1
|
Griffin KH, Sagheb IS, Coonan TP, Fierro FA, Randall RL, Leach JK. Macrophage and osteosarcoma cell crosstalk is dependent on oxygen tension and 3D culture. BIOMATERIALS ADVANCES 2025; 169:214154. [PMID: 39708660 DOI: 10.1016/j.bioadv.2024.214154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 11/29/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Osteosarcoma (OS), the most common form of primary bone cancer in young adults, has had no improvements in clinical outcomes in 50 years. This highlights a critical need to advance mechanistic understanding of OS to further therapeutic discovery, which will only be possible with accurate models of the disease. Compared to traditional monolayer studies and preclinical models, in vitro models that better replicate the three-dimensional (3D) bone marrow microenvironment will facilitate methodical investigations of the events and factors that drive OS progression. Herein, we use fibrin-alginate interpenetrating network (FA IPN) hydrogels to model the hematological bone marrow environment. We interrogated the effects of oxygen tension, 3D culture, and macrophage phenotype on OS behavior and specifically examine the immunomodulatory crosstalk between OS and macrophages. We observe that OS is more sensitive to oxygen tension when cultured in 3D. Specifically, both highly and less metastatic OS exhibit decreased changes in DNA content over time in 3D, but then demonstrate diverging behaviors in heterotypic culture with macrophages. OS response to macrophages differs as a function of metastatic potential, where highly metastatic OS shows increased immunosuppression that varies with oxygen tension but relies on direct coculture conditions. To our knowledge, this is among the first work to report the effects of 3D culture on the interplay between OS and macrophages in a coculture microenvironment. Together, these data introduce FA IPNs as a promising platform for cancer research and emphasize the importance of novel models for the mechanistic study of OS.
Collapse
Affiliation(s)
- Katherine H Griffin
- School of Veterinary Medicine, University of California, Davis, CA, USA; Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, USA
| | - Isabel S Sagheb
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Thomas P Coonan
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Fernando A Fierro
- Department of Cell Biology and Human Anatomy, UC Davis Health, Sacramento, CA, USA
| | - R Lor Randall
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, USA; Department of Biomedical Engineering, University of California, Davis, CA, USA.
| |
Collapse
|
2
|
Ringwalt EM, Currier MA, Glaspell AM, Chen CY, Cannon MV, Cam M, Gross AC, Gust M, Wang PY, Boon L, Biederman LE, Schwarz E, Rajappa P, Lee DA, Mardis ER, Carson WE, Roberts RD, Cripe TP. Trabectedin promotes oncolytic virus antitumor efficacy, viral gene expression, and immune effector function in models of bone sarcoma. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200886. [PMID: 39492947 PMCID: PMC11530761 DOI: 10.1016/j.omton.2024.200886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/13/2024] [Accepted: 09/24/2024] [Indexed: 11/05/2024]
Abstract
We previously reported that the DNA alkylator and transcriptional-blocking chemotherapeutic agent trabectedin enhances oncolytic herpes simplex viroimmunotherapy in human sarcoma xenograft models, though the mechanism remained to be elucidated. Here we report trabectedin disrupts the intrinsic cellular antiviral response which increases viral transcript presence in the human tumor cells. We also extended our synergy findings to syngeneic murine sarcoma models, which are poorly susceptible to virus infection. In the absence of robust virus replication, we found trabectedin enhanced viroimmunotherapy efficacy by reducing infiltrating immunosuppressive CD4 T and myeloid cells and stimulating granzyme expression in infiltrating T and natural killer cells to cause immune-mediated tumor regressions. Thus, trabectedin enhances both the direct virus-mediated killing of tumor cells and the viral-induced activation of cytotoxic effector lymphocytes to cause tumor regressions across models. Our data provide a strong rationale for clinical translation as both mechanisms should be simultaneously active in human patients.
Collapse
Affiliation(s)
- Emily M. Ringwalt
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Mark A. Currier
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Andrea M. Glaspell
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Chun-Yu Chen
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Matthew V. Cannon
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Maren Cam
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Amy C. Gross
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Matthew Gust
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Pin-Yi Wang
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | | | - Laura E. Biederman
- Department of Pathology, Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Emily Schwarz
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Prajwal Rajappa
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Pediatrics and Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Dean A. Lee
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Division of Hematology/Oncology/BMT, Nationwide Children’s Hospital, Columbus, OH 43205, USA
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Elaine R. Mardis
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - William E. Carson
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Department of Surgery, Division of Surgical Oncology, The Ohio State University, Columbus, OH 43210, USA
| | - Ryan D. Roberts
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Division of Hematology/Oncology/BMT, Nationwide Children’s Hospital, Columbus, OH 43205, USA
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Timothy P. Cripe
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Division of Hematology/Oncology/BMT, Nationwide Children’s Hospital, Columbus, OH 43205, USA
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
3
|
Sagheb IS, Coonan TP, Randall RL, Griffin KH, Leach JK. Extracellular matrix production and oxygen diffusion regulate chemotherapeutic response in osteosarcoma spheroids. Cancer Med 2024; 13:e70239. [PMID: 39300969 PMCID: PMC11413413 DOI: 10.1002/cam4.70239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/25/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Osteosarcoma (OS) survival rates and outcome have not improved in 50 years since the advent of modern chemotherapeutics. Thus, there is a critical need for an improved understanding of the tumor microenvironment to identify better therapies. Extracellular matrix (ECM) deposition and hypoxia are known to abrogate the efficacy of various chemical and cell-based therapeutics. Here, we aim to mechanistically investigate the combinatorial effects of hypoxia and matrix deposition with the use of OS spheroids. METHODS We use two murine OS cell lines with differential metastatic potential to form spheroids. We form spheroids of two sizes, use ascorbate-2-phosphate supplementation to enhance ECM deposition, and study cell response under standard (21% O2) and physiologic (5% O2) oxygen tensions. Finally, we examine chemotherapeutic responses to doxorubicin treatment. RESULTS ECM production and oxygen tension are key determinants of spheroid size through cell organization based on nutrient and oxygen distribution. Interestingly, highly metastatic OS is more susceptible to chemotherapeutics compared to less metastatic OS when matrix production increases. Together, these data suggest that dynamic interactions between ECM production and oxygen diffusion may result in distinct chemotherapeutic responses despite inherent tumor aggressiveness. CONCLUSION This work establishes OS spheroids as a valuable tool for early OS tumor formation investigation and holds potential for novel therapeutic target and prognostic indicator discovery.
Collapse
Affiliation(s)
- Isabel S. Sagheb
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Thomas P. Coonan
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - R. Lor Randall
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
| | - Katherine H. Griffin
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
- School of Veterinary MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - J. Kent Leach
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
| |
Collapse
|
4
|
Lu J, Rui J, Xu XY, Shen JK. Exploring the Role of Neutrophil-Related Genes in Osteosarcoma via an Integrative Analysis of Single-Cell and Bulk Transcriptome. Biomedicines 2024; 12:1513. [PMID: 39062086 PMCID: PMC11274533 DOI: 10.3390/biomedicines12071513] [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: 05/29/2024] [Revised: 06/29/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND The involvement of neutrophil-related genes (NRGs) in patients with osteosarcoma (OS) has not been adequately explored. In this study, we aimed to examine the association between NRGs and the prognosis as well as the tumor microenvironment of OS. METHODS The OS data were obtained from the TARGET-OS and GEO database. Initially, we extracted NRGs by intersecting 538 NRGs from single-cell RNA sequencing (scRNA-seq) data between aneuploid and diploid groups, as well as 161 up-regulated differentially expressed genes (DEGs) from the TARGET-OS datasets. Subsequently, we conducted Least Absolute Shrinkage and Selection Operator (Lasso) analyses to identify the hub genes for constructing the NRG-score and NRG-signature. To assess the prognostic value of the NRG signatures in OS, we performed Kaplan-Meier analysis and generated time-dependent receiver operating characteristic (ROC) curves. Gene enrichment analysis (GSEA) and gene set variation analysis (GSVA) were utilized to ascertain the presence of tumor immune microenvironments (TIMEs) and immunomodulators (IMs). Additionally, the KEGG neutrophil signaling pathway was evaluated using ssGSEA. Subsequently, PCR and IHC were conducted to validate the expression of hub genes and transcription factors (TFs) in K7M2-induced OS mice. RESULTS FCER1G and C3AR1 have been identified as prognostic biomarkers for overall survival. The findings indicate a significantly improved prognosis for OS patients. The effectiveness and precision of the NRG signature in prognosticating OS patients were validated through survival ROC curves and an external validation dataset. The results clearly demonstrate that patients with elevated NRG scores exhibit decreased levels of immunomodulators, stromal score, immune score, ESTIMATE score, and infiltrating immune cell populations. Furthermore, our findings substantiate the potential role of SPI1 as a transcription factor in the regulation of the two central genes involved in osteosarcoma development. Moreover, our analysis unveiled a significant correlation and activation of the KEGG neutrophil signaling pathway with FCER1G and C3AR1. Notably, PCR and IHC demonstrated a significantly higher expression of C3AR1, FCER1G, and SPI1 in Balb/c mice induced with K7M2. CONCLUSIONS Our research emphasizes the significant contribution of neutrophils within the TIME of osteosarcoma. The newly developed NRG signature could serve as a good instrument for evaluating the prognosis and therapeutic approach for OS.
