51
|
Jacobs JE, Davis L, McWeeney S. Single nucleotide variants in nuclear pore complex disassembly pathway associated with poor survival in osteosarcoma. Front Genet 2024; 15:1303404. [PMID: 38562379 PMCID: PMC10982431 DOI: 10.3389/fgene.2024.1303404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction The bone tumor, osteosarcoma, remains challenging to treat in children and young adults, especially when patients present with metastatic disease. Developing new therapies based on genomic data from sequencing projects has proven difficult given the lack of recurrent genetic lesions across tumors. MYC overexpression has been associated with poor outcomes in osteosarcoma. However, other genomic markers of disease severity are lacking. Materials and Methods We utilized whole genome sequencing of 106 tumors and matched normal controls in order to define genomic characteristics that correlate with overall survival. Single nucleotide variants were overlaid onto annotated molecular pathways in order to define aberrant pathway signatures specific to aggressive osteosarcoma. Additionally, we calculated differential gene expression in a subsample of 71 tumors. Differentially expressed genes were then queried for known MYC-responsive genes. Results Molecular pathways specific to nuclear pore complex disassembly (NPCD) show significant correlation with poor overall survival in osteosarcoma when mutations were present. Genes involved in immune response and immune regulation are enriched in the differential expression analysis of samples with and without NPCD pathway aberrations. Furthermore, neither MYC nor MYC-responsive genes show differential expression between NPCD-aberrant and non-aberrant groups. The NPCD pathway mutations are dominated by regulatory region variants rather than protein-altering mutations, suggesting that dysregulation of genetic regulatory networks may be the underlying mechanism for their relation to osteosarcoma phenotype. Discussion Overall survival is significantly worse in patients whose tumors show aberrations in the NPCD pathway. Moreover, this difference in survival is not driven by MYC-overexpression, suggesting a novel mechanism for some aggressive osteosarcomas. These findings add light to the evolving understanding of the drivers of osteosarcoma and may aid in the search for new treatments based on patient-specific genetic data.
Collapse
Affiliation(s)
- James E. Jacobs
- Oregon Health & Science University, Portland, OR, United States
| | | | | |
Collapse
|
52
|
Wang J, Ferrena A, Zhang R, Singh S, Viscarret V, Al-Harden W, Aldahamsheh O, Borjihan H, Singla A, Yaguare S, Tingling J, Zi X, Lo Y, Gorlick R, Schwartz EL, Zhao H, Yang R, Geller DS, Zheng D, Hoang BH. Targeted inhibition of SCF SKP2 confers anti-tumor activities resulting in a survival benefit in osteosarcoma. Oncogene 2024; 43:962-975. [PMID: 38355807 PMCID: PMC10959747 DOI: 10.1038/s41388-024-02942-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024]
Abstract
Osteosarcoma(OS) is a highly aggressive bone cancer for which treatment has remained essentially unchanged for decades. Although OS is characterized by extensive genomic heterogeneity and instability, RB1 and TP53 have been shown to be the most commonly inactivated tumor suppressors in OS. We previously generated a mouse model with a double knockout (DKO) of Rb1 and Trp53 within cells of the osteoblastic lineage, which largely recapitulates human OS with nearly complete penetrance. SKP2 is a repression target of pRb and serves as a substrate recruiting subunit of the SCFSKP2 complex. In addition, SKP2 plays a central role in regulating the cell cycle by ubiquitinating and promoting the degradation of p27. We previously reported the DKOAA transgenic model, which harbored a knock-in mutation in p27 that impaired its binding to SKP2. Here, we generated a novel p53-Rb1-SKP2 triple-knockout model (TKO) to examine SKP2 function and its potential as a therapeutic target in OS. First, we observed that OS tumorigenesis was significantly delayed in TKO mice and their overall survival was markedly improved. In addition, the loss of SKP2 also promoted an apoptotic microenvironment and reduced the stemness of DKO tumors. Furthermore, we found that small-molecule inhibitors of SKP2 exhibited anti-tumor activities in vivo and in OS organoids as well as synergistic effects when combined with a standard chemotherapeutic agent. Taken together, our results suggest that SKP2 inhibitors may reduce the stemness plasticity of OS and should be leveraged as next-generation adjuvants in this cancer.
Collapse
Affiliation(s)
- Jichuan Wang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Musculoskleletal Tumor Center, Beijing Key Laboratory for Musculoskeletal Tumors, Peking University People's Hospital, Beijing, China
| | - Alexander Ferrena
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Clinical and Translational Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ranxin Zhang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Swapnil Singh
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Valentina Viscarret
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Waleed Al-Harden
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Osama Aldahamsheh
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Orthopedic Department, Al-Balqa Applied University, As-Salt, Jordan
| | - Hasibagan Borjihan
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Amit Singla
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Simon Yaguare
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Janet Tingling
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xiaolin Zi
- Department of Urology, University of California, Irvine Medical Center, Orange, CA, USA
| | - Yungtai Lo
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Richard Gorlick
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edward L Schwartz
- Departments of Oncology, Molecular Pharmacology, and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hongling Zhao
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rui Yang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David S Geller
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Deyou Zheng
- Departments of Genetics, Neurology and Neuroscience. Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Bang H Hoang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
53
|
Schott CR, Koehne AL, Sayles LC, Young EP, Luck C, Yu K, Lee AG, Breese MR, Leung SG, Xu H, Shah AT, Liu HY, Spillinger A, Behroozfard IH, Marini KD, Dinh PT, Pons Ventura MV, Vanderboon EN, Hazard FK, Cho SJ, Avedian RS, Mohler DG, Zimel M, Wustrack R, Curtis C, Sirota M, Sweet-Cordero EA. Osteosarcoma PDX-Derived Cell Line Models for Preclinical Drug Evaluation Demonstrate Metastasis Inhibition by Dinaciclib through a Genome-Targeted Approach. Clin Cancer Res 2024; 30:849-864. [PMID: 37703185 PMCID: PMC10870121 DOI: 10.1158/1078-0432.ccr-23-0873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 03/26/2023] [Accepted: 08/08/2023] [Indexed: 09/15/2023]
Abstract
PURPOSE Models to study metastatic disease in rare cancers are needed to advance preclinical therapeutics and to gain insight into disease biology. Osteosarcoma is a rare cancer with a complex genomic landscape in which outcomes for patients with metastatic disease are poor. As osteosarcoma genomes are highly heterogeneous, multiple models are needed to fully elucidate key aspects of disease biology and to recapitulate clinically relevant phenotypes. EXPERIMENTAL DESIGN Matched patient samples, patient-derived xenografts (PDX), and PDX-derived cell lines were comprehensively evaluated using whole-genome sequencing and RNA sequencing. The in vivo metastatic phenotype of the PDX-derived cell lines was characterized in both an intravenous and an orthotopic murine model. As a proof-of-concept study, we tested the preclinical effectiveness of a cyclin-dependent kinase inhibitor on the growth of metastatic tumors in an orthotopic amputation model. RESULTS PDXs and PDX-derived cell lines largely maintained the expression profiles of the patient from which they were derived despite the emergence of whole-genome duplication in a subset of cell lines. The cell lines were heterogeneous in their metastatic capacity, and heterogeneous tissue tropism was observed in both intravenous and orthotopic models. Single-agent dinaciclib was effective at dramatically reducing the metastatic burden. CONCLUSIONS The variation in metastasis predilection sites between osteosarcoma PDX-derived cell lines demonstrates their ability to recapitulate the spectrum of the disease observed in patients. We describe here a panel of new osteosarcoma PDX-derived cell lines that we believe will be of wide use to the osteosarcoma research community.
Collapse
Affiliation(s)
- Courtney R. Schott
- Department of Pediatrics, University of California San Francisco, San Francisco, California
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Amanda L. Koehne
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Leanne C. Sayles
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Elizabeth P. Young
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Cuyler Luck
- Department of Pediatrics, University of California San Francisco, San Francisco, California
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, California
| | - Katherine Yu
- Department of Pediatrics, University of California San Francisco, San Francisco, California
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, California
| | - Alex G. Lee
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Marcus R. Breese
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Stanley G. Leung
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Hang Xu
- Departments of Genetics and Medicine, Stanford University School of Medicine, Stanford University, Stanford, California
| | - Avanthi Tayi Shah
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Heng-Yi Liu
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Aviv Spillinger
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Inge H. Behroozfard
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Kieren D. Marini
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Phuong T. Dinh
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - María V. Pons Ventura
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Emma N. Vanderboon
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Florette K. Hazard
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, California
| | - Soo-Jin Cho
- Department of Pathology, University of California San Francisco, San Francisco, California
| | - Raffi S. Avedian
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford University, Stanford, California
| | - David G. Mohler
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford University, Stanford, California
| | - Melissa Zimel
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California
| | - Rosanna Wustrack
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California
| | - Christina Curtis
- Departments of Genetics and Medicine, Stanford University School of Medicine, Stanford University, Stanford, California
| | - Marina Sirota
- Department of Pediatrics, University of California San Francisco, San Francisco, California
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, California
| | | |
Collapse
|
54
|
Zhang Y, Lu G, Guan Y, Xu T, Duan Z, Li G. LINC00960 affects osteosarcoma treatment and prognosis by regulating the tumor immune microenvironment. Heliyon 2024; 10:e24990. [PMID: 38352756 PMCID: PMC10862516 DOI: 10.1016/j.heliyon.2024.e24990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 11/29/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024] Open
Abstract
Background Osteosarcoma (OS), the commonest primary malignant bone tumor, is mainly seen in children and teenagers. LINC00960, a newly discovered long intergenic non-protein coding RNA, has been shown to be important in certain cancers. The objective of this study was to assess LINC00960's prognostic and therapeutic value and analyze its mechanism of action in osteosarcoma. Methods With the transcriptome information of 85 osteosarcomas from the TARGET database, the Cox regression analyses, K-M curve, and ROC curve, were conducted for survival and prognostic analysis. The functional analysis was conducted using GO, KEGG, GSEA, and GSVA. The ESTIMATE, ssGSEA, MCP-counter, ImmuCellAI algorithms, and immune checkpoint correlation analysis were performed for immune-related analysis. The single-cell RNA sequencing data of 6 osteosarcoma patients was obtained from the Gene Expression Omnibus database. The Tumor Immune Dysfunction and Exclusion algorithm and the "pRRophetic" R package were performed to predict the response to immunotherapy and chemotherapy. Results LINC00960 overexpression is associated with osteosarcoma metastasis and poor prognosis. Based on the LINC00960 expression, the nomogram prediction model was created, which showed good accuracy and precision to predict the overall survival of osteosarcoma. Single-cell and immune-related analysis showed that LINC00960 is mainly highly expressed in the tumor-exhausted CD8 T cells in osteosarcoma. In osteosarcoma, the expression of LIC00960 was favorably connected with immune checkpoint-related genes and negatively correlated with immune infiltration. TIDE analysis indicated that low LINC00960 expression patients might have a better response to immunotherapy. Drug sensitivity analysis showed that high LINC00960 expression patients might have better responses to Bleomycin and Doxorubicin. Conclusion LINC00960 has the potential to be a novel biomarker for predicting overall survival in osteosarcoma patients and to guide more individualized treatment and clinical decision-making.
Collapse
Affiliation(s)
- Yiwei Zhang
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Guanghua Lu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yonghao Guan
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Tianyang Xu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Zhengwei Duan
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Guodong Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| |
Collapse
|
55
|
Xia M, Tong S, Gao L. Identification of MDK as a Hypoxia- and Epithelial-Mesenchymal Transition-Related Gene Biomarker of Glioblastoma Based on a Novel Risk Model and In Vitro Experiments. Biomedicines 2024; 12:92. [PMID: 38255198 PMCID: PMC10813330 DOI: 10.3390/biomedicines12010092] [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: 09/19/2023] [Revised: 11/23/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Tumor cells are commonly exposed to a hypoxic environment, which can easily induce the epithelial-mesenchymal transition (EMT) of tumor cells, further affecting tumor proliferation, invasion, metastasis, and drug resistance. However, the predictive role of hypoxia and EMT-related genes in glioblastoma (GBM) has not been investigated. METHODS Intersection genes were identified by weighted correlation network analysis (WGCNA) and differential expression analyses, and a risk model was further constructed by LASSO and Cox analyses. Clinical, immune infiltration, tumor mutation, drug treatment, and enrichment profiles were analyzed based on the risk model. The expression level of the MDK gene was tested using RT-PCR, immunohistochemistry, and immunofluorescence. CCK8 and EdU were employed to determine the GBM cells' capacity for proliferation while the migration and invasion ability were detected by a wound healing assay and transwell assay, respectively. RESULTS Based on the GBM data of the TCGA and GTEx databases, 58 intersection genes were identified, and a risk model was constructed. The model was verified in the CGGA cohort, and its accuracy was confirmed by the ROC curve (AUC = 0.807). After combining clinical subgroups, univariate and multivariate Cox regression analyses showed that risk score and age were independent risk factors for GBM patients. Furthermore, our subsequent analysis of immune infiltration, tumor mutation, and drug treatment showed that risk score and high- and low-risk groups were associated with multiple immune cells, mutated genes, and drugs. Enrichment analysis indicated that the differences between high- and low-risk groups were manifested in tumor-related pathways, including the PI3K-AKT and JAK-STAT pathways. Finally, in vivo experiments proved that the hypoxia environment promoted the expression of MDK, and MDK knockdown reduced the proliferation, migration, and EMT of GBM cells induced by hypoxia. CONCLUSIONS Our novel prognostic correlation model provided more potential treatment strategies for GBM patients.
Collapse
Affiliation(s)
- Minqi Xia
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shiao Tong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ling Gao
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
56
|
Solomon PE, Bracken CJ, Carozza JA, Wang H, Young EP, Wellner A, Liu CC, Sweet-Cordero EA, Li L, Wells JA. Discovery of VH domains that allosterically inhibit ENPP1. Nat Chem Biol 2024; 20:30-41. [PMID: 37400538 PMCID: PMC10746542 DOI: 10.1038/s41589-023-01368-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/23/2023] [Indexed: 07/05/2023]
Abstract
Ectodomain phosphatase/phosphodiesterase-1 (ENPP1) is overexpressed on cancer cells and functions as an innate immune checkpoint by hydrolyzing extracellular cyclic guanosine monophosphate adenosine monophosphate (cGAMP). Biologic inhibitors have not yet been reported and could have substantial therapeutic advantages over current small molecules because they can be recombinantly engineered into multifunctional formats and immunotherapies. Here we used phage and yeast display coupled with in cellulo evolution to generate variable heavy (VH) single-domain antibodies against ENPP1 and discovered a VH domain that allosterically inhibited the hydrolysis of cGAMP and adenosine triphosphate (ATP). We solved a 3.2 Å-resolution cryo-electron microscopy structure for the VH inhibitor complexed with ENPP1 that confirmed its new allosteric binding pose. Finally, we engineered the VH domain into multispecific formats and immunotherapies, including a bispecific fusion with an anti-PD-L1 checkpoint inhibitor that showed potent cellular activity.
Collapse
Affiliation(s)
- Paige E Solomon
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Colton J Bracken
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Cartography Biosciences, South San Francisco, CA, USA
| | - Jacqueline A Carozza
- Department of Biochemistry, Stanford University Medical School, Stanford, CA, USA
| | - Haoqing Wang
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Macromolecular Structural Knowledge Center, Stanford University, Stanford, CA, USA
| | - Elizabeth P Young
- Division of Pediatric Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Alon Wellner
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - Chang C Liu
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
- Department of Chemistry, University of California, Irvine, CA, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - E Alejandro Sweet-Cordero
- Division of Pediatric Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Lingyin Li
- Department of Biochemistry, Stanford University Medical School, Stanford, CA, USA
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA.
