial Melanoma Specific Oncogenic Non-coding RNA (SAMMSON) is essential for UM cell survival and that antisense oligonucleotide (ASO)-mediated silencing of SAMMSON impaired cell viability and tumor growth in vitro and in vivo. By screening a library of 2911 clinical stage compounds, we identified the mammalian target of rapamycin (mTOR) inhibitor GDC-0349 to synergize with SAMMSON inhibition in UM. Mechanistic studies revealed that mTOR inhibition enhanced uptake and reduced lysosomal accumulation of lipid complexed SAMMSON ASOs, improving SAMMSON knockdown and further decreasing UM cell viability. We found mTOR inhibition to also enhance target knockdown in other cancer cell lines as well as normal cells when combined with lipid nanoparticle complexed or encapsulated ASOs or small interfering RNAs (siRNAs). Our results are relevant to nucleic acid treatment in general and highlight the potential of mTOR inhibition to enhance ASO and siRNA-mediated target knockdown.
Collapse
Affiliation(s)
- Shanna Dewaele
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Louis Delhaye
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Biotechnology, VIB-Ghent University, Ghent, Belgium
| | - Boel De Paepe
- Division of Pediatric Neurology and Metabolism, Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Bram Bogaert
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Laboratory for General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Ramiro Martinez
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jasper Anckaert
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Nurten Yigit
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Justine Nuytens
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Rudy Van Coster
- Division of Pediatric Neurology and Metabolism, Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Sven Eyckerman
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Biotechnology, VIB-Ghent University, Ghent, Belgium
| | - Koen Raemdonck
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Laboratory for General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Pieter Mestdagh
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
11
|
Croley CR, Pumarol J, Delgadillo BE, Cook AC, Day F, Kaceli T, Ward CC, Husain I, Husain A, Banerjee S, Bishayee A. Signaling pathways driving ocular malignancies and their targeting by bioactive phytochemicals. Pharmacol Ther 2023:108479. [PMID: 37330112 DOI: 10.1016/j.pharmthera.2023.108479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/05/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Ocular cancers represent a rare pathology. The American Cancer Society estimates that 3,360 cases of ocular cancer occur annually in the United States. The major types of cancers of the eye include ocular melanoma (also known as uveal melanoma), ocular lymphoma, retinoblastoma, and squamous cell carcinoma. While uveal melanoma is one of the primary intraocular cancers with the highest occurrence in adults, retinoblastoma remains the most common primary intraocular cancer in children, and squamous cell carcinoma presents as the most common conjunctival cancer. The pathophysiology of these diseases involves specific cell signaling pathways. Oncogene mutations, tumor suppressor mutations, chromosome deletions/translocations and altered proteins are all described as causal events in developing ocular cancer. Without proper identification and treatment of these cancers, vision loss, cancer spread, and even death can occur. The current treatments for these cancers involve enucleation, radiation, excision, laser treatment, cryotherapy, immunotherapy, and chemotherapy. These treatments present a significant burden to the patient that includes a possible loss of vision and a myriad of side effects. Therefore, alternatives to traditional therapy are urgently needed. Intercepting the signaling pathways for these cancers with the use of naturally occurring phytochemicals could be a way to relieve both cancer burden and perhaps even prevent cancer occurrence. This research aims to present a comprehensive review of the signaling pathways involved in various ocular cancers, discuss current therapeutic options, and examine the potential of bioactive phytocompounds in the prevention and targeted treatment of ocular neoplasms. The current limitations, challenges, pitfalls, and future research directions are also discussed.
Collapse
Affiliation(s)
- Courtney R Croley
- Healthcare Corporation of America, Department of Ophthalmology, Morsani College of Medicine, University of South Florida, Hudson, FL 34667, USA
| | - Joshua Pumarol
- Ross University School of Medicine, Miramar, FL 33027, USA
| | - Blake E Delgadillo
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Andrew C Cook
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Faith Day
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Tea Kaceli
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Caroline C Ward
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Imran Husain
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, USA
| | - Ali Husain
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, USA
| | - Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol 713 301, India
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| |
Collapse
|
12
|
Zhang C, Wu S. RUVBL1-modulated chromatin remodeling alters the transcriptional activity of oncogenic CTNNB1 in uveal melanoma. Cell Death Discov 2023; 9:132. [PMID: 37076452 PMCID: PMC10115834 DOI: 10.1038/s41420-023-01429-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 04/21/2023] Open
Abstract
Recent years have witnessed an increasing research interest in the therapeutic value of aberrant chromatin regulatory processes in carcinogenesis. Our study was performed to explore the possible carcinogenic mechanism of the chromatin regulator RuvB-like protein 1 (RUVBL1) in uveal melanoma (UVM). The expression pattern of RUVBL1 was retrieved in bioinformatics data. The correlation between RUVBL1 expression and the prognosis of patients with UVM was analyzed in publicly available database. The downstream target genes of RUVBL1 were predicted and further verified by co-immunoprecipitation. The bioinformatics analysis results showed that RUVBL1 may be associated with the transcriptional activity of CTNNB1 by regulating chromatin remodeling, and that RUVBL1 functioned as an independent prognostic factor for patients with UVM. The UVM cells manipulated with RUVBL1 knockdown were introduced for in vitro investigation. CCK-8 assay, flow cytometry, scratch assay, Transwell assay and Western blot analysis were used for detection on the resultant UVM cell proliferation, apoptosis, migration, invasion and cell cycle distribution. In vitro cell experimental data showed that RUVBL1 expression was significantly increased in UVM cells and RUVBL1 knockdown inhibited the proliferation, invasion and migration of UVM cells, accompanied by augmented apoptosis rate and blocked cell cycle progression. To sum up, RUVBL1 enhances the malignant biological characteristics of UVM cells by increasing the chromatin remodeling and subsequent transcription activity of CTNNB1.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Strabismus and Pediatric Ophthalmology, the Second Hospital of Jilin University, 130041, Changchun, P. R. China
| | - Shuai Wu
- Department of Orbital Disease and Ocular Plastic Surgery, the Second Hospital of Jilin University, 130041, Changchun, P. R. China.
| |
Collapse
|
13
|
Schrenk S, Bischoff LJ, Goines J, Cai Y, Vemaraju S, Odaka Y, Good SR, Palumbo JS, Szabo S, Reynaud D, Van Raamsdonk CD, Lang RA, Boscolo E. MEK inhibition reduced vascular tumor growth and coagulopathy in a mouse model with hyperactive GNAQ. Nat Commun 2023; 14:1929. [PMID: 37024491 PMCID: PMC10079932 DOI: 10.1038/s41467-023-37516-7] [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: 11/17/2021] [Accepted: 03/21/2023] [Indexed: 04/08/2023] Open
Abstract
Activating non-inherited mutations in the guanine nucleotide-binding protein G(q) subunit alpha (GNAQ) gene family have been identified in childhood vascular tumors. Patients experience extensive disfigurement, chronic pain and severe complications including a potentially lethal coagulopathy termed Kasabach-Merritt phenomenon. Animal models for this class of vascular tumors do not exist. This has severely hindered the discovery of the molecular consequences of GNAQ mutations in the vasculature and, in turn, the preclinical development of effective targeted therapies. Here we report a mouse model expressing hyperactive mutant GNAQ in endothelial cells. Mutant mice develop vascular and coagulopathy phenotypes similar to those seen in patients. Mechanistically, by transcriptomic analysis we demonstrate increased mitogen activated protein kinase signaling in the mutant endothelial cells. Targeting of this pathway with Trametinib suppresses the tumor growth by reducing vascular cell proliferation and permeability. Trametinib also prevents the development of coagulopathy and improves mouse survival.
Collapse
Affiliation(s)
- Sandra Schrenk
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lindsay J Bischoff
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jillian Goines
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yuqi Cai
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Shruti Vemaraju
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yoshinobu Odaka
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Biology, University of Cincinnati Blue Ash College, Blue Ash, OH, USA
| | - Samantha R Good
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Joseph S Palumbo
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sara Szabo
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Damien Reynaud
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Richard A Lang
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Ophthalmology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Elisa Boscolo
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
14
|
Zeng C, Long M, Lu Y. Monensin synergizes with chemotherapy in uveal melanoma through suppressing RhoA. Immunopharmacol Immunotoxicol 2023; 45:35-42. [PMID: 36043455 DOI: 10.1080/08923973.2022.2112219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Uveal melanoma (UM) is the common primary cancer of the eye and new treatments are needed. Substantial evidence has shown that an antibiotic monensin is an attractive candidate for the development of anti-cancer drug. In this study, we investigated the potential of repositioning monensin for the treatment of UM in the pre-clinical setting. MATERIALS AND METHODS Cellular activity assays were performed using multiple cell lines representing UM models with different cellular origins and genetic profiling and normal cells as control. Combination studies were performed using Chou-Talalay method. Mechanism studies were performed using immunoblotting and ELISA. RESULTS Monensin was effective against all tested UM cell lines and less effective against normal fibroblast cells. Monensin induced G0/G1 arrest and thus decreased S phase, leading to UM cell growth inhibition. It also inhibited migration and induced apoptosis in UM cells. In addition, the combination of monensin and dacarbazine was synergistic in targeting UM cells. Our mechanistic studies showed that monensin specifically decreased activity of RhoA without affecting other small GTPases, such as Ras and Rac1. Consistently, monensin decreased phosphorylation of downstream effectors of RhoA signaling, including ROCK, MYPT1 and MLC. Rescue studies using RhoA activator calpeptin showed that calpeptin significantly abolished the inhibitory effects of monensin on RhoA activity, proliferation, migration and survival, confirming that RhoA is the target of monensin in UM cells. CONCLUSIONS Our study demonstrates that monensin is a potent inhibitor of UM and synergizes with chemotherapy, via suppressing RhoA activity and RhoA-mediated signaling. Our findings suggest that monensin may be a potential lead compound for further development into a drug for UM treatment.
