1
|
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
|
2
|
Curcio A, Rocca R, Alcaro S, Artese A. The Histone Deacetylase Family: Structural Features and Application of Combined Computational Methods. Pharmaceuticals (Basel) 2024; 17:620. [PMID: 38794190 PMCID: PMC11124352 DOI: 10.3390/ph17050620] [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: 04/18/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Histone deacetylases (HDACs) are crucial in gene transcription, removing acetyl groups from histones. They also influence the deacetylation of non-histone proteins, contributing to the regulation of various biological processes. Thus, HDACs play pivotal roles in various diseases, including cancer, neurodegenerative disorders, and inflammatory conditions, highlighting their potential as therapeutic targets. This paper reviews the structure and function of the four classes of human HDACs. While four HDAC inhibitors are currently available for treating hematological malignancies, numerous others are undergoing clinical trials. However, their non-selective toxicity necessitates ongoing research into safer and more efficient class-selective or isoform-selective inhibitors. Computational techniques have greatly facilitated the discovery of HDAC inhibitors that achieve the desired potency and selectivity. These techniques encompass ligand-based strategies such as scaffold hopping, pharmacophore modeling, three-dimensional quantitative structure–activity relationships (3D-QSAR), and structure-based virtual screening (molecular docking). Additionally, advancements in molecular dynamics simulations, along with Poisson–Boltzmann/molecular mechanics generalized Born surface area (PB/MM-GBSA) methods, have enhanced the accuracy of predicting ligand binding affinity. In this review, we delve into the ways in which these methods have contributed to designing and identifying HDAC inhibitors.
Collapse
Affiliation(s)
- Antonio Curcio
- Dipartimento di Scienze della Salute, Campus “S. Venuta”, Università degli Studi “Magna Græcia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (A.C.); (S.A.); (A.A.)
| | - Roberta Rocca
- Dipartimento di Scienze della Salute, Campus “S. Venuta”, Università degli Studi “Magna Græcia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (A.C.); (S.A.); (A.A.)
- Net4Science S.r.l., Università degli Studi “Magna Græcia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Campus “S. Venuta”, Università degli Studi “Magna Græcia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (A.C.); (S.A.); (A.A.)
- Net4Science S.r.l., Università degli Studi “Magna Græcia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Anna Artese
- Dipartimento di Scienze della Salute, Campus “S. Venuta”, Università degli Studi “Magna Græcia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (A.C.); (S.A.); (A.A.)
- Net4Science S.r.l., Università degli Studi “Magna Græcia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| |
Collapse
|
3
|
Yang Q, Vafaei S, Falahati A, Khosh A, Bariani MV, Omran MM, Bai T, Siblini H, Ali M, He C, Boyer TG, Al-Hendy A. Bromodomain-Containing Protein 9 Regulates Signaling Pathways and Reprograms the Epigenome in Immortalized Human Uterine Fibroid Cells. Int J Mol Sci 2024; 25:905. [PMID: 38255982 PMCID: PMC10815284 DOI: 10.3390/ijms25020905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/26/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Bromodomain-containing proteins (BRDs) are involved in many biological processes, most notably epigenetic regulation of transcription, and BRD dysfunction has been linked to many diseases, including tumorigenesis. However, the role of BRDs in the pathogenesis of uterine fibroids (UFs) is entirely unknown. The present study aimed to determine the expression pattern of BRD9 in UFs and matched myometrium and further assess the impact of a BRD9 inhibitor on UF phenotype and epigenetic/epitranscriptomic changes. Our studies demonstrated that the levels of BRD9 were significantly upregulated in UFs compared to matched myometrium, suggesting that the aberrant BRD expression may contribute to the pathogenesis of UFs. We then evaluated the potential roles of BRD9 using its specific inhibitor, I-BRD9. Targeted inhibition of BRD9 suppressed UF tumorigenesis with increased apoptosis and cell cycle arrest, decreased cell proliferation, and extracellular matrix deposition in UF cells. The latter is the key hallmark of UFs. Unbiased transcriptomic profiling coupled with downstream bioinformatics analysis further and extensively demonstrated that targeted inhibition of BRD9 impacted the cell cycle- and ECM-related biological pathways and reprogrammed the UF cell epigenome and epitranscriptome in UFs. Taken together, our studies support the critical role of BRD9 in UF cells and the strong interconnection between BRD9 and other pathways controlling the UF progression. Targeted inhibition of BRDs might provide a non-hormonal treatment option for this most common benign tumor in women of reproductive age.