Collapse
Affiliation(s)
- Jing Lu
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou 215025, China;
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China; (J.R.); (X.-Y.X.)
| | - Jiang Rui
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China; (J.R.); (X.-Y.X.)
| | - Xiao-Yu Xu
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China; (J.R.); (X.-Y.X.)
| | - Jun-Kang Shen
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou 215025, China;
| |
Collapse
|
5
|
Roberts BK, Li DI, Somerville C, Matta B, Jha V, Steinke A, Brune Z, Blanc L, Soffer SZ, Barnes BJ. IRF5 suppresses metastasis through the regulation of tumor-derived extracellular vesicles and pre-metastatic niche formation. Sci Rep 2024; 14:15557. [PMID: 38969706 PMCID: PMC11226449 DOI: 10.1038/s41598-024-66168-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024] Open
Abstract
Metastasis is driven by extensive cooperation between a tumor and its microenvironment, resulting in the adaptation of molecular mechanisms that evade the immune system and enable pre-metastatic niche (PMN) formation. Little is known of the tumor-intrinsic factors that regulate these mechanisms. Here we show that expression of the transcription factor interferon regulatory factor 5 (IRF5) in osteosarcoma (OS) and breast carcinoma (BC) clinically correlates with prolonged survival and decreased secretion of tumor-derived extracellular vesicles (t-dEVs). Conversely, loss of intra-tumoral IRF5 establishes a PMN that supports metastasis. Mechanistically, IRF5-positive tumor cells retain IRF5 transcripts within t-dEVs that contribute to altered composition, secretion, and trafficking of t-dEVs to sites of metastasis. Upon whole-body pre-conditioning with t-dEVs from IRF5-high or -low OS and BC cells, we found increased lung metastatic colonization that replicated findings from orthotopically implanted cancer cells. Collectively, our findings uncover a new role for IRF5 in cancer metastasis through its regulation of t-dEV programming of the PMN.
Collapse
Affiliation(s)
- Bailey K Roberts
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Dan Iris Li
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Carter Somerville
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Bharati Matta
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Vaishali Jha
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | | | - Zarina Brune
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| | - Lionel Blanc
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
- Departments of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, 11549, USA
| | - Samuel Z Soffer
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
- Department of Pediatric Surgery, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, 11549, USA
| | - Betsy J Barnes
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
- Departments of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, 11549, USA.
| |
Collapse
|
6
|
Pearce SM, Cross NA, Smith DP, Clench MR, Flint LE, Hamm G, Goodwin R, Langridge JI, Claude E, Cole LM. Multimodal Mass Spectrometry Imaging of an Osteosarcoma Multicellular Tumour Spheroid Model to Investigate Drug-Induced Response. Metabolites 2024; 14:315. [PMID: 38921450 PMCID: PMC11205347 DOI: 10.3390/metabo14060315] [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: 04/18/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
A multimodal mass spectrometry imaging (MSI) approach was used to investigate the chemotherapy drug-induced response of a Multicellular Tumour Spheroid (MCTS) 3D cell culture model of osteosarcoma (OS). The work addresses the critical demand for enhanced translatable early drug discovery approaches by demonstrating a robust spatially resolved molecular distribution analysis in tumour models following chemotherapeutic intervention. Advanced high-resolution techniques were employed, including desorption electrospray ionisation (DESI) mass spectrometry imaging (MSI), to assess the interplay between metabolic and cellular pathways in response to chemotherapeutic intervention. Endogenous metabolite distributions of the human OS tumour models were complemented with subcellularly resolved protein localisation by the detection of metal-tagged antibodies using Imaging Mass Cytometry (IMC). The first application of matrix-assisted laser desorption ionization-immunohistochemistry (MALDI-IHC) of 3D cell culture models is reported here. Protein localisation and expression following an acute dosage of the chemotherapy drug doxorubicin demonstrated novel indications for mechanisms of region-specific tumour survival and cell-cycle-specific drug-induced responses. Previously unknown doxorubicin-induced metabolite upregulation was revealed by DESI-MSI of MCTSs, which may be used to inform mechanisms of chemotherapeutic resistance. The demonstration of specific tumour survival mechanisms that are characteristic of those reported for in vivo tumours has underscored the increasing value of this approach as a tool to investigate drug resistance.
Collapse
Affiliation(s)
- Sophie M. Pearce
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - Neil A. Cross
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - David P. Smith
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - Malcolm R. Clench
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - Lucy E. Flint
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, The Discovery Centre (DISC), Biomedical Campus, 1 Francis Crick Ave, Trumpington, Cambridge CB2 0AA, UK; (L.E.F.); (G.H.); (R.G.)
| | - Gregory Hamm
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, The Discovery Centre (DISC), Biomedical Campus, 1 Francis Crick Ave, Trumpington, Cambridge CB2 0AA, UK; (L.E.F.); (G.H.); (R.G.)
| | - Richard Goodwin
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, The Discovery Centre (DISC), Biomedical Campus, 1 Francis Crick Ave, Trumpington, Cambridge CB2 0AA, UK; (L.E.F.); (G.H.); (R.G.)
| | - James I. Langridge
- Waters Corporation, Stamford Avenue, Altrincham Road, Wilmslow, Cheshire SK9 4AX, UK; (J.I.L.); (E.C.)
| | - Emmanuelle Claude
- Waters Corporation, Stamford Avenue, Altrincham Road, Wilmslow, Cheshire SK9 4AX, UK; (J.I.L.); (E.C.)
| | - Laura M. Cole
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| |
Collapse
|
7
|
Bencze N, Scheich B, Szőke É, Wilhelm I, Körmöndi S, Botz B, Helyes Z. Osteosarcoma-Induced Pain Is Mediated by Glial Cell Activation in the Spinal Dorsal Horn, but Not Capsaicin-Sensitive Nociceptive Neurons: A Complex Functional and Morphological Characterization in Mice. Cancers (Basel) 2024; 16:1788. [PMID: 38791867 PMCID: PMC11120600 DOI: 10.3390/cancers16101788] [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: 02/15/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/26/2024] Open
Abstract
Bone cancer and its related chronic pain are huge clinical problems since the available drugs are often ineffective or cannot be used long term due to a broad range of side effects. The mechanisms, mediators and targets need to be identified to determine potential novel therapies. Here, we characterize a mouse bone cancer model induced by intratibial injection of K7M2 osteosarcoma cells using an integrative approach and investigate the role of capsaicin-sensitive peptidergic sensory nerves. The mechanical pain threshold was assessed by dynamic plantar aesthesiometry, limb loading by dynamic weight bearing, spontaneous pain-related behaviors via observation, knee diameter with a digital caliper, and structural changes by micro-CT and glia cell activation by immunohistochemistry in BALB/c mice of both sexes. Capsaicin-sensitive peptidergic sensory neurons were defunctionalized by systemic pretreatment with a high dose of the transient receptor potential vanilloid 1 (TRPV1) agonist resiniferatoxin (RTX). During the 14- and 28-day experiments, weight bearing on the affected limb and the paw mechanonociceptive thresholds significantly decreased, demonstrating secondary mechanical hyperalgesia. Signs of spontaneous pain and osteoplastic bone remodeling were detected both in male and female mice without any sex differences. Microglia activation was shown by the increased ionized calcium-binding adapter molecule 1 (Iba1) immunopositivity on day 14 and astrocyte activation by the enhanced glial fibrillary acidic protein (GFAP)-positive cell density on day 28 in the ipsilateral spinal dorsal horn. Interestingly, defunctionalization of the capsaicin-sensitive afferents representing approximately 2/3 of the nociceptive fibers did not alter any functional parameters. Here, we provide the first complex functional and morphological characterization of the K7M2 mouse osteosarcoma model. Bone-cancer-related chronic pain and hyperalgesia are likely to be mediated by central sensitization involving neuroinflammation via glial cell activation in the spinal dorsal horn, but not the capsaicin-sensitive sensory neuronal system.
Collapse
Affiliation(s)
- Noémi Bencze
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (N.B.); (É.S.); (B.B.)