Collapse
Affiliation(s)
- Chaoxia Zeng
- Hainan Eye Hospital and Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Haikou, PR China
| | - Mingxia Long
- Department of Nursing, Wuhan Third Hospital-Tongren Hospital of Wuhan University, Wuhan, PR China
| | - Ying Lu
- Department of Integrated Traditional Chinese and Western Medicine, Wuhan Third Hospital -Tongren Hospital of Wuhan University, Wuhan, PR China
| |
Collapse
|
15
|
Cutaneous Melanocytic Tumor With CRTC1::TRIM11 Translocation: An Emerging Entity Analyzed in a Series of 41 Cases. Am J Surg Pathol 2022; 46:1457-1466. [PMID: 35993578 DOI: 10.1097/pas.0000000000001952] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cutaneous melanocytic tumor with CRTC1::TRIM11 fusion (CMTCT) is a recently described dermally based neoplasm with melanocytic differentiation. It can easily be confused with clear cell sarcoma and metastatic melanoma. Our understanding of this lesion, including its potential for aggressive disease, has been limited by the small number of previously reported cases (13) and the limited clinical follow-up data. Here, we report a series of 41 CMTCT confirmed by molecular studies. We find that the lesion shows highly uniform and reproducible morphologic, immunohistochemical, and genetic features across a wide variety of anatomic locations and age groups. Among 22 cases with follow-up, 1 local recurrence and 1 nodal metastasis were identified. Our data support the classification of CMTCT as a unique nosologic entity and emphasize the importance of distinguishing this entity from its histologic mimics, especially clear cell sarcoma and metastatic melanoma, to guide therapy and establish accurate prognostic expectations.
Collapse
|
16
|
Jo VY, Russell-Goldman E, Yoon CH, Doyle LA, Hanna J. Melanoma arising in extracutaneous cellular blue nevus: report of two cases with comparison to cutaneous counterparts and uveal melanoma. Histopathology 2022; 81:625-634. [PMID: 35941700 DOI: 10.1111/his.14735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/29/2022]
Abstract
AIMS Blue nevi are benign melanocytic lesions that typically occur in the dermis. Melanoma arising in blue nevus is rare and shows a molecular profile distinct from conventional forms of cutaneous melanoma and more similar to uveal melanoma and central nervous system (CNS) melanocytomas. In contrast to conventional cutaneous melanoma, these tumor types typically show activating driver mutations in GNAQ or GNA11, a low mutational burden without evidence of a UV signature, and a reproducible pattern of chromosomal copy number changes. Blue nevi can also occur at extracutaneous sites. Here we report two cases of melanoma arising in extracutaneous blue nevus and compare their molecular features to cohorts of melanoma arising in cutaneous blue nevus (5 patients) and uveal melanoma (6 patients). METHODS AND RESULTS We describe the clinical, histomorphologic, immunohistochemical, and molecular findings in these two cases of melanoma arising in extracutaneous blue nevus. We compare their molecular profiles to melanomas arising in cutaneous blue nevus and uveal melanoma using a targeted next-generation DNA sequencing platform and find striking similarities between all three groups. CONCLUSIONS The close relationship between blue nevus-associated melanomas, regardless of their anatomic site, supports and validates the concept of melanoma arising in extracutaneous blue nevus and suggests that the two groups share common pathogenic mechanisms. The similarity of both groups to uveal melanoma in turn supports the close relationship between blue nevus-associated melanoma, uveal melanoma, and CNS melanocytoma, and their distinction from conventional UV-associated melanoma. These findings have important implications for prognosis and therapy.
Collapse
Affiliation(s)
- Vickie Y Jo
- Departments of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eleanor Russell-Goldman
- Departments of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Charles H Yoon
- Departments of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Leona A Doyle
- Departments of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - John Hanna
- Departments of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Cordycepin (3′-Deoxyadenosine) Suppresses Heat Shock Protein 90 Function and Targets Tumor Growth in an Adenosine Deaminase-Dependent Manner. Cancers (Basel) 2022; 14:cancers14133122. [PMID: 35804893 PMCID: PMC9264932 DOI: 10.3390/cancers14133122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
Alterations in metabolism and energy production are increasingly being recognized as important drivers of neoplasia, raising the possibility that metabolic analogs could disrupt oncogenic pathways. 3′-deoxyadenosine, also known as cordycepin, is an adenosine analog that inhibits the growth of several types of cancer. However, the effects of cordycepin have only been examined in a limited number of tumor types, and its mechanism of action is poorly understood. We found that cordycepin slows the growth and promotes apoptosis in uveal melanoma, as well as a range of other hard-to-treat malignancies, including retinoblastoma, atypical teratoid rhabdoid tumors, and diffuse midline gliomas. Interestingly, these effects were dependent on low adenosine deaminase (ADA) expression or activity. Inhibition of ADA using either siRNA or pharmacologic approaches sensitized tumors with higher ADA to cordycepin in vitro and in vivo, with increased apoptosis, reduced clonogenic capacity, and slower migration of neoplastic cells. Our studies suggest that ADA is both a biomarker predicting response to cordycepin and a target for combination therapy. We also describe a novel mechanism of action for cordycepin: competition with adenosine triphosphate (ATP) in binding to Hsp90, resulting in impaired processing of oncogenic Hsp90 client proteins.
Collapse
|
18
|
Farhoumand LS, Fiorentzis M, Kraemer MM, Sak A, Stuschke M, Rassaf T, Hendgen-Cotta U, Bechrakis NE, Berchner-Pfannschmidt U. The Adrenergic Receptor Antagonist Carvedilol Elicits Anti-Tumor Responses in Uveal Melanoma 3D Tumor Spheroids and May Serve as Co-Adjuvant Therapy with Radiation. Cancers (Basel) 2022; 14:cancers14133097. [PMID: 35804869 PMCID: PMC9264933 DOI: 10.3390/cancers14133097] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/26/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Uveal melanoma (UM) is the most common intraocular tumor in adults. Despite local tumor control, no effective therapy has been found to prevent metastasis, resulting in a high mortality rate. In the present study, we evaluated the anti-tumor potential of non-selective ß-blockers in 3D tumor spheroids grown from UM cell lines. Of the various ß-blockers tested, carvedilol and its enantiomers were most potent in decreasing the viability of Mel270 spheroids. Carvedilol at a concentration of 10–50 µM significantly elicited cytotoxicity and induced apoptosis in spheroid cells. In result, carvedilol inhibited tumor spheroid growth and compactness, and furthermore prevented the long-term survival and repopulation of spreading spheroid cells. The drug sensitivity of the different spheroids grown from Mel270, 92-1, UPMD2, or UPMM3 cell lines was dependent on 3D morphology rather than on high-risk cytogenetic profile or adrenergic receptor expression levels. In fact, the monosomy-3-containing UPMM3 cell line was most responsive to carvedilol treatment compared to the other cell lines. The concurrent treatment of UPMM3 spheroids with carvedilol and 5 or 10 Gy irradiation revealed additive cytotoxic effects that provided tumor control. Collectively, our data demonstrate the anti-tumor properties of carvedilol and its enantiomers, which may serve as candidates for the co-adjuvant therapy of UM.
Collapse
Affiliation(s)
- Lina S. Farhoumand
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (L.S.F.); (M.F.); (M.M.K.); (N.E.B.)
| | - Miltiadis Fiorentzis
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (L.S.F.); (M.F.); (M.M.K.); (N.E.B.)
| | - Miriam M. Kraemer
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (L.S.F.); (M.F.); (M.M.K.); (N.E.B.)
| | - Ali Sak
- Department of Radiotherapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (A.S.); (M.S.)
| | - Martin Stuschke
- Department of Radiotherapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (A.S.); (M.S.)
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (T.R.); (U.H.-C.)
| | - Ulrike Hendgen-Cotta
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (T.R.); (U.H.-C.)
| | - Nikolaos E. Bechrakis
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (L.S.F.); (M.F.); (M.M.K.); (N.E.B.)
| | - Utta Berchner-Pfannschmidt
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (L.S.F.); (M.F.); (M.M.K.); (N.E.B.)
- Correspondence: ; Tel.: +49-201-723-6028
| |
Collapse
|
19
|
Zhang H, Jin C, Zhang L, Peng B, Zhang Y, Liu Y, Li L, Ye M, Xiong W, Tan W. CD71-Specific Aptamer Conjugated with Monomethyl Auristatin E for the Treatment of Uveal Melanoma. ACS APPLIED MATERIALS & INTERFACES 2022; 14:32-40. [PMID: 34928139 DOI: 10.1021/acsami.1c13980] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Uveal melanoma (UM) is the most common primary intraocular malignancy among adults. Despite significant advances in diagnosis and treatment, the general mortality of UM remains alarmingly high. This calls for the development of new approaches for the treatment of UM, such as targeted cancer therapy. CD71, also known as transferrin receptor 1, is overexpressed in UM cell lines and tissues. Herein, we report the development of a CD71-specific aptamer targeting the XQ-2d-MMAE conjugate that can distinguish UM cells from normal human uveal melanocytes. The cytotoxic drug monomethyl auristatin E (MMAE) could be easily coupled onto XQ-2d, a DNA aptamer that specifically targets CD71, to achieve efficiently targeted cancer growth inhibition in a mouse xenograft model, thus implying that XQ-2d-MMAE might be developed into a promising novel anti-tumor agent for the treatment of UM. Collectively, our results demonstrated that CD71 is a reliable target for drug delivery in UM and could be utilized as a model to explore aptamer-mediated targeted UM treatment strategies.
Collapse
Affiliation(s)
- Hui Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Cheng Jin
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Lin Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Bo Peng
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Yibin Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Yan Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Ling Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Wei Xiong
- Department of Ophthalmology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
20
|
Dewaele S, Delhaye L, De Paepe B, de Bony EJ, De Wilde J, Vanderheyden K, Anckaert J, Yigit N, Nuytens J, Vanden Eynde E, Smet J, Verschoore M, Nemati F, Decaudin D, Rodrigues M, Zhao P, Jochemsen A, Leucci E, Vandesompele J, Van Dorpe J, Marine JC, Van Coster R, Eyckerman S, Mestdagh P. The long non-coding RNA SAMMSON is essential for uveal melanoma cell survival. Oncogene 2022; 41:15-25. [PMID: 34508176 PMCID: PMC8724009 DOI: 10.1038/s41388-021-02006-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 08/02/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022]
Abstract
Long non-coding RNAs (lncRNAs) can exhibit cell-type and cancer-type specific expression profiles, making them highly attractive as therapeutic targets. Pan-cancer RNA sequencing data revealed broad expression of the SAMMSON lncRNA in uveal melanoma (UM), the most common primary intraocular malignancy in adults. Currently, there are no effective treatments for UM patients with metastatic disease, resulting in a median survival time of 6-12 months. We aimed to investigate the therapeutic potential of SAMMSON inhibition in UM. Antisense oligonucleotide (ASO)-mediated SAMMSON inhibition impaired the growth and viability of a genetically diverse panel of uveal melanoma cell lines. These effects were accompanied by an induction of apoptosis and were recapitulated in two uveal melanoma patient derived xenograft (PDX) models through subcutaneous ASO delivery. SAMMSON pulldown revealed several candidate interaction partners, including various proteins involved in mitochondrial translation. Consequently, inhibition of SAMMSON impaired global, mitochondrial and cytosolic protein translation levels and mitochondrial function in uveal melanoma cells. The present study demonstrates that SAMMSON expression is essential for uveal melanoma cell survival. ASO-mediated silencing of SAMMSON may provide an effective treatment strategy to treat primary and metastatic uveal melanoma patients.