Collapse
Affiliation(s)
- Qiwei Yang
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (S.V.); (M.V.B.); (M.M.O.); (H.S.); (M.A.); (A.A.-H.)
| | - Somayeh Vafaei
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (S.V.); (M.V.B.); (M.M.O.); (H.S.); (M.A.); (A.A.-H.)
| | - Ali Falahati
- DNA GTx LAB, Dubai Healthcare City, Dubai 505262, United Arab Emirates;
| | - Azad Khosh
- Department of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.K.); (T.G.B.)
| | - Maria Victoria Bariani
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (S.V.); (M.V.B.); (M.M.O.); (H.S.); (M.A.); (A.A.-H.)
| | - Mervat M. Omran
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (S.V.); (M.V.B.); (M.M.O.); (H.S.); (M.A.); (A.A.-H.)
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Tao Bai
- Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Hiba Siblini
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (S.V.); (M.V.B.); (M.M.O.); (H.S.); (M.A.); (A.A.-H.)
| | - Mohamed Ali
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (S.V.); (M.V.B.); (M.M.O.); (H.S.); (M.A.); (A.A.-H.)
| | - Chuan He
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA;
| | - Thomas G. Boyer
- Department of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.K.); (T.G.B.)
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (S.V.); (M.V.B.); (M.M.O.); (H.S.); (M.A.); (A.A.-H.)
| |
Collapse
|
4
|
Azemin WA, Alias N, Ali AM, Shamsir MS. In silico analysis prediction of HepTH1-5 as a potential therapeutic agent by targeting tumour suppressor protein networks. J Biomol Struct Dyn 2023; 41:1141-1167. [PMID: 34935583 DOI: 10.1080/07391102.2021.2017349] [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: 01/18/2023]
Abstract
Many studies reported that the activation of tumour suppressor protein, p53 induced the human hepcidin expression. However, its expression decreased when p53 was silenced in human hepatoma cells. Contrary to Tilapia hepcidin TH1-5, HepTH1-5 was previously reported to trigger the p53 activation through the molecular docking approach. The INhibitor of Growth (ING) family members are also shown to directly interact with p53 and promote cell cycle arrest, senescence, apoptosis and participate in DNA replication and DNA damage responses to suppress the tumour initiation and progression. However, the interrelation between INGs and HepTH1-5 remains unknown. Therefore, this study aims to identify the mechanism and their protein interactions using in silico approaches. The finding revealed that HepTH1-5 and its ligands had interacted mostly on hotspot residues of ING proteins which involved in histone modifications via acetylation, phosphorylation, and methylation. This proves that HepTH1-5 might implicate in an apoptosis signalling pathway and preserve the protein structure and function of INGs by reducing the perturbation of histone binding upon oxidative stress response. This study would provide theoretical guidance for the design and experimental studies to decipher the role of HepTH1-5 as a potential therapeutic agent for cancer therapy. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Wan-Atirah Azemin
- Faculty of Bioresources and Food Industry, School of Agriculture Science and Biotechnology, Universiti Sultan Zainal Abidin, Besut, Malaysia.,Faculty of Science, Bioinformatics Research Group (BIRG), Department of Biosciences, Universiti Teknologi Malaysia, Skudai, Malaysia
| | - Nadiawati Alias
- Faculty of Bioresources and Food Industry, School of Agriculture Science and Biotechnology, Universiti Sultan Zainal Abidin, Besut, Malaysia
| | - Abdul Manaf Ali
- Faculty of Bioresources and Food Industry, School of Agriculture Science and Biotechnology, Universiti Sultan Zainal Abidin, Besut, Malaysia
| | - Mohd Shahir Shamsir
- Faculty of Science, Bioinformatics Research Group (BIRG), Department of Biosciences, Universiti Teknologi Malaysia, Skudai, Malaysia.,Faculty of Applied Sciences and Technology, Universiti Tun Hussein Onn Malaysia, Pagoh Higher Education Hub, Muar, Malaysia
| |
Collapse
|
5
|
Yang Q, Falahati A, Khosh A, Mohammed H, Kang W, Corachán A, Bariani MV, Boyer TG, Al-Hendy A. Targeting Class I Histone Deacetylases in Human Uterine Leiomyosarcoma. Cells 2022; 11:cells11233801. [PMID: 36497061 PMCID: PMC9735512 DOI: 10.3390/cells11233801] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/19/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Uterine leiomyosarcoma (uLMS) is the most frequent subtype of uterine sarcoma that presents a poor prognosis, high rates of recurrence, and metastasis. Currently, the molecular mechanism of the origin and development of uLMS is unknown. Class I histone deacetylases (including HDAC1, 2, 3, and 8) are one of the major classes of the HDAC family and catalyze the removal of acetyl groups from lysine residues in histones and cellular proteins. Class I HDACs exhibit distinct cellular and subcellular expression patterns and are involved in many biological processes and diseases through diverse signaling pathways. However, the link between class I HDACs and uLMS is still being determined. In this study, we assessed the expression panel of Class I HDACs in uLMS and characterized the role and mechanism of class I HDACs in the pathogenesis of uLMS. Immunohistochemistry analysis revealed that HDAC1, 2, and 3 are aberrantly upregulated in uLMS tissues compared to adjacent myometrium. Immunoblot analysis demonstrated that the expression levels of HDAC 1, 2, and 3 exhibited a graded increase from normal and benign to malignant uterine tumor cells. Furthermore, inhibition of HDACs with Class I HDACs inhibitor (Tucidinostat) decreased the uLMS proliferation in a dose-dependent manner. Notably, gene set enrichment analysis of differentially expressed genes (DEGs) revealed that inhibition of HDACs with Tucidinostat altered several critical pathways. Moreover, multiple epigenetic analyses suggested that Tucidinostat may alter the transcriptome via reprogramming the oncogenic epigenome and inducing the changes in microRNA-target interaction in uLMS cells. In the parallel study, we also determined the effect of DL-sulforaphane on the uLMS. Our study demonstrated the relevance of class I HDACs proteins in the pathogenesis of malignant uLMS. Further understanding the role and mechanism of HDACs in uLMS may provide a promising and novel strategy for treating patients with this aggressive uterine cancer.