- National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
| | - Bálint Scheich
- Department of Pathology and Experimental Cancer Research, Faculty of Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary;
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (N.B.); (É.S.); (B.B.)
- National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- Hungarian Research Network, Chronic Pain Research Group (HUN-REN-PTE), 7624 Pécs, Hungary
| | - Imola Wilhelm
- Institute of Biophysics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary;
| | - Sándor Körmöndi
- Department of Traumatology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | - Bálint Botz
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (N.B.); (É.S.); (B.B.)
- Department of Medical Imaging, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (N.B.); (É.S.); (B.B.)
- National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- Hungarian Research Network, Chronic Pain Research Group (HUN-REN-PTE), 7624 Pécs, Hungary
- PharmInVivo Ltd., Szondy György Str. 10, 7629 Pécs, Hungary
| |
Collapse
|
8
|
Fischer S, Creytens D, Gijsels S, Descamps B, Lapeire L, Hendrix A, Sys G, De Wever O. Generation of post-surgical minimal residual disease models to investigate metastasis in soft tissue sarcoma patient-derived orthotopic xenografts. STAR Protoc 2024; 5:102863. [PMID: 38421864 PMCID: PMC10910305 DOI: 10.1016/j.xpro.2024.102863] [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: 10/24/2023] [Revised: 12/20/2023] [Accepted: 01/18/2024] [Indexed: 03/02/2024] Open
Abstract
Despite optimal multimodal treatment including surgical resection, 50%-80% of high-grade soft tissue sarcoma (STS) patients metastasize. Here, we present a protocol for the generation and use of post-surgical minimal residual disease models to investigate metastatic relapse in STS patient-derived xenografts. We describe steps for orthotopic engraftment of high-grade STS patient-derived tumor tissue. We then detail procedures for primary tumor resection with broad, negative resection margins and follow-up until metastases using MRI. For complete details on the use and execution of this protocol, please refer to Fischer et al. (2023).1.
Collapse
Affiliation(s)
- Suzanne Fischer
- Laboratory of Experimental Cancer Research, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent University, Ghent, Belgium; Department of Gastro-Intestinal Surgery, Ghent University Hospital, Ghent, Belgium.
| | - David Creytens
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium; Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Stefanie Gijsels
- Laboratory of Experimental Cancer Research, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent University, Ghent, Belgium; Department of Gastro-Intestinal Surgery, Ghent University Hospital, Ghent, Belgium
| | - Benedicte Descamps
- Animalarium, Radiological and Radiobiological Techniques, Histology Core, Ghent University, Ghent, Belgium
| | - Lore Lapeire
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium; Department of Medical Oncology, Ghent University Hospital, Ghent, Belgium
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Gwen Sys
- Laboratory of Experimental Cancer Research, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent University, Ghent, Belgium; Department of Orthopedics and Traumatology, Ghent University Hospital, Ghent, Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.
| |
Collapse
|
9
|
Ringwalt EM, Currier MA, Glaspell AM, Chen CY, Cannon MV, Cam M, Gross AC, Gust M, Wang PY, Boon L, Biederman LE, Schwarz E, Rajappa P, Lee DA, Mardis ER, Carson WE, Roberts RD, Cripe TP. Trabectedin Enhances Oncolytic Virotherapy by Reducing Barriers to Virus Spread and Cytotoxic Immunity in Preclinical Pediatric Bone Sarcoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.02.582994. [PMID: 38464161 PMCID: PMC10925327 DOI: 10.1101/2024.03.02.582994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
We previously reported that the DNA alkylator and transcriptional-blocking chemotherapeutic agent trabectedin enhances oncolytic herpes simplex viroimmunotherapy in human sarcoma xenograft models, though the mechanism remained to be elucidated. Here we report trabectedin disrupts the intrinsic cellular anti-viral response which increases viral transcript spread throughout the human tumor cells. We also extended our synergy findings to syngeneic murine sarcoma models, which are poorly susceptible to virus infection. In the absence of robust virus replication, we found trabectedin enhanced viroimmunotherapy efficacy by reducing immunosuppressive macrophages and stimulating granzyme expression in infiltrating T and NK cells to cause immune-mediated tumor regressions. Thus, trabectedin enhances both the direct virus-mediated killing of tumor cells and the viral-induced activation of cytotoxic effector lymphocytes to cause tumor regressions across models. Our data provide a strong rationale for clinical translation as both mechanisms should be simultaneously active in human patients.
Collapse
|
10
|
Griffin KH, Thorpe SW, Sebastian A, Hum NR, Coonan TP, Sagheb IS, Loots GG, Randall RL, Leach JK. Engineered bone marrow as a clinically relevant ex vivo model for primary bone cancer research and drug screening. Proc Natl Acad Sci U S A 2023; 120:e2302101120. [PMID: 37729195 PMCID: PMC10523456 DOI: 10.1073/pnas.2302101120] [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: 02/07/2023] [Accepted: 08/17/2023] [Indexed: 09/22/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone cancer in children and adolescents. While numerous other cancers now have promising therapeutic advances, treatment options for OS have remained unchanged since the advent of standard chemotherapeutics and offer less than a 25% 5-y survival rate for those with metastatic disease. This dearth of clinical progress underscores a lack of understanding of OS progression and necessitates the study of this disease in an innovative system. Here, we adapt a previously described engineered bone marrow (eBM) construct for use as a three-dimensional platform to study how microenvironmental and immune factors affect OS tumor progression. We form eBM by implanting acellular bone-forming materials in mice and explanting the cellularized constructs after 8 wk for study. We interrogate the influence of the anatomical implantation site on eBM tissue quality, test ex vivo stability under normoxic (5% O2) and standard (21% O2) culture conditions, culture OS cells within these constructs, and compare them to human OS samples. We show that eBM stably recapitulates the composition of native bone marrow. OS cells exhibit differential behavior dependent on metastatic potential when cultured in eBM, thus mimicking in vivo conditions. Furthermore, we highlight the clinical applicability of eBM as a drug-screening platform through doxorubicin treatment and show that eBM confers a protective effect on OS cells that parallel clinical responses. Combined, this work presents eBM as a cellular construct that mimics the complex bone marrow environment that is useful for mechanistic bone cancer research and drug screening.
Collapse
Affiliation(s)
- Katherine H. Griffin
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA95817
- School of Veterinary Medicine, University of California, Davis, CA95616
| | - Steven W. Thorpe
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA95817
| | - Aimy Sebastian
- Physical and Life Sciences, Lawrence Livermore National Laboratory, Livermore, CA94550
| | - Nicholas R. Hum
- Physical and Life Sciences, Lawrence Livermore National Laboratory, Livermore, CA94550
| | - Thomas P. Coonan
- Department of Biomedical Engineering, University of California, Davis, CA95616
| | - Isabel S. Sagheb
- Department of Biomedical Engineering, University of California, Davis, CA95616
| | - Gabriela G. Loots
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA95817
- Physical and Life Sciences, Lawrence Livermore National Laboratory, Livermore, CA94550
| | - R. Lor Randall
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA95817
| | - J. Kent Leach
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA95817
- Department of Biomedical Engineering, University of California, Davis, CA95616
| |
Collapse
|
11
|
Ge J, Yang N, Yang Y, Yu H, Yang X, Wang Y, Wang T, Cheng S, Wang Y, Han Z, Teng Y, Zou J, Yang H, Cheng L. The combination of eddy thermal effect of biodegradable magnesium with immune checkpoint blockade shows enhanced efficacy against osteosarcoma. Bioact Mater 2023; 25:73-85. [PMID: 36733928 PMCID: PMC9883145 DOI: 10.1016/j.bioactmat.2023.01.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/02/2023] [Accepted: 01/12/2023] [Indexed: 01/24/2023] Open
Abstract
Osteosarcoma (OS) patients have a poor prognosis due to its high degree of heterogeneity and high rate of metastasis. Magnetic hyperthermia therapy (MHT) combined with immunotherapy is an effective strategy to treat solid and metastatic tumors. Here, we combined biodegradable magnesium (Mg) macroscale rods, which acted as an eddy thermo-magnetic agent under a low external alternating magnetic field, and immunotherapy to achieve a radical cure for OS. The eddy thermal effect (ETE) of the Mg rods (MgR) showed outstanding cytotoxic effects and enhanced the maturation of dendritic cells (DCs), and the mild MHT induced the immunogenic cell death (ICD) in the OS cells. Combined with immune checkpoint blockade (ICB) therapy, we obtained an excellent curative effect against OS, and a further evaluation demonstrated that the local MHT induced by the MgR increased T cells infiltration and the polarization of M1 macrophages. Interestingly, the biodegradable MgR also promoted bone osteogenesis. Our work highlighted the uneven ETE mediated by the biodegradable MgR induced a comprehensive immunologic activation in the OS tumor microenvironment (TME), which would inspire the application of MHT for the effective treatment of OS.
