1
|
Pasquali S, Moura DS, Danks MR, Manasterski PJ, Zaffaroni N, Stacchiotti S, Mondaza-Hernandez JL, Kerrison WGJ, Martin-Broto J, Huang PH, Brunton VG. Preclinical models of soft tissue sarcomas - generation and applications to enhance translational research. Crit Rev Oncol Hematol 2025; 207:104621. [PMID: 39824369 DOI: 10.1016/j.critrevonc.2025.104621] [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: 11/21/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025] Open
Abstract
Soft tissue sarcomas (STS) represent a large group of rare and ultra-rare tumors distinguished by unique morphological, molecular and clinical features. Patients with such rare cancers are generally underrepresented in clinical trials which has limited the introduction of new treatment options and subsequent improvement of patient outcomes. Preclinical models of STS that recapitulate the human disease can aid progress in identifying new effective treatments. However, due to the rarity of these tumors there are limited STS models available. Here we review the existing preclinical models of STS, including patient-derived cell lines and organoids, patient-derived xenografts and genetically engineered mouse models. We discuss the advantages and disadvantages of the different models and describe to what extent they have aided clinical translation. Finally, we consider what can be done in the future to enhance their predictivity in the preclinical setting.
Collapse
Affiliation(s)
- Sandro Pasquali
- Molecular Pharmacology, Department of Experimental Oncology, Fondazione IRCCS Instituto Nazionale dei Tumori di Milano, via G. Amadeo 42, Milano 20133, Italy
| | - David S Moura
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain; Department of Medical Oncology, Fundacion Jimenez Diaz University Hospital, Madrid, Spain; University Hospital General of Villalba, Madrid, Spain
| | - Molly R Danks
- Edinburgh Cancer Research, CRUK Scotland Centre, University of Edinburgh, Crewe Road South, Edinburgh EH4 2RX, UK
| | - Piotr J Manasterski
- Edinburgh Cancer Research, CRUK Scotland Centre, University of Edinburgh, Crewe Road South, Edinburgh EH4 2RX, UK
| | - Nadia Zaffaroni
- Molecular Pharmacology, Department of Experimental Oncology, Fondazione IRCCS Instituto Nazionale dei Tumori di Milano, via G. Amadeo 42, Milano 20133, Italy
| | - Silvia Stacchiotti
- Molecular Pharmacology, Department of Experimental Oncology, Fondazione IRCCS Instituto Nazionale dei Tumori di Milano, via G. Amadeo 42, Milano 20133, Italy
| | - Jose L Mondaza-Hernandez
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain; Department of Medical Oncology, Fundacion Jimenez Diaz University Hospital, Madrid, Spain; University Hospital General of Villalba, Madrid, Spain
| | - William G J Kerrison
- Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road Sutton, London, SM2 5NG, UK
| | - Javier Martin-Broto
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain; Department of Medical Oncology, Fundacion Jimenez Diaz University Hospital, Madrid, Spain; University Hospital General of Villalba, Madrid, Spain
| | - Paul H Huang
- Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road Sutton, London, SM2 5NG, UK
| | - Valerie G Brunton
- Edinburgh Cancer Research, CRUK Scotland Centre, University of Edinburgh, Crewe Road South, Edinburgh EH4 2RX, UK.
| |
Collapse
|
2
|
Stacchiotti S, Martini S, Pasquali S, Frezza AM, Beretta A, Percio S, Lecchi M, Tortoreto M, Barisella M, Collini P, Dagrada GP, Merlini A, Huang PH, Jenks A, Jones RL, Tap WD, Ingrosso M, Morosi C, Brich S, Giani C, Verderio P, Casali PG, Leonard H, Gronchi A, Zuco V, Zaffaroni N. GDF-15 Predicts Epithelioid Hemangioendothelioma Aggressiveness and Is Downregulated by Sirolimus through ATF4/ATF5 Suppression. Clin Cancer Res 2024; 30:5122-5137. [PMID: 39283723 PMCID: PMC11565171 DOI: 10.1158/1078-0432.ccr-23-3991] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/24/2024] [Accepted: 09/12/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Epithelioid hemangioendothelioma (EHE), an ultra-rare sarcoma, poses therapeutic challenges because of limited efficacy of conventional chemotherapy in advanced cases, necessitating exploration of new treatment avenues and identification of novel aggressive biomarkers. This study aimed at (i) utilizing a patient-derived xenograft model of EHE and its associated cell line to assess the efficacy of sirolimus and (ii) analyzing two distinct patient cohorts to pinpoint circulating biomarkers of EHE aggressiveness. EXPERIMENTAL DESIGN A patient-derived xenograft model and corresponding cell line were established from a patient with advanced EHE, demonstrating consistency with the original tumor in terms of histomorphology, WWTR1::CAMTA1 fusion presence, and genomic and transcriptomic profiles. Two independent patient series were employed to investigate the association between growth/differentiation factor 15 (GDF-15) serum levels and EHE aggressiveness. RESULTS ELISA analyses on EHE cell culture medium and blood from EHE-carrying mice revealed the release of GDF-15 by EHE cells. Sirolimus exhibited markedly higher antitumor activity compared with doxorubicin, concurrently reducing GDF-15 expression/release both in vivo and in vitro. This reduction was attributed to the drug-induced inhibition of phosphorylation/activation of 4E-BP1 and subsequent downregulation of the GDF-15 transcription factors ATF4 and ATF5. Blood sample analyses from two independent patient series showed a significant correlation between GDF-15 and EHE aggressiveness. CONCLUSIONS This study identifies GDF-15 as a novel biomarker of EHE aggressiveness and underscores the superior efficacy of sirolimus compared with doxorubicin in our experimental models. The observed inhibition of GDF-15 release by sirolimus suggests its potential as a biomarker for monitoring the drug's activity in patients.