Collapse
Affiliation(s)
- Qiwei Yang
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA
- Correspondence:
| | - Ali Falahati
- Department of Biology, Yazd University, Yazd 891581841, Iran
| | - Azad Khosh
- Department of Biology, Yazd University, Yazd 891581841, Iran
| | - Hanaa Mohammed
- Anatomy Department, Faculty of Medicine, Sohag University, Sohag 82524, Egypt
| | - Wenjun Kang
- Center for Research Informatics, University of Chicago, Chicago, IL 60637, USA
| | - Ana Corachán
- Department of Paediatrics, University of Valencia, Obstetrics and Gynecology, 46026 Valencia, Spain
| | | | - Thomas G. Boyer
- Department of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
6
|
Russi S, Sgambato A, Bochicchio AM, Zoppoli P, Aieta M, Capobianco AML, Ruggieri V, Zifarone E, Falco G, Laurino S. CHIR99021, trough GSK-3β Targeting, Reduces Epithelioid Sarcoma Cell Proliferation by Activating Mitotic Catastrophe and Autophagy. Int J Mol Sci 2021; 22:11147. [PMID: 34681807 PMCID: PMC8538073 DOI: 10.3390/ijms222011147] [Citation(s) in RCA: 8] [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: 09/22/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
Epithelioid sarcoma (ES) is a rare disease representing <1% of soft tissue sarcomas. Current therapies are based on anthracycline alone or in combination with ifosfamide or other cytotoxic drugs. ES is still characterized by a poor prognosis with high rates of recurrence. Indeed, for years, ES survival rates have remained stagnant, suggesting that conventional treatments should be revised and improved. New therapeutic approaches are focused to target the key regulators of signaling pathways, the causative markers of tumor pathophysiology. To this end, we selected, among the drugs to which an ES cell line is highly sensitive, those that target signaling pathways known to be dysregulated in ES. In particular, we found a key role for GSK-3β, which results in up-regulation in tumor versus normal tissue samples and associated to poor prognosis in sarcoma patients. Following this evidence, we evaluated CHIR99021, a GSK-3 inhibitor, as a potential drug for use in ES therapy. Our data highlight that, in ES cells, CHIR99021 induces cell cycle arrest, mitotic catastrophe (MC) and autophagic response, resulting in reduced cell proliferation. Our results support the potential efficacy of CHIR99021 in ES treatment and encourage further preclinical and clinical studies.
Collapse
Affiliation(s)
- Sabino Russi
- IRCCS CROB—Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy; (S.R.); (A.S.); (A.M.B.); (P.Z.); (M.A.); (A.M.L.C.); (V.R.); (E.Z.); (S.L.)
| | - Alessandro Sgambato
- IRCCS CROB—Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy; (S.R.); (A.S.); (A.M.B.); (P.Z.); (M.A.); (A.M.L.C.); (V.R.); (E.Z.); (S.L.)
| | - Anna Maria Bochicchio
- IRCCS CROB—Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy; (S.R.); (A.S.); (A.M.B.); (P.Z.); (M.A.); (A.M.L.C.); (V.R.); (E.Z.); (S.L.)
| | - Pietro Zoppoli
- IRCCS CROB—Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy; (S.R.); (A.S.); (A.M.B.); (P.Z.); (M.A.); (A.M.L.C.); (V.R.); (E.Z.); (S.L.)
| | - Michele Aieta
- IRCCS CROB—Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy; (S.R.); (A.S.); (A.M.B.); (P.Z.); (M.A.); (A.M.L.C.); (V.R.); (E.Z.); (S.L.)
| | - Alba Maria Lucia Capobianco
- IRCCS CROB—Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy; (S.R.); (A.S.); (A.M.B.); (P.Z.); (M.A.); (A.M.L.C.); (V.R.); (E.Z.); (S.L.)
| | - Vitalba Ruggieri
- IRCCS CROB—Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy; (S.R.); (A.S.); (A.M.B.); (P.Z.); (M.A.); (A.M.L.C.); (V.R.); (E.Z.); (S.L.)