Collapse
Affiliation(s)
- Jun Ge
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu, China
| | - Nailin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu, China
| | - Yuqi Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu, China
| | - Hao Yu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaoyuan Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu, China
| | - Yingjie Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Tianyi Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shuning Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu, China
| | - Yuanjie Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu, China
| | - Zhihui Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu, China
| | - Yun Teng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Zou
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Corresponding author.
| | - Huilin Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Corresponding author.
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu, China,Corresponding author.
| |
Collapse
|
12
|
Hebron KE, Wan X, Roth JS, Liewehr DJ, Sealover NE, Frye WJ, Kim A, Stauffer S, Perkins OL, Sun W, Isanogle KA, Robinson CM, James A, Awasthi P, Shankarappa P, Luo X, Lei H, Butcher D, Smith R, Edmondson EF, Chen JQ, Kedei N, Peer CJ, Shern JF, Figg WD, Chen L, Hall MD, Difilippantonio S, Barr FG, Kortum RL, Robey RW, Vaseva AV, Khan J, Yohe ME. The Combination of Trametinib and Ganitumab is Effective in RAS-Mutated PAX-Fusion Negative Rhabdomyosarcoma Models. Clin Cancer Res 2023; 29:472-487. [PMID: 36322002 PMCID: PMC9852065 DOI: 10.1158/1078-0432.ccr-22-1646] [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: 05/23/2022] [Revised: 09/22/2022] [Accepted: 10/31/2022] [Indexed: 11/05/2022]
Abstract
PURPOSE PAX-fusion negative rhabdomyosarcoma (FN RMS) is driven by alterations in the RAS/MAP kinase pathway and is partially responsive to MEK inhibition. Overexpression of IGF1R and its ligands is also observed in FN RMS. Preclinical and clinical studies have suggested that IGF1R is itself an important target in FN RMS. Our previous studies revealed preclinical efficacy of the MEK1/2 inhibitor, trametinib, and an IGF1R inhibitor, BMS-754807, but this combination was not pursued clinically due to intolerability in preclinical murine models. Here, we sought to identify a combination of an MEK1/2 inhibitor and IGF1R inhibitor, which would be tolerated in murine models and effective in both cell line and patient-derived xenograft models of RAS-mutant FN RMS. EXPERIMENTAL DESIGN Using proliferation and apoptosis assays, we studied the factorial effects of trametinib and ganitumab (AMG 479), a mAb with specificity for human and murine IGF1R, in a panel of RAS-mutant FN RMS cell lines. The molecular mechanism of the observed synergy was determined using conventional and capillary immunoassays. The efficacy and tolerability of trametinib/ganitumab was assessed using a panel of RAS-mutated cell-line and patient-derived RMS xenograft models. RESULTS Treatment with trametinib and ganitumab resulted in synergistic cellular growth inhibition in all cell lines tested and inhibition of tumor growth in four of six models of RAS-mutant RMS. The combination had little effect on body weight and did not produce thrombocytopenia, neutropenia, or hyperinsulinemia in tumor-bearing SCID beige mice. Mechanistically, ganitumab treatment prevented the phosphorylation of AKT induced by MEK inhibition alone. Therapeutic response to the combination was observed in models without a mutation in the PI3K/PTEN axis. CONCLUSIONS We demonstrate that combined trametinib and ganitumab is effective in a genomically diverse panel of RAS-mutated FN RMS preclinical models. Our data also show that the trametinib/ganitumab combination likely has a favorable tolerability profile. These data support testing this combination in a phase I/II clinical trial for pediatric patients with relapsed or refractory RAS-mutated FN RMS.
Collapse
Affiliation(s)
- Katie E. Hebron
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892,Laboratory of Cell and Developmental Signaling, Center for Cancer Research, 8560 Progress Drive, Frederick, MD 21701
| | - Xiaolin Wan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Jacob S. Roth
- Early Translation Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD 20850
| | - David J. Liewehr
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Nancy E. Sealover
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Services, Bethesda, MD 20814
| | - William J.E. Frye
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892
| | - Angela Kim
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, 8560 Progress Drive, Frederick, MD 21701
| | - Stacey Stauffer
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, 8560 Progress Drive, Frederick, MD 21701
| | - Olivia L. Perkins
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Wenyue Sun
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892
| | - Kristine A. Isanogle
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Christina M. Robinson
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Amy James
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Parirokh Awasthi
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Priya Shankarappa
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Xiaoling Luo
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Haiyan Lei
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Donna Butcher
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Roberta Smith
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Elijah F. Edmondson
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Jin-Qiu Chen
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Noemi Kedei
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Cody J. Peer
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Jack F. Shern
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - W. Douglas Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Lu Chen
- Early Translation Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD 20850
| | - Matthew D. Hall
- Early Translation Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD 20850
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Frederic G. Barr
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892
| | - Robert L. Kortum
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Services, Bethesda, MD 20814
| | - Robert W. Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892
| | - Angelina V. Vaseva
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, Texas, USA
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892,Co-corresponding authors Correspondence: Marielle Yohe, M.D., Ph.D., Center for Cancer Research, National Cancer Institute, 8560 Progress Drive Room D3026, Frederick, MD 27101, Phone: (240) 760-7436,
| | - Marielle E. Yohe
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892,Laboratory of Cell and Developmental Signaling, Center for Cancer Research, 8560 Progress Drive, Frederick, MD 21701,Co-corresponding authors Correspondence: Marielle Yohe, M.D., Ph.D., Center for Cancer Research, National Cancer Institute, 8560 Progress Drive Room D3026, Frederick, MD 27101, Phone: (240) 760-7436,
| |
Collapse
|
13
|
Beird HC, Bielack SS, Flanagan AM, Gill J, Heymann D, Janeway KA, Livingston JA, Roberts RD, Strauss SJ, Gorlick R. Osteosarcoma. Nat Rev Dis Primers 2022; 8:77. [PMID: 36481668 DOI: 10.1038/s41572-022-00409-y] [Citation(s) in RCA: 203] [Impact Index Per Article: 67.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2022] [Indexed: 12/13/2022]
Abstract
Osteosarcoma is the most common primary malignant tumour of the bone. Osteosarcoma incidence is bimodal, peaking at 18 and 60 years of age, and is slightly more common in males. The key pathophysiological mechanism involves several possible genetic drivers of disease linked to bone formation, causing malignant progression and metastasis. While there have been significant improvements in the outcome of patients with localized disease, with event-free survival outcomes exceeding 60%, in patients with metastatic disease, event-free survival outcomes remain poor at less than 30%. The suspicion of osteosarcoma based on radiographs still requires pathological evaluation of a bone biopsy specimen for definitive diagnosis and CT imaging of the chest should be performed to identify lung nodules. So far, population-based screening and surveillance strategies have not been implemented due to the rarity of osteosarcoma and the lack of reliable markers. Current screening focuses only on groups at high risk such as patients with genetic cancer predisposition syndromes. Management of osteosarcoma requires a multidisciplinary team of paediatric and medical oncologists, orthopaedic and general surgeons, pathologists, radiologists and specialist nurses. Survivors of osteosarcoma require specialized medical follow-up, as curative treatment consisting of chemotherapy and surgery has long-term adverse effects, which also affect the quality of life of patients. The development of osteosarcoma model systems and related research as well as the evaluation of new treatment approaches are ongoing to improve disease outcomes, especially for patients with metastases.
Collapse
Affiliation(s)
- Hannah C Beird
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stefan S Bielack
- Pediatric Oncology, Hematology, Immunology, Klinikum Stuttgart - Olgahospital, Stuttgart Cancer Center, Stuttgart, Germany
| | - Adrienne M Flanagan
- Research Department of Pathology, Cancer Institute, University College London, London, UK
| | - Jonathan Gill
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dominique Heymann
- Nantes Université, CNRS, UMR6286, US2B, Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Katherine A Janeway
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - J Andrew Livingston
- Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ryan D Roberts
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Sandra J Strauss
- University College London Hospitals NHS Foundation Trust, University College London, London, UK
| | - Richard Gorlick
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA. .,Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
14
|
Beck J, Ren L, Huang S, Berger E, Bardales K, Mannheimer J, Mazcko C, LeBlanc A. Canine and murine models of osteosarcoma. Vet Pathol 2022; 59:399-414. [PMID: 35341404 PMCID: PMC9290378 DOI: 10.1177/03009858221083038] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor in children. Despite efforts to develop and implement new therapies, patient outcomes have not measurably improved since the 1980s. Metastasis continues to be the main source of patient mortality, with 30% of cases developing metastatic disease within 5 years of diagnosis. Research models are critical in the advancement of cancer research and include a variety of species. For example, xenograft and patient-derived xenograft (PDX) mouse models provide opportunities to study human tumor cells in vivo while transgenic models have offered significant insight into the molecular mechanisms underlying OS development. A growing recognition of naturally occurring cancers in companion species has led to new insights into how veterinary patients can contribute to studies of cancer biology and drug development. The study of canine cases, including the use of diagnostic tissue archives and clinical trials, offers a potential mechanism to further canine and human cancer research. Advancement in the field of OS research requires continued development and appropriate use of animal models. In this review, animal models of OS are described with a focus on the mouse and tumor-bearing pet dog as parallel and complementary models of human OS.
