1
|
Lai J, Madan V, Qadri A, Danilova L, Yuan L, Jacobs V, Ogurtsova A, Engle LL, Sunshine JC. Lymphocyte Activation Gene 3 Expression, γδ T-Cell/Major Histocompatibility Complex Class I Interactions, and Prognosis in Merkel Cell Carcinoma. J Transl Med 2025; 105:102178. [PMID: 39510251 DOI: 10.1016/j.labinv.2024.102178] [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: 05/01/2024] [Revised: 10/08/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024] Open
Abstract
Merkel cell carcinoma (MCC) is an aggressive cutaneous malignancy with a poor prognosis. One of the major mechanisms of immune evasion in MCC involves downregulation of major histocompatibility complex class I (MHC-I). Anti-PD-1/programmed death ligand 1 checkpoint inhibitors have revolutionized treatment for MCC, producing objective responses in approximately 50% of patients, and are now the standard of care; however, a substantial proportion of patients either fail to respond or develop resistance to checkpoint inhibitors. Given these recent successes, identification of other targetable immune checkpoints in the MCC tumor microenvironment is of great interest. Additionally, γδ T cells may play critical roles in response to MHC-I-deficient cancers; therefore, evaluating γδ T cells as a prognostic biomarker is warranted. We characterized the expression of programmed death ligand 1, PD-1, CD3, CD8, lymphocyte activation gene 3 (LAG-3), MHC-I, and γδ T cells by immunohistochemistry in a preimmunotherapy retrospective cohort of 54 cases of MCC and quantified expression levels and marker density using HALO software. The increased density of LAG-3 and γδ T cells correlated with other markers of an inflamed tumor microenvironment, with significant positive associations across all 6 markers (P < .002). Reflective of their putative role in the response to MHC-I-suppressed cancers, cases with low human leukocyte antigen I density showed a trend toward a higher ratio of γδ T cells:CD3+ T cells (Spearman r = -0.1582; P = .21). Importantly, high CD3 density (hazard ratio [HR], 0.23; P = .002), LAG-3 density (HR, 0.47; P = .037), γδ T-cell density (HR, 0.26; P = .02), and CD8 density (HR, 0.27; P = .03) showed associations with improved progression-free survival. Conditional tree analysis demonstrated that high CD8 and TCRδ expression were nonsignificant predictors of improved progression-free survival and overall survival. Overall, LAG-3 is expressed in MCC infiltrates and is prognostic in preimmunotherapy MCC, suggesting a potential role for LAG-3 inhibition in MCC. Additionally, CD8 and γδ T cells may play a critical role in the response to MCC, and γδ T-cell density may represent a novel biomarker in MCC.
Collapse
MESH Headings
- Humans
- Carcinoma, Merkel Cell/mortality
- Carcinoma, Merkel Cell/metabolism
- Carcinoma, Merkel Cell/immunology
- Carcinoma, Merkel Cell/diagnosis
- Carcinoma, Merkel Cell/pathology
- Male
- Skin Neoplasms/mortality
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
- Skin Neoplasms/diagnosis
- Skin Neoplasms/pathology
- Female
- Prognosis
- Aged
- Lymphocyte Activation Gene 3 Protein
- Histocompatibility Antigens Class I/metabolism
- Aged, 80 and over
- Intraepithelial Lymphocytes/metabolism
- Intraepithelial Lymphocytes/immunology
- Retrospective Studies
- Middle Aged
- Antigens, CD/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Tumor Microenvironment
- Biomarkers, Tumor/metabolism
Collapse
Affiliation(s)
- Jonathan Lai
- Department of Dermatology, Johns Hopkins University, Baltimore, Maryland; Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Vrinda Madan
- Department of Dermatology, Johns Hopkins University, Baltimore, Maryland; Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Aasheen Qadri
- Department of Dermatology, Johns Hopkins University, Baltimore, Maryland; Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Ludmila Danilova
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland; Division of Quantitative Sciences, Johns Hopkins University, Baltimore, Maryland; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Long Yuan
- Department of Dermatology, Johns Hopkins University, Baltimore, Maryland; Department of Pathology, Johns Hopkins University, Baltimore, Maryland; The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, Maryland
| | - Victoria Jacobs
- Department of Dermatology, Johns Hopkins University, Baltimore, Maryland; Department of Pathology, Johns Hopkins University, Baltimore, Maryland; The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, Maryland
| | - Aleksandra Ogurtsova
- Department of Dermatology, Johns Hopkins University, Baltimore, Maryland; Department of Pathology, Johns Hopkins University, Baltimore, Maryland; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland; The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, Maryland; The Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland
| | - Logan L Engle
- Department of Dermatology, Johns Hopkins University, Baltimore, Maryland; Department of Pathology, Johns Hopkins University, Baltimore, Maryland; The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, Maryland; The Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland
| | - Joel C Sunshine
- Department of Dermatology, Johns Hopkins University, Baltimore, Maryland; Department of Pathology, Johns Hopkins University, Baltimore, Maryland; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland; The Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
2
|
Pedersen EA, Verhaegen ME, Joseph MK, Harms KL, Harms PW. Merkel cell carcinoma: updates in tumor biology, emerging therapies, and preclinical models. Front Oncol 2024; 14:1413793. [PMID: 39136002 PMCID: PMC11317257 DOI: 10.3389/fonc.2024.1413793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024] Open
Abstract
Merkel cell carcinoma (MCC) is an aggressive cutaneous neuroendocrine carcinoma thought to arise via either viral (Merkel cell polyomavirus) or ultraviolet-associated pathways. Surgery and radiotherapy have historically been mainstays of management, and immunotherapy has improved outcomes for advanced disease. However, there remains a lack of effective therapy for those patients who fail to respond to these established approaches, underscoring a critical need to better understand MCC biology for more effective prognosis and treatment. Here, we review the fundamental aspects of MCC biology and the recent advances which have had profound impact on management. The first genetically-engineered mouse models for MCC tumorigenesis provide opportunities to understand the potential MCC cell of origin and may prove useful for preclinical investigation of novel therapeutics. The MCC cell of origin debate has also been advanced by recent observations of MCC arising in association with a clonally related hair follicle tumor or squamous cell carcinoma in situ. These studies also suggested a role for epigenetics in the origin of MCC, highlighting a potential utility for this therapeutic avenue in MCC. These and other therapeutic targets form the basis for a wealth of ongoing clinical trials to improve MCC management. Here, we review these recent advances in the context of the existing literature and implications for future investigations.
Collapse
Affiliation(s)
| | | | - Mallory K. Joseph
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Kelly L. Harms
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Paul W. Harms
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
3
|
Mazziotta C, Badiale G, Cervellera CF, Morciano G, Di Mauro G, Touzé A, Pinton P, Tognon M, Martini F, Rotondo JC. All-trans retinoic acid exhibits anti-proliferative and differentiating activity in Merkel cell carcinoma cells via retinoid pathway modulation. J Eur Acad Dermatol Venereol 2024; 38:1419-1431. [PMID: 38450801 DOI: 10.1111/jdv.19933] [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: 09/21/2023] [Accepted: 01/22/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND The limited therapies available for treating Merkel cell carcinoma (MCC), a highly aggressive skin neoplasm, still pose clinical challenges, and novel treatments are required. Targeting retinoid signalling with retinoids, such as all-trans retinoic acid (ATRA), is a promising and clinically useful antitumor approach. ATRA drives tumour cell differentiation by modulating retinoid signalling, leading to anti-proliferative and pro-apoptotic effects. Although retinoid signalling is dysregulated in MCC, ATRA activity in this tumour is unknown. This study aimed to evaluate the impact of ATRA on the pathological phenotype of MCC cells. METHODS The effect of ATRA was tested in various Merkel cell polyomavirus-positive and polyomavirus-negative MCC cell lines in terms of cell proliferation, viability, migration and clonogenic abilities. In addition, cell cycle, apoptosis/cell death and the retinoid gene signature were evaluated upon ATRA treatments. RESULTS ATRA efficiently impaired MCC cell proliferation and viability in MCC cells. A strong effect in reducing cell migration and clonogenicity was determined in ATRA-treated cells. Moreover, ATRA resulted as strongly effective in arresting cell cycle and inducing apoptosis/cell death in all tested MCC cells. Enrichment analyses indicated that ATRA was effective in modulating the retinoid gene signature in MCC cells to promote cell differentiation pathways, which led to anti-proliferative and pro-apoptotic/cell death effects. CONCLUSIONS These results underline the potential of retinoid-based therapy for MCC management and might open the way to novel experimental approaches with other retinoids and/or combinatorial treatments.