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
57
|
Yang YT, Engleberg AI, Yuzbasiyan-Gurkan V. Establishment and Characterization of Cell Lines from Canine Metastatic Osteosarcoma. Cells 2023; 13:25. [PMID: 38201229 PMCID: PMC10778184 DOI: 10.3390/cells13010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/15/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024] Open
Abstract
Despite the advancements in treatments for other cancers, the outcomes for osteosarcoma (OSA) patients have not improved in the past forty years, especially in metastatic patients. Moreover, the major cause of death in OSA patients is due to metastatic lesions. In the current study, we report on the establishment of three cell lines derived from metastatic canine OSA patients and their transcriptome as compared to normal canine osteoblasts. All the OSA cell lines displayed significant upregulation of genes in the epithelial mesenchymal transition (EMT) pathway, and upregulation of key cytokines such as CXCL8, CXCL10 and IL6. The two most upregulated genes are MX1 and ISG15. Interestingly, ISG15 has recently been identified as a potential therapeutic target for OSA. In addition, there is notable downregulation of cell cycle control genes, including CDKN2A, CDKN2B and THBS1. At the protein level, p16INK4A, coded by CDKN2A, was undetectable in all the canine OSA cell lines, while expression of the tumor suppressor PTEN was variable, with one cell line showing complete absence and others showing low levels of expression. In addition, the cells express a variety of actionable genes, including KIT, ERBB2, VEGF and immune checkpoint genes. These findings, similar to those reported in human OSA, point to some genes that can be used for prognosis, targeted therapies and novel drug development for both canine and human OSA patients.
Collapse
Affiliation(s)
- Ya-Ting Yang
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA; (Y.-T.Y.); (A.I.E.)
| | - Alexander I. Engleberg
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA; (Y.-T.Y.); (A.I.E.)
| | - Vilma Yuzbasiyan-Gurkan
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA; (Y.-T.Y.); (A.I.E.)
- Department of Microbiology & Molecular Genetics, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
58
|
Makielski KM, Sarver AL, Henson MS, Stuebner KM, Borgatti A, Suksanpaisan L, Preusser C, Tabaran AF, Cornax I, O’Sullivan MG, Chehadeh A, Groschen D, Bergsrud K, Pracht S, Winter A, Mills LJ, Schwabenlander MD, Wolfe M, Farrar MA, Cutter GR, Koopmeiners JS, Russell SJ, Modiano JF, Naik S. Neoadjuvant systemic oncolytic vesicular stomatitis virus is safe and may enhance long-term survivorship in dogs with naturally occurring osteosarcoma. Mol Ther Oncolytics 2023; 31:100736. [PMID: 37965295 PMCID: PMC10641240 DOI: 10.1016/j.omto.2023.100736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
Osteosarcoma is a devastating bone cancer that disproportionally afflicts children, adolescents, and young adults. Standard therapy includes surgical tumor resection combined with multiagent chemotherapy, but many patients still suffer from metastatic disease progression. Neoadjuvant systemic oncolytic virus (OV) therapy has the potential to improve clinical outcomes by targeting primary and metastatic tumor sites and inducing durable antitumor immune responses. Here we describe the first evaluation of neoadjuvant systemic therapy with a clinical-stage recombinant oncolytic vesicular stomatitis virus (VSV), VSV-IFNβ-NIS, in naturally occurring cancer, specifically appendicular osteosarcoma in companion dogs. Canine osteosarcoma has a similar natural disease history as its human counterpart. VSV-IFNβ-NIS was administered prior to standard of care surgical resection, permitting microscopic and genomic analysis of tumors. Treatment was well-tolerated and a "tail" of long-term survivors (∼35%) was apparent in the VSV-treated group, a greater proportion than observed in two contemporary control cohorts. An increase in tumor inflammation was observed in VSV-treated tumors and RNA-seq analysis showed that all the long-term responders had increased expression of a T cell anchored immune gene cluster. We conclude that neoadjuvant VSV-IFNβ-NIS is safe and may increase long-term survivorship in dogs with naturally occurring osteosarcoma, particularly those that exhibit pre-existing antitumor immunity.
Collapse
Affiliation(s)
- Kelly M. Makielski
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Aaron L. Sarver
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael S. Henson
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Kathleen M. Stuebner
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Antonella Borgatti
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | | | - Caitlin Preusser
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | - Ingrid Cornax
- Department of Veterinary Population Medicine, St. Paul, MN 55108, USA
| | | | - Andrea Chehadeh
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Donna Groschen
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Kelly Bergsrud
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Sara Pracht
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Amber Winter
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Lauren J. Mills
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Pediatrics, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA
| | - Marc D. Schwabenlander
- Veterinary Diagnostic Laboratory, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Melissa Wolfe
- Veterinary Diagnostic Laboratory, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Michael A. Farrar
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Gary R. Cutter
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joseph S. Koopmeiners
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Division of Biostatistics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Stephen J. Russell
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
- Vyriad, Inc., 2900 37th St NW, Rochester, MN 55901, USA
| | - Jaime F. Modiano
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Engineering and Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Shruthi Naik
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
- Vyriad, Inc., 2900 37th St NW, Rochester, MN 55901, USA
| |
Collapse
|
59
|
Zhang Y, Wei J, Kong L, Song M, Zhang Y, Xiao X, Cao H, Jin Y. Network pharmacology-based research on the effect of Radix Astragali on osteosarcoma and the underlying mechanism. Sci Rep 2023; 13:22315. [PMID: 38102307 PMCID: PMC10724296 DOI: 10.1038/s41598-023-49597-x] [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: 05/02/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023] Open
Abstract
To explore the anti-tumor effects of Radix Astragali on osteosarcoma and its mechanism. We analyzed the PPI network of Radix Astragali's potential targets for treating osteosarcoma and got the hub targets. We used KM curves to screen hub targets that could prolong sarcoma patients' survival time. We performed GO and KEGG enrichment analysis of Radix Astragali's potential targets and predicted Radix Astragali's molecular mechanism and function in treating osteosarcoma. The binding process between the hub targets, which could prolong sarcoma patients' survival time, and Radix Astragali was simulated through molecular docking. PPI network analysis of potential therapeutic targets discriminated 25 hub targets. The KM curves of the hub targets showed there were 13 hub targets that were effective in improving the 5-year survival rate of sarcoma patients. GO and KEGG enrichment demonstrated that Radix Astragali regulates multiple signaling pathways of osteosarcoma. Molecular docking results indicated that Radix Astragali could bind freely to the hub target, which could prolong the sarcoma patient's survival time. Radix Astragali act on osteosarcoma by regulating a signaling network formed by hub targets connecting multiple signaling pathways. Radix Astragali has the potential to become a drug for treating osteosarcoma and prolonging the sarcoma patient's survival time.
Collapse
Affiliation(s)
- Yafang Zhang
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China
| | - Junqiang Wei
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China
| | - Lingwei Kong
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China
| | - Mingze Song
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China
| | - Yange Zhang
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China
| | - Xiangyu Xiao
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China
| | - Haiying Cao
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China
| | - Yu Jin
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China.
| |
Collapse
|
60
|
Raj M, Arnav A, Pal AK, Mondal S. Global Research Trends in Limb Salvage Surgery for Osteosarcoma: Findings from a Bibliometric and Visualized Analysis over 15 Years. Indian J Orthop 2023; 57:1927-1948. [PMID: 38009167 PMCID: PMC10673777 DOI: 10.1007/s43465-023-01005-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 09/21/2023] [Indexed: 11/28/2023]
Abstract
Background Limb salvage surgery in osteosarcoma requires a multidisciplinary team of experts, due to which research interest has remained limited globally. This article analyzes research trends over 15 years from 2007 to 2022. Materials and Methods Publications on limb salvage surgery in osteosarcoma were retrieved using the Web of Science. Bibliometric analysis of the publication metadata was done using R software. VOS viewer software was used to analyze the bibliographic coupling, co-citation, co-authorship, and co-occurrence to report the current trends in global research on limb salvage surgery in osteosarcoma. Results A total of 693 articles were retrieved. On applying the inclusion and exclusion criteria, a publication metadata of 276 articles was analyzed using the methodology mentioned. Annual scientific production on the subject has shown a steady rising trend globally. China has the highest number of publications on the topic; however, the USA has the highest citations globally. The Journal "Clinical Orthopedics and Related Research" remains the pioneer in the topic with the highest number of publications and H index among all journals. Most of the research interest is generated in the developed countries of the USA, Europe, and China. Keyword analysis suggested 4 clusters of surgical reconstruction, Survival, Chemotherapy, and general management related. Newer keywords such as biological reconstructions, allograft, metastases, cell, and chemotherapy suggest future research topics in the field. Conclusion Research interest in limb salvage surgery in osteosarcoma continues to grow with the introduction of concepts such as biological reconstructions and allografts. However, for more inclusive research on the topic, research interest must also be encouraged in underdeveloped and developing countries.
Collapse
Affiliation(s)
- Manish Raj
- Department of Orthopaedics, All India Institute of Medical Sciences, Deoghar, India
| | - Amiy Arnav
- Department of Surgical Oncology, All India Institute of Medical Sciences, Deoghar, India
| | - Arup Kumar Pal
- Department of Computer Science and Engineering, Indian Institute of Technology (ISM), Dhanbad, India
| | - Shukla Mondal
- Department of Computer Science and Engineering, Indian Institute of Technology (ISM), Dhanbad, India
| |
Collapse
|
61
|
Assi A, Farhat M, Hachem MCR, Zalaquett Z, Aoun M, Daher M, Sebaaly A, Kourie HR. Tyrosine kinase inhibitors in osteosarcoma: Adapting treatment strategiesa. J Bone Oncol 2023; 43:100511. [PMID: 38058514 PMCID: PMC10696463 DOI: 10.1016/j.jbo.2023.100511] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/19/2023] [Accepted: 11/01/2023] [Indexed: 12/08/2023] Open
Abstract
Osteosarcoma (OS) is an aggressive primary bone malignancy that metastasizes rapidly. The standard of care has changed little over the previous four decades, and survival rates have plateaued. In this context, tyrosine kinase inhibitors (TKIs) emerge as potential treatments. A literature search was conducted to collect data related to receptor tyrosine kinase genetic alterations and expression in OS specimens. Gene amplification and protein expression of these receptors were linked to prognosis and tumor behavior. Relevant TKIs were evaluated as monotherapies and as parts of combination therapies. Certain TKIs, such as apatinib, regorafenib, and cabozantinib, present a potential therapeutic avenue for OS patients, especially when combined with chemotherapy. Producing long-lasting responses and enhancing quality of life remain key goals in OS treatment. To this effect, optimizing the use of TKIs by identifying biomarkers predictive of response and assessing promising TKIs in larger-scale trials to validate the efficacy and safety outcomes relative to these drugs reported in phase II clinical trials. To this effect, it is necessary to identify biomarkers predictive of response to TKIs in larger-scale trials and to validate the efficacy and safety of these drugs reported in phase II clinical trials.
Collapse
Affiliation(s)
- Ahmad Assi
- Hematology-Oncology Department, Hotel Dieu de France, Beirut, Lebanon
| | - Mohamad Farhat
- Hematology-Oncology Department, Hotel Dieu de France, Beirut, Lebanon
| | | | - Ziad Zalaquett
- Hematology-Oncology Department, Hotel Dieu de France, Beirut, Lebanon
| | - Marven Aoun
- Orthopedics Department, Hotel Dieu de France, Beirut, Lebanon
| | - Mohammad Daher
- Orthopedics Department, Hotel Dieu de France, Beirut, Lebanon
- Orthopedics Department, Brown University, Providence, RI, USA
| | - Amer Sebaaly
- Orthopedics Department, Hotel Dieu de France, Beirut, Lebanon
| | | |
Collapse
|
62
|
Yang Y, Huang Z, Yuan M, Rui J, Chen R, Jin T, Sun Y, Deng Z, Shan H, Niu X, Liu W. Genomic and transcriptomic characterization of pre-operative chemotherapy response in patients with osteosarcoma. Sci Rep 2023; 13:20914. [PMID: 38017005 PMCID: PMC10684554 DOI: 10.1038/s41598-023-46857-8] [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: 07/05/2023] [Accepted: 11/06/2023] [Indexed: 11/30/2023] Open
Abstract
Osteosarcoma is a heterogeneous disease with regard to its chemotherapy response and clinical outcomes. This study aims to investigate the genomic and transcriptomic characteristics related to pre-operative chemotherapy response. Samples from 25 osteosarcoma patients were collected to perform both whole exome and transcriptome sequencing. Osteosarcoma had significant amount of chromosomal copy number variants (CNVs). Chemotherapy responders showed the higher chromosomal CNV burden than non-responders (p = 0.0775), but the difference was not significant. The percentage of COSMIC signature 3, associated with homologous recombination repair deficiency, was higher in responders (56%) than in non-responders (45%). Transcriptomic analysis suggested that 11 genes were significantly up-regulated in responders and 18 genes were up-regulated in non-responders. Both GSEA and KEGG enrichment analysis indicted that four pathways related to cardiomyopathy were up-regulated in responders, while neuroactive ligand - receptor interaction was up-regulated in non-responders. Finally, a previously published chemoresistant model was validated using our dataset, with the area under the curve of 0.796 (95% CI, 0.583-1.000). Osteosarcoma had the heterogeneous mutational profile with frequent occurrence of CNVs. Transcriptomic analysis identified several signaling pathways associated with chemotherapy responsiveness to osteosarcoma. Transcriptomic signatures provides a potential research direction for predicting the chemotherapy response.
Collapse
Affiliation(s)
- Yongkun Yang
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
- Fourth Medical College of Peking University, Beijing, 100035, China
| | - Zhen Huang
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
- Fourth Medical College of Peking University, Beijing, 100035, China
| | | | - Jinqiu Rui
- Geneplus-Beijing, Beijing, 102206, China
| | | | - Tao Jin
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
- Fourth Medical College of Peking University, Beijing, 100035, China
| | - Yang Sun
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
- Fourth Medical College of Peking University, Beijing, 100035, China
| | - Zhiping Deng
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
- Fourth Medical College of Peking University, Beijing, 100035, China
| | - Huachao Shan
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
- Fourth Medical College of Peking University, Beijing, 100035, China
| | - Xiaohui Niu
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
- Fourth Medical College of Peking University, Beijing, 100035, China
| | - Weifeng Liu
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China.