Collapse
Affiliation(s)
- Silvia Stacchiotti
- Medical Oncology Unit 2, Cancer Medicine Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale Tumori, Milan, Italy
| | - Silvia Martini
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Sandro Pasquali
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Anna M. Frezza
- Medical Oncology Unit 2, Cancer Medicine Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale Tumori, Milan, Italy
| | - Alessia Beretta
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Stefano Percio
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Mara Lecchi
- Unit of Bioinformatics and Biostatistics, Department Epidemiology and Data Science, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Tortoreto
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | - Paola Collini
- Soft Tissue Tumor Pathology Unit, Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Gian Paolo Dagrada
- Soft Tissue Tumor Pathology Unit, Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | - Paul H. Huang
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Andrew Jenks
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Robin L. Jones
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - William D. Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matilde Ingrosso
- Medical Oncology Unit 2, Cancer Medicine Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale Tumori, Milan, Italy
| | - Carlo Morosi
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Silvia Brich
- Soft Tissue Tumor Pathology Unit, Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Claudia Giani
- Medical Oncology Unit 2, Cancer Medicine Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale Tumori, Milan, Italy
| | - Paolo Verderio
- Unit of Bioinformatics and Biostatistics, Department Epidemiology and Data Science, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo G. Casali
- Medical Oncology Unit 2, Cancer Medicine Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale Tumori, Milan, Italy
| | - Hugh Leonard
- Chair of Trustees of the EHE Rare Cancer Charity UK, Charity Number 1162472, Kingston-Upon-Thames, United Kingdom
| | - Alessandro Gronchi
- Sarcoma Service, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Valentina Zuco
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| |
Collapse
|
3
|
Giusti V, Miserocchi G, Sbanchi G, Pannella M, Hattinger CM, Cesari M, Fantoni L, Guerrieri AN, Bellotti C, De Vita A, Spadazzi C, Donati DM, Torsello M, Lucarelli E, Ibrahim T, Mercatali L. Xenografting Human Musculoskeletal Sarcomas in Mice, Chick Embryo, and Zebrafish: How to Boost Translational Research. Biomedicines 2024; 12:1921. [PMID: 39200384 PMCID: PMC11352184 DOI: 10.3390/biomedicines12081921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Musculoskeletal sarcomas pose major challenges to researchers and clinicians due to their rarity and heterogeneity. Xenografting human cells or tumor fragments in rodents is a mainstay for the generation of cancer models and for the preclinical trial of novel drugs. Lately, though, technical, intrinsic and ethical concerns together with stricter regulations have significantly curbed the employment of murine patient-derived xenografts (mPDX). In alternatives to murine PDXs, researchers have focused on embryonal systems such as chorioallantoic membrane (CAM) and zebrafish embryos. These systems are time- and cost-effective hosts for tumor fragments and near-patient cells. The CAM of the chick embryo represents a unique vascularized environment to host xenografts with high engraftment rates, allowing for ease of visualization and molecular detection of metastatic cells. Thanks to the transparency of the larvae, zebrafish allow for the tracking of tumor development and metastatization, enabling high-throughput drug screening. This review will focus on xenograft models of musculoskeletal sarcomas to highlight the intrinsic and technically distinctive features of the different hosts, and how they can be exploited to elucidate biological mechanisms beneath the different phases of the tumor's natural history and in drug development. Ultimately, the review suggests the combination of different models as an advantageous approach to boost basic and translational research.
Collapse
Affiliation(s)
- Veronica Giusti
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Giacomo Miserocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (G.M.); (A.D.V.); (C.S.)
| | - Giulia Sbanchi
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Micaela Pannella
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Claudia Maria Hattinger
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Marilena Cesari
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Leonardo Fantoni
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Ania Naila Guerrieri
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Chiara Bellotti
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Alessandro De Vita
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (G.M.); (A.D.V.); (C.S.)
| | - Chiara Spadazzi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (G.M.); (A.D.V.); (C.S.)
| | - Davide Maria Donati
- Orthopaedic Oncology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Monica Torsello
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Enrico Lucarelli
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Laura Mercatali
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| |
Collapse
|
4
|
Lee JY, Guan P, Lim AH, Guo Z, Li Z, Kok JST, Lee ECY, Lim BY, Kannan B, Loh JW, Ng CCY, Lim KS, Teh BT, Ko TK, Chan JY. Establishment and characterization of a patient-derived solitary fibrous tumor/hemangiopericytoma cell line model. Hum Cell 2024; 37:310-322. [PMID: 38070062 DOI: 10.1007/s13577-023-01013-2] [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: 08/23/2023] [Accepted: 11/16/2023] [Indexed: 01/04/2024]
Abstract
Solitary fibrous tumor/Hemangiopericytoma (SFT/HPC) is a rare subtype of soft tissue sarcoma harboring NAB2-STAT6 gene fusions. Mechanistic studies and therapeutic development on SFT/HPC are impeded by scarcity and lack of system models. In this study, we established and characterized a novel SFT/HPC patient-derived cell line (PDC), SFT-S1, and screened for potential drug candidates that could be repurposed for the treatment of SFT/HPC. Immunohistochemistry profiles of the PDC was consistent with the patient's tumor sample (CD99+/CD34+/desmin-). RNA sequencing, followed by Sanger sequencing confirmed the pathognomonic NAB2exon3-STAT6exon18 fusion in both the PDC and the original tumor. Transcriptomic data showed strong enrichment for oncogenic pathways (epithelial-mesenchymal transition, FGF, EGR1 and TGFβ signaling pathways) in the tumor. Whole genome sequencing identified potentially pathogenic somatic variants such as MAGEA10 and ABCA2. Among a panel of 14 targeted agents screened, dasatinib was identified to be the most potent small molecule inhibitor against the PDC (IC50, 473 nM), followed by osimertinib (IC50, 730 nM) and sunitinib (IC50, 1765 nM). Methylation profiling of the tumor suggests that this specific variant of SFT/HPC could lead to genome-wide hypomethylation. In conclusion, we established a novel PDC model of SFT/HPC with comprehensive characterization of its genomic, epigenomic and transcriptomic landscape, which can facilitate future preclinical studies of SFT/HPC, such as in vitro drug screening and in vivo drug testing.
Collapse
Affiliation(s)
- Jing Yi Lee
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Peiyong Guan
- Agency for Science, Technology and Research (A*STAR), Genome Institute of Singapore, Singapore, Singapore
| | - Abner Herbert Lim
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Zexi Guo
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Zhimei Li
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Jessica Sook Ting Kok
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | | | - Boon Yee Lim
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Bavani Kannan
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Jui Wan Loh
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Cedric Chuan-Young Ng
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Kah Suan Lim
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Bin Tean Teh
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
- Agency for Science, Technology and Research (A*STAR), Genome Institute of Singapore, Singapore, Singapore
- Duke-NUS Medical School, Oncology Academic Clinical Program, Singapore, Singapore
| | - Tun Kiat Ko
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Jason Yongsheng Chan
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore.
- Duke-NUS Medical School, Oncology Academic Clinical Program, Singapore, Singapore.
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore, 168583, Singapore.
| |
Collapse
|
5
|
Janik AM, Terlecka A, Spałek MJ, Boye K, Szostakowski B, Chmiel P, Szumera-Ciećkiewicz A, Bobak K, Świtaj T, Rutkowski P, Czarnecka AM. Diagnostics and Treatment of Extrameningeal Solitary Fibrous Tumors. Cancers (Basel) 2023; 15:5854. [PMID: 38136399 PMCID: PMC10742263 DOI: 10.3390/cancers15245854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Solitary fibrous tumors (SFT) are rare mesenchymal neoplasms that account for less than 2% of all soft tissue masses. In the latest WHO 2020 Classification of Soft Tissue Tumors, extrameningeal SFT was listed as intermediate (rarely metastasizing) or malignant neoplasms. Due to the lack of characteristic clinical features, their diagnosis and treatment remain challenging. The pathogenesis of SFT is often associated with the presence of fusions of the NAB2-STAT6 gene on the 12q13 chromosome. Cytoplasmic CD34 positive staining is considerably characteristic for most SFTs; less frequently, factor XII, vimentin, bcl-2, and CD99 are present. A key factor in the diagnosis is the prevalent nuclear location of STAT6 expression. Radical resection is the mainstay of localized SFTs. In the case of unresectable disease, only radiotherapy or radio-chemotherapy may significantly ensure long-term local control of primary and metastatic lesions. To date, no practical guidelines have been published for the treatment of advanced or metastatic disease. Classical anthracycline-based chemotherapy is applicable. The latest studies suggest that antiangiogenic therapies should be considered after first-line treatment. Other drugs, such as imatinib, figitumumab, axitinib, and eribulin, are also being tested. Definitive radiotherapy appears to be a promising therapeutic modality. Since standards for the treatment of advanced and metastatic diseases are not available, further investigation of novel agents is necessary.