Collapse
Affiliation(s)
- Shanna Dewaele
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Louis Delhaye
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Biotechnology, VIB-Ghent University, Ghent, Belgium
| | - Boel De Paepe
- Department of Pediatrics, Division of Pediatric Neurology and Metabolism, Ghent University Hospital, Ghent, Belgium
| | - Eric James de Bony
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jilke De Wilde
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Department of pathology, Ghent University Hospital, Ghent, Belgium
| | - Katrien Vanderheyden
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jasper Anckaert
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Nurten Yigit
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Justine Nuytens
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Eveline Vanden Eynde
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Joél Smet
- Department of Pediatrics, Division of Pediatric Neurology and Metabolism, Ghent University Hospital, Ghent, Belgium
| | - Maxime Verschoore
- Department of Pediatrics, Division of Pediatric Neurology and Metabolism, Ghent University Hospital, Ghent, Belgium
| | - Fariba Nemati
- Institut Curie, Laboratory of Preclinical Investigation, Translational Research Department, PSL Research University, Paris, France
| | - Didier Decaudin
- Institut Curie, Laboratory of Preclinical Investigation, Translational Research Department, PSL Research University, Paris, France
- Institut Curie, Department of Medical Oncology, PSL Research University, Paris, France
| | - Manuel Rodrigues
- Institut Curie, Department of Medical Oncology, PSL Research University, Paris, France
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, Paris, 75005, France
| | - Peihua Zhao
- Center for Medical Biotechnology, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Aart Jochemsen
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Eleonora Leucci
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
- TRACE, LKI Leuven Cancer Institute, Leuven, Belgium
| | - Jo Vandesompele
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jo Van Dorpe
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of pathology, Ghent University Hospital, Ghent, Belgium
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Department of Oncology, KULeuven, Leuven, Belgium
| | - Rudy Van Coster
- Department of Pediatrics, Division of Pediatric Neurology and Metabolism, Ghent University Hospital, Ghent, Belgium
| | - Sven Eyckerman
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Biotechnology, VIB-Ghent University, Ghent, Belgium
| | - Pieter Mestdagh
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.
| |
Collapse
|
21
|
Lapadula D, Benovic JL. Targeting Oncogenic Gα q/11 in Uveal Melanoma. Cancers (Basel) 2021; 13:6195. [PMID: 34944815 PMCID: PMC8699590 DOI: 10.3390/cancers13246195] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 11/17/2022] Open
Abstract
Uveal melanoma is the most common intraocular cancer in adults and arises from the transformation of melanocytes in the uveal tract. While treatment of the primary tumor is often effective, 36-50% of patients develop metastatic disease primarily to the liver. While various strategies have been used to treat the metastatic disease, there remain no effective treatments that improve survival. Significant insight has been gained into the pathways that are altered in uveal melanoma, with mutually exclusive activating mutations in the GNAQ and GNA11 genes being found in over 90% of patients. These genes encode the alpha subunits of the hetetrotrimeric G proteins, Gq and G11, and mutations result in activation of several important signaling pathways, including phospholipase C and activation of the transcription factor YAP. In this review, we discuss current efforts to target various signaling pathways in the treatment of uveal melanoma including recent efforts to target Gq and G11 in mouse models. While selective targeting of Gq and G11 provides a potential therapeutic strategy to treat uveal melanoma, it is evident that improved inhibitors and methods of delivery are needed.
Collapse
Affiliation(s)
| | - Jeffrey L. Benovic
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| |
Collapse
|
22
|
Prognostic Values of G-Protein Mutations in Metastatic Uveal Melanoma. Cancers (Basel) 2021; 13:cancers13225749. [PMID: 34830903 PMCID: PMC8616238 DOI: 10.3390/cancers13225749] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/11/2021] [Accepted: 11/13/2021] [Indexed: 02/03/2023] Open
Abstract
Simple Summary Uveal melanoma (UM) is the most common primary intraocular malignancy in adults. More than 90% of UMs harbor mutually exclusive activating mutations in G-proteins. The mutations are early events in UM development and considered to be driver mutations in carcinogenesis. Even after treatment of primary uveal melanoma, up to 50% of patients subsequently develop recurrence, predominantly in the liver. GNAQ mutations are not reported to be correlated to survival, while the mutations in GNA11 are reported more frequently in metastatic UM. We investigated the correlation of survival after development of metastasis (Met-to-Death) of metastatic uveal melanoma (MUM) patients with GNA11 and GNAQ mutations. We identified that MUM with mutation patterns of Q209P vs. Q209L in GNA11 and GNAQ might predict survival of MUM patients. Abstract Uveal melanoma is the most common primary ocular malignancy in adults, characterized by gene mutations in G protein subunit alpha q (GNAQ) and G protein subunit alpha 11 (GNA11). Although they are considered to be driver mutations, their role in MUM remains elusive. We investigated key somatic mutations of MUM and their impact on patients’ survival after development of systemic metastasis (Met-to-Death). Metastatic lesions from 87 MUM patients were analyzed by next generation sequencing (NGS). GNA11 (41/87) and GNAQ (39/87) mutations were most predominantly seen in MUM. Most GNA11 mutations were Q209L (36/41), whereas GNAQ mutations comprised Q209L (14/39) and Q209P (21/39). Epigenetic pathway mutations BAP1 (42/66), SF3B1 (11/66), FBXW7 (2/87), PBRM1 (1/66), and SETD2 (1/66) were found. No specimen had the EIF1AX mutation. Interestingly, Met-to-Death was longer in patients with GNAQ Q209P compared to GNAQ/GNA11 Q209L mutations, suggesting the difference in mutation type in GNAQ/GNA11 might determine the prognosis of MUM. Structural alterations of the GNAQ/GNA11 protein and their impact on survival of MUM patients should be further investigated.
Collapse
|
23
|
Potential of miRNA-Based Nanotherapeutics for Uveal Melanoma. Cancers (Basel) 2021; 13:cancers13205192. [PMID: 34680340 PMCID: PMC8534265 DOI: 10.3390/cancers13205192] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Human uveal melanoma (UM) is the most common primary intraocular tumor with high metastatic risk in adults. Currently, no effective treatment is available for metastatic UM; therefore, new therapeutic approaches are needed to improve overall survival. Given the increased understanding of microRNAs (miRNAs) and their roles in UM tumorigenesis and metastasis, miRNA-based therapy may offer the hope of improving therapeutic outcomes. This review summarizes the actions of select miRNAs examined in preclinical studies using miRNAs as therapeutic targets in UM. The focus of this review is the application of established nanotechnology-assisted delivery systems to overcome the limitations of therapeutic miRNAs. A blend of therapeutic miRNAs and nanodelivery systems may facilitate the translation of miRNA therapies to clinical settings. Abstract Uveal melanoma (UM) is the most common adult intraocular cancer, and metastatic UM remains deadly and incurable. UM is a complex disease associated with the deregulation of numerous genes and redundant intracellular signaling pathways. As understanding of epigenetic dysregulation in the oncogenesis of UM has increased, the abnormal expression of microRNAs (miRNAs) has been found to be an epigenetic mechanism underlying UM tumorigenesis. A growing number of miRNAs are being found to be associated with aberrant signaling pathways in UM, and some have been investigated and functionally characterized in preclinical settings. This review summarizes the miRNAs with promising therapeutic potential for UM treatment, paying special attention to the therapeutic miRNAs (miRNA mimics or inhibitors) used to restore dysregulated miRNAs to their normal levels. However, several physical and physiological limitations associated with therapeutic miRNAs have prevented their translation to cancer therapeutics. With the advent of nanotechnology delivery systems, the development of effective targeted therapies for patients with UM has received great attention. Therefore, this review provides an overview of the use of nanotechnology drug delivery systems, particularly nanocarriers that can be loaded with therapeutic miRNAs for effective delivery into target cells. The development of miRNA-based therapeutics with nanotechnology-based delivery systems may overcome the barriers of therapeutic miRNAs, thereby enabling their translation to therapeutics, enabling more effective targeting of UM cells and consequently improving therapeutic outcomes.
Collapse
|
24
|
Patel RP, Thomas JR, Curt KM, Fitzsimmons CM, Batista PJ, Bates SE, Gottesman MM, Robey RW. Dual Inhibition of Histone Deacetylases and the Mechanistic Target of Rapamycin Promotes Apoptosis in Cell Line Models of Uveal Melanoma. Invest Ophthalmol Vis Sci 2021; 62:16. [PMID: 34533562 PMCID: PMC8458781 DOI: 10.1167/iovs.62.12.16] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Purpose Over 90% of uveal melanomas harbor pathogenic variants of the GNAQ or GNA11 genes that activate survival pathways. As previous studies found that Ras-mutated cell lines were vulnerable to a combination of survival pathway inhibitors and the histone-deacetylase inhibitor romidepsin, we investigated whether this combination would be effective in models of uveal melanoma. Methods A small-scale screen of inhibitors of bromodomain-containing protein 4 (BRD4; OTX-015), extracellular signal-related kinase (ERK; ulixertinib), mechanistic target of rapamycin (mTOR; AZD-8055), or phosphoinositide 3-kinase (PI3K; GDC-0941) combined with a clinically relevant administration of romidepsin was performed on a panel of uveal melanoma cell lines (92.1, Mel202, MP38, and MP41) and apoptosis was quantified by flow cytometry after 48 hours. RNA sequencing analysis was performed on Mel202 cells treated with romidepsin alone, AZD-8055 alone, or the combination, and protein changes were validated by immunoblot. Results AZD-8055 with romidepsin was the most effective combination in inducing apoptosis in the cell lines. Increased caspase-3 and PARP cleavage were noted in the cell lines when they were treated with romidepsin and mTOR inhibitors. RNA sequencing analysis of Mel202 cells revealed that apoptosis was the most affected pathway in the romidepsin/AZD-8055-treated cells. Increases in pro-apoptotic BCL2L11 and decreases in anti-apoptotic BIRC5 and BCL2L1 transcripts noted in the sequencing analysis were confirmed at the protein level in Mel202 cells. Conclusions Our data suggest that romidepsin in combination with mTOR inhibition could be an effective treatment strategy against uveal melanoma due in part to changes in apoptotic proteins.