- Fourth Medical College of Peking University, Beijing, 100035, China.
| |
Collapse
|
63
|
Zhong L, Wang J, Chen W, Lv D, Zhang R, Wang X, Zeng C, He X, Zheng L, Gao Y, Wang S, Li M, Wu Y, Yin J, Kang T, Liao D. Augmenting L3MBTL2-induced condensates suppresses tumor growth in osteosarcoma. SCIENCE ADVANCES 2023; 9:eadi0889. [PMID: 37992172 PMCID: PMC10664996 DOI: 10.1126/sciadv.adi0889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 10/24/2023] [Indexed: 11/24/2023]
Abstract
Osteosarcoma is a highly aggressive cancer and lacks effective therapeutic targets. We found that L3MBTL2 acts as a tumor suppressor by transcriptionally repressing IFIT2 in osteosarcoma. L3MBTL2 recruits the components of Polycomb repressive complex 1.6 to form condensates via both Pho-binding pockets and polybasic regions within carboxyl-terminal intrinsically disordered regions; the L3MBTL2-induced condensates are required for its tumor suppression. Multi-monoubiquitination of L3MBTL2 by UBE2O results in its proteasomal degradation, and the UBE2O/L3MBTL2 axis was crucial for osteosarcoma growth. There is a reverse correlation between L3MBTL2 and UBE2O in osteosarcoma tissues, and higher UBE2O and lower L3MBTL2 are associated with poorer prognosis in osteosarcoma. Pharmacological blockage of UBE2O by arsenic trioxide can enhance L3MBTL2-induced condensates and consequently suppress osteosarcoma growth. Our findings unveil a crucial biological function of L3MBTL2-induced condensates in mediating tumor suppression, proposing the UBE2O-L3MBTL2 axis as a potential cancer therapeutic target in osteosarcoma.
Collapse
Affiliation(s)
- Li Zhong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Center of Digestive Diseases, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jingxuan Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wanqi Chen
- Center of Digestive Diseases, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Dongming Lv
- Department of Musculoskeletal Oncology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ruhua Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Cuiling Zeng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaobo He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lisi Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ying Gao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shang Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Miao Li
- Center of Digestive Diseases, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuanzhong Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Junqiang Yin
- Department of Musculoskeletal Oncology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tiebang Kang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dan Liao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
64
|
Welch DL, Fridley BL, Cen L, Teer JK, Yoder SJ, Pettersson F, Xu L, Cheng CH, Zhang Y, Alexandrow MG, Xiang S, Robertson-Tessi M, Brown JS, Metts J, Brohl AS, Reed DR. Modeling phenotypic heterogeneity towards evolutionarily inspired osteosarcoma therapy. Sci Rep 2023; 13:20125. [PMID: 37978271 PMCID: PMC10656496 DOI: 10.1038/s41598-023-47412-1] [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: 08/10/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023] Open
Abstract
Osteosarcoma is the most common bone sarcoma in children and young adults. While universally delivered, chemotherapy only benefits roughly half of patients with localized disease. Increasingly, intratumoral heterogeneity is recognized as a source of therapeutic resistance. In this study, we develop and evaluate an in vitro model of osteosarcoma heterogeneity based on phenotype and genotype. Cancer cell populations vary in their environment-specific growth rates and in their sensitivity to chemotherapy. We present the genotypic and phenotypic characterization of an osteosarcoma cell line panel with a focus on co-cultures of the most phenotypically divergent cell lines, 143B and SAOS2. Modest environmental (pH, glutamine) or chemical perturbations dramatically shift the success and composition of cell lines. We demonstrate that in nutrient rich culture conditions 143B outcompetes SAOS2. But, under nutrient deprivation or conventional chemotherapy, SAOS2 growth can be favored in spheroids. Importantly, when the simplest heterogeneity state is evaluated, a two-cell line coculture, perturbations that affect the faster growing cell line have only a modest effect on final spheroid size. Thus the only evaluated therapies to eliminate the spheroids were by switching therapies from a first strike to a second strike. This extensively characterized, widely available system, can be modeled and scaled to allow for improved strategies to anticipate resistance in osteosarcoma due to heterogeneity.
Collapse
Affiliation(s)
- Darcy L Welch
- Adolescent and Young Adult Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Individualized Cancer Management, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Brooke L Fridley
- Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ling Cen
- Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jamie K Teer
- Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Sean J Yoder
- Molecular Genomics Core Facility, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Fredrik Pettersson
- Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Liping Xu
- Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Chia-Ho Cheng
- Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Yonghong Zhang
- Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Mark G Alexandrow
- Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Shengyan Xiang
- Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Mark Robertson-Tessi
- Integrative Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Cancer Biology and Evolution, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Joel S Brown
- Integrative Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Cancer Biology and Evolution, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jonathan Metts
- Sarcoma Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Andrew S Brohl
- Sarcoma Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Damon R Reed
- Adolescent and Young Adult Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
- Department of Individualized Cancer Management, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
- Integrative Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
- Cancer Biology and Evolution, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
65
|
Zhang L, Tian Y, Wang J, Deng S, Fan H. Network pharmacology-based research on the effect of Scutellaria baicalensis on osteosarcoma and the underlying mechanism. Medicine (Baltimore) 2023; 102:e35957. [PMID: 37986331 PMCID: PMC10659624 DOI: 10.1097/md.0000000000035957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 11/22/2023] Open
Abstract
To explore the anti-tumor effects of Scutellaria baicalensis on osteosarcoma and its mechanism. Network pharmacology and molecular docking techniques were applied to investigate the effect and mechanism of Scutellaria baicalensis on osteosarcoma (OS). We analyzed the protein-protein interaction (PPI) network for potential targets of Scutellaria baicalensis for treating osteosarcoma and identified hub targets. We used KM curves to screen for hub targets that could effectively prolong the survival time of OS patients. We systematically performed gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) enrichment analysis of the Scutellaria baicalensis potential targets and predicted the Scutellaria baicalensis molecular mechanism and function in treating osteosarcoma. Through molecular docking, the binding process between the hub targets, which could prolong the survival time of sarcoma patients, and Scutellaria baicalensis was simulated. PPI network analysis of potential therapeutic targets discriminated 12 hub targets. The KM curves of the hub targets showed that upregulation of RXRA, RELA, ESR1, TNF, IL6, IL1B, and RB1 expression, and downregulation of MAPK1, VEGFA, MAPK14, CDK1, and PPARG expression were effective in improving the 5-year survival rate of OS patients. GO and KEGG enrichment demonstrated that Scutellaria baicalensis regulated multiple signaling pathways of OS. Molecular docking results indicated that Scutellaria baicalensis could bind freely to the above hub target, which could prolong the survival time of sarcoma patients. Scutellaria baicalensis acted on osteosarcoma by regulating a signaling network formed by hub targets connecting multiple signaling pathways. Scutellaria baicalensis appears to have the potential to serve as a therapeutic drug for osteosarcoma and to prolong the survival of OS patients.
Collapse
Affiliation(s)
- Lijuan Zhang
- Department of Integrated Traditional and Western Medicine, Union Hospital, Tong ji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yushi Tian
- Department of Integrated Traditional and Western Medicine, Union Hospital, Tong ji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jingbo Wang
- Department of Integrated Traditional and Western Medicine, Union Hospital, Tong ji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuangjiao Deng
- Department of Integrated Traditional and Western Medicine, Union Hospital, Tong ji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Heng Fan
- Department of Integrated Traditional and Western Medicine, Union Hospital, Tong ji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
66
|
Mancarella C, Morrione A, Scotlandi K. PROTAC-Based Protein Degradation as a Promising Strategy for Targeted Therapy in Sarcomas. Int J Mol Sci 2023; 24:16346. [PMID: 38003535 PMCID: PMC10671294 DOI: 10.3390/ijms242216346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
Sarcomas are heterogeneous bone and soft tissue cancers representing the second most common tumor type in children and adolescents. Histology and genetic profiling discovered more than 100 subtypes, which are characterized by peculiar molecular vulnerabilities. However, limited therapeutic options exist beyond standard therapy and clinical benefits from targeted therapies were observed only in a minority of patients with sarcomas. The rarity of these tumors, paucity of actionable mutations, and limitations in the chemical composition of current targeted therapies hindered the use of these approaches in sarcomas. Targeted protein degradation (TPD) is an innovative pharmacological modality to directly alter protein abundance with promising clinical potential in cancer, even for undruggable proteins. TPD is based on the use of small molecules called degraders or proteolysis-targeting chimeras (PROTACs), which trigger ubiquitin-dependent degradation of protein of interest. In this review, we will discuss major features of PROTAC and PROTAC-derived genetic systems for target validation and cancer treatment and focus on the potential of these approaches to overcome major issues connected to targeted therapies in sarcomas, including drug resistance, target specificity, and undruggable targets. A deeper understanding of these strategies might provide new fuel to drive molecular and personalized medicine to sarcomas.
Collapse
Affiliation(s)
- Caterina Mancarella
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Katia Scotlandi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| |
Collapse
|
67
|
Kinnaman MD, Zaccaria S, Makohon-Moore A, Arnold B, Levine MF, Gundem G, Arango Ossa JE, Glodzik D, Rodríguez-Sánchez MI, Bouvier N, Li S, Stockfisch E, Dunigan M, Cobbs C, Bhanot UK, You D, Mullen K, Melchor JP, Ortiz MV, O'Donohue TJ, Slotkin EK, Wexler LH, Dela Cruz FS, Hameed MR, Glade Bender JL, Tap WD, Meyers PA, Papaemmanuil E, Kung AL, Iacobuzio-Donahue CA. Subclonal Somatic Copy-Number Alterations Emerge and Dominate in Recurrent Osteosarcoma. Cancer Res 2023; 83:3796-3812. [PMID: 37812025 PMCID: PMC10646480 DOI: 10.1158/0008-5472.can-23-0385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/14/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023]
Abstract
Multiple large-scale genomic profiling efforts have been undertaken in osteosarcoma to define the genomic drivers of tumorigenesis, therapeutic response, and disease recurrence. The spatial and temporal intratumor heterogeneity could also play a role in promoting tumor growth and treatment resistance. We conducted longitudinal whole-genome sequencing of 37 tumor samples from 8 patients with relapsed or refractory osteosarcoma. Each patient had at least one sample from a primary site and a metastatic or relapse site. Subclonal copy-number alterations were identified in all patients except one. In 5 patients, subclones from the primary tumor emerged and dominated at subsequent relapses. MYC gain/amplification was enriched in the treatment-resistant clones in 6 of 7 patients with multiple clones. Amplifications in other potential driver genes, such as CCNE1, RAD21, VEGFA, and IGF1R, were also observed in the resistant copy-number clones. A chromosomal duplication timing analysis revealed that complex genomic rearrangements typically occurred prior to diagnosis, supporting a macroevolutionary model of evolution, where a large number of genomic aberrations are acquired over a short period of time followed by clonal selection, as opposed to ongoing evolution. A mutational signature analysis of recurrent tumors revealed that homologous repair deficiency (HRD)-related SBS3 increases at each time point in patients with recurrent disease, suggesting that HRD continues to be an active mutagenic process after diagnosis. Overall, by examining the clonal relationships between temporally and spatially separated samples from patients with relapsed/refractory osteosarcoma, this study sheds light on the intratumor heterogeneity and potential drivers of treatment resistance in this disease. SIGNIFICANCE The chemoresistant population in recurrent osteosarcoma is subclonal at diagnosis, emerges at the time of primary resection due to selective pressure from neoadjuvant chemotherapy, and is characterized by unique oncogenic amplifications.
Collapse
Affiliation(s)
- Michael D. Kinnaman
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Simone Zaccaria
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, United Kingdom
| | - Alvin Makohon-Moore
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian Arnold
- Department of Computer Science, Princeton University, Princeton, New Jersey
- Center for Statistics and Machine Learning, Princeton University, Princeton, New Jersey
| | - Max F. Levine
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gunes Gundem
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Juan E. Arango Ossa
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dominik Glodzik
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Nancy Bouvier
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shanita Li
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Emily Stockfisch
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marisa Dunigan
- Integrated Genomics Operation Core, Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cassidy Cobbs
- Integrated Genomics Operation Core, Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Umesh K. Bhanot
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
- Precision Pathology Biobanking Center, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daoqi You
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katelyn Mullen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, New York, New York
| | - Jerry P. Melchor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael V. Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Tara J. O'Donohue
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Emily K. Slotkin
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Leonard H. Wexler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Filemon S. Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Meera R. Hameed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Julia L. Glade Bender
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William D. Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paul A. Meyers
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elli Papaemmanuil
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew L. Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christine A. Iacobuzio-Donahue
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
68
|
Hu J, Yang X, Ren J, Zhong S, Fan Q, Li W. Identification and verification of characteristic differentially expressed ferroptosis-related genes in osteosarcoma using bioinformatics analysis. Toxicol Mech Methods 2023; 33:781-795. [PMID: 37488941 DOI: 10.1080/15376516.2023.2240879] [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: 05/17/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND This study identified and verified the characteristic differentially expressed ferroptosis-related genes (CDEFRGs) in osteosarcoma (OS). METHODS We extracted ferroptosis-related genes (FRGs), identified differentially expressed FRGs (DEFRGs) in OS, and conducted correlation analysis between DEFRGs. Next, we conducted GO and KEGG analyses to explore the biological functions and pathways of DEFRGs. Furthermore, we used LASSO and SVM-RFE algorithms to screen CDEFRGs, and evaluated its accuracy in diagnosing OS through ROC curves. Then, we demonstrated the molecular function and pathway enrichment of CDEFRGs through GSEA analysis. In addition, we evaluated the differences in immune cell infiltration between OS and NC groups, as well as the correlation between CDEFRGs expressions and immune cell infiltrations. Finally, the expression of CDEFRGs was verified through qRT-PCR, western blotting, and immunohistochemistry experiments. RESULTS We identified 51 DEFRGs and the expression relationship between them. GO and KEGG analysis revealed their key functions and important pathways. Based on four CDEFRGs (PEX3, CPEB1, NOX1, and ALOX5), we built the OS diagnostic model, and verified its accuracy. GSEA analysis further revealed the important functions and pathways of CDEFRGs. In addition, there were differences in immune cell infiltration between OS group and NC group, and CDEFRGs showed significant correlation with certain infiltrating immune cells. Finally, we validated the differential expression levels of four CDEFRGs through external experiments. CONCLUSIONS This study has shed light on the molecular pathological mechanism of OS and has offered novel perspectives for the early diagnosis and immune-targeted therapy of OS patients.
Collapse
Affiliation(s)
- Jianhua Hu
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, P. R. China
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, P. R. China
| | - Xi Yang
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, P. R. China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, P. R. China
| | - Jing Ren
- Department of Spinal Surgery, Qujing No. 1 Hospital, Affiliated Qujing Hospital of Kunming Medical University, Qujing, P. R. China
| | - Shixiao Zhong
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, P. R. China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, P. R. China
| | - Qianbo Fan
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, P. R. China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, P. R. China
| | - Weichao Li
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, P. R. China
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, P. R. China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, P. R. China
| |
Collapse
|
69
|
van Ewijk R, Cleirec M, Herold N, le Deley MC, van Eijkelenburg N, Boudou-Rouquette P, Risbourg S, Strauss SJ, Palmerini E, Boye K, Kager L, Hecker-Nolting S, Marchais A, Gaspar N. A systematic review of recent phase-II trials in refractory or recurrent osteosarcoma: Can we inform future trial design? Cancer Treat Rev 2023; 120:102625. [PMID: 37738712 DOI: 10.1016/j.ctrv.2023.102625] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND/OBJECTIVE To analyze changes in recurrent/refractory osteosarcoma phase II trials over time to inform future trials in this population with poor prognosis. METHODS A systematic review of trials registered on trial registries between 01/01/2017-14/02/2022. Comparison of 98 trials identified between 2003 and 2016. Publication search/analysis for both periods, last update on 01/12/2022. RESULTS Between 2017 and 2022, 71 phase-II trials met our selection criteria (19 osteosarcoma-specific trials, 14 solid tumor trials with and 38 trials without an osteosarcoma-specific stratum). The trial number increased over time: 13.9 versus 7 trials/year (p = 0.06). Monotherapy remained the predominant treatment (62% vs. 62%, p = 1). Targeted therapies were increasingly evaluated (66% vs. 41%, P = 0.001). Heterogeneity persisted in the trial characteristics. The inclusion criteria were measurable disease (75%), evaluable disease (14%), and surgical remission (11%). 82% of the trials included pediatric or adolescent patients. Biomarker-driven trials accounted for 25% of the total trials. The survival endpoint use (rather than response) slightly increased (40% versus 31%), but the study H1/H0 hypotheses remained heterogeneous. Single-arm designs predominated over multiarm trials (n = 7). Available efficacy data on 1361 osteosarcoma patients in 58 trials remained disappointing, even though 21% of these trials were considered positive, predominantly those evaluating multi-targeted kinase inhibitors. CONCLUSION Despite observed changes in trial design and an increased number of trials investigating new therapies, high heterogeneity remained with respect to patient selection, study design, primary endpoints, and statistical hypotheses in recently registered phase II trials for osteosarcoma. Continued optimization of trial design informed by a deeper biological understanding should strengthen the development of new therapies.