Collapse
Affiliation(s)
- Anna Maria Janik
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.M.J.); (A.T.); (M.J.S.); (B.S.); (P.C.); (K.B.); (T.Ś.); (P.R.)
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Anna Terlecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.M.J.); (A.T.); (M.J.S.); (B.S.); (P.C.); (K.B.); (T.Ś.); (P.R.)
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Mateusz J. Spałek
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.M.J.); (A.T.); (M.J.S.); (B.S.); (P.C.); (K.B.); (T.Ś.); (P.R.)
- Department of Radiotherapy I, Maria Sklodowska-Curie National Research Institute of Oncology, 02-718 Warsaw, Poland
| | - Kjetil Boye
- Department of Oncology, Oslo University Hospital, 0372 Oslo, Norway;
| | - Bartłomiej Szostakowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.M.J.); (A.T.); (M.J.S.); (B.S.); (P.C.); (K.B.); (T.Ś.); (P.R.)
| | - Paulina Chmiel
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.M.J.); (A.T.); (M.J.S.); (B.S.); (P.C.); (K.B.); (T.Ś.); (P.R.)
| | - Anna Szumera-Ciećkiewicz
- Department of Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
- Diagnostic Hematology Department, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland
| | - Klaudia Bobak
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.M.J.); (A.T.); (M.J.S.); (B.S.); (P.C.); (K.B.); (T.Ś.); (P.R.)
| | - Tomasz Świtaj
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.M.J.); (A.T.); (M.J.S.); (B.S.); (P.C.); (K.B.); (T.Ś.); (P.R.)
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.M.J.); (A.T.); (M.J.S.); (B.S.); (P.C.); (K.B.); (T.Ś.); (P.R.)
| | - Anna M. Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.M.J.); (A.T.); (M.J.S.); (B.S.); (P.C.); (K.B.); (T.Ś.); (P.R.)
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute Polish Academy of Sciences, 02-106 Warsaw, Poland
| |
Collapse
|
6
|
Ghanim B, Baier D, Pirker C, Müllauer L, Sinn K, Lang G, Hoetzenecker K, Berger W. Trabectedin Is Active against Two Novel, Patient-Derived Solitary Fibrous Pleural Tumor Cell Lines and Synergizes with Ponatinib. Cancers (Basel) 2022; 14:cancers14225602. [PMID: 36428694 PMCID: PMC9688590 DOI: 10.3390/cancers14225602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/05/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022] Open
Abstract
Solitary fibrous tumor of the pleura (SFT) is a rare disease. Besides surgery combined with radiotherapy in nondisseminated stages, curative options are currently absent. Out of fourteen primo-cell cultures, established from surgical SFT specimens, two showed stable in vitro growth. Both cell models harbored the characteristic NAB2-STAT6 fusion and were further investigated by different preclinical methods assessing cell viability, clone formation, and protein regulation upon single-drug treatment or in response to selected treatment combinations. Both fusion-positive cell models showed-in line with the clinical experience and the literature-a low to moderate response to most of the tested cytotoxic and targeted agents. However, the multi-tyrosine kinase inhibitors ponatinib and dasatinib, as well as the anti-sarcoma compound trabectedin, revealed promising activity against SFT growth. Furthermore, both cell models spontaneously presented strong FGFR downstream signaling targetable by ponatinib. Most interestingly, the combination of either ponatinib or dasatinib with trabectedin showed synergistic effects. In conclusion, this study identified novel trabectedin-based treatment combinations with clinically approved tyrosine kinase inhibitors, using two newly established NAB2-STAT6 fusion-positive cell models. These findings can be the basis for anti-SFT drug repurposing approaches in this rare and therapy-refractory disease.
Collapse
Affiliation(s)
- Bahil Ghanim
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
- Department of General and Thoracic Surgery, Karl Landsteiner University of Health Sciences, University Hospital Krems, 3500 Krems, Austria
| | - Dina Baier
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
- Institute of Inorganic Chemistry, University of Vienna, 1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
- Correspondence:
| | - Christine Pirker
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria
| | - Katharina Sinn
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Gyoergy Lang
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Walter Berger
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
| |
Collapse
|
7
|
Martín-Broto J, Hindi N, Grignani G, Merlini A, Ibrahim T, Le Cesne A. Experience with second-line trabectedin in daily clinical practice: case studies. Future Oncol 2022; 18:23-32. [PMID: 36200932 DOI: 10.2217/fon-2022-0519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
As a recommended second-line option for advanced soft tissue sarcoma, trabectedin can provide the necessary balance between long-term tumor control and preserved quality of life. Three case studies illustrate the long-lasting responses that patients can achieve with second-line trabectedin. A female patient with metastatic leiomyosarcoma maintained disease control for 2 years with trabectedin (× 41 cycles) with excellent tolerability and no relevant adverse events. At the time of writing, a male patient with a metastatic solitary fibrous tumor was asymptomatic after 30 cycles of trabectedin and treatment was ongoing. A young male patient with a recurrent, nonresectable, retroperitoneal myxoid/round cell liposarcoma was able to continue his sporting activities (triathlons) over 2 years with trabectedin (× 14 cycles) plus watchful waiting.
Collapse
Affiliation(s)
- Javier Martín-Broto
- Medical Oncology Department, University Hospital Fundación Jimenez Diaz, 28040 Madrid, Spain.,University Hospital General de Villalba, 28400 Collado Villalba, Madrid, Spain.,Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), 28015 Madrid, Spain
| | - Nadia Hindi
- Medical Oncology Department, University Hospital Fundación Jimenez Diaz, 28040 Madrid, Spain.,University Hospital General de Villalba, 28400 Collado Villalba, Madrid, Spain.,Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), 28015 Madrid, Spain
| | - Giovanni Grignani
- Candiolo Cancer Institute, FPO-IRCCS, SP 142 Km 3.95, 10060 Candiolo, Italy
| | - Alessandra Merlini
- Candiolo Cancer Institute, FPO-IRCCS, SP 142 Km 3.95, 10060 Candiolo, Italy.,Department of Oncology, University of Turin, 10124 Turin, Italy
| | - Tony Ibrahim
- International Department of Medical Oncology, Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif, France
| | - Axel Le Cesne
- International Department of Medical Oncology, Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif, France
| |
Collapse
|
8
|
Kazazian K, Demicco EG, de Perrot M, Strauss D, Swallow CJ. Toward Better Understanding and Management of Solitary Fibrous Tumor. Surg Oncol Clin N Am 2022; 31:459-483. [PMID: 35715145 DOI: 10.1016/j.soc.2022.03.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Solitary fibrous tumor (SFT) comprises a histologic spectrum of soft tissue neoplasms that are characterized by the unique NAB2-STAT6 gene fusion. Changes in diagnostic terminology and site-specific classification over the past few decades have resulted in a disjointed literature. Complete surgical excision with preservation of function remains the mainstay of treatment. New risk stratification systems including risk factors such as mitotic rate, age, tumor size, and presence of necrosis, among others, can be used to predict risk of recurrence or metastasis. Long-term follow-up after surgical resection is recommended. The clinical manifestations, diagnosis, management, and prognosis of SFT are reviewed here.