Collapse
Affiliation(s)
- Ruchi P Patel
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Joanna R Thomas
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Katherine M Curt
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Christina M Fitzsimmons
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Pedro J Batista
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Susan E Bates
- Columbia University Medical Center, Division of Hematology/Oncology, New York, New York, United States
| | - Michael M Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Robert W Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
25
|
de Lange MJ, Nell RJ, van der Velden PA. Scientific and clinical implications of genetic and cellular heterogeneity in uveal melanoma. MOLECULAR BIOMEDICINE 2021; 2:25. [PMID: 35006486 PMCID: PMC8607395 DOI: 10.1186/s43556-021-00048-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 07/16/2021] [Indexed: 10/27/2022] Open
Abstract
Here, we discuss the presence and roles of heterogeneity in the development of uveal melanoma. Both genetic and cellular heterogeneity are considered, as their presence became undeniable due to single cell approaches that have recently been used in uveal melanoma analysis. However, the presence of precursor clones and immune infiltrate in uveal melanoma have been described as being part of the tumour already decades ago. Since uveal melanoma grow in the corpus vitreous, they present a unique tumour model because every cell present in the tumour tissue is actually part of the tumour and possibly plays a role. For an effective treatment of uveal melanoma metastasis, it should be clear whether precursor clones and normal cells play an active role in progression and metastasis. We propagate analysis of bulk tissue that allows analysis of tumour heterogeneity in a clinical setting.
Collapse
Affiliation(s)
- Mark J de Lange
- Department of Ophthalmology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Rogier J Nell
- Department of Ophthalmology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Pieter A van der Velden
- Department of Ophthalmology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
26
|
Shi X, Xia S, Chu Y, Yang N, Zheng J, Chen Q, Fen Z, Jiang Y, Fang S, Lin J. CARD11 is a prognostic biomarker and correlated with immune infiltrates in uveal melanoma. PLoS One 2021; 16:e0255293. [PMID: 34370778 PMCID: PMC8351993 DOI: 10.1371/journal.pone.0255293] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 07/13/2021] [Indexed: 12/15/2022] Open
Abstract
Uveal melanoma (UVM), the most common primary intraocular malignancy, has a high mortality because of a high propensity to metastasize. Our study analyzed prognostic value and immune-related characteristics of CARD11 in UVM, hoping to provide a potential management and research direction. The RNA-sequence data of 80 UVM patients were downloaded from The Cancer Genome Atlas database and divided them into high- and low-expression groups. We analyzed the differentially expressed genes, enrichment analyses and the infiltration of immune cells using the R package and Gene-Set Enrichment Analysis. A clinical prediction nomogram and protein-protein interaction network were constructed and the first 8 genes were considered as the hub-genes. Finally, we constructed a competing endogenous RNA (ceRNA) network by Cytoscape and analyzed the statistical data via the R software. Here we found that CARD11 expression had notable correlation with UVM clinicopathological features, which was also an independent predictor for overall survival (OS). Intriguingly, CARD11 had a positively correlation to autophagy, cellular senescence and apoptosis. Infiltration of monocytes was significantly higher in low CARD11 expression group, and infiltration of T cells regulatory was lower in the same group. Functional enrichment analyses revealed that CARD11 was positively related to T cell activation pathways and cell adhesion molecules. The expressions of hub-genes were all increased in the high CARD11 expression group and the ceRNA network showed the interaction among mRNA, miRNA and lncRNA. These findings show that high CARD11 expression in UVM is associated with poor OS, indicating that CARD11 may serve as a potential biomarker for the diagnosis and prognosis of the UVM.
Collapse
Affiliation(s)
- Xueying Shi
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Shilin Xia
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yingming Chu
- Department of Integrated Traditional Chinese Medicine, Peking University First Hospital, Beijing, China
| | - Nan Yang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jingyuan Zheng
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Qianyi Chen
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Zeng Fen
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Yuankuan Jiang
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shifeng Fang
- Department of Ophthalmology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jingrong Lin
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
27
|
Akin-Bali DF. Bioinformatics analysis of GNAQ, GNA11, BAP1, SF3B1,SRSF2, EIF1AX, PLCB4, and CYSLTR2 genes and their role in the pathogenesis of Uveal Melanoma. Ophthalmic Genet 2021; 42:732-743. [PMID: 34353217 DOI: 10.1080/13816810.2021.1961280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Uveal melanoma (UM) is the most common primary intraocular malignancy in adults, and its metastases are known to be fatal. It is critical to identify molecular markers to be used in potential prognostic evaluation for early diagnosis, treatment, and metastasis or to investigate all aspects of known genetic anomalies. Therefore, this study aimed to analyze the eight genes (GNAQ, GNA11, BAP1, SF3B1, SRSF2, EIF1AX, PLCB4, and CYSLTR2) that are associated with the most common genetic anomalies in UM from a molecular perspective. The genome sequences and expression profiles of 108 UM patients were obtained via bioinformatics tools that provide data from TCGA. The overall mutational load and the mutation patterns for eight genes, in particular, were thoroughly determined. Moreover, PolyPhen2 and SNAP2 tools were used to estimate the oncogenic/pathogenic properties of identified mutations for UM. In addition to the mutation profile, the effects of the presence of a mutation on gene expression and survival were determined. Finally, STRING network analysis was performed to better understand the functional relationships of mutated proteins in cellular processes. There were 27 missense mutations, 16 frameshift mutations, six nonsense mutations, and three splice region mutations among the 52 mutations found in eight genes, and 26 of them had pathogenic properties. BAP1 m-RNA expression was significantly lower in tumors with the mutant genotype (p = .001). The impact of gene expression, which has poor prognostic importance, on survival is statistically significant for high-expressed BAP1 (p = .0015) and low-expressed CYSLTR2 (p = .0021). To assess the current state of this potentially devastating disease, a molecular perspective has been evaluated. Defining this molecular perspective can be useful in developing targeted drug therapies and personalized medicine.
Collapse
|
28
|
Yong L, Croft DP, Troscianko J, Ramnarine IW, Wilson AJ. Sensory-based quantification of male colour patterns in Trinidadian guppies reveals no support for parallel phenotypic evolution in multivariate trait space. Mol Ecol 2021; 31:1337-1357. [PMID: 34170592 DOI: 10.1111/mec.16039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/29/2021] [Accepted: 06/16/2021] [Indexed: 12/01/2022]
Abstract
Parallel evolution, in which independent populations evolve along similar phenotypic trajectories, offers insights into the repeatability of adaptive evolution. Here, we revisit a classic example of parallelism, that of repeated evolution of brighter males in the Trinidadian guppy (Poecilia reticulata). In guppies, colonisation of low predation habitats is associated with emergence of 'more colourful' phenotypes since predator-induced viability selection for crypsis weakens while sexual selection by female preference for conspicuousness remains strong. Our study differs from previous investigations in three respects. First, we adopted a multivariate phenotyping approach to characterise parallelism in multitrait space. Second, we used ecologically-relevant colour traits defined by the visual systems of the two selective agents (i.e., guppy, predatory cichlid). Third, we estimated population genetic structure to test for adaptive (parallel) evolution against a model of neutral phenotypic divergence. We find strong phenotypic differentiation that is inconsistent with a neutral model but very limited support for the predicted pattern of greater conspicuousness at low predation. Effects of predation regime on each trait were in the expected direction, but weak, largely nonsignificant, and explained little among-population variation. In multitrait space, phenotypic trajectories of lineages colonising low from high predation regimes were not parallel. Our results are consistent with reduced predation risk facilitating adaptive differentiation, potentially by female choice, but suggest that this proceeds in independent directions of multitrait space across lineages. Pool-sequencing data also revealed SNPs showing greater differentiation than expected under neutrality, among which some are found in genes contributing to colour pattern variation, presenting opportunities for future genetic study.
Collapse
Affiliation(s)
- Lengxob Yong
- Centre for Ecology and Conservation, College of Life and Environmental Sciences, University of Exeter, Penryn, UK
| | - Darren P Croft
- Centre for Research in Animal Behaviour, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Jolyon Troscianko
- Centre for Ecology and Conservation, College of Life and Environmental Sciences, University of Exeter, Penryn, UK
| | - Indar W Ramnarine
- Department of Life Sciences, The University of The West Indies, St Augustine, Trinidad and Tobago
| | - Alastair J Wilson
- Centre for Ecology and Conservation, College of Life and Environmental Sciences, University of Exeter, Penryn, UK
| |
Collapse
|
29
|
Vergara IA, Wilmott JS, Long GV, Scolyer RA. Genetic drivers of non-cutaneous melanomas: Challenges and opportunities in a heterogeneous landscape. Exp Dermatol 2021; 31:13-30. [PMID: 33455025 DOI: 10.1111/exd.14287] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/16/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022]
Abstract
Non-cutaneous melanomas most frequently involve the uveal tract and mucosal membranes, including the conjunctiva. In contrast to cutaneous melanoma, they often present at an advanced clinical stage, are associated with worse clinical outcomes and show poorer responses to immunotherapy. The mutational load within most non-cutaneous melanomas reflects their lower ultraviolet light (UV) exposure. The genetic drivers within non-cutaneous melanomas are heterogeneous. Within ocular melanomas, posterior uveal tract melanomas typically harbour one of two distinct, sets of driver mutations and alterations of clinical and biological significance. In contrast to posterior uveal tract melanomas, anterior uveal tract melanomas of the iris and conjunctival melanomas frequently carry both a higher mutational burden and specific mutations linked with UV exposure. The genetic drivers in iris melanomas more closely resemble those of the posterior uveal tract, whereas conjunctival melanomas harbour similar genetic driver mutations to cutaneous melanomas. Mucosal melanomas occur in sun-shielded sites including sinonasal and oral cavities, nasopharynx, oesophagus, genitalia, anus and rectum, and their mutational landscape is frequently associated with a dominant process of spontaneous deamination and infrequent presence of UV mutation signatures. Genetic drivers of mucosal melanomas are diverse and vary with anatomic location. Further understanding of the causes of already identified recurrent molecular events in non-cutaneous melanomas, identification of additional drivers in specific subtypes, integrative multi-omics analyses and analysis of the tumor immune microenvironment will expand knowledge in this field. Furthermore, such data will likely uncover new therapeutic strategies which will lead to improved clinical outcomes in non-cutaneous melanoma patients.
