1
|
Lesovaya EA, Fetisov TI, Bokhyan BY, Maksimova VP, Kulikov EP, Belitsky GA, Kirsanov KI, Yakubovskaya MG. Genetic and Molecular Heterogeneity of Synovial Sarcoma and Associated Challenges in Therapy. Cells 2024; 13:1695. [PMID: 39451213 PMCID: PMC11506332 DOI: 10.3390/cells13201695] [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: 09/10/2024] [Revised: 09/26/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
Synovial sarcoma (SS) is one of the most common types of pediatric soft tissue sarcoma (STS) being far less frequent in adults. This STS type is characterized by one specific chromosomal translocation SS18-SSX and the associated changes in signaling. However, other genetic and epigenetic abnormalities in SS do not necessarily include SS18-SSX-related events, but abnormalities are more sporadic and do not correlate well with the prognosis and response to therapy. Currently, targeted therapy for synovial sarcoma includes a limited range of drugs, and surgical resection is the mainstay treatment for localized cancer with adjuvant or neoadjuvant chemotherapy and radiotherapy. Understanding the molecular characteristics of synovial sarcoma subtypes is becoming increasingly important for detecting new potential targets and developing innovative therapies. Novel approaches to treating synovial sarcoma include immune-based therapies (such as TCR-T cell therapy to NY-ESO-1, MAGE4, PRAME or using immune checkpoint inhibitors), epigenetic modifiers (HDAC inhibitors, EZH2 inhibitors, BRD disruptors), as well as novel or repurposed receptor tyrosine kinase inhibitors. In the presented review, we aimed to summarize the genetic and epigenetic landscape of SS as well as to find out the potential niches for the development of novel diagnostics and therapies.
Collapse
Affiliation(s)
- Ekaterina A. Lesovaya
- Department of Chemical Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (G.A.B.); (K.I.K.)
- Oncology Department, Ryazan State Medical University Named after Academician I.P. Pavlov, Ministry of Health of Russia, Ryazan 390026, Russia;
- Institute of Medicine, RUDN University, Moscow 117198, Russia
| | - Timur I. Fetisov
- Department of Chemical Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (G.A.B.); (K.I.K.)
| | - Beniamin Yu. Bokhyan
- Department of Chemical Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (G.A.B.); (K.I.K.)
| | - Varvara P. Maksimova
- Department of Chemical Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (G.A.B.); (K.I.K.)
| | - Evgeny P. Kulikov
- Oncology Department, Ryazan State Medical University Named after Academician I.P. Pavlov, Ministry of Health of Russia, Ryazan 390026, Russia;
| | - Gennady A. Belitsky
- Department of Chemical Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (G.A.B.); (K.I.K.)
| | - Kirill I. Kirsanov
- Department of Chemical Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (G.A.B.); (K.I.K.)
- Institute of Medicine, RUDN University, Moscow 117198, Russia
| | - Marianna G. Yakubovskaya
- Department of Chemical Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (G.A.B.); (K.I.K.)
- Institute of Medicine, RUDN University, Moscow 117198, Russia
| |
Collapse
|
2
|
Mastoraki A, Schizas D, Vlachou P, Melissaridou NM, Charalampakis N, Fioretzaki R, Kole C, Savvidou O, Vassiliu P, Pikoulis E. Assessment of Synergistic Contribution of Histone Deacetylases in Prognosis and Therapeutic Management of Sarcoma. Mol Diagn Ther 2020; 24:557-569. [PMID: 32696211 DOI: 10.1007/s40291-020-00487-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sarcomas are a rare group of neoplasms with a mesenchymal origin that are mainly characterized by the abnormal growth of connective tissue cells. The standard treatment for local control of sarcomas includes surgery and radiation, while for adjuvant and palliative therapy, chemotherapy has been strongly recommended. Despite the availability of multimodal therapies, the survival rate for patients with sarcoma is still not satisfactory. In recent decades, there has been a considerable effort to overcome chemotherapy resistance in sarcoma cells. This has led to the investigation of more cellular compounds implicated in gene expression and transcription processes. Furthermore, it has been discovered that histone acetylation/deacetylation equilibrium is affected in carcinogenesis, leading to a modified chromatin structure and therefore changes in gene expression. In addition, histone deacetylase inhibition is found to play a key role in limiting the tumor burden in sarcomas, as histone deacetylase inhibitors act on well-described oncogenic signaling pathways. Histone deacetylase inhibitors disrupt the increased cell motility and invasiveness of sarcoma cells, undermining their metastatic potential. Moreover, their activity on evoking cell arrest has been extensively described, with histone deacetylase inhibitors regulating the reactivation of tumor suppressor genes and induction of apoptosis. Promoting autophagy and increasing cellular reactive oxygen species are also included in the antitumor activity of histone deacetylase inhibitors. It should be noted that many studies revealed the synergy between histone deacetylase inhibitors and other drugs, leading to the enhancement of an antitumor effect in sarcomas. Therefore, there is an urgent need for therapeutic interventions modulated according to the distinct clinical and molecular characteristics of each sarcoma subtype. It is concluded that a better understanding of histone deacetylase and histone deacetylase inhibitors could provide patients with sarcoma with more targeted and efficient therapies, which may contribute to significant improvement of their survival potential.