Collapse
Affiliation(s)
- Roelof van Ewijk
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Morgane Cleirec
- Department of Pediatric Oncology, CHU Nantes, Nantes, France
| | - Nikolas Herold
- Paediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden, and Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Marie-Cécile le Deley
- Unité de Méthodologie et Biostatistiques, Centre Oscar Lambret, Lille, France; Université Paris-Saclay, Université Paris-Sud, UVSQ, CESP, INSERM, U1018 ONCOSTAT, F-94085 Villejuif, France
| | | | - Pascaline Boudou-Rouquette
- Department of Medical Oncology, Cochin Hospital, Cochin Institute, INSERMU1016, Paris Cancer Institute, CARPEM, AP-HP, Paris, France
| | - Séverine Risbourg
- Unité de Méthodologie et Biostatistiques, Centre Oscar Lambret, Lille, France
| | - Sandra J Strauss
- Department of Oncology, University College London Cancer Institute, London, UK
| | - Emanuela Palmerini
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Kjetil Boye
- Department of Oncology, Oslo University Hospital, Norway
| | - Leo Kager
- St. Anna Children's Hospital, Department of Pediatrics, Medical University Vienna, Vienna, Austria; St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | | | - Antonin Marchais
- Department of Oncology for Child and Adolescents, Gustave Roussy Cancer Center, Paris-Saclay University, Villejuif, France; National Institute for Health and Medical Research (INSERM) U1015, BiiOSTeam, Gustave Roussy Institute, Villejuif, France
| | - Nathalie Gaspar
- Department of Oncology for Child and Adolescents, Gustave Roussy Cancer Center, Paris-Saclay University, Villejuif, France; National Institute for Health and Medical Research (INSERM) U1015, BiiOSTeam, Gustave Roussy Institute, Villejuif, France.
| |
Collapse
|
70
|
Du X, Wei H, Zhang B, Pang LK, Zhao R, Zhang XD, Yao W. Unveiling the prognostic implications of RPLP1 upregulation in osteosarcoma. Am J Cancer Res 2023; 13:4822-4831. [PMID: 37970363 PMCID: PMC10636679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/26/2023] [Indexed: 11/17/2023] Open
Abstract
Osteosarcoma, a malignant bone tumor characterized by a high rate of metastasis and poor survival, presents a critical need for identifying novel biomarkers associated with metastasis. In this study, we conducted an extensive analysis utilizing transcriptional and clinical data sourced from databases such as GEO, TCGA, CCLE, R2, and Xena. And we discovered that Ribosomal protein LP1 (RPLP1) ranked among the top upregulated genes in relation to osteosarcoma metastasis. Notably, RPLP1 exhibited significant expression in both osteosarcoma cell lines and patient samples. Moreover, multiple osteosarcoma studies revealed a strong correlation between RPLP1 overexpression and worse metastasis-free survival as well as overall survival. Additionally, we observed a consistent association between dysregulation of RPLP1 and reduced overall survival across various tumor types. Knocking down of RPLP1 led to the down-regulation of MYL5 and functional enrichment toward cell cycle and cellular interaction. Based on these findings, we propose that RPLP1 has the potential to serve as a prognostic biomarker, indicating increased metastasis and worse survival outcomes in osteosarcoma. These insights contribute to a better understanding of the disease and may pave the way for future research and therapeutic approaches.
Collapse
Affiliation(s)
- Xinhui Du
- School of Basic Medical Sciences, Zhengzhou UniversityZhengzhou 450001, Henan, China
- Academy of Medical Sciences, Zhengzhou UniversityZhengzhou 450001, Henan, China
- Department of Bone and Soft Tissue, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhou 450008, Henan, China
| | - Hua Wei
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University1 East Jianshe Road, Zhongyuan District, Zhengzhou 450052, Henan, China
| | - Boya Zhang
- Department of Bone and Soft Tissue, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhou 450008, Henan, China
| | - Lon Kai Pang
- Baylor College of MedicineHouston, TX 77030, USA
| | - Ruiying Zhao
- Department of Integrative Biology & Pharmacology, McGovern Medical School, The University of Texas Health Science Center at HoustonHouston, TX 77030, USA
| | - Xu Dong Zhang
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Non-coding RNA and Cancer Metabolism, Henan International Join Laboratory of Non-coding RNA and Metabolism in Cancer, Henan Provincial People’s Hospital, Zhengzhou UniversityZhengzhou 450053, Henan, China
- School of Biomedical Sciences and Pharmacy, The University of NewcastleNSW 2308, Australia
| | - Weitao Yao
- Department of Bone and Soft Tissue, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhou 450008, Henan, China
| |
Collapse
|
71
|
Kusanishi T, Tanaka N, Itoi M, Ijiri M, Ebisawa K, Suzuka K, Hagiwara Y, Yonemoto T, Araki A, Itami M. Multidisciplinary management for primary uterine osteosarcoma, including gene panel testing: case report and literature review. Int Cancer Conf J 2023; 12:241-247. [PMID: 37577351 PMCID: PMC10421837 DOI: 10.1007/s13691-023-00613-x] [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: 01/12/2023] [Accepted: 04/28/2023] [Indexed: 08/15/2023] Open
Abstract
Primary osteosarcoma of the uterus (uOS) is rare, and its standard treatment has not yet been established. Herein, we present the case of a 50-year-old woman with uOS who demonstrated an improved prognosis after multiple surgeries to the metastatic sites. After the initial diagnosis of uOS, the patient showed recurrence and distant metastasis and hence expected to exhibit a poor prognosis. The patient underwent multiple surgical resections of the metastatic as well as primary tumors, which enabled the patient to survive for 24 months after the initial surgery. Considering that the median survival time of patients with uOS is approximately 6 months, the survival rate of our patient is noteworthy. Based on our observations, it is suggested that the resection of the primary and metastatic tumors might contribute to the extension of the survival period of the patient with chemo-resistant uOS.
Collapse
Affiliation(s)
- Takako Kusanishi
- Department of Gynecology, Chiba Cancer Center, 666-2, Nitona-cho, Chuo-ku, Chiba 260-8717 Japan
| | - Naotake Tanaka
- Department of Gynecology, Chiba Cancer Center, 666-2, Nitona-cho, Chuo-ku, Chiba 260-8717 Japan
| | - Mizue Itoi
- Department of Gynecology, Chiba Cancer Center, 666-2, Nitona-cho, Chuo-ku, Chiba 260-8717 Japan
| | - Miwa Ijiri
- Department of Gynecology, Chiba Cancer Center, 666-2, Nitona-cho, Chuo-ku, Chiba 260-8717 Japan
| | - Keiko Ebisawa
- Department of Gynecology, Chiba Cancer Center, 666-2, Nitona-cho, Chuo-ku, Chiba 260-8717 Japan
| | - Kiyomi Suzuka
- Department of Gynecology, Chiba Cancer Center, 666-2, Nitona-cho, Chuo-ku, Chiba 260-8717 Japan
| | - Youko Hagiwara
- Department of Orthopedics, Chiba Cancer Center, 666-2, Nitona-cho, Chuo-ku, Chiba 260-8717 Japan
| | - Tukasa Yonemoto
- Department of Orthopedics, Chiba Cancer Center, 666-2, Nitona-cho, Chuo-ku, Chiba 260-8717 Japan
| | - Akinobu Araki
- Department of Pathology, Chiba Cancer Center, 666-2, Nitona-cho, Chuo-ku, Chiba 260-8717 Japan
| | - Makiko Itami
- Department of Pathology, Chiba Cancer Center, 666-2, Nitona-cho, Chuo-ku, Chiba 260-8717 Japan
| |
Collapse
|
72
|
Huang R, Xu M, Guo W, Cheng M, Dong R, Tu J, Xu S, Zou C. Network pharmacology and experimental verification-based strategy for exploring the mechanisms of luteolin in the treatment of osteosarcoma. Cancer Cell Int 2023; 23:213. [PMID: 37749554 PMCID: PMC10521544 DOI: 10.1186/s12935-023-03046-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 08/29/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Luteolin is an active ingredient in various traditional Chinese medicines for the treatment of multiple tumors. However, the mechanisms of its inhibitory effect on osteosarcoma proliferation and metastasis remain unclear. PURPOSE To elucidate the anti-osteosarcoma mechanisms of luteolin based on network pharmacology and experimental verification. STUDY DESIGN Integrate network pharmacology predictions, scRNA-seq analysis, molecular docking, and experimental validation. METHODS Luteolin-related targets and osteosarcoma-associated targets were collected from several public databases. The luteolin against osteosarcoma targets were screened and a PPI network was constructed to identify the hub targets. The GO and KEGG enrichment of osteosarcoma-associated targets and luteolin against osteosarcoma targets were performed. And scRNA-seq analysis was performed to determine the distribution of the core target expression in OS tissues. Molecular docking, cell biological assays, and osteosarcoma orthotopic mouse model was performed to validate the inhibitory effect and mechanisms of luteolin on osteosarcoma proliferation and metastasis. RESULTS Network pharmacology showed that 251 luteolin against osteosarcoma targets and 8 hub targets including AKT1, ALB, CASP3, IL6, JUN, STAT3, TNF, and VEGFA, and the PI3K-AKT signaling pathway might play an important role in anti-osteosarcoma of luteolin. Analysis of public data revealed that AKT1, IL6, JUN, STAT3, TNF, and VEGFA expression in OS tissue was significantly higher than that in normal bones, and the diagnostic value of VEGFA for overall survival and metastasis was increased over time. scRNA-seq analysis revealed significantly higher expression of AKT1, STAT3, and VEGFA in MYC+ osteoblastic OS cells, especially in primary samples. Moreover, the docking activity between luteolin and the hub targets was excellent, as verified by molecular docking. Experimental results showed that luteolin could inhibit cell viability and significantly decrease the expression of AKT1, STAT3, IL6, TNF, and VEGFA, and luteolin could also inhibit osteosarcoma proliferation and metastasis in osteosarcoma orthotopic mouse model. CONCLUSION This study shows that luteolin may regulate multiple signaling pathways by targeting various genes like AKT1, STAT3, IL6, TNF, and VEGFA to inhibit osteosarcoma proliferation and metastasis.
Collapse
Affiliation(s)
- Renxuan Huang
- Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Guangzhou, 510080, China
| | - Mingxian Xu
- Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Guangzhou, 510080, China
| | - Weitang Guo
- Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Guangzhou, 510080, China
| | - Mingzhe Cheng
- Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Guangzhou, 510080, China
| | - Rui Dong
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Sciences, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Jian Tu
- Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Guangzhou, 510080, China
| | - Shao Xu
- Department of Stomatology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road, Guangzhou, 510630, China.
| | - Changye Zou
- Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Guangzhou, 510080, China.
| |
Collapse
|
73
|
Makielski KM, Sarver AL, Henson MS, Stuebner KM, Borgatti A, Suksanpaisan L, Preusser C, Tabaran AF, Cornax I, O'Sullivan MG, Chehadeh A, Groschen D, Bergsrud K, Pracht S, Winter A, Mills LJ, Schwabenlander MD, Wolfe M, Farrar MA, Cutter GR, Koopmeiners JS, Russell SJ, Modiano JF, Naik S. Neoadjuvant systemic oncolytic vesicular stomatitis virus is safe and may enhance long-term survivorship in dogs with naturally occurring osteosarcoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.16.533664. [PMID: 37131624 PMCID: PMC10153185 DOI: 10.1101/2023.04.16.533664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Osteosarcoma is a devastating bone cancer that disproportionally afflicts children, adolescents, and young adults. Standard therapy includes surgical tumor resection combined with multiagent chemotherapy, but many patients still suffer from metastatic disease progression. Neoadjuvant systemic oncolytic virus (OV) therapy has the potential to improve clinical outcomes by targeting primary and metastatic tumor sites and inducing durable antitumor immune responses. Here we described the first evaluation of neoadjuvant systemic therapy with a clinical-stage recombinant oncolytic Vesicular stomatitis virus (VSV), VSV-IFNβ-NIS, in naturally occurring cancer, specifically appendicular osteosarcoma in companion dogs. Canine osteosarcoma has a similar natural disease history as its human counterpart. VSV-IFNβ-NIS was administered prior to standard of care surgical resection, permitting microscopic and genomic analysis of tumors. Treatment was well-tolerated and a 'tail' of long-term survivors (~35%) was apparent in the VSV-treated group, a greater proportion than observed in two contemporary control cohorts. An increase in tumor inflammation was observed in VSV-treated tumors and RNAseq analysis showed that all the long-term responders had increased expression of a T-cell anchored immune gene cluster. We conclude that neoadjuvant VSV-IFNβ-NIS is safe and may increase long-term survivorship in dogs with naturally occurring osteosarcoma, particularly those that exhibit pre-existing antitumor immunity.
Collapse
|
74
|
Huang S, Ren L, Beck JA, Phelps TE, Olkowski C, Ton A, Roy J, White ME, Adler S, Wong K, Cherukuri A, Zhang X, Basuli F, Choyke PL, Jagoda EM, LeBlanc AK. Exploration of Imaging Biomarkers for Metabolically-Targeted Osteosarcoma Therapy in a Murine Xenograft Model. Cancer Biother Radiopharm 2023; 38:475-485. [PMID: 37253167 PMCID: PMC10623067 DOI: 10.1089/cbr.2022.0090] [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] [Indexed: 06/01/2023] Open
Abstract
Background: Osteosarcoma (OS) is an aggressive pediatric cancer with unmet therapeutic needs. Glutaminase 1 (GLS1) inhibition, alone and in combination with metformin, disrupts the bioenergetic demands of tumor progression and metastasis, showing promise for clinical translation. Materials and Methods: Three positron emission tomography (PET) clinical imaging agents, [18F]fluoro-2-deoxy-2-D-glucose ([18F]FDG), 3'-[18F]fluoro-3'-deoxythymidine ([18F]FLT), and (2S, 4R)-4-[18F]fluoroglutamine ([18F]GLN), were evaluated in the MG63.3 human OS xenograft mouse model, as companion imaging biomarkers after treatment for 7 d with a selective GLS1 inhibitor (CB-839, telaglenastat) and metformin, alone and in combination. Imaging and biodistribution data were collected from tumors and reference tissues before and after treatment. Results: Drug treatment altered tumor uptake of all three PET agents. Relative [18F]FDG uptake decreased significantly after telaglenastat treatment, but not within control and metformin-only groups. [18F]FLT tumor uptake appears to be negatively affected by tumor size. Evidence of a flare effect was seen with [18F]FLT imaging after treatment. Telaglenastat had a broad influence on [18F]GLN uptake in tumor and normal tissues. Conclusions: Image-based tumor volume quantification is recommended for this paratibial tumor model. The performance of [18F]FLT and [18F]GLN was affected by tumor size. [18F]FDG may be useful in detecting telaglenastat's impact on glycolysis. Exploration of kinetic tracer uptake protocols is needed to define clinically relevant patterns of [18F]GLN uptake in patients receiving telaglenastat.