Collapse
Affiliation(s)
- Ismael A Vergara
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and New South Wales Health Pathology, Sydney, NSW, Australia
| |
Collapse
|
30
|
Ortega MA, Fraile-Martínez O, García-Honduvilla N, Coca S, Álvarez-Mon M, Buján J, Teus MA. Update on uveal melanoma: Translational research from biology to clinical practice (Review). Int J Oncol 2020; 57:1262-1279. [PMID: 33173970 PMCID: PMC7646582 DOI: 10.3892/ijo.2020.5140] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
Uveal melanoma is the most common type of intraocular cancer with a low mean annual incidence of 5‑10 cases per million. Tumours are located in the choroid (90%), ciliary body (6%) or iris (4%) and of 85% are primary tumours. As in cutaneous melanoma, tumours arise in melanocytes; however, the characteristics of uveal melanoma differ, accounting for 3‑5% of melanocytic cancers. Among the numerous risk factors are age, sex, genetic and phenotypic predisposition, the work environment and dermatological conditions. Management is usually multidisciplinary, including several specialists such as ophthalmologists, oncologists and maxillofacial surgeons, who participate in the diagnosis, treatment and complex follow‑up of these patients, without excluding the management of the immense emotional burden. Clinically, uveal melanoma generates symptoms that depend as much on the affected ocular globe site as on the tumour size. The anatomopathological study of uveal melanoma has recently benefited from developments in molecular biology. In effect, disease classification or staging according to molecular profile is proving useful for the assessment of this type of tumour. Further, the improved knowledge of tumour biology is giving rise to a more targeted approach to diagnosis, prognosis and treatment development; for example, epigenetics driven by microRNAs as a target for disease control. In the present study, the main epidemiological, clinical, physiopathological and molecular features of this disease are reviewed, and the associations among all these factors are discussed.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid
- University Center for The Defense of Madrid (CUD-ACD), 28047 Madrid
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid
- University Center for The Defense of Madrid (CUD-ACD), 28047 Madrid
| | - Santiago Coca
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid
- University Center for The Defense of Madrid (CUD-ACD), 28047 Madrid
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid
- University Center for The Defense of Madrid (CUD-ACD), 28047 Madrid
- Internal and Oncology Service (CIBER-EHD), University Hospital Príncipe de Asturias, Alcalá de Henares, 28805 Madrid
| | - Julia Buján
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid
- University Center for The Defense of Madrid (CUD-ACD), 28047 Madrid
| | - Miguel A. Teus
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
- Ophthalmology Service, University Hospital Príncipe de Asturias, Alcalá de Henares, 28805 Madrid, Spain
| |
Collapse
|
31
|
Porcelli L, Mazzotta A, Garofoli M, Di Fonte R, Guida G, Guida M, Tommasi S, Azzariti A. Active notch protects MAPK activated melanoma cell lines from MEK inhibitor cobimetinib. Biomed Pharmacother 2020; 133:111006. [PMID: 33202284 DOI: 10.1016/j.biopha.2020.111006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/26/2020] [Accepted: 11/08/2020] [Indexed: 12/18/2022] Open
Abstract
The crosstalk between Notch and MAPK pathway plays a role in MEK inhibitor resistance in BRAFV600E metastatic melanoma (MM) and promotes migration in GNAQQ209L uveal melanoma (UM) cells. We determined the cytotoxicity of combinatorial inhibition of MEK and Notch by cobimetinib and γ-secretase inhibitor (GSI) nirogacestat, in BRAFV600E and BRAF wt MM and GNAQQ209L UM cells displaying different Erk1/2 and Notch activation status, with the aim to elucidate the impact of Notch signaling in the response to MEK inhibitor. Overall the combination was synergic in BRAFV600E MM and GNAQQ209L UM cells and antagonistic in BRAF wt one. Focusing on UM cells, we found that cobimetinib resulted in G0/G1 phase arrest and apoptosis induction, whereas the combination with GSI increased treatment efficacy by inducing a senescent-like state of cells and by blocking migration towards liver cancer cells. Mechanistically, this was reflected in a strong reduction of cyclin D1, in the inactivation of retinoblastoma protein and in the increase of p27KIP1 expression levels. Of note, each drug alone prevented Notch signaling activation resulting in inhibition of c-jun(Ser63) and Hes-1 expression. The combination achieved the strongest inhibition on Notch signaling and on both c-jun(Ser63) and Erk1/2 activation level. In conclusion we unveiled a coordinate action of MAPK and Notch signaling in promoting proliferation of BRAFV600E MM and GNAQQ209L UM cells. Remarkably, the simultaneous inhibition of MEK and Notch signaling highlighted a role for the second pathway in protecting cells against senescence in GNAQQ209L UM cells treated with the MEK inhibitor.
Collapse
Affiliation(s)
- Letizia Porcelli
- Experimental Pharmacology Laboratory, Italia, 70124, Bari, Italy
| | | | | | - Roberta Di Fonte
- Experimental Pharmacology Laboratory, Italia, 70124, Bari, Italy
| | - Gabriella Guida
- Department of Basic Medical Sciences Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare 11, 70124 Bari, Italy
| | | | - Stefania Tommasi
- Molecular Diagnostics and Pharmacogenetics Unit IRCCS Istituto Tumori "Giovanni Paolo II" di Bari, Italia, 70124, Bari, Italy
| | - Amalia Azzariti
- Experimental Pharmacology Laboratory, Italia, 70124, Bari, Italy.
| |
Collapse
|
32
|
Li Y, Shi J, Yang J, Ge S, Zhang J, Jia R, Fan X. Uveal melanoma: progress in molecular biology and therapeutics. Ther Adv Med Oncol 2020; 12:1758835920965852. [PMID: 33149769 PMCID: PMC7586035 DOI: 10.1177/1758835920965852] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Uveal melanoma (UM) is the most common intraocular malignancy in adults. So far, no systemic therapy or standard treatment exists to reduce the risk of metastasis and improve overall survival of patients. With the increased knowledge regarding the molecular pathways that underlie the oncogenesis of UM, it is expected that novel therapeutic approaches will be available to conquer this disease. This review provides a summary of the current knowledge of, and progress made in understanding, the pathogenesis, genetic mutations, epigenetics, and immunology of UM. With the advent of the omics era, multi-dimensional big data are publicly available, providing an innovation platform to develop effective targeted and personalized therapeutics for UM patients. Indeed, recently, a great number of therapies have been reported specifically for UM caused by oncogenic mutations, as well as other etiologies. In this review, special attention is directed to advancements in targeted therapies. In particular, we discuss the possibilities of targeting: GNAQ/GNA11, PLCβ, and CYSLTR2 mutants; regulators of G-protein signaling; the secondary messenger adenosine diphosphate (ADP)-ribosylation factor 6 (ARF6); downstream pathways, such as those involving mitogen-activated protein kinase/MEK/extracellular signal-related kinase, protein kinase C (PKC), phosphoinositide 3-kinase/Akt/mammalian target of rapamycin (mTOR), Trio/Rho/Rac/Yes-associated protein, and inactivated BAP1; and immune-checkpoint proteins cytotoxic T-lymphocyte antigen 4 and programmed cell-death protein 1/programmed cell-death ligand 1. Furthermore, we conducted a survey of completed and ongoing clinical trials applying targeted and immune therapies for UM. Although drug combination therapy based on the signaling pathways involved in UM has made great progress, targeted therapy is still an unmet medical need.
Collapse
Affiliation(s)
- Yongyun Li
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jiahao Shi
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jie Yang
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jianming Zhang
- National Research Center for Translational Medicine, Shanghai State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai 200001, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200001, China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai 200001, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, 833 Zhizaoju Road, Huangpu District, Shanghai 200001, China
| |
Collapse
|
33
|
Distinct Mutation Patterns Reveal Melanoma Subtypes and Influence Immunotherapy Response in Advanced Melanoma Patients. Cancers (Basel) 2020; 12:cancers12092359. [PMID: 32825510 PMCID: PMC7563780 DOI: 10.3390/cancers12092359] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/14/2020] [Accepted: 08/16/2020] [Indexed: 12/23/2022] Open
Abstract
The detection of somatic driver mutations by next-generation sequencing (NGS) is becoming increasingly important in the care of advanced melanoma patients. In our study, we evaluated the NGS results of 82 melanoma patients from clinical routine in 2017. Besides determining the tumor mutational burden (TMB) and annotation of all genetic driver alterations, we investigated their potential as a predictor for resistance to immune checkpoint inhibitors (ICI) and as a distinguishing feature between melanoma subtypes. Melanomas of unknown primary had a similar mutation pattern and TMB to cutaneous melanoma, which hints at its cutaneous origin. Besides the typical hotspot mutation in BRAF and NRAS, we frequently observed CDKN2A deletions. Acral and mucosal melanomas were dominated by CNV alterations affecting PDGFRA, KIT, CDK4, RICTOR, CCND2 and CHEK2. Uveal melanoma often had somatic SNVs in GNA11/Q and amplification of MYC in all cases. A significantly higher incidence of BRAF V600 mutations and EGFR amplifications, PTEN and TP53 deletions was found in patients with disease progression while on ICI. Thus, NGS might help to characterize melanoma subtypes more precisely and to identify possible resistance mechanisms to ICI therapy. Nevertheless, NGS based studies, including larger cohorts, are needed to support potential genetic ICI resistance mechanisms.
Collapse
|
34
|
A Phase II Study of Glembatumumab Vedotin for Metastatic Uveal Melanoma. Cancers (Basel) 2020; 12:cancers12082270. [PMID: 32823698 PMCID: PMC7465139 DOI: 10.3390/cancers12082270] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022] Open
Abstract
Glembatumumab vedotin (CDX-011, GV) is a fully human Immunoglobulin G2 monoclonal antibody directed against glycoprotein NMB coupled via a peptide linker to monomethyl auristatin E (MMAE), a potent cytotoxic microtubule inhibitor. This phase II study evaluated the overall response rate and safety of GV, glycoprotein NMB (GPNMB) expression, and survival in patients with metastatic uveal melanoma. Eligible patients with metastatic uveal melanoma who had not previously been treated with chemotherapy received GV 1.9 mg/kg every three weeks. The primary endpoint was the objective response rate (ORR). Secondary endpoints included GPNMB expression, progression-free survival (PFS), overall survival (OS), and toxicity analysis. GPNMB expression was assessed pre- and post-treatment via immunohistochemistry for patients with available tumor tissue. Out of 35 patients who received treatment, two patients had confirmed partial responses (PRs; 6%), and 18 patients had a stable disease (SD; 51%) as the best objective response. 38% of the patients had stable disease >100 days. The grade 3 or 4 toxicities that occurred in two or more patients were neutropenia, rash, hyponatremia, and vomiting. The median progression-free survival was 3.1 months (95% CI: 1.5–5.6), and the median overall survival was 11.9 months (95% CI 9.0–16.9) in the evaluable study population. GV is well-tolerated in metastatic uveal melanoma. The disease control rate was 57% despite a low objective response rate. Exploratory immune correlation studies are underway to provide insight into target saturation, combination strategies, and antigen release.