- UOC Clinical Pathology, Altamura Hospital, 70022 Altamura, Italy
| | - Emanuela Zifarone
- IRCCS CROB—Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy; (S.R.); (A.S.); (A.M.B.); (P.Z.); (M.A.); (A.M.L.C.); (V.R.); (E.Z.); (S.L.)
| | - Geppino Falco
- Department of Biology, University of Naples Federico II, 80133 Naples, Italy
- Biogem—Istituto di Biologia e Genetica Molecolare, 83031 Ariano Irpino, Italy
| | - Simona Laurino
- IRCCS CROB—Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy; (S.R.); (A.S.); (A.M.B.); (P.Z.); (M.A.); (A.M.L.C.); (V.R.); (E.Z.); (S.L.)
| |
Collapse
|
7
|
Harttrampf AC, da Costa MEM, Renoult A, Daudigeos-Dubus E, Geoerger B. Histone deacetylase inhibitor panobinostat induces antitumor activity in epithelioid sarcoma and rhabdoid tumor by growth factor receptor modulation. BMC Cancer 2021; 21:833. [PMID: 34281526 PMCID: PMC8290558 DOI: 10.1186/s12885-021-08579-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 07/10/2021] [Indexed: 01/06/2023] Open
Abstract
Background Epithelioid sarcomas and rhabdoid tumors are rare, aggressive malignancies with poor prognosis. Both are characterized by INI1 alterations and deregulation of growth factor receptors albeit their interaction has not been elucidated. Methods In this study, we investigated the activity of a panel of epigenetic modulators and receptor tyrosine kinase inhibitors in vitro on respective cell lines as well as on primary patient-derived epithelioid sarcoma cells, and in vivo on xenografted mice. Focusing on histone deacetylase (HDAC) inhibitors, we studied the mechanism of action of this class of agents, its effect on growth factor receptor regulation, and changes in epithelial-to-mesenchymal transition by using cell- and RT-qPCR-based assays. Results Pan-HDAC inhibitor panobinostat exhibited potent anti-proliferative activity at low nanomolar concentrations in A204 rhabdoid tumor, and VAESBJ/GRU1 epithelioid sarcoma cell lines, strongly induced apoptosis, and resulted in significant tumor growth inhibition in VAESBJ xenografts. It differentially regulated EGFR, FGFR1 and FGFR2, leading to downregulation of EGFR in epithelioid sarcoma and to mesenchymal-to-epithelial transition whereas in rhabdoid tumor cells, EGFR was strongly upregulated and reinforced the mesenchymal phenotype. All three cell lines were rendered more susceptible towards combination with EGFF inhibitor erlotinib, further enhancing apoptosis. Conclusions HDAC inhibitors exhibit significant anticancer activity due to their multifaceted actions on cytotoxicity, differentiation and drug sensitization. Our data suggest that the tailored, tissue-specific combination of HDAC inhibitors with therapeutics which target cellular salvage mechanisms might increase their therapeutic relevance. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08579-w.
Collapse
Affiliation(s)
- Anne Catherine Harttrampf
- Gustave Roussy Cancer Center, INSERM U1015, Université Paris-Saclay, Villejuif, France.,Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | | | - Aline Renoult
- Gustave Roussy Cancer Center, INSERM U1015, Université Paris-Saclay, Villejuif, France.,Present address: Institute of Research in Immunology and Cancer, Dr Trang Hoang Laboratory, Université de Montréal, Montreal, Québec, Canada
| | - Estelle Daudigeos-Dubus
- Gustave Roussy Cancer Center, INSERM U1015, Université Paris-Saclay, Villejuif, France.,Present address: AP-HP Nord, DMU Neurosciences, Service de Neurologie, FHU NeuroVasc, Université de Paris, Paris, France
| | - Birgit Geoerger
- Gustave Roussy Cancer Center, INSERM U1015, Université Paris-Saclay, Villejuif, France. .,Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, 114 Rue Edouard Vaillant, 94805, Villejuif, France.