Collapse
Affiliation(s)
- Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- Center for Studies on Gender Medicine-Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Giada Badiale
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | | | - Giulia Di Mauro
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Antoine Touzé
- Biologie des infections à Polyomavirus team, UMR INRA ISP 1282, University of Tours, Tours, France
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- Center for Studies on Gender Medicine-Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- Center for Studies on Gender Medicine-Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
4
|
Durand MA, Drouin A, Bachiri K, Durand L, Berthon P, Houben R, Schrama D, Coyaud E, Samimi M, Touzé A, Kervarrec T. Enhancer of Zeste Homolog 2 Inhibition Induces HLA Class I Re-Expression in Merkel Cell Carcinoma. J Invest Dermatol 2024; 144:1398-1401.e1. [PMID: 38052265 DOI: 10.1016/j.jid.2023.10.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 10/13/2023] [Accepted: 10/31/2023] [Indexed: 12/07/2023]
Affiliation(s)
- Marie-Alice Durand
- Team "Biologie des Infections à Polyomavirus", UMR INRAE ISP 1282, University of Tours, Tours, France
| | - Aurélie Drouin
- Team "Biologie des Infections à Polyomavirus", UMR INRAE ISP 1282, University of Tours, Tours, France
| | - Kamel Bachiri
- Department of Biology, Univ. Lille, Inserm, CHU Lille, U1192-Protéomique Réponse Inflammatoire Spectrométrie de Masse (PRISM), Lille, France
| | - Laurine Durand
- Team "Biologie des Infections à Polyomavirus", UMR INRAE ISP 1282, University of Tours, Tours, France
| | - Patricia Berthon
- Team "Biologie des Infections à Polyomavirus", UMR INRAE ISP 1282, University of Tours, Tours, France
| | - Roland Houben
- Department of Dermatology, Venerology and Allergology, University Hospital of Würzburg, Würzburg, Germany
| | - David Schrama
- Department of Dermatology, Venerology and Allergology, University Hospital of Würzburg, Würzburg, Germany
| | - Etienne Coyaud
- Department of Biology, Univ. Lille, Inserm, CHU Lille, U1192-Protéomique Réponse Inflammatoire Spectrométrie de Masse (PRISM), Lille, France
| | - Mahtab Samimi
- Team "Biologie des Infections à Polyomavirus", UMR INRAE ISP 1282, University of Tours, Tours, France; Department of Dermatology, University Hospital Center (CHRU), University of Tours, Chambray-les-Tours, France
| | - Antoine Touzé
- Team "Biologie des Infections à Polyomavirus", UMR INRAE ISP 1282, University of Tours, Tours, France
| | - Thibault Kervarrec
- Team "Biologie des Infections à Polyomavirus", UMR INRAE ISP 1282, University of Tours, Tours, France; Department of Pathology, University Hospital Center (CHRU), University of Tours, Chambray-les-Tours, France.
| |
Collapse
|
5
|
Durand MA, Drouin A, Mouchard A, Durand L, Esnault C, Berthon P, Tallet A, Le Corre Y, Hainaut-Wierzbicka E, Blom A, Saiag P, Beneton N, Bens G, Nardin C, Aubin F, Dinulescu M, Collin C, Fromont-Hankard G, Cribier B, Laurent-Roussel S, Cokelaere K, Houben R, Schrama D, Peixoto P, Hervouet E, Bachiri K, Kantar D, Coyaud E, Guyétant S, Samimi M, Touzé A, Kervarrec T. Distinct Regulation of EZH2 and its Repressive H3K27me3 Mark in Polyomavirus-Positive and -Negative Merkel Cell Carcinoma. J Invest Dermatol 2023; 143:1937-1946.e7. [PMID: 37037414 DOI: 10.1016/j.jid.2023.02.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 04/12/2023]
Abstract
Merkel cell carcinoma (MCC) is an aggressive skin cancer for which Merkel cell polyomavirus integration and expression of viral oncogenes small T and Large T have been identified as major oncogenic determinants. Recently, a component of the PRC2 complex, the histone methyltransferase enhancer of zeste homolog 2 (EZH2) that induces H3K27 trimethylation as a repressive mark has been proposed as a potential therapeutic target in MCC. Because divergent results have been reported for the levels of EZH2 and trimethylation of lysine 27 on histone 3, we analyzed these factors in a large MCC cohort to identify the molecular determinants of EZH2 activity in MCC and to establish MCC cell lines' sensitivity to EZH2 inhibitors. Immunohistochemical expression of EZH2 was observed in 92% of MCC tumors (156 of 170), with higher expression levels in virus-positive than virus-negative tumors (P = 0.026). For the latter, we showed overexpression of EZHIP, a negative regulator of the PRC2 complex. In vitro, ectopic expression of the large T antigen in fibroblasts led to the induction of EZH2 expression, whereas the knockdown of T antigens in MCC cell lines resulted in decreased EZH2 expression. EZH2 inhibition led to selective cytotoxicity on virus-positive MCC cell lines. This study highlights the distinct mechanisms of EZH2 induction between virus-negative and -positive MCC.