Collapse
Affiliation(s)
- Aikaterini Mastoraki
- Third Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 1 Rimini Street, Chaidari, Athens, Greece.
| | - Dimitrios Schizas
- First Department of Surgery, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Pigi Vlachou
- Third Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 1 Rimini Street, Chaidari, Athens, Greece
| | - Nikoleta Maria Melissaridou
- Third Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 1 Rimini Street, Chaidari, Athens, Greece
| | | | | | - Christo Kole
- First Department of Surgery, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Olga Savvidou
- First Department of Orthopedics, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Pantelis Vassiliu
- Fourth Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Emmanouil Pikoulis
- Third Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 1 Rimini Street, Chaidari, Athens, Greece
| |
Collapse
|
3
|
The Utility of NKX2.2 and TLE1 Immunohistochemistry in the Differentiation of Ewing Sarcoma and Synovial Sarcoma. Appl Immunohistochem Mol Morphol 2019; 27:174-179. [DOI: 10.1097/pai.0000000000000573] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
4
|
Xing Z, Wei L, Jiang X, Conroy J, Glenn S, Bshara W, Yu T, Pao A, Tanaka S, Kawai A, Choi C, Wang J, Liu S, Morrison C, Yu YE. Analysis of mutations in primary and metastatic synovial sarcoma. Oncotarget 2018; 9:36878-36888. [PMID: 30627328 PMCID: PMC6305143 DOI: 10.18632/oncotarget.26416] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/16/2018] [Indexed: 02/06/2023] Open
Abstract
Synovial sarcoma is the most common pediatric non-rhabdomyosarcoma soft tissue sarcoma and accounts for about 8-10% of all soft tissue sarcoma in childhood and adolescence. The presence of a chromosomal translocation-associated SS18-SSX-fusion gene is causally linked to development of primary synovial sarcoma. Metastases occur in approximately 50-70% of synovial sarcoma cases with yet unknown mechanisms, which led to about 70-80% mortality rate in five years. To explore the possibilities to investigate metastatic mechanisms of synovial sarcoma, we carried out the first genome-wide search for potential genetic biomarkers and drivers associated with metastasis by comparative mutational profiling of 18 synovial sarcoma samples isolated from four patients carrying the primary tumors and another four patients carrying the metastatic tumors through whole exome sequencing. Selected from the candidates yielded from this effort, we examined the effect of the multiple missense mutations of ADAM17, which were identified solely in metastatic synovial sarcoma. The mutant alleles as well as the wild-type control were expressed in the mammalian cells harboring the SS18-SSX1 fusion gene. The ADAM17-P729H mutation was shown to enhance cell migration, a phenotype associated with metastasis. Therefore, like ADAM17-P729H, other mutations we identified solely in metastatic synovial sarcoma may also have the potential to serve as an entry point for unraveling the metastatic mechanisms of synovial sarcoma.