Collapse
Affiliation(s)
| | - Ling Ren
- National Cancer Institute, Bethesda, MD
| | | | | | - Kathleen Bardales
- National Cancer Institute, Bethesda, MD
- University of Pennsylvania, Philadelphia, PA
| | | | | | | |
Collapse
|
15
|
Impact of limb amputation and cisplatin chemotherapy on metastatic progression in mouse models of osteosarcoma. Sci Rep 2021; 11:24435. [PMID: 34952927 PMCID: PMC8709858 DOI: 10.1038/s41598-021-04018-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/08/2021] [Indexed: 12/23/2022] Open
Abstract
Development of animal models that accurately recapitulate human cancer is an ongoing challenge. This is particularly relevant in the study of osteosarcoma (OS), a highly malignant bone tumor diagnosed in approximately 1000 pediatric/adolescent patients each year. Metastasis is the leading cause of patient death underscoring the need for relevant animal models of metastatic OS. In this study, we describe how existing OS mouse models can be interrogated in a time-course context to determine the kinetics of spontaneous metastasis from an orthotopically implanted primary tumor. We evaluated four highly metastatic OS cell lines (3 human, 1 mouse) to establish a timeline for metastatic progression in immune deficient NSG mice. To discern the effects of therapy on tumor development and metastasis in these models, we investigated cisplatin therapy and surgical limb amputation at early and late timepoints. These data help define the appropriate observational periods for studies of metastatic progression in OS and further our understanding of existing mouse models. Efforts to advance the study of metastatic OS are critical for facilitating the identification of novel therapeutics and for improving patient survival.
Collapse
|
16
|
Yu X, Yustein JT, Xu J. Research models and mesenchymal/epithelial plasticity of osteosarcoma. Cell Biosci 2021; 11:94. [PMID: 34022967 PMCID: PMC8141200 DOI: 10.1186/s13578-021-00600-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022] Open
Abstract
Most osteosarcomas (OSs) develop from mesenchymal cells at the bone with abnormal growth in young patients. OS has an annual incidence of 3.4 per million people and a 60-70% 5-year surviving rate. About 20% of OS patients have metastasis at diagnosis, and only 27% of patients with metastatic OS survive longer than 5 years. Mutation of tumor suppressors RB1, TP53, REQL4 and INK4a and/or deregulation of PI3K/mTOR, TGFβ, RANKL/NF-κB and IGF pathways have been linked to OS development. However, the agents targeting these pathways have yielded disappointing clinical outcomes. Surgery and chemotherapy remain the main treatments of OS. Recurrent and metastatic OSs are commonly resistant to these therapies. Spontaneous canine models, carcinogen-induced rodent models, transgenic mouse models, human patient-derived xenograft models, and cell lines from animal and human OSs have been developed for studying the initiation, growth and progression of OS and testing candidate drugs of OS. The cell plasticity regulated by epithelial-to-mesenchymal transition transcription factors (EMT-TFs) such as TWIST1, SNAIL, SLUG, ZEB1 and ZEB2 plays an important role in maintenance of the mesenchymal status and promotion of cell invasion and metastasis of OS cells. Multiple microRNAs including miR-30/9/23b/29c/194/200, proteins including SYT-SSX1/2 fusion proteins and OVOL2, and other factors that inhibit AMF/PGI and LRP5 can suppress either the expression or activity of EMT-TFs to increase epithelial features and inhibit OS metastasis. Further understanding of the molecular mechanisms that regulate OS cell plasticity should provide potential targets and therapeutic strategies for improving OS treatment.
Collapse
Affiliation(s)
- Xiaobin Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Jason T Yustein
- Department of Pediatrics, Texas Children's Cancer and Hematology Center, and The Faris D. Virani Ewing Sarcoma Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
17
|
Hennessy M, Wahba A, Felix K, Cabrera M, Segura MG, Kundra V, Ravoori MK, Stewart J, Kleinerman ES, Jensen VB, Gopalakrishnan V, Pena R, Quach P, Kim G, Kivimäe S, Madakamutil L, Overwijk WW, Zalevsky J, Gordon N. Bempegaldesleukin (BEMPEG; NKTR-214) efficacy as a single agent and in combination with checkpoint-inhibitor therapy in mouse models of osteosarcoma. Int J Cancer 2021; 148:1928-1937. [PMID: 33152115 PMCID: PMC7984260 DOI: 10.1002/ijc.33382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/04/2020] [Accepted: 10/02/2020] [Indexed: 12/30/2022]
Abstract
Survival of patients with relapsed/refractory osteosarcoma has not improved in the last 30 years. Several immunotherapeutic approaches have shown benefit in murine osteosarcoma models, including the anti-programmed death-1 (anti-PD-1) and anti-cytotoxic T-lymphocyte antigen-4 (anti-CTLA-4) immune checkpoint inhibitors. Treatment with the T-cell growth factor interleukin-2 (IL-2) has shown some clinical benefit but has limitations due to poor tolerability. Therefore, we evaluated the efficacy of bempegaldesleukin (BEMPEG; NKTR-214), a first-in-class CD122-preferential IL-2 pathway agonist, alone and in combination with anti-PD-1 or anti-CTLA-4 immune checkpoint inhibitors in metastatic and orthotopic murine models of osteosarcoma. Treatment with BEMPEG delayed tumor growth and increased overall survival of mice with K7M2-WT osteosarcoma pulmonary metastases. BEMPEG also inhibited primary tumor growth and metastatic relapse in lungs and bone in the K7M3 orthotopic osteosarcoma mouse model. In addition, it enhanced therapeutic activity of anti-CTLA-4 and anti-PD-1 checkpoint blockade in the DLM8 subcutaneous murine osteosarcoma model. Finally, BEMPEG strongly increased accumulation of intratumoral effector T cells and natural killer cells, but not T-regulatory cells, resulting in improved effector:inhibitory cell ratios. Collectively, these data in multiple murine models of osteosarcoma provide a path toward clinical evaluation of BEMPEG-based regimens in human osteosarcoma.
Collapse
Affiliation(s)
| | - Andrew Wahba
- Children's Memorial Hermann HospitalUT Health Science CenterHoustonTexasUSA
| | - Kumar Felix
- Department of Pharmaceutical SciencesHampton UniversityHamptonVirginiaUSA
| | - Mariella Cabrera
- Department of PediatricsLincoln Medical and Mental Health CenterNew YorkNew YorkUSA
| | | | - Vikas Kundra
- Division of Pediatrics, Department of Pediatrics ResearchThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Murali K. Ravoori
- Division of Pediatrics, Department of Pediatrics ResearchThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - John Stewart
- Division of Pediatrics, Department of Pediatrics ResearchThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Eugenie S. Kleinerman
- Division of Pediatrics, Department of Pediatrics ResearchThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Vanessa Behrana Jensen
- Division of Pediatrics, Department of Pediatrics ResearchThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Vidya Gopalakrishnan
- Division of Pediatrics, Department of Pediatrics ResearchThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | | | - Phi Quach
- Nektar TherapeuticsSan FranciscoCaliforniaUSA
| | - Grace Kim
- Nektar TherapeuticsSan FranciscoCaliforniaUSA
- Verge GenomicsSouth San FranciscoCaliforniaUSA
| | | | - Loui Madakamutil
- Nektar TherapeuticsSan FranciscoCaliforniaUSA
- InvivoscribeSan DiegoCAUSA
| | | | | | - Nancy Gordon
- Division of Pediatrics, Department of Pediatrics ResearchThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
18
|
Long SA, Huang S, Kambala A, Ren L, Wilson J, Goetz M, Hao X, Yang X, Goncharova EI, Jia L, LeBlanc A, Khanna C, Henrich CJ, Beutler JA. Identification of potential modulators of osteosarcoma metastasis by high-throughput cellular screening of natural products. Chem Biol Drug Des 2021; 97:77-86. [PMID: 32666679 PMCID: PMC8808376 DOI: 10.1111/cbdd.13762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/26/2020] [Accepted: 07/05/2020] [Indexed: 12/17/2022]
Abstract
A high-throughput screening assay was developed and applied to a large library of natural product extract samples, in order to identify compounds which preferentially inhibited the in vitro 2D growth of a highly metastatic osteosarcoma cell line (MG63.3) compared to a cognate parental cell line (MG63) with low metastatic potential. Evaluation of differentially active natural product extracts with bioassay-guided fractionation led to the identification of lovastatin (IC50 = 11 µm) and the limonoid toosendanin (IC50 = 26 nm). Other statins and limonoids were then tested, and cerivastatin was identified as a particularly potent (IC50 < 0.1 µm) and selective agent. These compounds potently and selectively induced apoptosis in MG63.3 cells, but not MG63. Assays with other cell pairs were used to examine the generality of these results. Statins and limonoids may represent unexplored opportunities for development of modulators of osteosarcoma metastasis. As cerivastatin was previously approved for clinical use, it could be considered for repurposing in osteosarcoma, pending validation in further models.