| |
Collapse
|
8
|
Duan YC, Zhang SJ, Shi XJ, Jin LF, Yu T, Song Y, Guan YY. Research progress of dual inhibitors targeting crosstalk between histone epigenetic modulators for cancer therapy. Eur J Med Chem 2021; 222:113588. [PMID: 34107385 DOI: 10.1016/j.ejmech.2021.113588] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/09/2021] [Accepted: 05/25/2021] [Indexed: 12/13/2022]
Abstract
Abnormal epigenetics is a critical hallmark of human cancers. Anticancer drug discovery directed at histone epigenetic modulators has gained impressive advances with six drugs available for cancer therapy and numerous other candidates undergoing clinical trials. However, limited therapeutic profile, drug resistance, narrow safety margin, and dose-limiting toxicities pose intractable challenges for their clinical utility. Because histone epigenetic modulators undergo intricate crosstalk and act cooperatively to shape an aberrant epigenetic profile, co-targeting histone epigenetic modulators with a different mechanism of action has rapidly emerged as an attractive strategy to overcome the limitations faced by the single-target epigenetic inhibitors. In this review, we summarize in detail the crosstalk of histone epigenetic modulators in regulating gene transcription and the progress of dual epigenetic inhibitors targeting this crosstalk.
Collapse
Affiliation(s)
- Ying-Chao Duan
- School of Pharmacy, Xinxiang Medical University, 453003, Xinxiang, Henan Province, PR China.
| | - Shao-Jie Zhang
- School of Pharmacy, Xinxiang Medical University, 453003, Xinxiang, Henan Province, PR China
| | - Xiao-Jing Shi
- Laboratory Animal Center, Academy of Medical Science, Zhengzhou University, 450052, Zhengzhou, Henan Province, PR China
| | - Lin-Feng Jin
- School of Pharmacy, Xinxiang Medical University, 453003, Xinxiang, Henan Province, PR China
| | - Tong Yu
- School of Pharmacy, Xinxiang Medical University, 453003, Xinxiang, Henan Province, PR China
| | - Yu Song
- School of Pharmacy, Xinxiang Medical University, 453003, Xinxiang, Henan Province, PR China
| | - Yuan-Yuan Guan
- School of Pharmacy, Xinxiang Medical University, 453003, Xinxiang, Henan Province, PR China.
| |
Collapse
|
9
|
Epithelioid Sarcoma-From Genetics to Clinical Practice. Cancers (Basel) 2020; 12:cancers12082112. [PMID: 32751241 PMCID: PMC7463637 DOI: 10.3390/cancers12082112] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 11/20/2022] Open
Abstract
Epithelioid sarcoma is a mesenchymal soft tissue sarcoma often arising in the extremities, usually in young adults with a pick of incidence at 35 years of age. Epithelioid sarcoma (ES) is characterized by the loss of SMARCB1/INI1 (integrase interactor 1) or other proteins of the SWI/SNF complex. Two distinct types, proximal and distal, with varying biology and treatment outcomes, are distinguished. ES is known for aggressive behavior, including a high recurrence rate and regional lymph node metastases. An optimal long-term management strategy is still to be defined. The best treatment of localized ES is wide surgical resection. Neo-adjuvant or adjuvant radiotherapy may be recommended, as it reduces the local recurrence rate. Sentinel lymph node biopsy should be considered in ES patients. Patients with metastatic ES have a poor prognosis with an expected median overall survival of about a year. Doxorubicin-based regimens are recommended for advanced ES. Tazemetostat, an EZH2 methyltransferase, has shown promising results in ES patients. Novel therapies, including immunotherapy, are still needed.
Collapse
|
10
|
Li Y, Cao G, Tao X, Guo J, Wu S, Tao Y. Clinicopathologic features of epithelioid sarcoma: report of seventeen cases and review of literature. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:3042-3048. [PMID: 31934143 PMCID: PMC6949731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/26/2019] [Indexed: 06/10/2023]
Abstract
Epithelioid sarcoma (ES) is a rare malignant soft tissue tumor, which is characterized by nodular aggregates of epithelioid cells and immunoreactivity of cytokeratin (CK) as well as epithelial membrane antigen (EMA) and often with CD34. It can be divided into proximal and distal subtypes. Classic ES has a microscopic nodular appearance that is composed of large polygonal epithelioid cells combined with central necrosis, and presents as a subcutaneous or deep dermal mass in the distal extremities of young adults. The proximal variant preferentially occurs in proximal limbs and limb girdles and the midline of the trunk, and is composed of more atypical cells with variable rhabdoid morphology. In this study we investigated the clinicopathologic features of 17 patients diagnosed with ES. In addition, we reviewed relevant literature and discussed some diagnostic and differential diagnostic points of this disease.