Collapse
Affiliation(s)
| | - Elizabeth G Demicco
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Marc de Perrot
- Department of Surgery, University of Toronto, Toronto, Canada; Division of Thoracic Surgery, Princess Margaret Cancer Centre/University Health Network, 200 Elizabeth Street, Toronto, Ontario M5G2C4, Canada
| | - Dirk Strauss
- Sarcoma Unit, Department of Academic Surgery, Royal Marsden Hospital, Royal Marsden NHS Foundation Trust, Fulham Road, London SW3 6JJ, England
| | - Carol J Swallow
- Department of Surgery, University of Toronto, Toronto, Canada; Department of Surgical Oncology, Princess Margaret Cancer Centre/Mount Sinai Hospital, Toronto, Canada.
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW To review current knowledge and recent advances in retroperitoneal sarcoma management. RECENT FINDINGS Surgery, radiotherapy, and medical treatments of retroperitoneal sarcomas should take into account the peculiarities of each histotype and the unique anatomical site. Surgery remains the mainstay of treatment and the only chance of cure for these diseases. In low-grade retroperitoneal sarcomas, like well differentiated liposarcoma, where the leading cause of death is dominated by local rather than distant relapses, treatment of the primary tumor encompasses extended surgery with multiorgan resection and evaluation of preoperative radiotherapy. Conversely, surgery is usually more conservative and without radiotherapy in those retroperitoneal sarcomas, such as leiomyosarcoma, characterized by a high risk of metastatic spread that prompted also the evaluation of neoadjuvant, histotype-driven chemotherapy. Surgery might have a role also for relapsed disease, despite long-term disease control probability declines at each recurrence. In advanced stages, anthracyclines still retain a key role and all medical treatment strategies should follow the specific chemosensitivity of each histotype to improve patient's outcomes. SUMMARY The rarity and heterogeneity in biological behavior and clinical presentation of retroperitoneal sarcomas deserves a multidisciplinary and histotype-driven treatment at all stages of the disease to be performed in highly specialized centers.
Collapse
|
10
|
Hayenga HN, Cai C, Fetzer D, White S, Kuban J, Wardak Z, Benjamin RS, Pan E, Strauss J, Gao B, Minna J, Martin-Broto J, Hinshaw JL. Rare solid and cystic presentation of hemangiopericytoma/ solitary fibrous tumor: a case report. CURRENT PROBLEMS IN CANCER: CASE REPORTS 2022. [DOI: 10.1016/j.cpccr.2022.100149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
11
|
Martin-Broto J, Mondaza-Hernandez JL, Moura DS, Hindi N. A Comprehensive Review on Solitary Fibrous Tumor: New Insights for New Horizons. Cancers (Basel) 2021; 13:cancers13122913. [PMID: 34200924 PMCID: PMC8230482 DOI: 10.3390/cancers13122913] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Solitary fibrous tumor (SFT) is a malignant condition that exhibits different clinical behaviors ranging from low to high aggressive SFT, with dedifferentiated SFT (DD-SFT) being the fastest-growing subtype. Even when surgery alone provides curation rates above 60%, recurrences do occur in a fraction of patients where surgery is unable to provide disease control. Among the systemic therapeutic options, antiangiogenic compounds have shown higher efficacy than chemotherapy by indirect comparisons. Furthermore, rotating different antiangiogenics, at the progression time, has been shown to be effective. The exception is DD-SFT since it is resistant to antiangiogenics but can respond to chemotherapy. This comprehensive review also analyzes the underlying molecular components that play a key role in SFT origin and aggressiveness. The discovery in 2013 of anomalous fusion genes between NAB2 and STAT6 was determinant to increase the knowledge on the molecular drivers in SFT that could be potential targets for future therapies. Abstract Solitary fibrous tumor (SFT) is a rare mesenchymal, ubiquitous tumor, with an incidence of 1 new case/million people/year. In the 2020 WHO classification, risk stratification models were recommended as a better tool to determine prognosis in SFT, to the detriment of “typical” or “malignant” classic terms. The risk for metastasis is up to 35–45%, or even greater, in series with a longer follow-up. Over the last few decades, advances in immunohistochemistry and molecular diagnostics identified STAT6 nuclear protein expression and the NAB2–STAT6 fusion gene as more precise tools for SFT diagnosis. Recent evidence taken from retrospective series and from two prospective phase II clinical trials showed that antiangiogenics are active and their sequential use from first line should be considered, except for dedifferentiated SFT for which chemotherapy is the best option. Since the fusion transcript driver’s first description in 2013, new insights have been brought on key molecular events in SFT. This comprehensive review mainly focuses on the superior efficacy of antiangiogenics over chemotherapeutic agents in SFT, provides the current knowledge of key molecules that could co-drive the SFT behavior, and suggests new target candidates that deserve to be explored in preclinical and clinical research in SFT.
Collapse
Affiliation(s)
- Javier Martin-Broto
- Fundacion Jimenez Díaz University Hospital, 28040 Madrid, Spain;
- General de Villalba University Hospital, Collado Villalba, 28400 Madrid, Spain
- Fundación Jiménez Díaz Institute for Medical Research (IIS/FJD), 28040 Madrid, Spain
- Correspondence:
| | - Jose L. Mondaza-Hernandez
- Institute of Biomedicine of Seville (IBiS, CSIC, US and HUVR), 41013 Sevilla, Spain; (J.L.M.-H.); (D.S.M.)
| | - David S. Moura
- Institute of Biomedicine of Seville (IBiS, CSIC, US and HUVR), 41013 Sevilla, Spain; (J.L.M.-H.); (D.S.M.)
| | - Nadia Hindi
- Fundacion Jimenez Díaz University Hospital, 28040 Madrid, Spain;
- General de Villalba University Hospital, Collado Villalba, 28400 Madrid, Spain
- Fundación Jiménez Díaz Institute for Medical Research (IIS/FJD), 28040 Madrid, Spain
| |
Collapse
|
12
|
Wang W, Bao B, Hu A, Zhu X, Chen Q. Two case reports of rare diseases occurring in rare parts: splenic vein solitary fibrous tumor and liver solitary fibrous tumor. AME Case Rep 2021; 5:17. [PMID: 33912806 DOI: 10.21037/acr-20-142] [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: 09/15/2020] [Accepted: 02/06/2021] [Indexed: 11/06/2022]
Abstract
Solitary fibrous tumor (SFT) is a rare soft tissue tumor originating from mesenchymal cells. Here we report two new cases of SFT. One case was a 37-year-old female patient whose primary tumor site was located in the splenic vein and the primary tumor resulted in splenomegaly and hypersplenism; its recurred for many times after surgical resection and eventually transferred to the liver, 4 operations were performed during 10 years of follow-up, and the patient is in a good condition right now. The second case was a 54-year-old male patient whose primary tumor site was located in the liver, spleen and left side of the chest wall. We performed two operations to remove these tumors, totally. Six years later, SFT recurred in the liver, given that the tumor was too large to be surgical resected completely, we chose orthotopic liver transplantation (OLT), and no tumor recurred during 6 years' follow-up, he is also in a good condition right now. The reports of these two cases of SFT are exceedingly rare, especially the splenic vein SFT is the first report case, which helps expand the understanding of SFT. Although the current mainstream treatment of SFT is surgical resection, liver transplantation may be a new option treatment for the huge liver SFT.