Collapse
Affiliation(s)
- Sarah A. Long
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Shan Huang
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Anusha Kambala
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Ling Ren
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jennifer Wilson
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Michael Goetz
- Natural Products Discovery Institute, Doylestown, PA, USA
| | - Xiaojiang Hao
- Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China
| | - Xiaosheng Yang
- Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China
| | - Ekaterina I. Goncharova
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Libin Jia
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - Amy LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Chand Khanna
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Curtis J. Henrich
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
- Basic Research Program, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - John A. Beutler
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| |
Collapse
|
19
|
Targetable Intercellular Signaling Pathways Facilitate Lung Colonization in Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 32767237 DOI: 10.1007/978-3-030-43085-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
Outcomes for young people diagnosed with osteosarcoma hinge almost exclusively on whether they develop lung metastasis. The striking predilection that osteosarcoma shows for metastatic spread to lung suggests properties and/or lung interactions that generate tissue-specific survival and proliferation advantages. While these mechanisms remain overall poorly defined, studies have begun to describe biological elements important to metastasis. Mechanisms described to date include both cell-autonomous adaptations that allow disseminated tumor cells to survive the stressors imposed by metastasis and intercellular signaling networks that tumor cells exploit to pirate needed signals from surrounding tissues or to recruit other cells that create a more favorable niche. Evidence suggests that cell-autonomous changes are largely driven by epigenetic reprogramming of disseminated tumor cells that facilitates resistance to late apoptosis, manages endoplasmic reticulum (ER) stressors, promotes translation of complex transcripts, and activates clotting pathways. Tumor-host signaling pathways important for lung colonization drive interactions with lung epithelium, mesenchymal stem cells, and mediators of innate and adaptive immunity. In this chapter, we highlight one particular pathway that integrates cell-autonomous adaptations with lung-specific tumor-host interactions. In this mechanism, aberrant ΔNp63 expression primes tumor cells to produce IL6 and CXCL8 upon interaction with lung epithelial cells. This tumor-derived IL6 and CXCL8 then initiates autocrine, osteosarcoma-lung paracrine, and osteosarcoma-immune paracrine interactions that facilitate metastasis. Importantly, many of these pathways appear targetable with clinically feasible therapeutics. Ongoing work to better understand metastasis is driving efforts to improve outcomes by targeting the most devastating complication of this disease.
Collapse
|
20
|
Zalacain M, Bunuales M, Marrodan L, Labiano S, Gonzalez-Huarriz M, Martinez-Vélez N, Laspidea V, Puigdelloses M, García-Moure M, Gonzalez-Aparicio M, Hernandez-Alcoceba R, Alonso MM, Patiño-García A. Local administration of IL-12 with an HC vector results in local and metastatic tumor control in pediatric osteosarcoma. MOLECULAR THERAPY-ONCOLYTICS 2020; 20:23-33. [PMID: 33575468 PMCID: PMC7851487 DOI: 10.1016/j.omto.2020.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 11/17/2020] [Indexed: 10/28/2022]
Abstract
Osteosarcoma is the most frequent and aggressive bone tumor in children and adolescents, with a long-term survival rate of 30%. Interleukin-12 (IL-12) is a potent cytokine that bridges innate and adaptive immunity, triggers antiangiogenic responses, and achieves potent antitumor effects. In this work, we evaluated the antisarcoma effect of a high-capacity adenoviral vector encoding mouse IL-12. This vector harbored a mifepristone-inducible system for controlled expression of IL-12 (High-Capacity adenoviral vector enconding the EF1α promoter [HCA-EFZP]-IL-12). We found that local administration of the vector resulted in a reduction in the tumor burden, extended overall survival, and tumor eradication. Moreover, long-term survivors exhibited immunological memory when rechallenged with the same tumor cells. Treatment with HCA-EFZP-IL-12 also resulted in a significant decrease in lung metastasis. Immunohistochemical analyses showed profound remodeling of the osteosarcoma microenvironment with decreases in angiogenesis and macrophage and myeloid cell numbers. In summary, our data underscore the potential therapeutic value of IL-12 in the context of a drug-inducible system that allows controlled expression of this cytokine, which can trigger a potent antitumor immune response in primary and metastatic pediatric osteosarcoma.
Collapse
Affiliation(s)
- Marta Zalacain
- Health Research Institute of Navarra (IDISNA), 31008 Pamplona, Navarra, Spain.,Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Navarra, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - María Bunuales
- Health Research Institute of Navarra (IDISNA), 31008 Pamplona, Navarra, Spain.,Program in Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Navarra, Spain
| | - Lucía Marrodan
- Health Research Institute of Navarra (IDISNA), 31008 Pamplona, Navarra, Spain.,Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Navarra, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Sara Labiano
- Health Research Institute of Navarra (IDISNA), 31008 Pamplona, Navarra, Spain.,Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Navarra, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Marisol Gonzalez-Huarriz
- Health Research Institute of Navarra (IDISNA), 31008 Pamplona, Navarra, Spain.,Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Navarra, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Naiara Martinez-Vélez
- Health Research Institute of Navarra (IDISNA), 31008 Pamplona, Navarra, Spain.,Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Navarra, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Virginia Laspidea
- Health Research Institute of Navarra (IDISNA), 31008 Pamplona, Navarra, Spain.,Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Navarra, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Montse Puigdelloses
- Health Research Institute of Navarra (IDISNA), 31008 Pamplona, Navarra, Spain.,Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Navarra, Spain
| | - Marc García-Moure
- Health Research Institute of Navarra (IDISNA), 31008 Pamplona, Navarra, Spain.,Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Navarra, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Manuela Gonzalez-Aparicio
- Health Research Institute of Navarra (IDISNA), 31008 Pamplona, Navarra, Spain.,Program in Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Navarra, Spain
| | - Rubén Hernandez-Alcoceba
- Health Research Institute of Navarra (IDISNA), 31008 Pamplona, Navarra, Spain.,Program in Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Navarra, Spain
| | - Marta M Alonso
- Health Research Institute of Navarra (IDISNA), 31008 Pamplona, Navarra, Spain.,Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Navarra, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Ana Patiño-García
- Health Research Institute of Navarra (IDISNA), 31008 Pamplona, Navarra, Spain.,Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Navarra, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
21
|
Kowalczyk JT, Wan X, Hernandez ER, Luo R, Lyons GC, Wilson KM, Gallardo DC, Isanogle KA, Robinson CM, Mendoza A, Heske CM, Chen JQ, Luo X, Kelly AE, Difilippantinio S, Robey RW, Thomas CJ, Sackett DL, Morrison DK, Randazzo PA, Jenkins LMM, Yohe ME. Rigosertib Induces Mitotic Arrest and Apoptosis in RAS-Mutated Rhabdomyosarcoma and Neuroblastoma. Mol Cancer Ther 2020; 20:307-319. [PMID: 33158997 DOI: 10.1158/1535-7163.mct-20-0525] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/16/2020] [Accepted: 10/30/2020] [Indexed: 11/16/2022]
Abstract
Relapsed pediatric rhabdomyosarcomas (RMS) and neuroblastomas (NBs) have a poor prognosis despite multimodality therapy. In addition, the current standard of care for these cancers includes vinca alkaloids that have severe toxicity profiles, further underscoring the need for novel therapies for these malignancies. Here, we show that the small-molecule rigosertib inhibits the growth of RMS and NB cell lines by arresting cells in mitosis, which leads to cell death. Our data indicate that rigosertib, like the vinca alkaloids, exerts its effects mainly by interfering with mitotic spindle assembly. Although rigosertib has the ability to inhibit oncogenic RAS signaling, we provide evidence that rigosertib does not induce cell death through inhibition of the RAS pathway in RAS-mutated RMS and NB cells. However, the combination of rigosertib and the MEK inhibitor trametinib, which has efficacy in RAS-mutated tumors, synergistically inhibits the growth of an RMS cell line, suggesting a new avenue for combination therapy. Importantly, rigosertib treatment delays tumor growth and prolongs survival in a xenograft model of RMS. In conclusion, rigosertib, through its impact on the mitotic spindle, represents a potential therapeutic for RMS.