Collapse
Affiliation(s)
- Shan Huang
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ling Ren
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jessica A. Beck
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Tim E. Phelps
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Colleen Olkowski
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Anita Ton
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jyoti Roy
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Margaret E. White
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephen Adler
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Bethesda, Maryland, USA
| | - Karen Wong
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Aswini Cherukuri
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Xiang Zhang
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter L. Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Elaine M. Jagoda
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Amy K. LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
75
|
Wang Y, Ming G, Gao B. A potential prognostic prediction model for metastatic osteosarcoma based on bioinformatics analysis. Acta Orthop Belg 2023; 89:373-380. [PMID: 37935218 DOI: 10.52628/89.2.10491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Osteosarcoma (OS) is a malignant primary bone tumor with a high incidence. This study aims to construct a prognostic prediction model by screening the prognostic mRNA of metastatic OS. Data on four eligible expression profiles from the National Center for Biotechnology Information Gene Expression Omnibus repository were obtained based on inclusion criteria and defined as the training set or the validation set. The differentially expressed genres (DEGs) between meta- static and non-metastatic OS samples in the training set were first identified, and DEGs related to prognosis were screened by univariate Cox regression analysis. In total, 107 DEGs related to the prognosis of metastatic OS were identified. Then, 46 DEGs were isolated as the optimized prognostic gene signature, and a metastatic-OS discriminating classifier was constructed, which had a high accuracy in distinguishing metastatic from non-metastatic OS samples. Furthermore, four optimized prognostic gene signatures (ALOX5AP, COL21A1, HLA-DQB1, and LDHB) were further screened, and the prognostic prediction model for metastatic OS was constructed. This model possesses a relatively satisfying prediction ability both in the training set and validation set. The prognostic prediction model that was constructed based on the four prognostic mRNA signatures has a high predictive ability for the prognosis of metastatic OS.
Collapse
|
76
|
Kobayashi K, Hanai N, Yoshimoto S, Saito Y, Homma A. Current topics and management of head and neck sarcomas. Jpn J Clin Oncol 2023; 53:743-756. [PMID: 37309253 PMCID: PMC10533342 DOI: 10.1093/jjco/hyad048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/18/2023] [Indexed: 06/14/2023] Open
Abstract
Given the low incidence, variety of histological types, and heterogeneous biological features of head and neck sarcomas, there is limited high-quality evidence available to head and neck oncologists. For resectable sarcomas, surgical resection followed by radiotherapy is the principle of local treatment, and perioperative chemotherapy is considered for chemotherapy-sensitive sarcomas. They often originate in anatomical border areas such as the skull base and mediastinum, and they require a multidisciplinary treatment approach considering functional and cosmetic impairment. Moreover, head and neck sarcomas may exhibit different behaviour and characteristics than sarcomas of other areas. In recent years, the molecular biological features of sarcomas have been used for the pathological diagnosis and development of novel agents. This review describes the historical background and recent topics that head and neck oncologists should know about this rare tumour from the following five perspectives: (i) epidemiology and general characteristics of head and neck sarcomas; (ii) changes in histopathological diagnosis in the genomic era; (iii) current standard treatment by histological type and clinical questions specific to head and neck; (iv) new drugs for advanced and metastatic soft tissue sarcomas; and (v) proton and carbon ion radiotherapy for head and neck sarcomas.
Collapse
Affiliation(s)
- Kenya Kobayashi
- Department of Otolaryngology–Head and Neck Surgery, University of Tokyo, Tokyo
| | - Nobuhiro Hanai
- Department of Head and Neck Surgery, Aichi Cancer Center Hospital, Nagoya
| | - Seiichi Yoshimoto
- Department of Head and Neck Surgery, National Cancer Center Hospital, Tokyo
| | - Yuki Saito
- Department of Otolaryngology–Head and Neck Surgery, University of Tokyo, Tokyo
| | - Akihiro Homma
- Department of Otolaryngology–Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
77
|
Mannheimer JD, Tawa G, Gerhold D, Braisted J, Sayers CM, McEachron TA, Meltzer P, Mazcko C, Beck JA, LeBlanc AK. Transcriptional profiling of canine osteosarcoma identifies prognostic gene expression signatures with translational value for humans. Commun Biol 2023; 6:856. [PMID: 37591946 PMCID: PMC10435536 DOI: 10.1038/s42003-023-05208-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 08/03/2023] [Indexed: 08/19/2023] Open
Abstract
Canine osteosarcoma is increasingly recognized as an informative model for human osteosarcoma. Here we show in one of the largest clinically annotated canine osteosarcoma transcriptional datasets that two previously reported, as well as de novo gene signatures devised through single sample Gene Set Enrichment Analysis (ssGSEA), have prognostic utility in both human and canine patients. Shared molecular pathway alterations are seen in immune cell signaling and activation including TH1 and TH2 signaling, interferon signaling, and inflammatory responses. Virtual cell sorting to estimate immune cell populations within canine and human tumors showed similar trends, predominantly for macrophages and CD8+ T cells. Immunohistochemical staining verified the increased presence of immune cells in tumors exhibiting immune gene enrichment. Collectively these findings further validate naturally occurring osteosarcoma of the pet dog as a translationally relevant patient model for humans and improve our understanding of the immunologic and genomic landscape of the disease in both species.
Collapse
Affiliation(s)
- Joshua D Mannheimer
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gregory Tawa
- Division of Preclinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - David Gerhold
- Division of Preclinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - John Braisted
- Division of Preclinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Carly M Sayers
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Troy A McEachron
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paul Meltzer
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jessica A Beck
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
78
|
chen F, Liu J, Yang T, Sun J, He X, Fu X, Qiao S, An J, Yang J. Analysis of intercellular communication in the osteosarcoma microenvironment based on single cell sequencing data. J Bone Oncol 2023; 41:100493. [PMID: 37501717 PMCID: PMC10368934 DOI: 10.1016/j.jbo.2023.100493] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/17/2023] [Accepted: 07/06/2023] [Indexed: 07/29/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary bone cancer in children and young adults, patient survival rates have not improved in recent decades. To further understand the interrelationship between different cell types in the tumor microenvironment of osteosarcoma, we comprehensively analyzed single-cell sequencing data from six patients with untreated osteosarcoma. Nine major cell types were identified from a total of 46,046 cells based on unbiased clustering of gene expression profiles and canonical markers. Osteosarcoma from different patients display heterogeneity in cellular composition. Myeloid cells were the most commonly represented cell type, followed by osteoblastic and TILs. Copy number variation (CNV) results identified amplifications and deletions in malignant osteoblastic cells and fibroblasts. Trajectory analysis based on RNA velocity showed that osteoclasts in the OS microenvironment could be differentiated from myeloid cells. Furthermore, we explored the intercellular communications in OS microenvironment and identified multiple ligand-receptor pairs between myeloid cells, osteoblastic cells and their cells, including 21 ligand-receptor pair genes that significantly associated with survival outcomes. Importantly, we found chemotherapy may have an effect on cellular communication in the OS microenvironment by analyzing single-cell sequencing data from seven primary osteosarcoma patients who received chemotherapy. We believe these observations will improve our understanding of potential mechanisms of microenvironment contributions to OS progression and help identify potential targets for new treatment development in the future.
Collapse
Affiliation(s)
- Fangyi chen
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jiao Liu
- Department of Clinical Nutrition, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Ting Yang
- School of Pharmacy, Yancheng Teachers University, Yancheng, Jiangsu, China
| | - Jianwei Sun
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xianwei He
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xinjie Fu
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, China
| | - Shigang Qiao
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, China
| | - Jianzhong An
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, China
| | - Jiao Yang
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, China
| |
Collapse
|
79
|
Zhang W, Qi L, Liu Z, He S, Wang C, Wu Y, Han L, Liu Z, Fu Z, Tu C, Li Z. Integrated multiomic analysis and high-throughput screening reveal potential gene targets and synergetic drug combinations for osteosarcoma therapy. MedComm (Beijing) 2023; 4:e317. [PMID: 37457661 PMCID: PMC10338795 DOI: 10.1002/mco2.317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/14/2023] [Accepted: 05/22/2023] [Indexed: 07/18/2023] Open
Abstract
Although great advances have been made over the past decades, therapeutics for osteosarcoma are quite limited. We performed long-read RNA sequencing and tandem mass tag (TMT)-based quantitative proteome on osteosarcoma and the adjacent normal tissues, next-generation sequencing (NGS) on paired osteosarcoma samples before and after neoadjuvant chemotherapy (NACT), and high-throughput drug combination screen on osteosarcoma cell lines. Single-cell RNA sequencing data were analyzed to reveal the heterogeneity of potential therapeutic target genes. Additionally, we clarified the synergistic mechanisms of doxorubicin (DOX) and HDACs inhibitors for osteosarcoma treatment. Consequently, we identified 2535 osteosarcoma-specific genes and several alternative splicing (AS) events with osteosarcoma specificity and/or patient heterogeneity. Hundreds of potential therapeutic targets were identified among them, which showed the core regulatory roles in osteosarcoma. We also identified 215 inhibitory drugs and 236 synergistic drug combinations for osteosarcoma treatment. More interestingly, the multiomic analysis pointed out the pivotal role of HDAC1 and TOP2A in osteosarcoma. HDAC inhibitors synergized with DOX to suppress osteosarcoma both in vitro and in vivo. Mechanistically, HDAC inhibitors synergized with DOX by downregulating SP1 to transcriptionally modulate TOP2A expression. This study provided a comprehensive view of molecular features, therapeutic targets, and synergistic drug combinations for osteosarcoma.
Collapse
Affiliation(s)
- Wenchao Zhang
- Department of OrthopedicsThe Second Xiangya HospitalCentral South UniversityChangshaChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya HospitalChangshaChina
| | - Lin Qi
- Department of OrthopedicsThe Second Xiangya HospitalCentral South UniversityChangshaChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya HospitalChangshaChina
| | - Zhongyue Liu
- Department of OrthopedicsThe Second Xiangya HospitalCentral South UniversityChangshaChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya HospitalChangshaChina
| | - Shasha He
- Department of OncologyThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | | | - Ying Wu
- MegaRobo Technologies Co., LtdSuzhouChina
| | | | | | - Zheng Fu
- MegaRobo Technologies Co., LtdSuzhouChina
| | - Chao Tu
- Department of OrthopedicsThe Second Xiangya HospitalCentral South UniversityChangshaChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya HospitalChangshaChina
| | - Zhihong Li
- Department of OrthopedicsThe Second Xiangya HospitalCentral South UniversityChangshaChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya HospitalChangshaChina
| |
Collapse
|
80
|
Hidalgo L, Somovilla-Crespo B, Garcia-Rodriguez P, Morales-Molina A, Rodriguez-Milla MA, Garcia-Castro J. Switchable CAR T cell strategy against osteosarcoma. Cancer Immunol Immunother 2023; 72:2623-2633. [PMID: 37062034 PMCID: PMC10361906 DOI: 10.1007/s00262-023-03437-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 03/22/2023] [Indexed: 04/17/2023]
Abstract
Immunotherapy with chimeric antigen receptor T (CAR T) cells has changed the treatment of hematological malignances, but they are still a challenge for solid tumors, including pediatric sarcomas. Here, we report a switchable CAR T cell strategy based on anti-FITC CAR T cells and a switch molecule conjugated with FITC for targeting osteosarcoma (OS) tumors. As a potential target, we analyzed the expression of B7-H3, an immune checkpoint inhibitor, in OS cell lines. In addition, we evaluate the capacity of an anti-B7-H3 monoclonal antibody conjugated with FITC (anti-B7-H3-FITC mAb) to control the antitumor activity of anti-FITC CAR T cells. The effector functions of anti-FITC CAR T cells against OS, measured in vitro by tumor cell killing activity and cytokine production, are dependent on the presence of the anti-B7-H3-FITC mAb switch. Moreover, OS cells stimulate anti-FITC CAR T cells migration. In vivo, anti-B7-H3 mAb penetrates in the tumor and binds 143B OS tumor cells. Furthermore, anti-FITC CAR T cells reach tumor region and exert antitumor effect in an OS NSG mouse model only in the presence of the switch molecule. We demonstrate that anti-B7-H3-FITC mAb redirects the cytotoxic activity of anti-FITC CAR T cells against OS tumors suggesting that switchable CAR T cell platforms might be a plausible strategy against OS.
Collapse
Affiliation(s)
- Laura Hidalgo
- Cellular Biotechnology Unit, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III (ISCIII), 28220, Madrid, Spain.
| | - Beatriz Somovilla-Crespo
- Cellular Biotechnology Unit, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III (ISCIII), 28220, Madrid, Spain
| | - Patricia Garcia-Rodriguez
- Cellular Biotechnology Unit, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III (ISCIII), 28220, Madrid, Spain
- Universidad Nacional de Educación a Distancia (UNED), 28015, Madrid, Spain
| | - Alvaro Morales-Molina
- Cellular Biotechnology Unit, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III (ISCIII), 28220, Madrid, Spain
| | - Miguel Angel Rodriguez-Milla
- Cellular Biotechnology Unit, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III (ISCIII), 28220, Madrid, Spain
| | - Javier Garcia-Castro
- Cellular Biotechnology Unit, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III (ISCIII), 28220, Madrid, Spain.
| |
Collapse
|
81
|
Kim YI, Tseng YC, Ayaz G, Wang S, Yan H, du Bois W, Yang H, Zhen T, Lee MP, Liu P, Kaplan RN, Huang J. SOX9 is a key component of RUNX2-regulated transcriptional circuitry in osteosarcoma. Cell Biosci 2023; 13:136. [PMID: 37491298 PMCID: PMC10367263 DOI: 10.1186/s13578-023-01088-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 07/18/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND The absence of prominent, actionable genetic alternations in osteosarcomas (OS) implies that transcriptional and epigenetic mechanisms significantly contribute to the progression of this life-threatening form of cancer. Therefore, the identification of potential transcriptional events that promote the survival of OS cells could be key in devising targeted therapeutic approaches for OS. We have previously shown that RUNX2 is a transcription factor (TF) essential for OS cell survival. Unfortunately, the transcriptional network or circuitry regulated by RUNX2 in OS cells is still largely unknown. METHODS The TFs that are in the RUNX2 transcriptional circuitry were identified by analyzing RNAseq and ChIPseq datasets of RUNX2. To evaluate the effect of SOX9 knockdown on the survival of osteosarcoma cells in vitro, we employed cleaved caspase-3 immunoblotting and propidium iodide staining techniques. The impact of SOX9 and JMJD1C depletion on OS tumor growth was examined in vivo using xenografts and immunohistochemistry. Downstream targets of SOX9 were identified and dissected using RNAseq, pathway analysis, and gene set enrichment analysis. Furthermore, the interactome of SOX9 was identified using BioID and validated by PLA. RESULT Our findings demonstrate that SOX9 is a critical TF that is induced by RUNX2. Both in vitro and in vivo experiments revealed that SOX9 plays a pivotal role in the survival of OS. RNAseq analysis revealed that SOX9 activates the transcription of MYC, a downstream target of RUNX2. Mechanistically, our results suggest a transcriptional network involving SOX9, RUNX2, and MYC, with SOX9 binding to RUNX2. Moreover, we discovered that JMJD1C, a chromatin factor, is a novel binding partner of SOX9, and depletion of JMJD1C impairs OS tumor growth. CONCLUSION The findings of this study represent a significant advancement in our understanding of the transcriptional network present in OS cells, providing valuable insights that may contribute to the development of targeted therapies for OS.