Collapse
Affiliation(s)
- Wenjing Wang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Banghe Bao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anbin Hu
- Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaofeng Zhu
- Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qing Chen
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Zuco V, Pasquali S, Tortoreto M, Brich S, Percio S, Dagrada GP, Colombo C, Sanfilippo R, Lauricella C, Gounder M, El Bezawy R, Barisella M, Dei Tos AP, Casali PG, Gronchi A, Stacchiotti S, Zaffaroni N. Selinexor versus doxorubicin in dedifferentiated liposarcoma PDXs: evidence of greater activity and apoptotic response dependent on p53 nuclear accumulation and survivin down-regulation. J Exp Clin Cancer Res 2021; 40:83. [PMID: 33648535 PMCID: PMC7923610 DOI: 10.1186/s13046-021-01886-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/17/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Dedifferentiated liposarcoma (DDLPS), a tumor that lacks effective treatment strategies and is associated with poor outcomes, expresses amplified MDM2 in the presence of wild-type p53. MDM2 ubiquitination of p53 facilitates its XPO1-mediated nuclear export, thus limiting p53 tumor suppressor functions. Consequently, nuclear export is a rational target in DDLPS. We directly compared the antitumor activity of the first-in class XPO1 inhibitor selinexor and doxorubicin, the standard front-line therapy in sarcomas, in DDLPS patient-derived xenografts (PDXs) and primary cell lines. METHODS Drug activity was assessed in three PDXs (and two corresponding cell lines) established from the dedifferentiated component of primary untreated retroperitoneal DDLPS with myogenic (N = 2) and rhabdomyoblastic (N = 1) differentiation from patients who underwent surgery. These models were marked by amplification of MDM2, CDK4 and HMGA2 genes. RESULTS Selinexor was moderately active in the three PDXs but achieved greater tumor response compared to doxorubicin (maximum tumor volume inhibition: 46-80 % vs. 37-60 %). The PDX harboring rhabdomyoblastic dedifferentiation showed the highest sensitivity to both agents. PDX response to selinexor and doxorubicin was not associated with the extent of MDM2 and CDK4 gene amplification. Interestingly, the most chemosensitive PDX model showed the lowest extent of HMGA2 amplification. Selinexor was also more efficient than doxorubicinin in inducing an apoptotic response in PDXs and cell lines. Consistently, an increased nuclear accumulation of p53 was seen in all selinexor-treated models. In addition, a time-dependent decrease of survivin expression, with an almost complete abrogation of the cytoplasmic anti-apoptotic pool of this protein, was observed as a consequence of the decreased acetylation/activation of STAT3 and the increased ubiquitination of nuclear survivin. CONCLUSIONS Selinexor showed a moderate antitumor activity in three DDLPS PDXs, which was, however, consistently higher than doxorubicin across all different models regardless the extent of MDM2 amplification and the histological differentiation. The depletion of survivin protein seems to significantly contribute to the induction of apoptosis through which selinexor exerts its antitumor activity.
Collapse
Affiliation(s)
- Valentina Zuco
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Sandro Pasquali
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Monica Tortoreto
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Silvia Brich
- Department of Pathology, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133, Milan, Italy
| | - Stefano Percio
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Gian Paolo Dagrada
- Department of Pathology, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133, Milan, Italy
| | - Chiara Colombo
- Sarcoma Service, Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133, Milan, Italy
| | - Roberta Sanfilippo
- Adult Mesenchymal Tumor and Rare Cancer Unit, Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133, Milan, Italy
| | | | - Mrinal Gounder
- Sarcoma Medical Oncology and Early Drug Development, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, 10065, New York, NY, USA
| | - Rihan El Bezawy
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Marta Barisella
- Department of Pathology, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133, Milan, Italy
| | - Angelo Paolo Dei Tos
- Department of Medicine, University of Padua School of Medicine, Via Giustiniani 2, 35128, Padua, Italy
| | - Paolo Giovanni Casali
- Adult Mesenchymal Tumor and Rare Cancer Unit, Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133, Milan, Italy
- Department of Biomedical and Clinical Sciences L. Sacco, University of Milan, Via Grassi 74, 20157, Milan, Italy
| | - Alessandro Gronchi
- Sarcoma Service, Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133, Milan, Italy
| | - Silvia Stacchiotti
- Adult Mesenchymal Tumor and Rare Cancer Unit, Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133, Milan, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, 20133, Milan, Italy.
| |
Collapse
|
14
|
Martín-Broto J, Reichardt P, Jones RL, Stacchiotti S. Different approaches to advanced soft tissue sarcomas depending on treatment line, goal of therapy and histological subtype. Expert Rev Anticancer Ther 2020; 20:15-28. [PMID: 32349558 DOI: 10.1080/14737140.2020.1753510] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Introduction: Although rapid evolution over the past few years in advanced soft tissue sarcoma (STS) management has not been without its challenges, it has brought clarity in several areas.Areas covered: This article summarizes the proceedings of the third edition of the Soft Tissue Sarcoma: Evidence and Experience symposium held March 2019 in Madrid, Spain. An update is provided of current approaches to advanced STS management. Case studies illustrate the role of trabectedin in advanced STS management.Expert opinion: First-line treatment of advanced STS requires distinct therapeutic strategies depending on goal: tumor shrinkage or tumor control. Since all sarcoma patients benefit from active treatment irrespective of age or line of therapy, oncologists have a duty to offer active systemic therapies unless the patient is unfit for treatment or chooses to end active treatment. Beyond the first line, histology becomes increasingly relevant for treatment selection. Agents with activity in specific sarcoma subtypes have been identified. Rare tumors represent a substantial medical need requiring strong international collaboration between research groups, pharmaceutical companies, regulatory agencies, and patients to identify active drugs per subtype. Multidisciplinary care in an expert sarcoma center is the primary means of reducing morbidity and mortality in patients with sarcoma.
Collapse
Affiliation(s)
- Javier Martín-Broto
- Medical Oncology Department in University Hospital Virgen del Rocio and Institute of Biomedicine of Sevilla (IBIS), (HUVR, CSIC, University of Sevilla), Sevilla, Spain
| | - Peter Reichardt
- Department of Oncology, Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Robin L Jones
- Royal Marsden Hospital, Institute of Cancer Research, London, UK
| | - Silvia Stacchiotti
- Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| |
Collapse
|
15
|
Comparative Assessment of Antitumor Effects and Autophagy Induction as a Resistance Mechanism by Cytotoxics and EZH2 Inhibition in INI1-Negative Epithelioid Sarcoma Patient-Derived Xenograft. Cancers (Basel) 2019; 11:cancers11071015. [PMID: 31331120 PMCID: PMC6678245 DOI: 10.3390/cancers11071015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/10/2019] [Accepted: 07/16/2019] [Indexed: 12/31/2022] Open
Abstract
Epithelioid sarcoma (ES) is a rare mesenchymal malignancy marked by SMARCB1/INI1 deficiency. Retrospective clinical data report on the activity of anthracycline- and gemcitabine-based regimens. EZH2 inhibitors are currently being tested in clinical trials. Since comparisons of these agents are unlikely to be prospectively evaluated in the clinics, we took advantage of an INI1-deficient proximal-type ES patient-derived xenograft (PDX ES-1) to comparatively assess its preclinical antitumor activity. Mice were treated with doxorubicin and ifosfamide, singly or in combination, gemcitabine, and the EZH2 inhibitor EPZ-011989. Comparable antitumor activity (max tumor volume inhibition: ~90%) was caused by gemcitabine, EPZ-011989, and the doxorubicin-ifosfamide combination. The integration of RNAseq data, generated on tumors obtained from untreated and EPZ-011989-treated mice, and results from functional studies, carried out on the PDX-derived ES-1 cell line, revealed autophagy induction as a possible survival mechanism in residual tumor cells following EPZ-011989 treatment and identified HMGA2 as a main player in this process. Our data support the clinical use of gemcitabine and the doxorubicin-ifosfamide combination, confirm EZH2 as a therapeutic target in proximal-type ES, and suggest autophagy as a cytoprotective mechanism against EZH2 inhibition.