Collapse
Affiliation(s)
- Yijie Li
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical CollegeBengbu, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeBengbu, Anhui Province, China
| | - Gaoxiang Cao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical CollegeBengbu, Anhui Province, China
| | - Xiaoying Tao
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical CollegeBengbu, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeBengbu, Anhui Province, China
| | - Jiannan Guo
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical CollegeBengbu, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeBengbu, Anhui Province, China
| | - Shiwu Wu
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical CollegeBengbu, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeBengbu, Anhui Province, China
| | - Yisheng Tao
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical CollegeBengbu, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeBengbu, Anhui Province, China
| |
Collapse
|
11
|
SARC018_SPORE02: Phase II Study of Mocetinostat Administered with Gemcitabine for Patients with Metastatic Leiomyosarcoma with Progression or Relapse following Prior Treatment with Gemcitabine-Containing Therapy. Sarcoma 2018; 2018:2068517. [PMID: 30473623 PMCID: PMC6220374 DOI: 10.1155/2018/2068517] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/07/2018] [Accepted: 10/02/2018] [Indexed: 02/07/2023] Open
Abstract
Histone deacetylase inhibitors (HDACi) can reverse chemoresistance, enhance chemotherapy-induced cytotoxicity, and reduce sarcoma proliferation in cell lines and animal models. We sought to determine the safety and toxicity of mocetinostat and its ability to reverse chemoresistance when administered with gemcitabine in patients with metastatic leiomyosarcoma resistant to prior gemcitabine-containing therapy. Participants with metastatic leiomyosarcoma received mocetinostat orally, 70 mg per day, three days per week, increasing to 90 mg after three weeks if well tolerated. Gemcitabine was administered at 1,000 mg/m2 intravenously at 10 mg/m2/minute on days five and 12 of every 21-day cycle. Disease response was evaluated with CT or MRI. Twenty participants with leiomyosarcoma were evaluated for toxicity. Median time to disease progression was 2.0 months (95% CI 1.54–3.12). Eighteen participants were evaluated for radiologic response by RECIST 1.1. Best responses included one PR and 12 SD. Tumor size reduced in 3 patients. Most common toxicities were fatigue, thrombocytopenia, anemia, nausea, and anorexia. One patient experienced a significant pericardial adverse event. No study-related deaths were observed. Rechallenging with gemcitabine by adding mocetinostat was feasible and demonstrated modest activity in patients with leiomyosarcoma. Further studies are needed to better define the role of HDAC inhibitors in patients with metastatic leiomyosarcoma.
Collapse
|
12
|
Histone deacetylase inhibitor ITF2357 leads to apoptosis and enhances doxorubicin cytotoxicity in preclinical models of human sarcoma. Oncogenesis 2018; 7:20. [PMID: 29472530 PMCID: PMC5833676 DOI: 10.1038/s41389-018-0026-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/26/2017] [Accepted: 11/09/2017] [Indexed: 12/14/2022] Open
Abstract
Sarcomas are rare tumors with generally poor prognosis, for which current therapies have shown limited efficacy. Histone deacetylase inhibitors (HDACi) are emerging anti-tumor agents; however, little is known about their effect in sarcomas. By using established and patient-derived sarcoma cells with different subtypes, we showed that the pan-HDACi, ITF2357, potently inhibited in vitro survival in a p53-independent manner. ITF2357-mediated cell death implied the activation of mitochondrial apoptosis, as attested by induction of pro-apoptotic BH3-only proteins and a caspases-dependent mechanism. ITF2357 also induced autophagy, which protected sarcoma cells from apoptotic cell death. ITF2357 activated forkhead box (FOXO) 1 and 3a transcription factors and their downstream target genes, however, silencing of both FOXO1 and 3a did not protect sarcoma cells against ITF2357-induced apoptosis and upregulated FOXO4 and 6. Notably, ITF2357 synergized with Doxorubicin to induce cell death of established and patient-derived sarcoma cells. Furthermore, combination treatment strongly impaired xenograft tumor growth in vivo, when compared to single treatments, suggesting that combination of ITF2357 with Doxorubicin has the potential to enhance sensitization in different preclinical models of sarcoma. Overall, our study highlights the therapeutic potential of ITF2357, alone or in rational combination therapies, for bone and soft tissue sarcomas management.