Collapse
Affiliation(s)
- Young-Im Kim
- Cancer and Stem Cell Epigenetics Group, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Yu-Chou Tseng
- Cancer and Stem Cell Epigenetics Group, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Gamze Ayaz
- Cancer and Stem Cell Epigenetics Group, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Shasha Wang
- Cancer and Stem Cell Epigenetics Group, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Hualong Yan
- Cancer and Stem Cell Epigenetics Group, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Wendy du Bois
- Cancer and Stem Cell Epigenetics Group, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Howard Yang
- High-Dimension Data Analysis Group, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Tao Zhen
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Maxwell P Lee
- High-Dimension Data Analysis Group, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Paul Liu
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Rosandra N Kaplan
- Tumor Microenvironment Section, Pediatric Oncology Branch, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jing Huang
- Cancer and Stem Cell Epigenetics Group, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
82
|
Nirala BK, Yamamichi T, Yustein JT. Deciphering the Signaling Mechanisms of Osteosarcoma Tumorigenesis. Int J Mol Sci 2023; 24:11367. [PMID: 37511127 PMCID: PMC10379831 DOI: 10.3390/ijms241411367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Osteosarcoma (OS) is the predominant primary bone tumor in the pediatric and adolescent populations. It has high metastatic potential, with the lungs being the most common site of metastasis. In contrast to many other sarcomas, OS lacks conserved translocations or genetic mutations; instead, it has heterogeneous abnormalities, including somatic DNA copy number alteration, ploidy, chromosomal amplification, and chromosomal loss and gain. Unfortunately, clinical outcomes have not significantly improved in over 30 years. Currently, no effective molecularly targeted therapies are available for this disease. Several genomic studies showed inactivation in the tumor suppressor genes, including p53, RB, and ATRX, and hyperactivation of the tumor promoter genes, including MYC and MDM2, in OS. Alterations in the major signaling pathways, including the PI3K/AKT/mTOR, JAK/STAT, Wnt/β-catenin, NOTCH, Hedgehog/Gli, TGF-β, RTKs, RANK/RANKL, and NF-κB signaling pathways, have been identified in OS development and metastasis. Although OS treatment is currently based on surgical excision and systematic multiagent therapies, several potential targeted therapies are in development. This review focuses on the major signaling pathways of OS, and we propose a biological rationale to consider novel and targeted therapies in the future.
Collapse
Affiliation(s)
| | | | - Jason T. Yustein
- Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA 30322, USA; (B.K.N.); (T.Y.)
| |
Collapse
|
83
|
Nirala BK, Patel TD, Kurenbekova L, Shuck R, Dasgupta A, Rainusso N, Coarfa C, Yustein JT. MYC regulates CSF1 expression via microRNA 17/20a to modulate tumor-associated macrophages in osteosarcoma. JCI Insight 2023; 8:e164947. [PMID: 37279073 PMCID: PMC10371352 DOI: 10.1172/jci.insight.164947] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 05/25/2023] [Indexed: 06/07/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary bone tumor of childhood. Approximately 20%-30% of OSs carry amplification of chromosome 8q24, which harbors the oncogene c-MYC and correlates with a poor prognosis. To understand the mechanisms that underlie the ability of MYC to alter both the tumor and its surrounding tumor microenvironment (TME), we generated and molecularly characterized an osteoblast-specific Cre-Lox-Stop-Lox-c-MycT58A p53fl/+ knockin genetically engineered mouse model (GEMM). Phenotypically, the Myc-knockin GEMM had rapid tumor development with a high incidence of metastasis. MYC-dependent gene signatures in our murine model demonstrated significant homology to the human hyperactivated MYC OS. We established that hyperactivation of MYC led to an immune-depleted TME in OS demonstrated by the reduced number of leukocytes, particularly macrophages. MYC hyperactivation led to the downregulation of macrophage colony-stimulating factor 1, through increased microRNA 17/20a expression, causing a reduction of macrophage population in the TME of OS. Furthermore, we developed cell lines from the GEMM tumors, including a degradation tag-MYC model system, which validated our MYC-dependent findings both in vitro and in vivo. Our studies utilized innovative and clinically relevant models to identify a potentially novel molecular mechanism through which MYC regulates the profile and function of the OS immune landscape.
Collapse
Affiliation(s)
- Bikesh K. Nirala
- Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center
| | - Tajhal D. Patel
- Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center
| | - Lyazat Kurenbekova
- Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center
| | - Ryan Shuck
- Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center
| | - Atreyi Dasgupta
- Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center
| | - Nino Rainusso
- Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center
| | - Cristian Coarfa
- Department of Molecular & Human Genetics, and
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Jason T. Yustein
- Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
84
|
Han Q, Yan P, Song R, Liu F, Tian Q. HOXC13-driven TIMM13 overexpression promotes osteosarcoma cell growth. Cell Death Dis 2023; 14:398. [PMID: 37407582 DOI: 10.1038/s41419-023-05910-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 06/11/2023] [Accepted: 06/19/2023] [Indexed: 07/07/2023]
Abstract
TIMM13 (translocase of inner mitochondrial membrane 13) located at the mitochondrial intermembrane space is vital for the integrity and function of mitochondria. We found that the mitochondrial protein TIMM13 is upregulated in human OS tissues and cells. In patient-derived primary OS cells and established cell lines, TIMM13 shRNA or knockout provoked mitochondrial dysfunction, causing mitochondrial depolarization, reactive oxygen species production, and oxidative injury, as well as lipid peroxidation, DNA damage, and ATP depletion. Moreover, TIMM13 depletion provoked OS cell apoptosis and inhibited cell proliferation and migration. Conversely, ectopic TIMM13 overexpression increased ATP contents, enhancing OS cell proliferation and migration. Moreover, we discovered that Akt-mTOR activation was inhibited with TIMM13 depletion in primary OS cells. Further studies revealed that HOXC13 (Homeobox C13)-dependent TIMM13 transcription was significantly increased in OS tissues and cells. Whereas TIMM13 transcription and expression were decreased following HOXC13 silencing in primary OS cells. In vivo, TIMM13 KO potently inhibited OS xenograft growth in the proximal tibia of nude mice. TIMM13 KO also induced Akt-mTOR inactivation, ATP depletion, oxidative injury, and apoptosis in the in situ OS tumors. Together, upregulation of the mitochondrial protein TIMM13 is important for OS cell growth, representing a novel and promising therapeutic target.
Collapse
Affiliation(s)
- Qicai Han
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Penghui Yan
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruipeng Song
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feifei Liu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qing Tian
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
85
|
Zhu M, Sun Y, Xue H, Wu G, Wang Z, Shi J, Ma J, Gu B, Yan X. NEK6 Promotes the Progression of Osteosarcoma Through Activating STAT3 Signaling Pathway by Down-Regulation of miR-26a-5p. Int J Gen Med 2023; 16:2831-2848. [PMID: 37426517 PMCID: PMC10329465 DOI: 10.2147/ijgm.s413461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/19/2023] [Indexed: 07/11/2023] Open
Abstract
Background Osteosarcoma is a malignant tumor originating from the skeletal system. There is no effective treatment other than surgery and chemotherapy, which seriously endangers the health of children and adolescents. NEK6 is a novel discovered Serine/Threonine protein kinase that can regulate cell cycle and activate several oncogenic pathways. Methods NEK6 expression in pan-cancer including sarcoma was evaluated using analysis tools of TIMER, UALCNA and GEPIA with TCGA database, and its association with overall survival in patients with sarcoma was also analyzed. TargetScan, tarbase, microT-CDS and Starbase online software were used to predict NEK6-targeted miRNAs, including miR-26a-5p. Tumor tissues from patients with osteosarcoma were collected for NEK6 and miRNA detection using RT-qPCR. NEK6 down-regulated by siRNAs or miR-26a-5p in osteosarcoma cells was detected by RT-qPCR, Western blot and Immunofluorescence staining assays. Effects of NEK6 knockdown on proliferation, migration, invasion and apoptosis of osteosarcoma cells were detected by CCK-8, wound healing, transwell and flow cytometry, respectively. The expressions of STAT3, metastasis and apoptosis-related genes were detected by Western blot. Results High expression of NEK6 and low expression of miR-26a-5p were lowly expressed in osteosarcoma and they were negative correlation. NEK6 has been confirmed as a direct target for miR-26a-5p. In addition, NEK6 down-regulated by siRNAs or miR-26a-5p led to inhibition of cell proliferation, migration and invasion while promoting cell apoptosis. The levels of phosphorylated STAT3 and metastasis genes (MMP-2, MMP-9) were inhibited, while apoptotic gene Bax was promoted and Bcl2 was inhibited by miR-26a-5p upregulation. Conclusion NEK6 can promote osteosarcoma progression via activating STAT3 signaling pathway, which is inhibited by miR-26a-5p, suggesting that NEK6 is a potential oncogene and miR-26a-5p is a suppressor of osteosarcoma. The strategy of inhibiting of NEK6 by miR-26a-5p may be an effective approach for osteosarcoma therapy.
Collapse
Affiliation(s)
- Min Zhu
- Department of Spine Surgery, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, People’s Republic of China
| | - Yuyu Sun
- Department of Spine Surgery, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, People’s Republic of China
| | - Huawei Xue
- Department of Spine Surgery, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, People’s Republic of China
| | - Gang Wu
- Department of Spine Surgery, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, People’s Republic of China
| | - Zhen Wang
- Department of Spine Surgery, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, People’s Republic of China
| | - Junfeng Shi
- Department of Spine Surgery, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, People’s Republic of China
| | - Jiye Ma
- Department of Spine Surgery, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, People’s Republic of China
| | - Baorong Gu
- Department of Spine Surgery, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, People’s Republic of China
| | - Xiaoling Yan
- Department of Chemotherapy, Affiliated Hospital of Nantong University, Nantong, People’s Republic of China
| |
Collapse
|
86
|
da Costa MEM, Droit R, Khneisser P, Gomez-Brouchet A, Adam-de-Beaumais T, Nolla M, Signolles N, Torrejon J, Lombard B, Loew D, Ayrault O, Scoazec JY, Geoerger B, Vassal G, Marchais A, Gaspar N. Longitudinal characterization of primary osteosarcoma and derived subcutaneous and orthotopic relapsed patient-derived xenograft models. Front Oncol 2023; 13:1166063. [PMID: 37377921 PMCID: PMC10291137 DOI: 10.3389/fonc.2023.1166063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/25/2023] [Indexed: 06/29/2023] Open
Abstract
Osteosarcoma is a rare bone cancer in adolescents and young adults with a dismal prognosis because of metastatic disease and chemoresistance. Despite multiple clinical trials, no improvement in outcome has occurred in decades. There is an urgent need to better understand resistant and metastatic disease and to generate in vivo models from relapsed tumors. We developed eight new patient-derived xenograft (PDX) subcutaneous and orthotopic/paratibial models derived from patients with recurrent osteosarcoma and compared the genetic and transcriptomic landscapes of the disease progression at diagnosis and relapse with the matching PDX. Whole exome sequencing showed that driver and copy-number alterations are conserved from diagnosis to relapse, with the emergence of somatic alterations of genes mostly involved in DNA repair, cell cycle checkpoints, and chromosome organization. All PDX patients conserve most of the genetic alterations identified at relapse. At the transcriptomic level, tumor cells maintain their ossification, chondrocytic, and trans-differentiation programs during progression and implantation in PDX models, as identified at the radiological and histological levels. A more complex phenotype, like the interaction with immune cells and osteoclasts or cancer testis antigen expression, seemed conserved and was hardly identifiable by histology. Despite NSG mouse immunodeficiency, four of the PDX models partially reconstructed the vascular and immune-microenvironment observed in patients, among which the macrophagic TREM2/TYROBP axis expression, recently linked to immunosuppression. Our multimodal analysis of osteosarcoma progression and PDX models is a valuable resource to understand resistance and metastatic spread mechanisms, as well as for the exploration of novel therapeutic strategies for advanced osteosarcoma.
Collapse
Affiliation(s)
- Maria Eugenia Marques da Costa
- INSERM U1015, Université Paris-Saclay, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Robin Droit
- INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Pierre Khneisser
- Department of Medical Biology and Pathology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Anne Gomez-Brouchet
- Department of Pathology, IUCT-Oncopole, CHU Toulouse and University Toulouse, Pharmacology and Structural Biology Institute, CNRS UMR5089, Toulouse, France
| | - Tiphaine Adam-de-Beaumais
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Marie Nolla
- Department of Pediatric Hemato-oncology, CHU Toulouse, Toulouse, France
| | - Nicolas Signolles
- Department of Medical Biology and Pathology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Jacob Torrejon
- Institut Curie, PSL Research University, CNRS UMR, INSERM, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR, INSERM, Orsay, France
| | - Bérangère Lombard
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Paris, France
| | - Damarys Loew
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Paris, France
| | - Olivier Ayrault
- Institut Curie, PSL Research University, CNRS UMR, INSERM, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR, INSERM, Orsay, France
| | - Jean-Yves Scoazec
- Department of Medical Biology and Pathology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Birgit Geoerger
- INSERM U1015, Université Paris-Saclay, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Gilles Vassal
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Antonin Marchais
- INSERM U1015, Université Paris-Saclay, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Nathalie Gaspar
- INSERM U1015, Université Paris-Saclay, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
87
|
Zhang Y, Wei J, Kong L, Song M, Zhang Y, Xiao X, Cao H, Li Z, Yang N, Jin Y. Network pharmacology-based research on the effect of angelicin on osteosarcoma and the underlying mechanism. Aging (Albany NY) 2023; 15:204786. [PMID: 37301545 PMCID: PMC10292874 DOI: 10.18632/aging.204786] [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: 12/28/2022] [Accepted: 05/09/2023] [Indexed: 06/12/2023]
Abstract
To explore the antitumor effects of angelicin on osteosarcoma and the underlying mechanism. We aimed to elucidate the mechanism by network pharmacology, molecular docking, and in vitro experiments. We analyzed a PPI network of potential angelicin targets in the treatment of osteosarcoma and identified hub targets. We systematically performed GO and KEGG enrichment analyses of the potential targets of angelicin, and we predicted it function in osteosarcoma treatment and the underlying molecular mechanism. Through molecular docking, the interactions between hub targets and angelicin were simulated, and then, the hub targets of angelicin were identified. Based on these results, we validated the effects of angelicin on osteosarcoma cells by conducting in vitro experiments. The PPI network analysis of potential therapeutic targets identified four apoptosis-related hub targets, namely, BCL-2, Casp9, BAX and BIRC 2. GO and KEGG enrichment analyses demonstrated that angelicin regulates osteosarcoma cell apoptosis. Molecular docking results indicated that angelicin can freely bind to the hub targets listed above. In vitro experiments showed that angelicin promoted osteosarcoma cell apoptosis in a dose-dependent manner and inhibited osteosarcoma cell migration and proliferation in a time- and dose-dependent manner. The RT-PCR results showed that angelicin simultaneously promoted the mRNA expression of Bcl-2 and Casp9 and inhibited the mRNA expression of BAX and BIRC 2. Angelicin promotes osteosarcoma cell apoptosis and inhibits osteosarcoma cell proliferation and migration by activating a signaling network that is composed of hub targets that link multiple signaling pathways. Angelicin could become an alternative drug for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Yafang Zhang
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Junqiang Wei
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Lingwei Kong
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Mingze Song
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Yange Zhang
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Xiangyu Xiao
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Haiying Cao
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Zhehong Li
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Ning Yang
- Central Laboratory, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Yu Jin
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| |
Collapse
|
88
|
Crane JN, Graham DS, Mona CE, Nelson SD, Samiei A, Dawson DW, Dry SM, Masri MG, Crompton JG, Benz MR, Czernin J, Eilber FC, Graeber TG, Calais J, Federman NC. Fibroblast Activation Protein Expression in Sarcomas. Sarcoma 2023; 2023:2480493. [PMID: 37333052 PMCID: PMC10275689 DOI: 10.1155/2023/2480493] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 05/03/2023] [Accepted: 05/23/2023] [Indexed: 06/20/2023] Open
Abstract
Objectives Fibroblast activation protein alpha (FAP) is highly expressed by cancer-associated fibroblasts in multiple epithelial cancers. The aim of this study was to characterize FAP expression in sarcomas to explore its potential utility as a diagnostic and therapeutic target and prognostic biomarker in sarcomas. Methods Available tissue samples from patients with bone or soft tissue tumors were identified at the University of California, Los Angeles. FAP expression was evaluated via immunohistochemistry (IHC) in tumor samples (n = 63), adjacent normal tissues (n = 30), and positive controls (n = 2) using semiquantitative systems for intensity (0 = negative; 1 = weak; 2 = moderate; and 3 = strong) and density (none, <25%, 25-75%; >75%) in stromal and tumor/nonstromal cells and using a qualitative overall score (not detected, low, medium, and high). Additionally, RNA sequencing data in publicly available databases were utilized to compare FAP expression in samples (n = 10,626) from various cancer types and evaluate the association between FAP expression and overall survival (OS) in sarcoma (n = 168). Results The majority of tumor samples had FAP IHC intensity scores ≥2 and density scores ≥25% for stromal cells (77.7%) and tumor cells (50.7%). All desmoid fibromatosis, myxofibrosarcoma, solitary fibrous tumor, and undifferentiated pleomorphic sarcoma samples had medium or high FAP overall scores. Sarcomas were among cancer types with the highest mean FAP expression by RNA sequencing. There was no significant difference in OS in patients with sarcoma with low versus high FAP expression. Conclusion The majority of the sarcoma samples showed FAP expression by both stromal and tumor/nonstromal cells. Further investigation of FAP as a potential diagnostic and therapeutic target in sarcomas is warranted.