Collapse
|
16
|
Cornillie J, Wozniak A, Li H, Wang Y, Boeckx B, Gebreyohannes YK, Wellens J, Vanleeuw U, Hompes D, Stas M, Sinnaeve F, Wafa H, Lambrechts D, Debiec-Rychter M, Sciot R, Schöffski P. Establishment and Characterization of Histologically and Molecularly Stable Soft-tissue Sarcoma Xenograft Models for Biological Studies and Preclinical Drug Testing. Mol Cancer Ther 2019; 18:1168-1178. [PMID: 30962320 DOI: 10.1158/1535-7163.mct-18-1045] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/13/2019] [Accepted: 04/01/2019] [Indexed: 11/16/2022]
Abstract
Soft-tissue sarcomas (STS) represent a heterogeneous group of rare, malignant tumors of mesenchymal origin. Reliable in vivo sarcoma research models are scarce. We aimed to establish and characterize histologically and molecularly stable patient-derived xenograft (PDX) models from a broad variety of STS subtypes. A total of 188 fresh tumor samples from consenting patients with localized or advanced STS were transplanted subcutaneously in NMRI-nu/nu-immunodeficient mice. Once tumor growth was observed, the material was passaged to a next generation of mice. A patient-derived tumor sample was considered "successfully engrafted" whenever the sample was transplanted to passage 1. A PDX model was considered "established" when observing stable morphologic and molecular features for at least two passages. With every passage, histologic and molecular analyses were performed. Specific genomic alterations and copy-number profile were assessed by FISH and low coverage whole-genome sequencing. The tumor engraftment rate was 32% (61/188) and 188 patient samples generated a total of 32 PDX models, including seven models of myxofibrosarcoma, five dedifferentiated liposarcoma, five leiomyosarcoma, three undifferentiated pleomorphic sarcoma, two malignant peripheral nerve sheet tumor models, and single models of synovial sarcoma and some other (ultra)rare subtypes. Seventeen additional models are in early stages of engraftment (passage 1-2). Histopathologic and molecular features were compared with the original donor tumor and were stable throughout passaging. The platform is used for studies on sarcoma biology and suited for in vivo preclinical drug testing as illustrated by a number of completed and ongoing laboratory studies.
Collapse
Affiliation(s)
- Jasmien Cornillie
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Agnieszka Wozniak
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Haifu Li
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Yannick Wang
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Bram Boeckx
- Laboratory for Translational Genetics, KU Leuven, Leuven, Belgium.,VIB Center for Cancer Biology, Leuven, Belgium
| | - Yemarshet K Gebreyohannes
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Jasmien Wellens
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium.
| | - Ulla Vanleeuw
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Daphne Hompes
- Department of Surgical Oncology, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Marguerite Stas
- Department of Surgical Oncology, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Friedl Sinnaeve
- Department of Orthopedic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Hazem Wafa
- Department of Orthopedic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, KU Leuven, Leuven, Belgium.,VIB Center for Cancer Biology, Leuven, Belgium
| | - Maria Debiec-Rychter
- Department of Human Genetics, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Raf Sciot
- Department of Pathology, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Patrick Schöffski
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| |
Collapse
|
17
|
Martin-Broto J, Stacchiotti S, Lopez-Pousa A, Redondo A, Bernabeu D, de Alava E, Casali PG, Italiano A, Gutierrez A, Moura DS, Peña-Chilet M, Diaz-Martin J, Biscuola M, Taron M, Collini P, Ranchere-Vince D, Garcia Del Muro X, Grignani G, Dumont S, Martinez-Trufero J, Palmerini E, Hindi N, Sebio A, Dopazo J, Dei Tos AP, LeCesne A, Blay JY, Cruz J. Pazopanib for treatment of advanced malignant and dedifferentiated solitary fibrous tumour: a multicentre, single-arm, phase 2 trial. Lancet Oncol 2018; 20:134-144. [PMID: 30578023 DOI: 10.1016/s1470-2045(18)30676-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/23/2018] [Accepted: 08/31/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND A solitary fibrous tumour is a rare soft-tissue tumour with three clinicopathological variants: typical, malignant, and dedifferentiated. Preclinical experiments and retrospective studies have shown different sensitivities of solitary fibrous tumour to chemotherapy and antiangiogenics. We therefore designed a trial to assess the activity of pazopanib in a cohort of patients with malignant or dedifferentiated solitary fibrous tumour. The clinical and translational results are presented here. METHODS In this single-arm, phase 2 trial, adult patients (aged ≥ 18 years) with histologically confirmed metastatic or unresectable malignant or dedifferentiated solitary fibrous tumour at any location, who had progressed (by RECIST and Choi criteria) in the previous 6 months and had an ECOG performance status of 0-2, were enrolled at 16 third-level hospitals with expertise in sarcoma care in Spain, Italy, and France. Patients received pazopanib 800 mg once daily, taken orally without food, at least 1 h before or 2 h after a meal, until progression or intolerance. The primary endpoint of the study was overall response measured by Choi criteria in the subset of the intention-to-treat population (patients who received at least 1 month of treatment with at least one radiological assessment). All patients who received at least one dose of the study drug were included in the safety analyses. This study is registered with ClinicalTrials.gov, number NCT02066285, and with the European Clinical Trials Database, EudraCT number 2013-005456-15. FINDINGS From June 26, 2014, to Nov 24, 2016, of 40 patients assessed, 36 were enrolled (34 with malignant solitary fibrous tumour and two with dedifferentiated solitary fibrous tumour). Median follow-up was 27 months (IQR 16-31). Based on central radiology review, 18 (51%) of 35 evaluable patients had partial responses, nine (26%) had stable disease, and eight (23%) had progressive disease according to Choi criteria. Further enrolment of patients with dedifferentiated solitary fibrous tumour was stopped after detection of early and fast progressions in a planned interim analysis. 51% (95% CI 34-69) of 35 patients achieved an overall response according to Choi criteria. Ten (29%) of 35 patients died. There were no deaths related to adverse events and the most frequent grade 3 or higher adverse events were hypertension (11 [31%] of 36 patients), neutropenia (four [11%]), increased concentrations of alanine aminotransferase (four [11%]), and increased concentrations of bilirubin (three [8%]). INTERPRETATION To our knowledge, this is the first trial of pazopanib for treatment of malignant solitary fibrous tumour showing activity in this patient group. The manageable toxicity profile and the activity shown by pazopanib suggests that this drug could be an option for systemic treatment of advanced malignant solitary fibrous tumour, and provides a benchmark for future trials. FUNDING Spanish Group for Research on Sarcomas (GEIS), Italian Sarcoma Group (ISG), French Sarcoma Group (FSG), GlaxoSmithKline, and Novartis.