Collapse
Affiliation(s)
- Marie-Alice Durand
- Team "Biologie des Infections à Polyomavirus", ISP UMR 1282, INRAE, University of Tours, Tours, France
| | - Aurélie Drouin
- Team "Biologie des Infections à Polyomavirus", ISP UMR 1282, INRAE, University of Tours, Tours, France
| | - Alice Mouchard
- Team "Biologie des Infections à Polyomavirus", ISP UMR 1282, INRAE, University of Tours, Tours, France; Department of Dermatology, CHRU of Tours, University of Tours, Chambray-lès-Tours, France
| | - Laurine Durand
- Team "Biologie des Infections à Polyomavirus", ISP UMR 1282, INRAE, University of Tours, Tours, France
| | - Clara Esnault
- Team "Biologie des Infections à Polyomavirus", ISP UMR 1282, INRAE, University of Tours, Tours, France
| | - Patricia Berthon
- Team "Biologie des Infections à Polyomavirus", ISP UMR 1282, INRAE, University of Tours, Tours, France
| | - Anne Tallet
- Platform of Somatic Tumor Molecular Genetics, CHU of Tours, University of Tours, Tours, France
| | - Yannick Le Corre
- Dermatology Department, CHU of Angers, LUNAM University, Angers, France
| | | | - Astrid Blom
- Department of General and Oncologic Dermatology, Ambroise-Paré Hospital, CARADERM Network, Boulogne-Billancourt, France; Research unit EA 4340, University of Versailles-Saint-Quentin-en-Yvelines, Paris-Saclay University, Boulogne-Billancourt, France
| | - Philippe Saiag
- Department of General and Oncologic Dermatology, Ambroise-Paré Hospital, CARADERM Network, Boulogne-Billancourt, France; Research unit EA 4340, University of Versailles-Saint-Quentin-en-Yvelines, Paris-Saclay University, Boulogne-Billancourt, France
| | - Nathalie Beneton
- Dermatology Department, CHU of Le Mans, University of Le Mans, Le Mans, France
| | - Guido Bens
- Dermatology department, CHR Orleans, Orleans, France
| | - Charlee Nardin
- Dermatology, CHU Besançon, Besançon, France; INSERM 1098, Université Bourgogne Franche-Comté, Besançon, France
| | - François Aubin
- Dermatology, CHU Besançon, Besançon, France; INSERM 1098, Université Bourgogne Franche-Comté, Besançon, France
| | - Monica Dinulescu
- Dermatology department, CHU Rennes, Institut Dermatologique du Grand Ouest (IDGO), Rennes, France
| | - Christine Collin
- Platform of Somatic Tumor Molecular Genetics, CHU of Tours, University of Tours, Tours, France
| | | | - Bernard Cribier
- Dermatology Department, CHU of Strasbourg, University of Strasbourg, Strasbourg, France
| | | | | | - Roland Houben
- Department of Dermatology, Venerology and Allergology, University Hospital of Würzburg, Würzburg, Germany
| | - David Schrama
- Department of Dermatology, Venerology and Allergology, University Hospital of Würzburg, Würzburg, Germany
| | - Paul Peixoto
- INSERM, EFS-BFC, UMR 1098 RIGHT, University Bourgogne-Franche-Comté, Besançon, France; EPIgenetics and GENe Expression Technical Platform (EPIGENExp), University Bourgogne Franche-Comté, Besançon, France
| | - Eric Hervouet
- INSERM, EFS-BFC, UMR 1098 RIGHT, University Bourgogne-Franche-Comté, Besançon, France; EPIgenetics and GENe Expression Technical Platform (EPIGENExp), University Bourgogne Franche-Comté, Besançon, France
| | - Kamel Bachiri
- Department of Biology, Inserm U1192, Protéomique Réponse Inflammatoire Spectrométrie de Masse-PRISM, CHU Lille, Université de Lille, Lille, France
| | - Diala Kantar
- Department of Biology, Inserm U1192, Protéomique Réponse Inflammatoire Spectrométrie de Masse-PRISM, CHU Lille, Université de Lille, Lille, France
| | - Etienne Coyaud