Collapse
Affiliation(s)
- Jacquelyn N. Crane
- Department of Pediatrics, Division of Pediatric Hematology, Oncology, Stem Cell Transplantation & Regenerative Medicine, Stanford University School of Medicine, 1000 Welch Rd, Suite 300, Palo Alto, CA 94304, USA
| | - Danielle S. Graham
- University of California Los Angeles, Department of Surgery, Los Angeles, CA, USA
| | - Christine E. Mona
- University of California Los Angeles, Department of Molecular and Medical Pharmacology, Los Angeles, CA, USA
| | - Scott D. Nelson
- University of California Los Angeles, Department of Pathology and Laboratory Medicine, Los Angeles, CA, USA
- University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Alireza Samiei
- University of California Los Angeles, Department of Pathology and Laboratory Medicine, Los Angeles, CA, USA
| | - David W. Dawson
- University of California Los Angeles, Department of Pathology and Laboratory Medicine, Los Angeles, CA, USA
- University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Sarah M. Dry
- University of California Los Angeles, Department of Pathology and Laboratory Medicine, Los Angeles, CA, USA
- University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Marwan G. Masri
- University of California Los Angeles, Department of Molecular and Medical Pharmacology, Los Angeles, CA, USA
| | - Joseph G. Crompton
- University of California Los Angeles, Department of Surgery, Los Angeles, CA, USA
- University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Matthias R. Benz
- University of California Los Angeles, Department of Molecular and Medical Pharmacology, Los Angeles, CA, USA
- University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Johannes Czernin
- University of California Los Angeles, Department of Molecular and Medical Pharmacology, Los Angeles, CA, USA
- University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Fritz C. Eilber
- University of California Los Angeles, Department of Surgery, Los Angeles, CA, USA
- University of California Los Angeles, Department of Molecular and Medical Pharmacology, Los Angeles, CA, USA
- University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Thomas G. Graeber
- University of California Los Angeles, Department of Molecular and Medical Pharmacology, Los Angeles, CA, USA
- University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Jeremie Calais
- University of California Los Angeles, Department of Molecular and Medical Pharmacology, Los Angeles, CA, USA
| | - Noah C. Federman
- University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
- University of California Los Angeles, Department of Pediatrics, Los Angeles, CA, USA
| |
Collapse
|
89
|
Fan D, Zhang C, Wang H, Wei Q, Cai H, Wei F, Bian Z, Liu W, Wang X, Liu Z. Fabrication of a composite 3D-printed titanium alloy combined with controlled in situ drug release to prevent osteosarcoma recurrence. Mater Today Bio 2023; 20:100683. [PMID: 37346395 PMCID: PMC10279918 DOI: 10.1016/j.mtbio.2023.100683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 04/28/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Abstract
Osteosarcoma is a malignant bone tumor occurring in adolescents. Surgery combined with adjuvant or neoadjuvant chemotherapy is the standard treatment. However, systemic chemotherapy is associated with serious side effects and a high risk of postoperative tumor recurrence, leading to a high amputation rate and mortality in cancer patients. Implant materials that can simultaneously repair large bone defects and prevent osteosarcoma recurrence are in urgent need. Herein, an intelligent system comprising 3D-printed titanium scaffold (TS) and pH-responsive PEGylated paclitaxel prodrugs was fabricated for bone defect reconstruction and recurrence prevention following osteosarcoma surgery. The drug-loaded implants exhibited excellent stability and biocompatibility for supporting the activity of bone stem cells under normal body fluid conditions and the rapid release of drugs in response to faintly acidic environments. An in vitro study demonstrated that five human osteosarcoma cell lines could be efficiently eradicated by paclitaxel released in an acidic microenvironment. Using mice models, we demonstrated that the drug-loaded TS can enable a pH-responsive treatment of postoperative tumors and effectively prevent osteosarcoma recurrence. Therefore, local implantation of this composite scaffold may be a promising topical therapeutic method to prevent osteosarcoma recurrence.
Collapse
Affiliation(s)
- Daoyang Fan
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan Hospital, Peking University, Beijing, 100035, China
| | - Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hufei Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qingguang Wei
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - Hong Cai
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - Feng Wei
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - Zhilei Bian
- Department of Hematology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Weifeng Liu
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan Hospital, Peking University, Beijing, 100035, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhongjun Liu
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| |
Collapse
|
90
|
Liu D, Peng Y, Li X, Zhu Z, Mi Z, Zhang Z, Fan H. Comprehensive landscape of TGFβ-related signature in osteosarcoma for predicting prognosis, immune characteristics, and therapeutic response. J Bone Oncol 2023; 40:100484. [PMID: 37234254 PMCID: PMC10205544 DOI: 10.1016/j.jbo.2023.100484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
Osteosarcoma (OS) is a highly heterogeneous malignant bone tumor, and its tendency to metastasize leads to a poor prognosis. TGFβ is an important regulator in the tumor microenvironment and is closely associated with the progression of various types of cancer. However, the role of TGFβ-related genes in OS is still unclear. In this study, we identified 82 TGFβ DEGs based on RNA-seq data from the TARGET and GETx databases and classified OS patients into two TGFβ subtypes. The KM curve showed that the Cluster 2 patients had a substantially poorer prognosis than the Cluster 1 patients. Subsequently, a novel TGFβ prognostic signatures (MYC and BMP8B) were developed based on the results of univariate, LASSO, and multifactorial Cox analyses. These signatures showed robust and reliable predictive performance for the prognosis of OS in the training and validation cohorts. To predict the three-year and five-year survival rate of OS, a nomogram that integrated clinical features and risk scores was also developed. The GSEA analysis showed that the different subgroups analyzed had distinct functions, particularly, the low-risk group was associated with high immune activity and a high infiltration abundance of CD8 T cells. Moreover, our results indicated that low-risk cases had higher sensitivity to immunotherapy, while high-risk cases were more sensitive to sorafenib and axitinib. scRNA-Seq analysis further revealed that MYC and BMP8B were strongly expressed mainly in tumor stromal cells. Finally, in this study, we confirmed the expression of MYC and BMP8B by performing qPCR, WB, and IHC analyses. To conclude, we developed and validated a TGFβ-related signature to accurately predict the prognosis of OS. Our findings might contribute to personalized treatment and making better clinical decisions for OS patients.
Collapse
Affiliation(s)
- Dong Liu
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Ye Peng
- Department of Orthopaedics, Air Force Medical Center, PLA, Beijing 100142, China
| | - Xian Li
- Department of Orthopaedics, Shenzhen University General Hospital, Shenzhen, China
| | - Zhijie Zhu
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Zhenzhou Mi
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Zhao Zhang
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Hongbin Fan
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
91
|
Ji T, Shi Q, Mei S, Xu J, Liang H, Xie L, Ren T, Sun K, Li D, Tang X, Zhang P, Guo W. Integrated analysis of single-cell and bulk RNA sequencing data reveals an immunostimulatory microenvironment in tumor thrombus of osteosarcoma. Oncogenesis 2023; 12:31. [PMID: 37244923 DOI: 10.1038/s41389-023-00474-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/29/2023] Open
Abstract
Tumor thrombus of bone sarcomas represents a unique reservoir for various types of cancer and immune cells, however, the investigation of tumor thrombus at a single-cell level is very limited. And it is still an open question to identify the thrombus-specific tumor microenvironment that is associated with the tumor-adaptive immune response. Here, by analyzing bulk tissue and single-cell level transcriptome from the paired thrombus and primary tumor samples of osteosarcoma (OS) patients, we define the immunostimulatory microenvironment in tumor thrombus of OS with a higher proportion of tumor-associated macrophages with M1-like states (TAM-M1) and TAM-M1 with high expression of CCL4. OS tumor thrombus is found to have upregulated IFN-γ and TGF-β signalings that are related to immune surveillance of circulating tumor cells in blood circulation. Further multiplexed immunofluorescence staining of the CD3/CD4/CD8A/CD68/CCL4 markers validates the immune-activated state in the tumor thrombus samples. Our study first reports the transcriptome differences at a single-cell level between tumor thrombus and primary tumor in sarcoma.
Collapse
Affiliation(s)
- Tao Ji
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Qianyu Shi
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Song Mei
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiuhui Xu
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Haijie Liang
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Lu Xie
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Tingting Ren
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Kunkun Sun
- Department of Pathology, People's Hospital, Peking University, Beijing, 100044, China
| | - Dasen Li
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Xiaodong Tang
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Rare Disease Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Wei Guo
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China.
| |
Collapse
|
92
|
Wood GE, Graves LA, Rubin EM, Reed DR, Riedel RF, Strauss SJ. Bad to the Bone: Emerging Approaches to Aggressive Bone Sarcomas. Am Soc Clin Oncol Educ Book 2023; 43:e390306. [PMID: 37220319 DOI: 10.1200/edbk_390306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Bone sarcomas are rare heterogeneous tumors that affect patients of all ages including children, adolescent young adults, and older adults. They include many aggressive subtypes and patient groups with poor outcomes, poor access to clinical trials, and lack of defined standard therapeutic strategies. Conventional chondrosarcoma remains a surgical disease, with no defined role for cytotoxic therapy and no approved targeted systemic therapies. Here, we discuss promising novel targets and strategies undergoing evaluation in clinical trials. Multiagent chemotherapy has greatly improved outcomes for patients with Ewing sarcoma (ES) and osteosarcoma, but management of those with high-risk or recurrent disease remains challenging and controversial. We describe the impact of international collaborative trials, such as the rEECur study, that aim to define optimal treatment strategies for those with recurrent, refractory ES, and evidence for high-dose chemotherapy with stem-cell support. We also discuss current and emerging strategies for other small round cell sarcomas, such as CIC-rearranged, BCOR-rearranged tumors, and the evaluation of emerging novel therapeutics and trial designs that may offer a new paradigm to improve survival in these aggressive tumors with notoriously bad (to the bone) outcomes.
Collapse
Affiliation(s)
- Georgina E Wood
- Department of Oncology, University College London Hospitals NHS Trust, UCL Cancer Institute, London, United Kingdom
| | - Laurie A Graves
- Division of Hematology/Oncology, Department of Pediatrics, Duke University, Durham, NC
| | - Elyssa M Rubin
- Division of Oncology, Children's Hospital of Orange County, Orange, CA
| | - Damon R Reed
- Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa, FL
| | - Richard F Riedel
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Durham, NC
| | - Sandra J Strauss
- Department of Oncology, University College London Hospitals NHS Trust, UCL Cancer Institute, London, United Kingdom
| |
Collapse
|
93
|
Xie L, Cai Z, Lu H, Meng F, Zhang X, Luo K, Su X, Lei Y, Xu J, Lou J, Wang H, Du Z, Wang Y, Li Y, Ren T, Xu J, Sun X, Tang X, Guo W. Distinct genomic features between osteosarcomas firstly metastasing to bone and to lung. Heliyon 2023; 9:e15527. [PMID: 37205995 PMCID: PMC10189180 DOI: 10.1016/j.heliyon.2023.e15527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 03/26/2023] [Accepted: 04/12/2023] [Indexed: 05/21/2023] Open
Abstract
Background Osteosarcoma initially metastasing to bone only shows distinct biological features compared to osteosarcoma that firstly metastasizes to the lung, which suggests us underlying different genomic pathogenetic mechanism. Methods We analyzed whole-exome sequencing (WES) data for 38 osteosarcoma with paired samples in different relapse patterns. We also sought to redefine disease subclassifications for osteosarcoma based on genetic alterations and correlate these genetic profiles with clinical treatment courses to elucidate potential evolving cladograms. Results We investigated WES of 12/38 patients with high-grade osteosarcoma (31.6%) with initial bone metastasis (group A) and 26/38 (68.4%) with initial pulmonary metastasis (group B), of whom 15/38 (39.5%) had paired samples of primary lesions and metastatic lesions. We found that osteosarcoma in group A mainly carries single-nucleotide variations displaying higher tumor mutation burden and neoantigen load and more tertiary lymphoid structures, while those in group B mainly exhibits structural variants. High conservation of reported genetic sequencing over time in their evolving cladograms. Conclusions Osteosarcoma with mainly single-nucleotide variations other than structural variants might exhibit biological behavior predisposing toward bone metastases as well as better immunogenicity in tumor microenvironment.