Collapse
|
13
|
Tang F, Choy E, Tu C, Hornicek F, Duan Z. Therapeutic applications of histone deacetylase inhibitors in sarcoma. Cancer Treat Rev 2017; 59:33-45. [PMID: 28732326 DOI: 10.1016/j.ctrv.2017.06.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 02/05/2023]
Abstract
Sarcomas are a rare group of malignant tumors originating from mesenchymal stem cells. Surgery, radiation and chemotherapy are currently the only standard treatments for sarcoma. However, their response rates to chemotherapy are quite low. Toxic side effects and multi-drug chemoresistance make treatment even more challenging. Therefore, better drugs to treat sarcomas are needed. Histone deacetylase inhibitors (HDAC inhibitors, HDACi, HDIs) are epigenetic modifying agents that can inhibit sarcoma growth in vitro and in vivo through a variety of pathways, including inducing tumor cell apoptosis, causing cell cycle arrest, impairing tumor invasion and preventing metastasis. Importantly, preclinical studies have revealed that HDIs can not only sensitize sarcomas to chemotherapy and radiotherapy, but also increase treatment responses when combined with other chemotherapeutic drugs. Several phase I and II clinical trials have been conducted to assess the efficacy of HDIs either as monotherapy or in combination with standard chemotherapeutic agents or targeted therapeutic drugs for sarcomas. Combination regimen for sarcomas appear to be more promising than monotherapy when using HDIs. This review summarizes our current understanding and therapeutic applications of HDIs in sarcomas.
Collapse
Affiliation(s)
- Fan Tang
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, USA; Department of Orthopedics, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, Sichuan 610041, China
| | - Edwin Choy
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, USA
| | - Chongqi Tu
- Department of Orthopedics, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, Sichuan 610041, China
| | - Francis Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, USA
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, USA.
| |
Collapse
|
14
|
|
15
|
Steppan DA, Pratilas CA, Loeb DM. Targeted therapy for soft tissue sarcomas in adolescents and young adults. Adolesc Health Med Ther 2017; 8:41-55. [PMID: 28408855 PMCID: PMC5384699 DOI: 10.2147/ahmt.s70377] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Soft tissue sarcomas (STSs) are a heterogeneous group of tumors originating from the mesenchyme. Even though they affect individuals in all age groups, the prevalence of subtypes of STSs changes significantly from childhood through adolescence into adulthood. The mainstay of therapy is surgery, with or without the addition of chemotherapy and/or radiation therapy. These treatment modalities are associated, in many cases, with significant morbidity and, given the heterogeneity of tumor histologies encompassed by the term "STS", have not uniformly improved outcomes. Moreover, some subgroups of STSs appear to be more, and others less, responsive to conventional chemotherapy agents. Over the last two decades, our understanding of the biology of STSs is slowly increasing, allowing for the development of more targeted therapies. We review the new treatment modalities that have been tested on patients with STSs, with a special focus on adolescents and young adults, a group of patients that is often underrepresented in clinical trials and has not received the dedicated attention it deserves, given the significant differences in biology and treatment response in comparison to children and adults.
Collapse
Affiliation(s)
- Diana A Steppan
- Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christine A Pratilas
- Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David M Loeb
- Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Zewdu A, Lopez G, Braggio D, Kenny C, Constantino D, Bid HK, Batte K, Iwenofu OH, Oberlies NH, Pearce CJ, Strohecker AM, Lev D, Pollock RE. Verticillin A Inhibits Leiomyosarcoma and Malignant Peripheral Nerve Sheath Tumor Growth via Induction of Apoptosis. ACTA ACUST UNITED AC 2016; 6. [PMID: 28184331 PMCID: PMC5295762 DOI: 10.4172/2161-1459.1000221] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Objective The heterogeneity of soft tissue sarcoma (STS) represents a major challenge for the development of effective therapeutics. Comprised of over 50 different histology subtypes of various etiologies, STS subsets are further characterized as either karyotypically simple or complex. Due to the number of genetic anomalies associated with genetically complex STS, development of therapies demonstrating potency against this STS cluster is especially challenging and yet greatly needed. Verticillin A is a small molecule natural product with demonstrated anticancer activity; however, the efficacy of this agent has never been evaluated in STS. Therefore, the goal of this study was to explore verticillin A as a potential STS therapeutic. Methods We performed survival (MTS) and clonogenic analyses to measure the impact of this agent on the viability and colony formation capability of karyotypically complex STS cell lines: malignant peripheral nerve sheath tumor (MPNST) and leiomyosarcoma (LMS). The in vitro effects of verticillin A on apoptosis were investigated through annexin V/PI flow cytometry analysis and by measuring fluorescently-labeled cleaved caspase 3/7 activity. The impact on cell cycle progression was assessed via cytometric measurement of propidium iodide intercalation. In vivo studies were performed using MPNST xenograft models. Tumors were processed and analyzed using immunohistochemistry (IHC) for verticillin A effects on growth (Ki67) and apoptosis (cleaved caspase 3). Results Treatment with verticillin A resulted in decreased STS growth and an increase in apoptotic levels after 24 h. 100 nM verticillin A induced significant cellular growth abrogation after 24 h (96.7, 88.7, 72.7, 57, and 39.7% reduction in LMS1, S462, ST88, SKLMS1, and MPNST724, respectively). We observed no arrest in cell cycle, elevated annexin, and a nearly two-fold increase in cleaved caspase 3/7 activity in all MPNST and LMS cell lines. Control normal human Schwann (HSC) and aortic smooth muscle (HASMC) cells displayed higher tolerance to verticillin A treatment compared to sarcoma cell lines, although toxicity was seen in HSC at the highest treatment dose. In vivo studies mirrored the in vitro results: by day 11, tumor size was significantly reduced in MPNST724 xenograft models with treatment of 0.25 and 0.5 mg/kg verticillin A. Additionally, IHC assessment of tumors demonstrated increased cleaved caspase 3 and decreased proliferation (Ki67) following treatment with verticillin A. Conclusion Advancement in the treatment of karyotypically complex STS is confounded by the high level of genetic abnormalities found in these diseases. Consequently, the identification and investigation of novel therapies is greatly needed. Our data suggest that verticillin A selectively inhibits MPNST and LMS growth via induction of apoptosis while exhibiting minimal to moderate effects on normal cells, pointing to verticillin A as a potential treatment for MPNST and LMS, after additional preclinical validation.