- Department of Biology, Inserm U1192, Protéomique Réponse Inflammatoire Spectrométrie de Masse-PRISM, CHU Lille, Université de Lille, Lille, France
| | - Serge Guyétant
- Pathology Department, CHU of Tours, University of Tours, Tours, France; Team "Biologie des Infections à Polyomavirus", ISP UMR 1282, INRAE, University of Tours, Tours, France
| | - Mahtab Samimi
- Team "Biologie des Infections à Polyomavirus", ISP UMR 1282, INRAE, University of Tours, Tours, France; Department of Dermatology, CHRU of Tours, University of Tours, Chambray-lès-Tours, France
| | - Antoine Touzé
- Team "Biologie des Infections à Polyomavirus", ISP UMR 1282, INRAE, University of Tours, Tours, France
| | - Thibault Kervarrec
- Team "Biologie des Infections à Polyomavirus", ISP UMR 1282, INRAE, University of Tours, Tours, France; Pathology Department, CHU of Tours, University of Tours, Tours, France.
| |
Collapse
|
6
|
Srinivas N, Song L, Lei KC, Gravemeyer J, Furtmann F, Gambichler T, Becker JC, Sriram A. The HDAC inhibitor domatinostat induces type I interferon α in Merkel cell carcinoma by HES1 repression. J Cancer Res Clin Oncol 2023; 149:8267-8277. [PMID: 37071208 PMCID: PMC10374800 DOI: 10.1007/s00432-023-04733-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND Class I selective histone deacetylase inhibitors (HDACi) have been previously demonstrated to not only increase major histocompatibility complex class I surface expression in Merkel cell carcinoma (MCC) cells by restoring the antigen processing and presentation machinery, but also exert anti-tumoral effect by inducing apoptosis. Both phenomena could be due to induction of type I interferons (IFN), as has been described for HDACi. However, the mechanism of IFN induction under HDACi is not fully understood because the expression of IFNs is regulated by both activating and inhibitory signaling pathways. Our own preliminary observations suggest that this may be caused by suppression of HES1. METHODS The effect of the class I selective HDACi domatinostat and IFNα on cell viability and the apoptosis of MCPyV-positive (WaGa, MKL-1) and -negative (UM-MCC 34) MCC cell lines, as well as, primary fibroblasts were assessed by colorimetric methods or measuring mitochondrial membrane potential and intracellular caspase-3/7, respectively. Next, the impact of domatinostat on IFNA and HES1 mRNA expression was measured by RT-qPCR; intracellular IFNα production was detected by flow cytometry. To confirm that the expression of IFNα induced by HDACi was due to the suppression of HES1, it was silenced by RNA interference and then mRNA expression of IFNA and IFN-stimulated genes was assessed. RESULTS Our studies show that the previously reported reduction in viability of MCC cell lines after inhibition of HDAC by domatinostat is accompanied by an increase in IFNα expression, both of mRNA and at the protein level. We confirmed that treatment of MCC cells with external IFNα inhibited their proliferation and induced apoptosis. Re-analysis of existing single-cell RNA sequencing data indicated that induction of IFNα by domatinostat occurs through repression of HES1, a transcriptional inhibitor of IFNA; this was confirmed by RT-qPCR. Finally, siRNA-mediated silencing of HES1 in the MCC cell line WaGa not only increased mRNA expression of IFNA and IFN-stimulated genes but also decreased cell viability. CONCLUSION Our results demonstrate that the direct anti-tumor effect of HDACi domatinostat on MCC cells is at least in part mediated via decreased HES1 expression allowing the induction of IFNα, which in turn causes apoptosis.