Collapse
Affiliation(s)
- Zhuo Xing
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program, Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Lei Wei
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Xiaoling Jiang
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program, Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Jeffrey Conroy
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.,OmniSeq Inc., Buffalo, NY, USA
| | - Sean Glenn
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.,OmniSeq Inc., Buffalo, NY, USA
| | - Wiam Bshara
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Tao Yu
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program, Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Medical Genetics, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Annie Pao
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program, Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Shinya Tanaka
- Department of Cancer Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Christopher Choi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Carl Morrison
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.,OmniSeq Inc., Buffalo, NY, USA.,Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Y Eugene Yu
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program, Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Genetics, Genomics and Bioinformatics Program, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
5
|
Natarajan V, Ramanathan P, Gopisetty G, Ramachandran B, Thangarajan R, Kesavan S. In silico and in vitro screening of small molecule Inhibitors against SYT-SSX1 fusion protein in synovial sarcoma. Comput Biol Chem 2018; 77:36-43. [PMID: 30219714 DOI: 10.1016/j.compbiolchem.2018.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 09/04/2018] [Accepted: 09/04/2018] [Indexed: 12/30/2022]
Abstract
Synovial sarcoma (SS) is characterized by a tumour specific chromosomal translocation t(X;18) (p11;q11) which results in the formation of SYT-SSX1 fusion protein. This fusion protein represents a clear therapeutic target and molecules specifically targeting SYT-SSX1 fusion protein are currently not available. In this study, SYT-SSX1 fusion protein sequence was retrieved from Uniprot and 3D structure was generated using I-TASSER modeling program. A structure based computational screening approach has been employed using Glide docking software to identify potential SYT-SSX1 small molecule inhibitors that bind to the junction region of the fusion protein. The obtained inhibitors were further filtered based on the docking score and ADME/T properties. Ten best fit compounds were chosen for in vitro studies. The anti-proliferative activities of these 10 compounds were screened in Yamato, ASKA (carries SYT-SSX1 fusion protein) and other sarcoma cell lines such as A673, 143B to understand the specificity of inhibition of the chosen compounds. The in vitro activity was compared against HEK293 cell lines. The compound 5-fluoro-3-(1-phenyl-1H-tetraazol-5-yl)-1H-indole (FPTI) was found to be selectively cytotoxic in synovial sarcoma cell lines (Yamato and ASKA) and this compound also showed insignificant anti proliferative activity on other cell lines. Further, target gene expression study confirmed that FPTI treatment down-regulated SYT-SSX1 and modulated its downstream target genes. Cell cycle analysis revealed the involvement of an apoptotic mechanism of cell death. Further experimental validations may elucidate the therapeutic potentials of FPTI against SYT-SSX1 fusion protein.
Collapse
Affiliation(s)
- Valliyammai Natarajan
- Dept of Molecular Oncology, Dr. S. Krishnamurthi Campus, Cancer Institute (WIA), Guindy, Chennai, 600036, India
| | - Priya Ramanathan
- Dept of Molecular Oncology, Dr. S. Krishnamurthi Campus, Cancer Institute (WIA), Guindy, Chennai, 600036, India
| | - Gopal Gopisetty
- Dept of Molecular Oncology, Dr. S. Krishnamurthi Campus, Cancer Institute (WIA), Guindy, Chennai, 600036, India
| | - Balaji Ramachandran
- Dept of Molecular Oncology, Dr. S. Krishnamurthi Campus, Cancer Institute (WIA), Guindy, Chennai, 600036, India
| | - Rajkumar Thangarajan
- Dept of Molecular Oncology, Dr. S. Krishnamurthi Campus, Cancer Institute (WIA), Guindy, Chennai, 600036, India
| | - Sabitha Kesavan
- Dept of Molecular Oncology, Dr. S. Krishnamurthi Campus, Cancer Institute (WIA), Guindy, Chennai, 600036, India.
| |
Collapse
|
6
|
Katz D, Palmerini E, Pollack SM. More Than 50 Subtypes of Soft Tissue Sarcoma: Paving the Path for Histology-Driven Treatments. Am Soc Clin Oncol Educ Book 2018; 38:925-938. [PMID: 30231352 DOI: 10.1200/edbk_205423] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Sarcomas are a diverse group of cancers with mesenchymal origin. Although sarcomas comprise less than 1% of cancers, there are more than 50 different subtypes that are quite different from one another in terms of both their biology and clinical behavior. Historically, the need for adequate patient numbers in clinical trials has pushed sarcoma researchers to lump these very different malignancies together and treat the patients using a "one-size-fits-all" approach. However, with improvements in our scientific understanding, we are finally ready for a histology-tailored therapeutic approach to these complex diseases. In this review, we discuss key advances in our understanding of the biology underlying selected sarcoma subtypes and how targeting these subtypes is relevant therapeutically with respect to both molecularly targeted agents as well as immunotherapy.