Collapse
Affiliation(s)
- A Zewdu
- Department of Surgical Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; The James Cancer Center, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - G Lopez
- Department of Surgical Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; The James Cancer Center, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - D Braggio
- Department of Surgical Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; The James Cancer Center, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - C Kenny
- Department of Surgical Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; The James Cancer Center, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - D Constantino
- Department of Surgical Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; The James Cancer Center, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - H K Bid
- Department of Surgical Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; The James Cancer Center, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; Resonant Therapeutics, Inc., Ann Arbor, Michigan, USA
| | - K Batte
- Department of Surgical Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; The James Cancer Center, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - O H Iwenofu
- Department of Surgical Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; The James Cancer Center, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; Department of Pathology, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, USA
| | - N H Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina, USA
| | - C J Pearce
- Mycosynthetix, Inc., Hillsborough, North Carolina, USA
| | - A M Strohecker
- Department of Surgical Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; The James Cancer Center, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; Department of Cancer Biology and Genetics, Ohio State University, Columbus, Ohio, USA
| | - D Lev
- Surgery B, Sheba Medical Center, Tel Aviv, Israel
| | - R E Pollock
- Department of Surgical Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; The James Cancer Center, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
17
|
Establishment and characterization of a new human myxoid liposarcoma cell line (DL-221) with the FUS-DDIT3 translocation. J Transl Med 2016; 96:885-94. [PMID: 27270875 PMCID: PMC4965313 DOI: 10.1038/labinvest.2016.64] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 04/18/2016] [Indexed: 12/19/2022] Open
Abstract
Myxoid liposarcoma has the pathognomonic fusion oncogene FUS-DDIT3 encoding a chimeric transcription factor. Metastatic risk is higher with an increased round cell component and has been linked to aberrations involving the IGFR/PI3K/AKT pathway. These molecular insights have yet to translate to targeted therapies, and the lack of experimental models is a major hindrance. We describe the initial in-depth characterization of a new cell line (DL-221) and establishment of a mouse xenograft model. The cell line DL-221 was derived from a metastatic pleural lesion showing myxoid and round cell histology. This newly established cell line was characterized for phenotypic properties and molecular cytogenetic profile, using PCR, COBRA-FISH, and western blot. Next-generation whole-exome sequencing was performed to further characterize the cell line and the parent tumor. NOD-SCID-IL2R gamma knockout mice were xenograft hosts. DL-221 cells grew an adhering monolayer and COBRA-FISH showed an aneuploid karyotype with t(12;16)(q13;p11) and several other rearrangements; RT-PCR demonstrated a FUS-DDIT3 fusion transcript type 1. Both the cell line and the original tumor harbored a TP53 compound heterozygous mutation in exon 4 and 7, and were wild-type for PIK3CA. Moreover, among the 1254 variants called by whole-exome sequencing, there was 77% concordance between the cell line and parent tumor. The recently described hotspot mutation in the TERT promoter region in myxoid liposarcomas was also found at C228T in DL-221. Xenografts suitable for additional preclinical studies were successfully established in mice after subcutaneous injection. The established DL-221 cell line is the only published available myxoid liposarcoma cell line that underwent spontaneous immortalization, without requiring SV40 transformation. The cell line and its xenograft model are unique and helpful tools to study the biology and novel potential-targeted treatment approaches for myxoid liposarcoma.
Collapse
|