Collapse
Affiliation(s)
- Javier Martin-Broto
- Department of Medical Oncology, University Hospital Virgen del Rocío, Sevilla, Spain; Institute of Biomedicine of Sevilla, Sevilla, Spain.
| | - Silvia Stacchiotti
- Adult Mesenchymal and Rare Tumor Unit, Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Andres Redondo
- Department of Medical Oncology, University Hospital La Paz, Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Daniel Bernabeu
- Musculoskeletal Imaging Section, University Hospital La Paz, Madrid, Spain
| | - Enrique de Alava
- Department of Pathology, University Hospital Virgen del Rocío, Sevilla, Spain; Institute of Biomedicine of Sevilla, Sevilla, Spain; CIBERONC, Madrid, Spain
| | - Paolo G Casali
- Adult Mesenchymal and Rare Tumor Unit, Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Antonio Gutierrez
- Department of Hematology, University Hospital Son Espases, Palma, Balearic Islands, Spain
| | | | - Maria Peña-Chilet
- Clinical Bioinformatics Area, Fundación Progreso y Salud, University Hospital Virgen del Rocío, Sevilla, Spain
| | | | - Michele Biscuola
- Department of Pathology, University Hospital Virgen del Rocío, Sevilla, Spain; CIBERONC, Madrid, Spain
| | - Miguel Taron
- Institute of Biomedicine of Sevilla, Sevilla, Spain
| | - Paola Collini
- Soft Tissue and Bone Pathology, Histopathology and Pediatric Pathology Unit, Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Xavier Garcia Del Muro
- Catalan Institute of Oncology, Barcelona, Spain; Institut d'Investigació Biomédica de Bellvitge, University of Barcelona, Barcelona, Spain
| | - Giovanni Grignani
- Sarcoma Unit, Division of Medical Oncology, Candiolo Cancer Institute, FPO, IRCCS, Candiolo, Italy
| | - Sarah Dumont
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | | | | | - Nadia Hindi
- Department of Medical Oncology, University Hospital Virgen del Rocío, Sevilla, Spain; Institute of Biomedicine of Sevilla, Sevilla, Spain
| | - Ana Sebio
- Department of Medical Oncology, Sant Pau Hospital, Barcelona, Spain
| | - Joaquin Dopazo
- Clinical Bioinformatics Area, Fundación Progreso y Salud, University Hospital Virgen del Rocío, Sevilla, Spain
| | | | - Axel LeCesne
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard, Université Claude Bernard Lyon, Lyon, France
| | - Josefina Cruz
- Department of Medical Oncology, University Hospital of Canarias, Tenerife, Spain
| |
Collapse
|
18
|
Dobson THW, Gopalakrishnan V. Preclinical Models of Pediatric Brain Tumors-Forging Ahead. Bioengineering (Basel) 2018; 5:E81. [PMID: 30279402 PMCID: PMC6315787 DOI: 10.3390/bioengineering5040081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/22/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022] Open
Abstract
Approximately five out of 100,000 children from 0 to 19 years old are diagnosed with a brain tumor. These children are treated with medication designed for adults that are highly toxic to a developing brain. Those that survive are at high risk for a lifetime of limited physical, psychological, and cognitive abilities. Despite much effort, not one drug exists that was designed specifically for pediatric patients. Stagnant government funding and the lack of economic incentives for the pharmaceutical industry greatly limits preclinical research and the development of clinically applicable pediatric brain tumor models. As more data are collected, the recognition of disease sub-groups based on molecular heterogeneity increases the need for designing specific models suitable for predictive drug screening. To overcome these challenges, preclinical approaches will need continual enhancement. In this review, we examine the advantages and shortcomings of in vitro and in vivo preclinical pediatric brain tumor models and explore potential solutions based on past, present, and future strategies for improving their clinical relevancy.
Collapse
Affiliation(s)
- Tara H W Dobson
- Department of Pediatrics, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Vidya Gopalakrishnan
- Department of Pediatrics, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
- Department of Molecular & Cellular Oncology, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
- Brain Tumor Center, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
- Center for Cancer Epigenetics, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
- Graduate School of Biomedical Sciences UT-Health Science Center, Houston, TX 77030, USA.
| |
Collapse
|
19
|
Kiyuna T, Tome Y, Murakami T, Kawaguchi K, Igarashi K, Miyake K, Miyake M, Li Y, Nelson SD, Dry SM, Singh AS, Russell TA, Elliott I, Singh SR, Kanaya F, Eilber FC, Hoffman RM. Trabectedin arrests a doxorubicin-resistant PDGFRA-activated liposarcoma patient-derived orthotopic xenograft (PDOX) nude mouse model. BMC Cancer 2018; 18:840. [PMID: 30126369 PMCID: PMC6102848 DOI: 10.1186/s12885-018-4703-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 07/30/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Pleomorphic liposarcoma (PLPS) is a rare, heterogeneous and an aggressive variant of liposarcoma. Therefore, individualized therapy is urgently needed. Our recent reports suggest that trabectedin (TRAB) is effective against several patient-derived orthotopic xenograft (PDOX) mouse models. Here, we compared the efficacy of first-line therapy, doxorubicin (DOX), and TRAB in a platelet-derived growth factor receptor-α (PDGFRA)-amplified PLPS. METHODS We used a fresh sample of PLPS tumor derived from a 68-year-old male patient diagnosed with a recurrent PLPS. Subcutaneous implantation of tumor tissue was performed in a nude mouse. After three weeks of implantation, tumor tissues were isolated and cut into small pieces. To match the patient a PDGFRA-amplified PLPS PDOX was created in the biceps femoris of nude mice. Mice were randomized into three groups: Group 1 (G1), control (untreated); Group 2 (G2), DOX-treated; Group 3 (G3), TRAB-treated. Measurement was done twice a week for tumor width, length, and mouse body weight. RESULTS The PLPS PDOX showed resistance towards DOX. However, TRAB could arrest the PLPS (p < 0.05 compared to control; p < 0.05 compared to DOX) without any significant changes in body-weight. CONCLUSIONS The data presented here suggest that for the individual patient the PLPS PDOX model could specifically distinguish both effective and ineffective drugs. This is especially crucial for PLPS because effective first-line therapy is harder to establish if it is not individualized.
Collapse
Affiliation(s)
- Tasuku Kiyuna
- AntiCancer Inc., San Diego, CA, USA.,Department of Surgery, University of California, San Diego, CA, USA.,Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Yasunori Tome
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan.
| | - Takashi Murakami
- AntiCancer Inc., San Diego, CA, USA.,Department of Surgery, University of California, San Diego, CA, USA
| | - Kei Kawaguchi
- AntiCancer Inc., San Diego, CA, USA.,Department of Surgery, University of California, San Diego, CA, USA
| | - Kentaro Igarashi
- AntiCancer Inc., San Diego, CA, USA.,Department of Surgery, University of California, San Diego, CA, USA
| | - Kentaro Miyake
- AntiCancer Inc., San Diego, CA, USA.,Department of Surgery, University of California, San Diego, CA, USA
| | - Masuyo Miyake
- AntiCancer Inc., San Diego, CA, USA.,Department of Surgery, University of California, San Diego, CA, USA
| | - Yunfeng Li
- Department of Surgery, University of California, Los Angeles, CA, USA
| | - Scott D Nelson
- Department of Surgery, University of California, Los Angeles, CA, USA
| | - Sarah M Dry
- Department of Surgery, University of California, Los Angeles, CA, USA
| | - Arun S Singh
- Division of Hematology-Oncology, University of California, Los Angeles, CA, USA
| | - Tara A Russell
- Division of Surgical Oncology, University of California, Los Angeles, CA, USA
| | - Irmina Elliott
- Division of Surgical Oncology, University of California, Los Angeles, CA, USA
| | - Shree Ram Singh
- Basic Research Laboratory, National Cancer Institute, Frederick, MD, USA.