Collapse
Affiliation(s)
- Daniela Katz
- From the Institute of Oncology, Assaf Harofeh Medical Center, Zrifin, Beer Yaakov, Israel; Medical Oncology, Istituto Ortopedico Rizzoli, Department of Experimental, Diagnostic, and Specialty Medicine, Bologna University, Bologna, Italy; Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
| | - Emanuela Palmerini
- From the Institute of Oncology, Assaf Harofeh Medical Center, Zrifin, Beer Yaakov, Israel; Medical Oncology, Istituto Ortopedico Rizzoli, Department of Experimental, Diagnostic, and Specialty Medicine, Bologna University, Bologna, Italy; Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
| | - Seth M Pollack
- From the Institute of Oncology, Assaf Harofeh Medical Center, Zrifin, Beer Yaakov, Israel; Medical Oncology, Istituto Ortopedico Rizzoli, Department of Experimental, Diagnostic, and Specialty Medicine, Bologna University, Bologna, Italy; Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
| |
Collapse
|
7
|
Feng Q, Guo P, Wang J, Zhang X, Yang HC, Feng JG. High expression of SDF-1 and VEGF is associated with poor prognosis in patients with synovial sarcomas. Exp Ther Med 2018; 15:2597-2603. [PMID: 29456663 DOI: 10.3892/etm.2018.5684] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 03/17/2017] [Indexed: 01/18/2023] Open
Abstract
Stromal cell-derived factor-1 (SDF-1) predicts poor clinical outcomes of certain types of cancer. Furthermore, vascular endothelial growth factor (VEGF) promotes the growth and metastasis of solid tumors. The aim of the present study was to examine the expression of SDF-1 and VEGF in patients with synovial sarcoma and to determine their expression is correlated with unfavorable outcomes. Levels of SDF-1 and VEGF proteins were evaluated in 54 patients with synovial sarcoma using immunohistochemical and immunofluorescence staining. Potential associations between the expression of SDF-1 and VEGF and various clinical parameters were analyzed using Pearson's χ2 test and the Spearman-rho test. Additionally, univariate and multivariate Cox regression analyses were used to identify potential prognostic factors, and the Kaplan-Meier method was used to analyze the overall survival rates of patients. Low SDF-1 and VEGF expression was detected in 20.4% (11/54) and 22.2% (12/54) of patients with synovial sarcoma; moderate expression was detected in 35.2% (19/54) and 37.0% (20/54) of patients and high expression was detected in 44.4% (24 of 54) and 40.7% (22 of 54) of patients, respectively. Levels of SDF-1 and VEGF proteins were significantly associated with histological grade (P<0.05), metastasis (P<0.05) and American Joint Committee on Cancer staging (P<0.05). In addition, levels of SDF-1 and VEGF expression were positively correlated with each other (P<0.001). Univariate analysis also indicated that VEGF expression was associated with shorter overall survival rates in (P<0.05), whereas multivariate analysis demonstrated that SDF-1 expression was associated with shorter patient survival rates (P<0.05). Finally, both SDF-1 and VEGF expression were associated with various characteristics of synovial sarcoma. Therefore, SDF-1 expression may be a potential independent prognostic indicator in patients with synovial sarcomas.
Collapse
Affiliation(s)
- Qi Feng
- Department of Orthopedics, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Peng Guo
- Department of Orthopedics, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jin Wang
- Department of Orthopedics, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Xiaoyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Hui-Chai Yang
- Department of Pathology, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jian-Gang Feng
- Department of Orthopedics, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
8
|
Prince AC, McGee AS, Siegel H, Rosenthal EL, Behnke NK, Warram JM. Evaluation of fluorescence-guided surgery agents in a murine model of soft tissue fibrosarcoma. J Surg Oncol 2017; 117:1179-1187. [PMID: 29284070 DOI: 10.1002/jso.24950] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 11/10/2017] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND OBJECTIVES Soft tissue sarcomas (STS) are mesenchymal malignancies. Treatment mainstay is surgical resection with negative margins ± adjuvant treatment. Fluorescence-guided surgical (FGS) resection can delineate intraoperative margins; FGS has improved oncologic outcomes in other malignancies. This novel strategy may minimize resection-associated morbidity while improving local tumor control. METHODS We evaluate the tumor-targeting specificity and utility of fluorescence-imaging agents to provide disease-specific contrast. Mice with HT1080 fibrosarcoma tumors received one of five probes: cetuximab-IRDye800CW (anti-EGFR), DC101-IRDye800CW (anti-VEGFR-2), IgG-IRDye800CW, the cathepsin-activated probe Prosense750EX, or the small molecule probe IntegriSense750. Tumors were imaged daily using open- and closed-field fluorescence imaging systems. Tumor-to-background ratios (TBR) were evaluated. On peak TBR days, probe sensitivity was evaluated. Tumors were stained and imaged microscopically. RESULTS At peak, closed-field imaging TBR of cetuximab-IRDye800CW (16.8) was significantly greater (P < 0.0001) than Integrisense750 (7.0), Prosense750EX (5.8), and DC101-IRDye800CW (3.7). All agents successfully localized as little as 1.0 mg of tumor tissue in the post-resection bed; cetuximab-IRDye800CW generated the greatest contrast (2.5). Cetuximab-IRDye800CW revealed strong tumor affinity microscopically; tumor fluorescence intensity was significantly greater (P < 0.0004) than 0.2 mm away from tumor border. CONCLUSION This study demonstrates cetuximab-IRDye800CW superiority. FGS has the potential to improve post-resection morbidity and mortality by improving disease detection.