Collapse
|
35
|
MicroRNAs and Uveal Melanoma: Understanding the Diverse Role of These Small Molecular Regulators. Int J Mol Sci 2020; 21:ijms21165648. [PMID: 32781746 PMCID: PMC7460624 DOI: 10.3390/ijms21165648] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/31/2020] [Accepted: 08/04/2020] [Indexed: 12/12/2022] Open
Abstract
Uveal melanoma (UM) is a rare tumour of the eye, characterised by a high propensity to metastasise in half of all patients, most frequently to the liver. Although there are effective treatment options for the primary tumour, once metastasis has occurred prognosis is poor, with overall survival limited to months. Currently, there are no effective treatments for metastatic UM, despite the tumour having a well-defined signalling pathway to which many therapies have been directed. In an effort to develop novel treatment approaches, understanding the role of other signalling molecules, such as microRNAs, is fundamental. MicroRNAs (miRNAs) are small non-coding RNA molecules involved in posttranscriptional gene regulation, resulting in reduced target gene expression and subsequent protein translation. In UM, several dysregulated miRNAs have been proposed to play a functional role in disease progression, whereas others have been put forward as clinical biomarkers of high-risk disease following isolation from blood, plasma and exosomes. Most recently, analyses of large datasets have identified promising prognostic miRNA signatures and panels. This review navigates the plethora of aberrant miRNAs disclosed so far in UM, and maps these to signalling pathways, which could be targeted in future therapies for the disseminated disease.
Collapse
|
36
|
Rago F, Elliott G, Li A, Sprouffske K, Kerr G, Desplat A, Abramowski D, Chen JT, Farsidjani A, Xiang KX, Bushold G, Feng Y, Shirley MD, Bric A, Vattay A, Möbitz H, Nakajima K, Adair CD, Mathieu S, Ntaganda R, Smith T, Papillon JPN, Kauffmann A, Ruddy DA, Bhang HEC, Castelletti D, Jagani Z. The Discovery of SWI/SNF Chromatin Remodeling Activity as a Novel and Targetable Dependency in Uveal Melanoma. Mol Cancer Ther 2020; 19:2186-2195. [PMID: 32747420 DOI: 10.1158/1535-7163.mct-19-1013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/15/2019] [Accepted: 07/21/2020] [Indexed: 11/16/2022]
Abstract
Uveal melanoma is a rare and aggressive cancer that originates in the eye. Currently, there are no approved targeted therapies and very few effective treatments for this cancer. Although activating mutations in the G protein alpha subunits, GNAQ and GNA11, are key genetic drivers of the disease, few additional drug targets have been identified. Recently, studies have identified context-specific roles for the mammalian SWI/SNF chromatin remodeling complexes (also known as BAF/PBAF) in various cancer lineages. Here, we find evidence that the SWI/SNF complex is essential through analysis of functional genomics screens and further validation in a panel of uveal melanoma cell lines using both genetic tools and small-molecule inhibitors of SWI/SNF. In addition, we describe a functional relationship between the SWI/SNF complex and the melanocyte lineage-specific transcription factor Microphthalmia-associated Transcription Factor, suggesting that these two factors cooperate to drive a transcriptional program essential for uveal melanoma cell survival. These studies highlight a critical role for SWI/SNF in uveal melanoma, and demonstrate a novel path toward the treatment of this cancer.
Collapse
Affiliation(s)
- Florencia Rago
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - GiNell Elliott
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Ailing Li
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | - Grainne Kerr
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Aurore Desplat
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | - Julie T Chen
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Ali Farsidjani
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Kay X Xiang
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Geoffrey Bushold
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Yun Feng
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Matthew D Shirley
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Anka Bric
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Anthony Vattay
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Henrik Möbitz
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | | | - Simon Mathieu
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Rukundo Ntaganda
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Troy Smith
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | | | - David A Ruddy
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Hyo-Eun C Bhang
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | - Zainab Jagani
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts.
| |
Collapse
|
37
|
Jin E, Burnier JV. Liquid Biopsy in Uveal Melanoma: Are We There Yet? Ocul Oncol Pathol 2020; 7:1-16. [PMID: 33796511 DOI: 10.1159/000508613] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
In the era of precision oncology, major strides are being made to use individual tumor information for clinical decision-making. Differing from traditional biopsy methods, the emerging practice of liquid biopsy provides a minimally invasive way of obtaining tumor cells and derived molecules. Liquid biopsy provides a means to detect and monitor disease progression, recurrence, and treatment response in a noninvasive way, and to potentially complement classical biopsy. Uveal melanoma (UM) is a unique malignancy, with diagnosis heavily reliant on imaging, few repeat biopsies, and a high rate of metastasis, which occurs hematogenously and often many years after diagnosis. In this disease setting, a noninvasive biomarker to detect, monitor, and study the disease in real time could lead to better disease understanding and patient care. While advances have been made in the detection of tumor-disseminated components, sensitivity and specificity remain important challenges. Ambiguity remains in how to interpret current findings and in how liquid biopsy can have a place in clinical practice. Related publications in UM are few compared to other cancers, but with further studies we may be able to uncover more about the biology of disseminated molecules and the mechanisms involved in the progression to metastasis.
Collapse
Affiliation(s)
- Eva Jin
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| |
Collapse
|
38
|
Thornton S, Kalirai H, Aughton K, Coupland SE. Unpacking the genetic etiology of uveal melanoma. EXPERT REVIEW OF OPHTHALMOLOGY 2020. [DOI: 10.1080/17469899.2020.1785872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Sophie Thornton
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Liverpool Clinical Laboratories, Liverpool University Hospitals Foundation Trusts, Liverpool, UK
| | - Helen Kalirai
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Liverpool Clinical Laboratories, Liverpool University Hospitals Foundation Trusts, Liverpool, UK
| | - Karen Aughton
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Sarah E. Coupland
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Liverpool Clinical Laboratories, Liverpool University Hospitals Foundation Trusts, Liverpool, UK
| |
Collapse
|
39
|
Hanbazazh M, Dryja TP. Molecular Genetics of Intraocular Tumors. Semin Ophthalmol 2020; 35:174-181. [PMID: 32507011 DOI: 10.1080/08820538.2020.1776343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
PURPOSE To explore the value of molecular technologies in the pathologic evaluation, diagnosis, and treatment of retinoblastoma and uveal melanoma. METHODS Review of the peer-reviewed literature on the molecular pathology of primary intraocular tumors. CONCLUSION Molecular tests are playing an increasingly important role in the diagnosis of intraocular tumors. They provide information valuable for diagnosis, prognosis, screening regimens, genetic counselling, and treatment. These technologies are becoming easier, faster, and with higher sensitivity and accuracy.
Collapse
Affiliation(s)
- Mehenaz Hanbazazh
- David G Cogan Laboratory of Ophthalmic Pathology, Massachusetts Eye and Ear Infirmary, Harvard Medical School , Boston, MA, USA
| | - Thaddeus P Dryja
- David G Cogan Laboratory of Ophthalmic Pathology, Massachusetts Eye and Ear Infirmary, Harvard Medical School , Boston, MA, USA
| |
Collapse
|
40
|
Bustamante P, Piquet L, Landreville S, Burnier JV. Uveal melanoma pathobiology: Metastasis to the liver. Semin Cancer Biol 2020; 71:65-85. [PMID: 32450140 DOI: 10.1016/j.semcancer.2020.05.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022]
Abstract
Uveal melanoma (UM) is a type of intraocular tumor with a propensity to disseminate to the liver. Despite the identification of the early driver mutations during the development of the pathology, the process of UM metastasis is still not fully comprehended. A better understanding of the genetic, molecular, and environmental factors participating to its spread and metastatic outgrowth could provide additional approaches for UM treatment. In this review, we will discuss the advances made towards the understanding of the pathogenesis of metastatic UM, summarize the current and prospective treatments, and introduce some of the ongoing research in this field.
Collapse
Affiliation(s)
- Prisca Bustamante
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Canada; Experimental Pathology Unit, Department of Pathology, McGill University, Montréal, Canada
| | - Léo Piquet
- Département d'ophtalmologie et d'ORL-CCF, Faculté de médecine, Université Laval, Quebec City, Canada; CUO-Recherche and Axe médecine régénératrice, Centre de recherche du CHU de Québec-Université Laval, Quebec City, Canada; Centre de recherche sur le cancer de l'Université Laval, Quebec City, Canada; Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, Canada
| | - Solange Landreville
- Département d'ophtalmologie et d'ORL-CCF, Faculté de médecine, Université Laval, Quebec City, Canada; CUO-Recherche and Axe médecine régénératrice, Centre de recherche du CHU de Québec-Université Laval, Quebec City, Canada; Centre de recherche sur le cancer de l'Université Laval, Quebec City, Canada; Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Canada; Experimental Pathology Unit, Department of Pathology, McGill University, Montréal, Canada; Gerald Bronfman Department Of Oncology, McGill University, Montréal, Canada.
| |
Collapse
|
41
|
Roy V, Magne B, Vaillancourt-Audet M, Blais M, Chabaud S, Grammond E, Piquet L, Fradette J, Laverdière I, Moulin VJ, Landreville S, Germain L, Auger FA, Gros-Louis F, Bolduc S. Human Organ-Specific 3D Cancer Models Produced by the Stromal Self-Assembly Method of Tissue Engineering for the Study of Solid Tumors. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6051210. [PMID: 32352002 PMCID: PMC7178531 DOI: 10.1155/2020/6051210] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 02/07/2020] [Accepted: 02/28/2020] [Indexed: 12/24/2022]
Abstract
Cancer research has considerably progressed with the improvement of in vitro study models, helping to understand the key role of the tumor microenvironment in cancer development and progression. Over the last few years, complex 3D human cell culture systems have gained much popularity over in vivo models, as they accurately mimic the tumor microenvironment and allow high-throughput drug screening. Of particular interest, in vitrohuman 3D tissue constructs, produced by the self-assembly method of tissue engineering, have been successfully used to model the tumor microenvironment and now represent a very promising approach to further develop diverse cancer models. In this review, we describe the importance of the tumor microenvironment and present the existing in vitro cancer models generated through the self-assembly method of tissue engineering. Lastly, we highlight the relevance of this approach to mimic various and complex tumors, including basal cell carcinoma, cutaneous neurofibroma, skin melanoma, bladder cancer, and uveal melanoma.