Collapse
Affiliation(s)
| | - Xiaolin Wan
- National Cancer Institute, Bethesda, Maryland
| | | | - Ruibai Luo
- National Cancer Institute, Bethesda, Maryland
| | | | - Kelli M Wilson
- National Center for Advancing Translational Sciences, Rockville, Maryland
| | | | - Kristine A Isanogle
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Christina M Robinson
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | | | | | | | | | | | - Simone Difilippantinio
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | | | - Craig J Thomas
- National Center for Advancing Translational Sciences, Rockville, Maryland
| | - Dan L Sackett
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland
| | | | | | | | | |
Collapse
|
22
|
Mochizuki Y, Tazawa H, Demiya K, Kure M, Kondo H, Komatsubara T, Sugiu K, Hasei J, Yoshida A, Kunisada T, Urata Y, Kagawa S, Ozaki T, Fujiwara T. Telomerase-specific oncolytic immunotherapy for promoting efficacy of PD-1 blockade in osteosarcoma. Cancer Immunol Immunother 2020; 70:1405-1417. [PMID: 33151368 DOI: 10.1007/s00262-020-02774-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/19/2020] [Indexed: 12/20/2022]
Abstract
Immune checkpoint inhibitors including anti-programmed cell death 1 (PD-1) antibody have recently improved clinical outcome in certain cancer patients; however, osteosarcoma (OS) patients are refractory to PD-1 blockade. Oncolytic virotherapy has emerged as novel immunogenic therapy to augment antitumor immune response. We developed a telomerase-specific replication-competent oncolytic adenovirus OBP-502 that induces lytic cell death via binding to integrins. In this study, we assessed the combined effect of PD-1 blockade and OBP-502 in OS cells. The expression of coxsackie and adenovirus receptor (CAR), integrins αvβ3 and αvβ5, and programmed cell death ligand 1 (PD-L1) was analyzed in two murine OS cells (K7M2, NHOS). The cytopathic activity of OBP-502 in both cells was analyzed using the XTT assay. OBP-502-induced immunogenic cell death was assessed by analyzing the level of extracellular ATP and high-mobility group box protein B1 (HMGB1). Subcutaneous tumor models for K7M2 and NHOS cells were used to evaluate the antitumor effect and number of tumor-infiltrating CD8+ cells in combination therapy. K7M2 and NHOS cells showed high expression of integrins αvβ3 and αvβ5, but not CAR. OBP-502 significantly suppressed the viability of both cells, in which PD-L1 expression and the release of ATP and HMGB1 were significantly increased. Intratumoral injection of OBP-502 significantly augmented the efficacy of PD-1 blockade on subcutaneous K2M2 and NHOS tumor models via enhancement of tumor-infiltrating CD8+ T cells. Our results suggest that telomerase-specific oncolytic virotherapy is a promising antitumor strategy to promote the efficacy of PD-1 blockade in OS.
Collapse
Affiliation(s)
- Yusuke Mochizuki
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Hiroshi Tazawa
- Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan. .,Center for Innovative Clinical Medicine, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Koji Demiya
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Miho Kure
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Hiroya Kondo
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Tadashi Komatsubara
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Kazuhisa Sugiu
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Joe Hasei
- Sports Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, 700-8558, Japan
| | - Aki Yoshida
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Toshiyuki Kunisada
- Medical Materials for Musculoskeletal Reconstruction, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Yasuo Urata
- Oncolys BioPharma, Inc, Tokyo, 105-0001, Japan
| | - Shunsuke Kagawa
- Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan.,Minimally Invasive Therapy Center, Okayama University Hospital, Okayama, 700-8558, Japan
| | - Toshifumi Ozaki
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Toshiyoshi Fujiwara
- Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| |
Collapse
|
23
|
The Role of ALDH in the Metastatic Potential of Osteosarcoma Cells and Potential ALDH Targets. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 32767240 DOI: 10.1007/978-3-030-43085-6_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
Aldehyde dehydrogenases are a family of enzymes that oxidize aldehydes to carboxylic acids. These enzymes are important in cellular homeostasis during oxidative stress by the elimination of toxic aldehyde by-products from various cellular processes. In osteosarcoma, aldehyde dehydrogenase 1A1has been described as a cancer stem cell marker. Its activity has been found to correlate with metastatic potential and the metastatic phenotype. As such, a more complete understanding of aldehyde dehydrogenase in osteosarcoma will give us a deeper knowledge of its impact on osteosarcoma metastatic potential. Our hope is that this knowledge can be translated into novel antimetastatic therapeutic strategies and thus improve osteosarcoma prognoses.
Collapse
|
24
|
Byrgazov K, Anderson C, Salzer B, Bozsaky E, Larsson R, Gullbo J, Lehner M, Lehmann F, Slipicevic A, Kager L, Fryknäs M, Taschner-Mandl S. Targeting aggressive osteosarcoma with a peptidase-enhanced cytotoxic melphalan flufenamide. Ther Adv Med Oncol 2020; 12:1758835920937891. [PMID: 32774473 PMCID: PMC7391428 DOI: 10.1177/1758835920937891] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/22/2020] [Indexed: 12/28/2022] Open
Abstract
Background: Low survival rates in metastatic high-grade osteosarcoma (HGOS) have remained stagnant for the last three decades. This study aims to investigate the role of aminopeptidase N (ANPEP) in HGOS progression and its targeting with a novel lipophilic peptidase-enhanced cytotoxic compound melphalan flufenamide (melflufen) in HGOS. Methods: Meta-analysis of publicly available gene expression datasets was performed to determine the impact of ANPEP gene expression on metastasis-free survival of HGOS patients. The efficacy of standard-of-care anti-neoplastic drugs and a lipophilic peptidase-enhanced cytotoxic conjugate melflufen was investigated in patient-derived HGOS ex vivo models and cell lines. The kinetics of apoptosis and necrosis induced by melflufen and doxorubicin were compared. Anti-neoplastic effects of melflufen were investigated in vivo. Results: Elevated ANPEP expression in diagnostic biopsies of HGOS patients was found to significantly reduce metastasis-free survival. In drug sensitivity assays, melflufen has shown an anti-proliferative effect in HGOS ex vivo samples and cell lines, including those resistant to methotrexate, etoposide, doxorubicin, and PARP inhibitors. Further, HGOS cells treated with melflufen displayed a rapid induction of apoptosis and this sensitivity correlated with high expression of ANPEP. In combination treatments, melflufen demonstrated synergy with doxorubicin in killing HGOS cells. Finally, Melflufen displayed anti-tumor growth and anti-metastatic effects in vivo. Conclusion: This study may pave the way for use of melflufen as an adjuvant to doxorubicin in improving the therapeutic efficacy for the treatment of metastatic HGOS.
Collapse
Affiliation(s)
| | - Claes Anderson
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala, Sweden
| | - Benjamin Salzer
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Eva Bozsaky
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Rolf Larsson
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala, Sweden
| | | | - Manfred Lehner
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | | | | | - Leo Kager
- Department of Pediatrics, St. Anna Children's Hospital, Medical University of Vienna and Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Mårten Fryknäs
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
25
|
McGuire JJ, Nerlakanti N, Lo CH, Tauro M, Utset-Ward TJ, Reed DR, Lynch CC. Histone deacetylase inhibition prevents the growth of primary and metastatic osteosarcoma. Int J Cancer 2020; 147:2811-2823. [PMID: 32599665 DOI: 10.1002/ijc.33046] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/18/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022]
Abstract
Overall survival rates for patients with advanced osteosarcoma have remained static for over three decades. An in vitro analysis of osteosarcoma cell lines for sensitivity to an array of approved cancer therapies revealed that panobinostat, a broad spectrum histone deacetalyase (HDAC) inhibitor, is highly effective at triggering osteosarcoma cell death. Using in vivo models of orthotopic and metastatic osteosarcoma, here we report that panobinostat impairs the growth of primary osteosarcoma in bone and spontaneous metastasis to the lung, the most common site of metastasis for this disease. Further, pretreatment of mice with panobinostat prior to tail vein inoculation of osteosarcoma prevents the seeding and growth of lung metastases. Additionally, panobinostat impaired the growth of established lung metastases and improved overall survival, and these effects were also manifest in the lung metastatic SAOS2-LM7 model. Mechanistically, the efficacy of panobinostat was linked to high expression of HDAC1 and HDAC2 in osteosarcoma, and silencing of HDAC1 and 2 greatly reduced osteosarcoma growth in vitro. In accordance with these findings, treatment with the HDAC1/2 selective inhibitor romidepsin compromised the growth of osteosarcoma in vitro and in vivo. Analysis of patient-derived xenograft osteosarcoma cell lines further demonstrated the sensitivity of the disease to panobinostat or romidepsin. Collectively, these studies provide rationale for clinical trials in osteosarcoma patients using the approved therapies panobinostat or romidepsin.