Collapse
Affiliation(s)
- Lu Xie
- Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Zhenyu Cai
- Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Hezhe Lu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, No. A3 Datun Road, Chaoyang District, Beijing 100101, China
| | - Fanfei Meng
- Shanghai OrigiMed Co., Ltd, Shanghai, No. 3576 Zhaolou Road, Minhang District, Shanghai, 201112, China
| | - Xin Zhang
- Shanghai OrigiMed Co., Ltd, Shanghai, No. 3576 Zhaolou Road, Minhang District, Shanghai, 201112, China
| | - Kun Luo
- Shanghai OrigiMed Co., Ltd, Shanghai, No. 3576 Zhaolou Road, Minhang District, Shanghai, 201112, China
| | - Xiaoxing Su
- Berry Oncology Corporation, Fuzhou, 350200, China
| | - Yan Lei
- Berry Oncology Corporation, Fuzhou, 350200, China
| | - Jiuhui Xu
- Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Jingbing Lou
- Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Han Wang
- Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Zhiye Du
- Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Yunfan Wang
- Pathology Department, Peking University Shougang Hospital, No. 9 Jinyuanzhuang Road, Shijingshan District, Beijing, 100144, China
| | - Yuan Li
- Radiology Department & Nuclear Medicine Department, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Tingting Ren
- Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Jie Xu
- Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Xin Sun
- Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Xiaodong Tang
- Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
- Corresponding author.
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
- Corresponding author.
| |
Collapse
|
94
|
Rajan S, Franz EM, McAloney CA, Vetter TA, Cam M, Gross AC, Taslim C, Wang M, Cannon MV, Oles A, Roberts RD. Osteosarcoma tumors maintain intra-tumoral transcriptional heterogeneity during bone and lung colonization. BMC Biol 2023; 21:98. [PMID: 37106386 PMCID: PMC10142502 DOI: 10.1186/s12915-023-01593-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Tumors are complex tissues containing collections of phenotypically diverse malignant and nonmalignant cells. We know little of the mechanisms that govern heterogeneity of tumor cells nor of the role heterogeneity plays in overcoming stresses, such as adaptation to different microenvironments. Osteosarcoma is an ideal model for studying these mechanisms-it exhibits widespread inter- and intra-tumoral heterogeneity, predictable patterns of metastasis, and a lack of clear targetable driver mutations. Understanding the processes that facilitate adaptation to primary and metastatic microenvironments could inform the development of therapeutic targeting strategies. RESULTS We investigated single-cell RNA-sequencing profiles of 47,977 cells obtained from cell line and patient-derived xenograft models as cells adapted to growth within primary bone and metastatic lung environments. Tumor cells maintained phenotypic heterogeneity as they responded to the selective pressures imposed during bone and lung colonization. Heterogenous subsets of cells defined by distinct transcriptional profiles were maintained within bone- and lung-colonizing tumors, despite high-level selection. One prominent heterogenous feature involving glucose metabolism was clearly validated using immunofluorescence staining. Finally, using concurrent lineage tracing and single-cell transcriptomics, we found that lung colonization enriches for multiple clones with distinct transcriptional profiles that are preserved across cellular generations. CONCLUSIONS Response to environmental stressors occurs through complex and dynamic phenotypic adaptations. Heterogeneity is maintained, even in conditions that enforce clonal selection. These findings likely reflect the influences of developmental processes promoting diversification of tumor cell subpopulations, which are retained, even in the face of selective pressures.
Collapse
Affiliation(s)
- Sanjana Rajan
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH USA
- Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Emily M. Franz
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH USA
- Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Camille A. McAloney
- Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH USA
| | - Tatyana A. Vetter
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Maren Cam
- Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Amy C. Gross
- Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Cenny Taslim
- Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Meng Wang
- Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Matthew V. Cannon
- Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Alexander Oles
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC USA
| | - Ryan D. Roberts
- Center for Childhood Cancer, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
- Division of Pediatric Hematology, Oncology, and BMT, Nationwide Children’s Hospital, Columbus, OH USA
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH USA
| |
Collapse
|
95
|
Rajan S, Zaccaria S, Cannon MV, Cam M, Gross AC, Raphael BJ, Roberts RD. Structurally Complex Osteosarcoma Genomes Exhibit Limited Heterogeneity within Individual Tumors and across Evolutionary Time. CANCER RESEARCH COMMUNICATIONS 2023; 3:564-575. [PMID: 37066022 PMCID: PMC10093779 DOI: 10.1158/2767-9764.crc-22-0348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/18/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023]
Abstract
Osteosarcoma is an aggressive malignancy characterized by high genomic complexity. Identification of few recurrent mutations in protein coding genes suggests that somatic copy-number aberrations (SCNA) are the genetic drivers of disease. Models around genomic instability conflict-it is unclear whether osteosarcomas result from pervasive ongoing clonal evolution with continuous optimization of the fitness landscape or an early catastrophic event followed by stable maintenance of an abnormal genome. We address this question by investigating SCNAs in >12,000 tumor cells obtained from human osteosarcomas using single-cell DNA sequencing, with a degree of precision and accuracy not possible when inferring single-cell states using bulk sequencing. Using the CHISEL algorithm, we inferred allele- and haplotype-specific SCNAs from this whole-genome single-cell DNA sequencing data. Surprisingly, despite extensive structural complexity, these tumors exhibit a high degree of cell-cell homogeneity with little subclonal diversification. Longitudinal analysis of patient samples obtained at distant therapeutic timepoints (diagnosis, relapse) demonstrated remarkable conservation of SCNA profiles over tumor evolution. Phylogenetic analysis suggests that the majority of SCNAs were acquired early in the oncogenic process, with relatively few structure-altering events arising in response to therapy or during adaptation to growth in metastatic tissues. These data further support the emerging hypothesis that early catastrophic events, rather than sustained genomic instability, give rise to structural complexity, which is then preserved over long periods of tumor developmental time. Significance Chromosomally complex tumors are often described as genomically unstable. However, determining whether complexity arises from remote time-limited events that give rise to structural alterations or a progressive accumulation of structural events in persistently unstable tumors has implications for diagnosis, biomarker assessment, mechanisms of treatment resistance, and represents a conceptual advance in our understanding of intratumoral heterogeneity and tumor evolution.
Collapse
Affiliation(s)
- Sanjana Rajan
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, Ohio
- Center for Childhood Cancers and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Simone Zaccaria
- Department of Computer Science, Princeton University, Princeton, New Jersey
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Matthew V. Cannon
- Center for Childhood Cancers and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Maren Cam
- Center for Childhood Cancers and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Amy C. Gross
- Center for Childhood Cancers and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Benjamin J. Raphael
- Department of Computer Science, Princeton University, Princeton, New Jersey
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Ryan D. Roberts
- Center for Childhood Cancers and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- The Ohio State University James Comprehensive Cancer Center, Columbus, Ohio
- Division of Pediatric Hematology, Oncology, and BMT, Nationwide Children's Hospital, Columbus, Ohio
| |
Collapse
|
96
|
Umeda K, Sakamoto A, Noguchi T, Uchihara Y, Kobushi H, Akazawa R, Ogata H, Saida S, Kato I, Hiramatsu H, Uto M, Mizowaki T, Haga H, Date H, Okamoto T, Watanabe K, Adachi S, Toguchida J, Matsuda S, Takita J. Clinical Outcomes of Patients With Osteosarcoma Experiencing Relapse or Progression: A Single-institute Experience. J Pediatr Hematol Oncol 2023; 45:e356-e362. [PMID: 35973000 DOI: 10.1097/mph.0000000000002521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/28/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Patients with osteosarcoma who experience relapse or progression [R/P] have a poor prognosis. METHODS Data from 30 patients who experienced R/P among 59 with a diagnosis of high-grade osteosarcoma, who were younger than 40 years old between 2000 and 2019, were retrospectively analyzed to identify prognostic and therapeutic factors influencing their outcomes. RESULTS The 5-year overall survival [OS] rates after the last R/P of patients experiencing first [n=30], second [n=14], and third [n=9] R/P were 50.3%, 51.3%, and 46.7%, respectively. Multivariate analysis did not identify any independent risk factors affecting OS. The 5-year PFS rate of the 30 patients after first R/P was 22.4%, and multivariate analysis identified histologic subtype and curative local surgery as independent risk factors influencing PFS. Long [>6 mo] partial response was observed in three patients treated using temozolomide+etoposide, irinotecan+carboplatin, or regorafenib. CONCLUSIONS OS rate in the patients with osteosarcoma experiencing R/P included in this study was markedly higher than that reported previously, mainly due to the surgical total removal of tumors, even after subsequent R/P. The recent establishment of salvage chemotherapy or molecular targeted therapy may also increase survival rates in a subgroup of patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Megumi Uto
- Radiation Oncology and Image-Applied Therapy
| | | | | | | | - Takeshi Okamoto
- Department of Orthopedic Surgery, Otsu Red Cross Hospital, Otsu, Japan
| | - Kenichiro Watanabe
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Souichi Adachi
- Department of Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junya Toguchida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Japan
| | | | | |
Collapse
|
97
|
Fleuren EDG, Vlenterie M, van der Graaf WTA. Recent advances on anti-angiogenic multi-receptor tyrosine kinase inhibitors in osteosarcoma and Ewing sarcoma. Front Oncol 2023; 13:1013359. [PMID: 36994209 PMCID: PMC10040783 DOI: 10.3389/fonc.2023.1013359] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 02/06/2023] [Indexed: 03/16/2023] Open
Abstract
Osteosarcoma (OS) and Ewing sarcoma (ES) are the two most common types of primary bone cancer that predominantly affect the young. Despite aggressive multimodal treatment, survival has not improved significantly over the past four decades. Clinical efficacy has historically been observed for some mono-Receptor Tyrosine Kinase (RTK) inhibitors, albeit in small subsets of OS and ES patients. Clinical efficacy in larger groups of OS or ES patients was reported recently with several newer generation multi-RTK inhibitors. All these inhibitors combine a strong anti-angiogenic (VEGFRs) component with simultaneous inhibition of other key RTKs implicated in OS and ES progression (PDGFR, FGFR, KIT and/or MET). However, despite interesting clinical data, none of these agents have obtained a registration for these indications and are thus difficult to implement in routine OS and ES patient care. It is at present also unclear which of these drugs, with largely overlapping molecular inhibition profiles, would work best for which patient or subtype, and treatment resistance almost uniformly occurs. Here, we provide a critical assessment and systemic comparison on the clinical outcomes to the six most tested drugs in this field in OS and ES to date, including pazopanib, sorafenib, regorafenib, anlotinib, lenvatinib and cabozantinib. We pay special attention to clinical response evaluations in bone sarcomas and provide drug comparisons, including drug-related toxicity, to put these drugs into context for OS and ES patients, and describe how future trials utilizing anti-angiogenic multi-RTK targeted drugs could be designed to ultimately improve response rates and decrease toxicity.
Collapse
Affiliation(s)
- Emmy D. G. Fleuren
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
- Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
- *Correspondence: Emmy D. G. Fleuren,
| | - Myrella Vlenterie
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Winette T. A. van der Graaf
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
98
|
Kamolphiwong R, Kanokwiroon K, Wongrin W, Chaiyawat P, Klangjorhor J, Settakorn J, Teeyakasem P, Sangphukieo A, Pruksakorn D. Potential target identification for osteosarcoma treatment: Gene expression re-analysis and drug repurposing. Gene X 2023; 856:147106. [PMID: 36513192 DOI: 10.1016/j.gene.2022.147106] [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: 02/15/2022] [Revised: 11/18/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Survival rate of osteosarcoma has remained plateaued for the past three decades. New treatment is needed to improve survival rate. Drug repurposing, a method to identify new indications of previous drugs, which saves time and cost compared to the de novo drug discovery. Data mining from gene expression profile was carried out and new potential targets were identified by using drug repurposing strategy. Selected data were newly categorized as pathophysiology and metastasis groups. Data were normalized and calculated the differential gene expression. Genes with log fold change ≥ 2 and adjusted p-value ≤ 0.05 were selected as primary candidate genes (PCGs). PCGs were further enriched to determine the secondary candidate genes (SCGs) by protein interaction analysis, upstream transcription factor and related-protein kinase identification. PCGs and SCGs were further matched with gene targeted of corresponding drugs from the Drug Repurposing Hub. A total of 778 targets were identified (360 from PCGs, and 418 from SCGs). This newly identified KLHL13 is a new candidate target based on its molecular function. KLHL13 was upregulated in clinical samples. We found 256 drugs from matching processes (50anti-cancerand206non-anticancerdrugs). Clinical trials of anti-cancer drugs from 5 targets (CDK4, BCL-2, JUN, SRC, PIK3CA) are being performed for osteosarcoma treatment. Niclosamide and synthetic PPARɣ ligands are candidates for repurposing due to the possibility based on their mechanism and pharmacology properties. Re-analysis of gene expression profile could identify new potential targets, confirm a current implication, and expand the chance of repurposing drugs for osteosarcoma treatment.
Collapse
Affiliation(s)
- Rawikant Kamolphiwong
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Kanyanatt Kanokwiroon
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand.
| | - Weerinrada Wongrin
- Department of Statistics, Faculty of Science, Chiang Mai University, Chiang Mai, Thailand
| | - Parunya Chaiyawat
- Musculoskeletal Science and Translational Research Center, Department of Orthopaedics, Chiang Mai University, Chiang Mai, Thailand; Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Thailand
| | - Jeerawan Klangjorhor
- Musculoskeletal Science and Translational Research Center, Department of Orthopaedics, Chiang Mai University, Chiang Mai, Thailand; Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Thailand
| | - Jongkolnee Settakorn
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pimpisa Teeyakasem
- Musculoskeletal Science and Translational Research Center, Department of Orthopaedics, Chiang Mai University, Chiang Mai, Thailand
| | - Apiwat Sangphukieo
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Thailand
| | - Dumnoensun Pruksakorn
- Musculoskeletal Science and Translational Research Center, Department of Orthopaedics, Chiang Mai University, Chiang Mai, Thailand; Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Thailand.
| |
Collapse
|
99
|
Zhang L, Liu YX, Yao YT, Zhou TJ, Jiang HL, Li CJ. Injectable rhein-assisted crosslinked hydrogel for efficient local osteosarcoma chemotherapy. Int J Pharm 2023; 634:122637. [PMID: 36702387 DOI: 10.1016/j.ijpharm.2023.122637] [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: 09/14/2022] [Revised: 12/29/2022] [Accepted: 01/18/2023] [Indexed: 01/24/2023]
Abstract
Osteosarcoma (OS) is the most common malignant tumor of the bone that affects children and adolescents, and its treatment usually involves doxorubicin hydrochloride (DOX). However, the drug resistance and side effects caused by high-dose DOX infusion greatly hinder its therapeutic effects. To achieve efficient OS treatment with low toxicity, an injectable rhein (RH)-assisted crosslinked hydrogel (PVA@RH@DOX hydrogel, PRDH) was designed, which was prepared by loading DOX and RH into a polyvinyl alcohol (PVA) solution. The cytotoxicity assay and live/dead staining results showed that the combination of RH and DOX more effectively killed OS cells, producing excellent effects at low concentrations of DOX. The wound healing and transwell test results proved that PRDH could significantly inhibit the metastasis and invasion of OS cells. PRDH showed a long-lasting antitumor effect after injection of a single dose, significantly suppressing the proliferation and metastasis of OS and achieving the strategy of a single administration for long-term treatment. Excitingly, RH facilitated hydrogel formation by assisting with PVA crosslinking. This system provides an alternative regimen and broadens the horizon for the clinical treatment of OS.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, PR China
| | - Ying-Xuan Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Ya-Ting Yao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Cheng-Jun Li
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, PR China.
| |
Collapse
|
100
|
Heydari SR, Samadi M, Shirangi A, Farokhi M, Moradi A, Bafkary R, Atyabi F, Mottaghitalab F, Dinarvand R. Dual responsive hydroxyapatite capped mesoporous silica nanoparticles for controlled delivery of Palbociclib to treat osteosarcoma. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|