Collapse
Affiliation(s)
- Vincent Roy
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Brice Magne
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Maude Vaillancourt-Audet
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Mathieu Blais
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Stéphane Chabaud
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Emil Grammond
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Léo Piquet
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
| | - Julie Fradette
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Isabelle Laverdière
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
- Faculty of Pharmacy, Université Laval and CHU de Québec-Université Laval Research Center, Oncology Division, Québec, QC, Canada
| | - Véronique J. Moulin
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Solange Landreville
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
- Department of Ophthalmology, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Lucie Germain
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - François A. Auger
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - François Gros-Louis
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Stéphane Bolduc
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
42
|
Hou C, Xiao L, Ren X, Tang F, Guo B, Zeng W, Liang C, Yan N. Mutations of GNAQ, GNA11, SF3B1, EIF1AX, PLCB4 and CYSLTR in Uveal Melanoma in Chinese Patients. Ophthalmic Res 2019; 63:358-368. [DOI: 10.1159/000502888] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 08/21/2019] [Indexed: 11/19/2022]
|
43
|
Piquet L, Dewit L, Schoonjans N, Millet M, Bérubé J, Gerges PRA, Bordeleau F, Landreville S. Synergic Interactions Between Hepatic Stellate Cells and Uveal Melanoma in Metastatic Growth. Cancers (Basel) 2019; 11:cancers11081043. [PMID: 31344830 PMCID: PMC6721369 DOI: 10.3390/cancers11081043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 12/15/2022] Open
Abstract
Uveal melanoma (UM) is a malignant intraocular tumor that spreads to the liver in half of the cases. Since hepatic cells could play a role in the therapeutic resistance of metastatic UM, the purpose of our study was to investigate the pro-invasive role of hepatic stellate cells (HSteCs) in metastatic UM at the micro- and macro-metastatic stages. We first performed an immunostaining with the alpha-smooth muscle actin (αSMA) to localize activated HSteCs in UM liver macro-metastases from four patients. Their accumulation of collagen was assessed with Masson’s Trichrome stain. Next, we inoculated metastatic UM cells alone or with human HSteCs in triple-immunodeficient mice, in order to determine if HSteCs are recruited as early as the micro-metastatic stage. The growth of metastatic foci was imaged in the liver by ex vivo fluorescence imaging. Histological analyses were performed with Masson’s Trichrome and Picrosirius Red stains, and antibodies against Melan-A and αSMA. The collagen content was measured in xenografts by quantitative polarization microscopy. In patient hepatectomy samples, activated HSteCs and their pathological matrix were localized surrounding the malignant lesions. In the mouse xenograft model, the number of hepatic metastases was increased when human HSteCs were co-inoculated. Histological analyses revealed a significant recruitment of HSteCs near the micro/macrolesions, and an increase in fibrillar collagen production. Our results show that HSteCs can provide a permissive microenvironment and might increase the therapeutic resistance of metastatic UM.
Collapse
Affiliation(s)
- Léo Piquet
- Faculté de médecine, Université Laval, Quebec City, QC G1V 0A6, Canada
- Centre de recherche du CHU de Québec-Université Laval, Quebec City, QC G1S 4L8, Canada
- Centre de recherche sur le cancer de l'Université Laval, Quebec City, QC G1R 3S3, Canada
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
| | - Louise Dewit
- Centre de recherche du CHU de Québec-Université Laval, Quebec City, QC G1S 4L8, Canada
- Centre de recherche sur le cancer de l'Université Laval, Quebec City, QC G1R 3S3, Canada
| | - Nathan Schoonjans
- Centre de recherche du CHU de Québec-Université Laval, Quebec City, QC G1S 4L8, Canada
- Centre de recherche sur le cancer de l'Université Laval, Quebec City, QC G1R 3S3, Canada
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
| | - Martial Millet
- Centre de recherche du CHU de Québec-Université Laval, Quebec City, QC G1S 4L8, Canada
- Centre de recherche sur le cancer de l'Université Laval, Quebec City, QC G1R 3S3, Canada
| | - Julie Bérubé
- Centre de recherche du CHU de Québec-Université Laval, Quebec City, QC G1S 4L8, Canada
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
| | - Peter R A Gerges
- Centre de recherche du CHU de Québec-Université Laval, Quebec City, QC G1S 4L8, Canada
| | - François Bordeleau
- Faculté de médecine, Université Laval, Quebec City, QC G1V 0A6, Canada
- Centre de recherche du CHU de Québec-Université Laval, Quebec City, QC G1S 4L8, Canada
- Centre de recherche sur le cancer de l'Université Laval, Quebec City, QC G1R 3S3, Canada
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
| | - Solange Landreville
- Faculté de médecine, Université Laval, Quebec City, QC G1V 0A6, Canada.
- Centre de recherche du CHU de Québec-Université Laval, Quebec City, QC G1S 4L8, Canada.
- Centre de recherche sur le cancer de l'Université Laval, Quebec City, QC G1R 3S3, Canada.
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada.
| |
Collapse
|
44
|
Croce M, Ferrini S, Pfeffer U, Gangemi R. Targeted Therapy of Uveal Melanoma: Recent Failures and New Perspectives. Cancers (Basel) 2019; 11:E846. [PMID: 31216772 PMCID: PMC6628160 DOI: 10.3390/cancers11060846] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/23/2022] Open
Abstract
Among Uveal Melanoma (UM) driver mutations, those involving GNAQ or GNA11 genes are the most frequent, while a minor fraction of tumors bears mutations in the PLCB4 or CYSLTR2 genes. Direct inhibition of constitutively active oncoproteins deriving from these mutations is still in its infancy in UM, whereas BRAFV600E-targeted therapy has obtained relevant results in cutaneous melanoma. However, UM driver mutations converge on common downstream signaling pathways such as PKC/MAPK, PI3K/AKT, and YAP/TAZ, which are presently considered as actionable targets. In addition, BAP1 loss, which characterizes UM metastatic progression, affects chromatin structure via histone H2A deubiquitylation that may be counteracted by histone deacetylase inhibitors. Encouraging results of preclinical studies targeting signaling molecules such as MAPK and PKC were unfortunately not confirmed in early clinical studies. Indeed, a general survey of all clinical trials applying new targeted and immune therapy to UM displayed disappointing results. This paper summarizes the most recent studies of UM-targeted therapies, analyzing the possible origins of failures. We also focus on hyperexpressed molecules involved in UM aggressiveness as potential new targets for therapy.
Collapse
Affiliation(s)
- Michela Croce
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| | | | - Ulrich Pfeffer
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| | | |
Collapse
|
45
|
Rossi E, Pagliara MM, Orteschi D, Dosa T, Sammarco MG, Caputo CG, Petrone G, Rindi G, Zollino M, Blasi MA, Cassano A, Bria E, Tortora G, Schinzari G. Pembrolizumab as first-line treatment for metastatic uveal melanoma. Cancer Immunol Immunother 2019; 68:1179-1185. [PMID: 31175402 PMCID: PMC6584707 DOI: 10.1007/s00262-019-02352-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 05/30/2019] [Indexed: 01/24/2023]
Abstract
Background No standard treatment has been defined for metastatic uveal melanoma (mUM). Although clinical trials testing Nivolumab/Pembrolizumab for cutaneous melanoma did not include mUM, anti PD-1 agents are commonly used for this disease. Patients and methods In this prospective observational cohort single arm study, we investigated efficacy and safety of Pembrolizumab as first-line therapy for mUM. The efficacy was evaluated in terms of progression-free survival (PFS), response rate and overall survival (OS). Toxicity was also assessed. Results Seventeen patients were enrolled. A median of 8 cycles were administered (range 2–28). Two patients achieved partial response (11.7%), 6 a disease stabilization (35.3%), whereas 9 (53%) had a progression. No complete response was observed. PFS of the overall population was 3.8 months. PFS was 9.7 months for patients with an interval higher than 5 years from diagnosis of primary tumor to metastatic disease and 2.6 months for patients with an interval lower than 5 years [p = 0.039, HR 0.2865 (95% CI 0.0869–0.9443)]. Median OS was not reached. The two responding patients were still on treatment with Pembrolizumab at the time of data analysis. Survival was 12.8 months for patients with clinical benefit, while OS for progressive patients was 3.1 months. PD-L1 expression and genomic abnormalities predictive of relapse after diagnosis of primary tumor were not associated with PFS. Toxicity was mild, without grade 3–4 side effects. Conclusions The efficacy of Pembrolizumab does not seem particularly different when compared to other agents for mUM, but responding patients had a remarkable disease control.
Collapse
Affiliation(s)
- Ernesto Rossi
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy.
| | - Monica Maria Pagliara
- Department of Ophthalmology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy
| | - Daniela Orteschi
- Institute of Genomic Medicine, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy
| | - Tommaso Dosa
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy
| | - Maria Grazia Sammarco
- Department of Ophthalmology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy
| | - Carmela Grazia Caputo
- Department of Ophthalmology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy
| | - Gianluigi Petrone
- Department of Pathology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy
| | - Guido Rindi
- Department of Pathology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy
| | - Marcella Zollino
- Institute of Genomic Medicine, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy
| | - Maria Antonietta Blasi
- Department of Ophthalmology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy
| | - Alessandra Cassano
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy
| | - Emilio Bria
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy
| | - Giampaolo Tortora
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy
| | - Giovanni Schinzari
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168, Rome, Italy
| |
Collapse
|
46
|
Mini E, Lapucci A, Perrone G, D'Aurizio R, Napoli C, Brugia M, Landini I, Tassi R, Picariello L, Simi L, Mancini I, Messerini L, Magi A, Pinzani P, Mazzei T, Tonelli F, Nobili S. RNA sequencing reveals PNN and KCNQ1OT1 as predictive biomarkers of clinical outcome in stage III colorectal cancer patients treated with adjuvant chemotherapy. Int J Cancer 2019; 145:2580-2593. [PMID: 30973654 DOI: 10.1002/ijc.32326] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/13/2019] [Accepted: 04/01/2019] [Indexed: 12/16/2022]
Abstract
Five-year overall survival of stage III colorectal cancer (CRC) patients treated with standard adjuvant chemotherapy (ACHT) is highly variable. Genomic biomarkers and/or transcriptomic profiles identified lack of adequate validation. Aim of our study was to identify and validate molecular biomarkers predictive of ACHT response in stage III CRC patients by a transcriptomic approach. From a series of CRC patients who received ACHT, two stage III extreme cohorts (unfavorable vs. favorable prognosis) were selected. RNA-sequencing was performed from fresh frozen explants. Tumors were characterized for somatic mutations. Validation was performed in stage III CRC patients extracted from two GEO datasets. According to disease-free survival (DFS), 108 differentially expressed genes (104/4 up/downregulated in the unfavorable prognosis group) were identified. Among 104 upregulated genes, 42 belonged to olfactory signaling pathways, 62 were classified as pseudogenes (n = 17), uncharacterized noncoding RNA (n = 10), immune response genes (n = 4), microRNA (n = 1), cancer-related genes (n = 14) and cancer-unrelated genes (n = 16). Three out of four down-regulated genes were cancer-related. Mutational status (i.e., RAS, BRAF, PIK3CA) did not differ among the cohorts. In the validation cohort, multivariate analysis showed high PNN and KCNQ1OT1 expression predictive of shorter DFS in ACHT treated patients (p = 0.018 and p = 0.014, respectively); no difference was observed in untreated patients. This is the first study that identifies by a transcriptomic approach and validates PNN and KCNQ1OT1 as molecular biomarkers predictive of chemotherapy response in stage III CRC patients. After a further validation in an independent cohort, PNN and KCNQ1OT1 evaluation could be proposed to prospectively identify stage III CRC patients benefiting from ACHT.