Collapse
Affiliation(s)
- Nalini Srinivas
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Lina Song
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kuan Cheok Lei
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Gravemeyer
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frauke Furtmann
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thilo Gambichler
- Skin Cancer Center, Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - Jürgen C. Becker
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Ashwin Sriram
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
7
|
Celikdemir B, Houben R, Kervarrec T, Samimi M, Schrama D. Current and preclinical treatment options for Merkel cell carcinoma. Expert Opin Biol Ther 2023; 23:1015-1034. [PMID: 37691397 DOI: 10.1080/14712598.2023.2257603] [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: 03/28/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Merkel cell carcinoma (MCC) is a rare, highly aggressive form of skin cancer with neuroendocrine features. The origin of this cancer is still unclear, but research in the last 15 years has demonstrated that MCC arises via two distinct etiologic pathways, i.e. virus and UV-induced. Considering the high mortality rate and the limited therapeutic options available, this review aims to highlight the significance of MCC research and the need for advancement in MCC treatment. AREAS COVERED With the advent of the immune checkpoint inhibitor therapies, we now have treatment options providing a survival benefit for patients with advanced MCC. However, the issue of primary and acquired resistance to these therapies remains a significant concern. Therefore, ongoing efforts seeking additional therapeutic targets and approaches for MCC therapy are a necessity. Through a comprehensive literature search, we provide an overview on recent preclinical and clinical studies with respect to MCC therapy. EXPERT OPINION Currently, the only evidence-based therapy for MCC is immune checkpoint blockade with anti-PD-1/PD-L1 for advanced patients. Neoadjuvant, adjuvant and combined immune checkpoint blockade are promising treatment options.
Collapse
Affiliation(s)
- Büke Celikdemir
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Roland Houben
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Thibault Kervarrec
- Department of Pathology, Centre Hospitalier Universitaire De Tours, Tours, France
| | - Mahtab Samimi
- Department of Dermatology, University Hospital of Tours, Tours, France
| | - David Schrama
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
8
|
Robb TJ, Ward Z, Houseman P, Woodhouse B, Patel R, Fitzgerald S, Tsai P, Lawrence B, Parker K, Print CG, Blenkiron C. Chromosomal Aberrations Accumulate during Metastasis of Virus-Negative Merkel Cell Carcinoma. J Invest Dermatol 2023; 143:1168-1177.e2. [PMID: 36736454 DOI: 10.1016/j.jid.2023.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/13/2022] [Accepted: 01/01/2023] [Indexed: 02/04/2023]
Abstract
Merkel cell carcinoma is a rare, aggressive skin tumor initiated by polyomavirus integration or UV light DNA damage. In New Zealand, there is a propensity toward the UV-driven form (31 of 107, 29% virus positive). Using archival formalin-fixed, paraffin-embedded tissues, we report targeted DNA sequencing covering 246 cancer genes on 71 tumor tissues and 38 nonmalignant tissues from 37 individuals, with 33 of 37 being negative for the virus. Somatic variants of New Zealand virus-negative Merkel cell carcinomas partially overlapped with those reported overseas, including TP53 variants in all tumors and RB1, LRP1B, NOTCH1, and EPHA3/7 variants each found in over half of the cohort. Variants in genes not analyzed or reported in previous studies were also found. Cataloging variants in TP53 and RB1 from published datasets revealed a broad distribution across these genes. Chr 1p gain and Chr 3p loss were identified in around 50% of New Zealand virus-negative Merkel cell carcinomas, and RB1 loss of heterozygosity was found in 90% of cases. Copy number variants accumulate in most metastases. Virus-negative Merkel cell carcinomas have complex combinations of somatic DNA-sequence variants and copy number variants. They likely carry the small genomic changes permissive for metastasis from early tumor development; however, chromosomal alterations may contribute to driving metastatic progression.