| | - Fuminori Kanaya
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Fritz C Eilber
- Division of Surgical Oncology, University of California, Los Angeles, CA, USA.
| | - Robert M Hoffman
- AntiCancer Inc., San Diego, CA, USA. .,Department of Surgery, University of California, San Diego, CA, USA.
| |
Collapse
|
20
|
Ray-Coquard I, Serre D, Reichardt P, Martín-Broto J, Bauer S. Options for treating different soft tissue sarcoma subtypes. Future Oncol 2018; 14:25-49. [DOI: 10.2217/fon-2018-0076] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Management of soft tissue sarcoma is increasingly subtype-dependent. Surgery is recommended for uterine leiomyosarcoma, with trabectedin being the preferred option for advanced disease when the treatment goal is long-term tumor stabilization. Liposarcoma subgroups are characterized by distinctive morphologies and genetics, different patterns of disease progression and clinical behavior, and variable responses to treatment. Genetic analysis of sarcomas has provided insights into pathogenesis with potential for developing new molecular targets. At the cytogenetic level, soft tissue sarcomas are categorized into specific, balanced translocations and those due to massive chromosomal rearrangements. For subtypes such as undifferentiated sarcomas, angiosarcomas, alveolar soft part sarcomas and clear cell sarcomas, evidence is especially limited, although it is known that these tumors display markedly different sensitivities to chemotherapeutic and targeted agents.
Collapse
Affiliation(s)
- Isabelle Ray-Coquard
- GINECO/TMRO and NETSARC Network, Center Léon Bérard & Université Claude Bernard, Lyon, France
| | - Delphine Serre
- GINECO/TMRO and NETSARC Network, Center Léon Bérard & Université Claude Bernard, Lyon, France
| | - Peter Reichardt
- Department of Oncology, Helios Klinikum Berlin-Buch, Schwanebecker Chaussee 50, 13125, Berlin, Germany
| | - Javier Martín-Broto
- Medical Oncology Department, Hospital Virgen del Rocio & Instituto Biomedicina de Sevilla (IBIS), Sevilla, Spain
| | - Sebastian Bauer
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
21
|
Lu W, Chao T, Ruiqi C, Juan S, Zhihong L. Patient-derived xenograft models in musculoskeletal malignancies. J Transl Med 2018; 16:107. [PMID: 29688859 PMCID: PMC5913806 DOI: 10.1186/s12967-018-1487-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/17/2018] [Indexed: 12/17/2022] Open
Abstract
Successful oncological drug development for bone and soft tissue sarcoma is grossly stagnating. A major obstacle in this process is the lack of appropriate animal models recapitulating the complexity and heterogeneity of musculoskeletal malignancies, resulting in poor efficiency in translating the findings of basic research to clinical applications. In recent years, patient-derived xenograft (PDX) models generated by directly engrafting patient-derived tumor fragments into immunocompromised mice have recaptured the attention of many researchers due to their properties of retaining the principle histopathology, biological behaviors, and molecular and genetic characteristics of the original tumor, showing promising future in both basic and clinical studies of bone and soft tissue sarcoma. Despite several limitations including low take rate and long take time in PDX generation, deficient immune system and heterologous tumor microenvironment of the host, PDXs offer a more advantageous platform for preclinical drug screening, biomarker identification and clinical therapeutic decision guiding. Here, we provide a timely review of the establishment and applications of PDX models for musculoskeletal malignancies and discuss current challenges and future directions of this approach.
Collapse
Affiliation(s)
- Wan Lu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410010, Hunan, People's Republic of China
| | - Tu Chao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410010, Hunan, People's Republic of China
| | - Chen Ruiqi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410010, Hunan, People's Republic of China
| | - Su Juan
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410010, Hunan, People's Republic of China
| | - Li Zhihong
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410010, Hunan, People's Republic of China.
| |
Collapse
|
22
|
Miserocchi G, Mercatali L, Liverani C, De Vita A, Spadazzi C, Pieri F, Bongiovanni A, Recine F, Amadori D, Ibrahim T. Management and potentialities of primary cancer cultures in preclinical and translational studies. J Transl Med 2017; 15:229. [PMID: 29116016 PMCID: PMC5688825 DOI: 10.1186/s12967-017-1328-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/27/2017] [Indexed: 02/07/2023] Open
Abstract
The use of patient-derived primary cell cultures in cancer preclinical assays has increased in recent years. The management of resected tumor tissue remains complex and a number of parameters must be respected to obtain complete sample digestion and optimal vitality yield. We provide an overview of the benefits of correct primary cell culture management using different preclinical methodologies, and describe the pros and cons of this model with respect to other kinds of samples. One important advantage is that the heterogeneity of the cell populations composing a primary culture partially reproduces the tumor microenvironment and crosstalk between malignant and healthy cells, neither of which is possible with cell lines. Moreover, the use of patient-derived specimens in innovative preclinical technologies, such as 3D systems or bioreactors, represents an important opportunity to improve the translational value of the results obtained. In vivo models could further our understanding of the crosstalk between tumor and other tissues as they enable us to observe the systemic and biological interactions of a complete organism. Although engineered mice are the most common model used in this setting, the zebrafish (Danio rerio) species has recently been recognized as an innovative experimental system. In fact, the transparent body and incomplete immune system of zebrafish embryos are especially useful for evaluating patient-derived tumor tissue interactions in healthy hosts. In conclusion, ex vivo systems represent an important tool for cancer research, but samples require correct manipulation to maximize their translational value.
Collapse
Affiliation(s)
- Giacomo Miserocchi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Laura Mercatali
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy.
| | - Chiara Liverani
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Alessandro De Vita
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Chiara Spadazzi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Federica Pieri
- Pathology Unit, Morgagni-Pierantoni Hospital, Via Carlo Forlanini 34, 47121, Forlì, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Federica Recine
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Dino Amadori
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| |
Collapse
|
23
|
Martín-Broto J, Reichardt P, Stacchiotti S, Blay JY. Review of past and present clinical cases with a view to future treatment options. Future Oncol 2017; 13:11-28. [DOI: 10.2217/fon-2017-0120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In the 10 years that have elapsed since trabectedin was first granted approval in Europe for treatment of advanced soft tissue sarcoma (STS), other cytotoxic agents and targeted therapies have been approved and numerous randomized controlled trials have been completed or are underway. As an academic exercise and positive proof of the advances in STS management that have occurred during the past decade, it is interesting to compare current and future treatment approaches. In this review, present and future treatment approaches are examined by case study for three STS subtypes: uterine leiomyosarcoma, abdominal dedifferentiated liposarcoma and malignant solitary fibrous tumor.
Collapse
Affiliation(s)
| | - Peter Reichardt
- Department of Interdisciplinary Oncology, HELIOS Klinikum Berlin-Buch, Schwanebecker Chaussee 50, 13125 Berlin, Germany
| | - Silvia Stacchiotti
- Medical Oncology Unit 2 – Adult Mesenchymal Tumors & Rare Cancers, Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| |
Collapse
|