Collapse
Affiliation(s)
- Andrew C Prince
- University of Alabama School of Medicine, Birmingham, Alabama
| | - Andrew S McGee
- University of Alabama School of Medicine, Birmingham, Alabama
| | - Herrick Siegel
- Department of Orthopedic Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Eben L Rosenthal
- Department of Otolaryngology, Stanford University, Stanford, California
| | - Nicole K Behnke
- Department of Orthopedic Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jason M Warram
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
9
|
Cassinelli G, Dal Bo L, Favini E, Cominetti D, Pozzi S, Tortoreto M, De Cesare M, Lecis D, Scanziani E, Minoli L, Naggi A, Vlodavsky I, Zaffaroni N, Lanzi C. Supersulfated low-molecular weight heparin synergizes with IGF1R/IR inhibitor to suppress synovial sarcoma growth and metastases. Cancer Lett 2017; 415:187-197. [PMID: 29225052 DOI: 10.1016/j.canlet.2017.12.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 01/26/2023]
Abstract
Synovial sarcoma (SS) is an aggressive tumor with propensity for lung metastases which significantly impact patients' prognosis. New therapeutic approaches are needed to improve treatment outcome. Targeting the heparanase/heparan sulfate proteoglycan system by heparin derivatives which act as heparanase inhibitors/heparan sulfate mimetics is emerging as a therapeutic approach that can sensitize the tumor response to chemotherapy. We investigated the therapeutic potential of a supersulfated low molecular weight heparin (ssLMWH) in preclinical models of SS. ssLMWH showed a potent anti-heparanase activity, dose-dependently inhibited SS colony growth and cell invasion, and downregulated the activation of receptor tyrosine kinases including IGF1R and IR. The combination of ssLMWH and the IGF1R/IR inhibitor BMS754807 synergistically inhibited proliferation of cells exhibiting IGF1R hyperactivation, also abrogating cell motility and promoting apoptosis in association with PI3K/AKT pathway inhibition. The drug combination strongly enhanced the antitumor effect against the CME-1 model, as compared to single agent treatment, abrogating orthotopic tumor growth and significantly repressing spontaneous lung metastatic dissemination in treated mice. These findings provide a strong preclinical rationale for developing drug regimens combining heparanase inhibitors/HS mimetics with IGF1R antagonists for treatment of metastatic SS.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Apoptosis/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Drug Synergism
- Glucuronidase/antagonists & inhibitors
- Glucuronidase/metabolism
- Heparin, Low-Molecular-Weight/administration & dosage
- Heparin, Low-Molecular-Weight/metabolism
- Heparin, Low-Molecular-Weight/pharmacology
- Humans
- Mice, SCID
- Neoplasm Metastasis
- Pyrazoles/administration & dosage
- Pyrazoles/pharmacology
- Receptor, IGF Type 1
- Receptors, Somatomedin/antagonists & inhibitors
- Receptors, Somatomedin/metabolism
- Sarcoma, Synovial/drug therapy
- Sarcoma, Synovial/metabolism
- Sarcoma, Synovial/pathology
- Sulfates
- Triazines/administration & dosage
- Triazines/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Giuliana Cassinelli
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| | - Laura Dal Bo
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Enrica Favini
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Denis Cominetti
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Sabina Pozzi
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Monica Tortoreto
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Michelandrea De Cesare
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Daniele Lecis
- Research Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Eugenio Scanziani
- Department of Veterinary Medicine, Università Degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; Mouse and Animal Pathology Laboratory, Fondazione Filarete, Viale Ortles 22/4, 20139 Milan, Italy
| | - Lucia Minoli
- Department of Veterinary Medicine, Università Degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; Mouse and Animal Pathology Laboratory, Fondazione Filarete, Viale Ortles 22/4, 20139 Milan, Italy
| | - Annamaria Naggi
- G. Ronzoni Institute for Chemical and Biochemical Research, Via G. Colombo 81, 20133 Milan, Italy
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research Center, Bruce Rappaport Faculty of Medicine, Technion, P.O. Box 9649, Haifa 31096, Israel
| | - Nadia Zaffaroni
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Cinzia Lanzi
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| |
Collapse
|