Collapse
Affiliation(s)
- Tamsin J Robb
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Hosted by the University of Auckland, Auckland, New Zealand
| | - Zoe Ward
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Pascalene Houseman
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Braden Woodhouse
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Department of Oncology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Rachna Patel
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sandra Fitzgerald
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Hosted by the University of Auckland, Auckland, New Zealand
| | - Peter Tsai
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Hosted by the University of Auckland, Auckland, New Zealand
| | - Ben Lawrence
- Maurice Wilkins Centre for Molecular Biodiscovery, Hosted by the University of Auckland, Auckland, New Zealand; Department of Oncology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kate Parker
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Department of Oncology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Cristin G Print
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Hosted by the University of Auckland, Auckland, New Zealand
| | - Cherie Blenkiron
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Hosted by the University of Auckland, Auckland, New Zealand; Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
9
|
MacLennan SA, Marra MA. Oncogenic Viruses and the Epigenome: How Viruses Hijack Epigenetic Mechanisms to Drive Cancer. Int J Mol Sci 2023; 24:ijms24119543. [PMID: 37298494 DOI: 10.3390/ijms24119543] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Globally, viral infections substantially contribute to cancer development. Oncogenic viruses are taxonomically heterogeneous and drive cancers using diverse strategies, including epigenomic dysregulation. Here, we discuss how oncogenic viruses disrupt epigenetic homeostasis to drive cancer and focus on how virally mediated dysregulation of host and viral epigenomes impacts the hallmarks of cancer. To illustrate the relationship between epigenetics and viral life cycles, we describe how epigenetic changes facilitate the human papillomavirus (HPV) life cycle and how changes to this process can spur malignancy. We also highlight the clinical impact of virally mediated epigenetic changes on cancer diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Signe A MacLennan
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - Marco A Marra
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| |
Collapse
|
10
|
Gravemeyer J, Spassova I, Verhaegen ME, Dlugosz AA, Hoffmann D, Lange A, Becker JC. DNA-methylation patterns imply a common cellular origin of virus- and UV-associated Merkel cell carcinoma. Oncogene 2022; 41:37-45. [PMID: 34667274 PMCID: PMC8724008 DOI: 10.1038/s41388-021-02064-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023]
Abstract
Merkel cell carcinoma (MCC) is a neuroendocrine tumor either induced by integration of the Merkel cell polyomavirus into the cell genome or by accumulation of UV-light-associated mutations (VP-MCC and UV-MCC). Whether VP- and UV-MCC have the same or different cellular origins is unclear; with mesenchymal or epidermal origins discussed. DNA-methylation patterns have a proven utility in determining cellular origins of cancers. Therefore, we used this approach to uncover evidence regarding the cell of origin of classical VP- and UV-MCC cell lines, i.e., cell lines with a neuroendocrine growth pattern (n = 9 and n = 4, respectively). Surprisingly, we observed high global similarities in the DNA-methylation of UV- and VP-MCC cell lines. CpGs of lower methylation in VP-MCC cell lines were associated with neuroendocrine marker genes such as SOX2 and INSM1, or linked to binding sites of EZH2 and SUZ12 of the polycomb repressive complex 2, i.e., genes with an impact on carcinogenesis and differentiation of neuroendocrine cancers. Thus, the observed differences appear to be rooted in viral compared to mutation-driven carcinogenesis rather than distinct cells of origin. To test this hypothesis, we used principal component analysis, to compare DNA-methylation data from different epithelial and non-epithelial neuroendocrine cancers and established a scoring model for epithelial and neuroendocrine characteristics. Subsequently, we applied this scoring model to the DNA-methylation data of the VP- and UV-MCC cell lines, revealing that both clearly scored as epithelial cancers. In summary, our comprehensive analysis of DNA-methylation suggests a common epithelial origin of UV- and VP-MCC cell lines.
Collapse
Affiliation(s)
- Jan Gravemeyer
- Translational Skin Cancer Research (TSCR), University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK) & German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ivelina Spassova
- Translational Skin Cancer Research (TSCR), University Duisburg-Essen, Essen, Germany
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | | | - Andrzej A Dlugosz
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel Hoffmann
- Bioinformatics & Computational Biophysics, University Duisburg-Essen, Essen, Germany
| | - Anja Lange
- Bioinformatics & Computational Biophysics, University Duisburg-Essen, Essen, Germany
| | - Jürgen C Becker
- Translational Skin Cancer Research (TSCR), University Duisburg-Essen, Essen, Germany.
- German Cancer Consortium (DKTK) & German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Dermatology, University Hospital Essen, Essen, Germany.
| |
Collapse
|