Collapse
Affiliation(s)
- Enrico Mini
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Andrea Lapucci
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Gabriele Perrone
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Romina D'Aurizio
- Institute of Informatics and Telematics (IIT), National Research Council (CNR), Pisa, Italy
| | - Cristina Napoli
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Marco Brugia
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Ida Landini
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Renato Tassi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lucia Picariello
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Lisa Simi
- Molecular and Clinical Biochemistry Laboratory, Careggi University Hospital, Florence, Italy
| | - Irene Mancini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Luca Messerini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alberto Magi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Pamela Pinzani
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.,Molecular and Clinical Biochemistry Laboratory, Careggi University Hospital, Florence, Italy
| | - Teresita Mazzei
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Francesco Tonelli
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Stefania Nobili
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| |
Collapse
|
47
|
Violanti SS, Bononi I, Gallenga CE, Martini F, Tognon M, Perri P. New Insights into Molecular Oncogenesis and Therapy of Uveal Melanoma. Cancers (Basel) 2019; 11:E694. [PMID: 31109147 PMCID: PMC6562554 DOI: 10.3390/cancers11050694] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/15/2022] Open
Abstract
Uveal melanoma (UM), which is the most common cancer of the eye, was investigated in recent years by many teams in the field of biomedical sciences and eye clinicians. New knowledge was acquired on molecular pathways found to be dysregulated during the multistep process of oncogenesis, whereas novel therapeutic approaches gave significant results in the clinical applications. Uveal melanoma-affected patients greatly benefited from recent advances of the research in this eye cancer. Tumour biology, genetics, epigenetics and immunology contributed significantly in elucidating the role of different genes and related pathways during uveal melanoma onset/progression and UM treatments. Indeed, these investigations allowed identification of new target genes and to develop new therapeutic strategies/compounds to cure this aggressive melanoma of the eye. Unfortunately, the advances reported in the treatment of cutaneous melanoma have not produced analogous benefits in metastatic uveal melanoma. Nowadays, no systemic adjuvant therapy has been shown to improve overall survival or reduce the risk of metastasis. However, the increasing knowledge of this disease, and the encouraging results seen in clinical trials, offer promise for future effective therapies. Herein, different pathways/genes involved in uveal melanoma onset/progression were taken into consideration, together with novel therapeutic approaches.
Collapse
Affiliation(s)
- Sara Silvia Violanti
- Department of Biomedical Sciences and Specialized Surgeries, School of Medicine, University of Ferrara and Eye Unit of University Hospital of Ferrara, 44124 Ferrara, Italy.
| | - Ilaria Bononi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Carla Enrica Gallenga
- Department of Biomedical Sciences and Specialized Surgeries, School of Medicine, University of Ferrara and Eye Unit of University Hospital of Ferrara, 44124 Ferrara, Italy.
| | - Fernanda Martini
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Mauro Tognon
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Paolo Perri
- Department of Biomedical Sciences and Specialized Surgeries, School of Medicine, University of Ferrara and Eye Unit of University Hospital of Ferrara, 44124 Ferrara, Italy.
| |
Collapse
|
48
|
Wang LX, Wan C, Dong ZB, Wang BH, Liu HY, Li Y. Integrative Analysis of Long Noncoding RNA (lncRNA), microRNA (miRNA) and mRNA Expression and Construction of a Competing Endogenous RNA (ceRNA) Network in Metastatic Melanoma. Med Sci Monit 2019; 25:2896-2907. [PMID: 31004080 PMCID: PMC6487673 DOI: 10.12659/msm.913881] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Worldwide, metastatic melanoma of the skin has an aggressive course with high morbidity and mortality. Therefore, an increased understanding of the pathogenesis of metastatic melanoma has gained increasing attention, including the role of epigenetic modification and competing endogenous RNA (ceRNA). This study aimed to used bioinformatics data to undertake an integrative analysis of long noncoding RNA (lncRNA), microRNA (miRNA) and mRNA expression to construct a ceRNA network in metastatic melanoma. Data from the Cancer Genome Atlas (TCGA), the Gene Ontology (GO) database, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway were analyzed. There were 471 cases that included 103 primary solid tumors and 368 cases of metastatic melanoma that included transcriptome sequencing data (including lncRNA and mRNA); 452 cases had miRNA sequencing data. Analysis of chip data identified 85 6 mRNAs, 67 miRNAs, and 250 lncRNAs that were differentially expressed in cases of metastatic melanoma, of which 25 miRNAs, 18 lncRNAs, and 18 mRNAs participated in the formation of ceRNAs. Survival analysis identified seven differentially expressed mRNAs, five differentially expressed miRNAs (miRNA-29c, miRNA-100, miR-142-3p, miR-150, miR-516a-2), and six differentially expressed lncRNAs (AC068594.1, C7orf71, FAM41C, GPC5-AS1, MUC19, LINC00402) that were correlated with survival time in patients with metastatic melanoma. Bioinformatics data and integrative analysis identified lncRNA, miRNA, and mRNA expression to construct a ceRNA and patient survival network in metastatic melanoma. These findings support the need for further studies on the mechanisms involved in the regulation of metastatic melanoma by ceRNAs.
Collapse
Affiliation(s)
- Li-Xin Wang
- Department of Dermatology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Chuan Wan
- Department of Dermatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Zheng-Bang Dong
- Department of Dermatology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China (mainland)
| | - Bai-He Wang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China (mainland)
| | - Hong-Ye Liu
- Department of Dermatology, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Yang Li
- Department of Dermatology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| |
Collapse
|
49
|
Rabbie R, Ferguson P, Molina‐Aguilar C, Adams DJ, Robles‐Espinoza CD. Melanoma subtypes: genomic profiles, prognostic molecular markers and therapeutic possibilities. J Pathol 2019; 247:539-551. [PMID: 30511391 PMCID: PMC6492003 DOI: 10.1002/path.5213] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 12/24/2022]
Abstract
Melanoma is characterised by its ability to metastasise at early stages of tumour development. Current clinico-pathologic staging based on the American Joint Committee on Cancer criteria is used to guide surveillance and management in early-stage disease, but its ability to predict clinical outcome has limitations. Herein we review the genomics of melanoma subtypes including cutaneous, acral, uveal and mucosal, with a focus on the prognostic and predictive significance of key molecular aberrations. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Roy Rabbie
- Experimental Cancer GeneticsThe Wellcome Sanger InstituteHinxtonUK
- Cambridge Cancer CentreCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Peter Ferguson
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred HospitalSydneyAustralia
- Melanoma Institute Australia, The University of SydneySydneyAustralia
| | - Christian Molina‐Aguilar
- Laboratorio Internacional de Investigación sobre el Genoma HumanoUniversidad Nacional Autónoma de MéxicoSantiago de QuerétaroMexico
| | - David J Adams
- Experimental Cancer GeneticsThe Wellcome Sanger InstituteHinxtonUK
| | - Carla D Robles‐Espinoza
- Experimental Cancer GeneticsThe Wellcome Sanger InstituteHinxtonUK
- Laboratorio Internacional de Investigación sobre el Genoma HumanoUniversidad Nacional Autónoma de MéxicoSantiago de QuerétaroMexico
| |
Collapse
|
50
|
Zhang D, Lin J, Chao Y, Zhang L, Jin L, Li N, He R, Ma B, Zhao W, Han C. Regulation of the adaptation to ER stress by KLF4 facilitates melanoma cell metastasis via upregulating NUCB2 expression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:176. [PMID: 30055641 PMCID: PMC6064624 DOI: 10.1186/s13046-018-0842-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/13/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Adaptation to ER stress has been indicated to play an important role in resistance to therapy in human melanoma. However, the relationship between adaptation to ER stress and cell metastasis in human melanoma remains unclear. METHODS The relationship of adaptation to ER stress and cell metastasis was investigated using transwell and mouse metastasis assays. The potential molecular mechanism of KLF4 in regulating the adaptation to ER stress and cell metastasis was investigated using RNA sequencing analysis, q-RT-PCR and western blot assays. The transcriptional regulation of nucleobindin 2 (NUCB2) by KLF4 was identified using bioinformatic analysis, luciferase assay, and chromatin immunoprecipitation (ChIP). The clinical significance of KLF4 and NUCB2 was based on human tissue microarray (TMA) analysis. RESULTS Here, we demonstrated that KLF4 was induced by ER stress in melanoma cells, and increased KLF4 inhibited cell apoptosis and promoted cell metastasis. Further mechanistic studies revealed that KLF4 directly bound to the promoter of NUCB2, facilitating its transcription. Additionally, an increase in KLF4 promoted melanoma ER stress resistance, tumour growth and cell metastasis by regulating NCUB2 expression in vitro and in vivo. Elevated KLF4 was found in human melanoma tissues, which was associated with NUCB2 expression. CONCLUSION Our data revealed that the promotion of ER stress resistance via the KLF4-NUCB2 axis is essential for melanoma cell metastasis, and KLF4 may be a promising specific target for melanoma therapy.
Collapse
Affiliation(s)
- Dongmei Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China.,Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Jingrong Lin
- Department of Dermatology, the First Affiliated Hospital, Dalian Medical University, Liaoning, 116027, China
| | - Yulin Chao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Lu Zhang
- Department of Orthopedics, Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China
| | - Lei Jin
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Na Li
- Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ruiping He
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Binbin Ma
- Department of Orthopedics, Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China
| | - Wenzhi Zhao
- Department of Orthopedics, Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China.
| | - Chuanchun Han
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|