Collapse
Affiliation(s)
- Jeremy J McGuire
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, Florida, USA.,Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Niveditha Nerlakanti
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, Florida, USA.,Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Chen Hao Lo
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, Florida, USA.,Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Marilena Tauro
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Thomas J Utset-Ward
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.,Department of Orthopaedic Surgery and Rehabilitation Medicine, University of Chicago, Chicago, Illinois, USA
| | - Damon R Reed
- Sarcoma Department & Department of Interdisciplinary Cancer Management (DICaM), H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Conor C Lynch
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| |
Collapse
|
26
|
Maloney C, Kallis MP, Edelman M, Tzanavaris C, Lesser M, Soffer SZ, Symons M, Steinberg BM. Gefitinib Inhibits Invasion and Metastasis of Osteosarcoma via Inhibition of Macrophage Receptor Interacting Serine-Threonine Kinase 2. Mol Cancer Ther 2020; 19:1340-1350. [PMID: 32371577 DOI: 10.1158/1535-7163.mct-19-0903] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/18/2019] [Accepted: 03/26/2020] [Indexed: 11/16/2022]
Abstract
Most patients with osteosarcoma have subclinical pulmonary micrometastases at diagnosis. Mounting evidence suggests that macrophages facilitate metastasis. As the EGFR has been implicated in carcinoma-macrophage cross-talk, in this study, we asked whether gefitinib, an EGFR inhibitor, reduces osteosarcoma invasion and metastatic outgrowth using the K7M2-Balb/c syngeneic murine model. Macrophages enhanced osteosarcoma invasion in vitro, which was suppressed by gefitinib. Oral gefitinib inhibited tumor extravasation in the lung and reduced the size of metastatic foci, resulting in reduced metastatic burden. Gefitinib also altered pulmonary macrophage phenotype, increasing MHCII and decreasing CD206 expression compared with controls. Surprisingly, these effects are mediated through inhibition of macrophage receptor interacting protein kinase 2 (RIPK2), rather than EGFR. Supporting this, lapatinib, a highly specific EGFR inhibitor that does not inhibit RIPK2, had no effect on macrophage-promoted invasion, and RIPK2-/- macrophages failed to promote invasion. The selective RIPK2 inhibitor WEHI-345 blocked tumor cell invasion in vitro and reduced metastatic burden in vivo In conclusion, our results indicate that gefitinib blocks macrophage-promoted invasion and metastatic extravasation by reprogramming macrophages through inhibition of RIPK2.
Collapse
Affiliation(s)
- Caroline Maloney
- The Elmezzi Graduate School of Molecular Medicine, Northwell Health, Manhasset, New York
- Karches Center for Oncology, The Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York
| | - Michelle P Kallis
- The Elmezzi Graduate School of Molecular Medicine, Northwell Health, Manhasset, New York
- Karches Center for Oncology, The Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York
| | - Morris Edelman
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York
| | - Christopher Tzanavaris
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York
| | - Martin Lesser
- The Elmezzi Graduate School of Molecular Medicine, Northwell Health, Manhasset, New York
- The Feinstein Institutes for Medical Research and Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York
| | - Samuel Z Soffer
- The Elmezzi Graduate School of Molecular Medicine, Northwell Health, Manhasset, New York
- Karches Center for Oncology, The Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York
| | - Marc Symons
- The Elmezzi Graduate School of Molecular Medicine, Northwell Health, Manhasset, New York
- Karches Center for Oncology, The Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York
| | - Bettie M Steinberg
- The Elmezzi Graduate School of Molecular Medicine, Northwell Health, Manhasset, New York.
- Karches Center for Oncology, The Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York
| |
Collapse
|
27
|
Wolf-Dennen K, Gordon N, Kleinerman ES. Exosomal communication by metastatic osteosarcoma cells modulates alveolar macrophages to an M2 tumor-promoting phenotype and inhibits tumoricidal functions. Oncoimmunology 2020; 9:1747677. [PMID: 32313728 PMCID: PMC7153823 DOI: 10.1080/2162402x.2020.1747677] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 03/11/2020] [Accepted: 03/14/2020] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma metastasizes to the lung, and there is a link between the predominance of tumor-promoting immunosuppressive M2 macrophages in the metastases and poor patient survival. By contrast, M1 macrophage predominance correlates with longer survival. M2 macrophages can be induced by various stimuli in the tumor microenvironment, including exosomes, which are 40- to 150-nm vesicles that are involved in intercellular communication and contribute to tumor progression and immune evasion. Recognizing that tumor cells can influence the tumor microenvironment to make it more permissive and because of the link between M2 dominance and curtailed patient survival, we evaluated the effect of exosomes from non-metastatic K7 and Dunn osteosarcoma cells and the metastatic sublines K7M3 and DLM8 on macrophage phenotype and function. Incubating MHS mouse alveolar macrophages with K7M3 and DLM8 exosomes induced expression of IL10, TGFB2, and CCL22 mRNA (markers of M2 macrophages) and decreased phagocytosis, efferocytosis, and macrophage-mediated tumor cell killing. In contrast, exosomes from non-metastatic K7 or Dunn cells did not inhibit phagocytosis, efferocytosis, and macrophage-mediated cytotoxicity or induce increased expression of IL10, TGFB2 or CCL22 mRNA. In addition, metastatic osteosarcoma cell exosomes significantly increased the secretion of TGFB2, a key signaling pathway associated with tumor- mediated immune suppression. Finally, the inhibition of TGFB2 reversed the suppressive activity of alveolar macrophages exposed to metastatic osteosarcoma cell exosomes. Our data suggest that the exosomes from metastatic osteosarcoma cells can modulate cellular signaling of tumor-associated macrophages, thereby promoting the M2 phenotype and creating an immunosuppressive, tumor-promoting microenvironment through the production of TGFB2.
Collapse
Affiliation(s)
- Kerri Wolf-Dennen
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nancy Gordon
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugenie S Kleinerman
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
28
|
Ren L, Ruiz-Rodado V, Dowdy T, Huang S, Issaq SH, Beck J, Wang H, Tran Hoang C, Lita A, Larion M, LeBlanc AK. Glutaminase-1 (GLS1) inhibition limits metastatic progression in osteosarcoma. Cancer Metab 2020; 8:4. [PMID: 32158544 PMCID: PMC7057558 DOI: 10.1186/s40170-020-0209-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 01/08/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteosarcoma (OS) is a malignant bone tumor that often develops during the period of rapid growth associated with adolescence. Despite successful primary tumor control accompanied by adjuvant chemotherapy, death from pulmonary metastases occurs in approximately 30% of patients within 5 years. As overall survival in patients remains unchanged over the last 30 years, urgent needs for novel therapeutic strategies exist. Cancer metastasis is characterized by complex molecular events which result from alterations in gene and protein expression/function. Recent studies suggest that metabolic adaptations, or "metabolic reprogramming," may similarly contribute to cancer metastasis. The goal of this study was to specifically interrogate the metabolic vulnerabilities of highly metastatic OS cell lines in a series of in vitro and in vivo experiments, in order to identify a tractable metabolically targeted therapeutic strategy for patients. METHODS Nutrient deprivation and drug treatment experiments were performed in MG63.3, 143B, and K7M2 OS cell lines to identify the impact of glutaminase-1 (GLS1) inhibition and metformin treatment on cell proliferation. We functionally validated the impact of drug treatment with extracellular flux analysis, nuclear magnetic resonance (NMR) spectroscopy, and mass spectrometry. 13C-glucose and 13C-glutamine tracing was employed to identify specific contributions of these nutrients to the global metabolic profiles generated with GLS1 inhibition and metformin treatment in vivo. RESULTS Highly metastatic OS cell lines require glutamine for proliferation, and exposure to CB-839, in combination with metformin, induces both primary tumor growth inhibition and a distinct reduction in metastatic outgrowth in vivo. Further, combination-treated OS cells showed a reduction in cellular mitochondrial respiration, while NMR confirmed the pharmacodynamic effects of glutaminase inhibition in tumor tissues. We observed global decreases in glycolysis and tricarboxylic acid (TCA) cycle functionality, alongside an increase in fatty acid oxidation and pyrimidine catabolism. CONCLUSIONS This data suggests combination-treated cells cannot compensate for metformin-induced electron transport chain inhibition by upregulating glutaminolysis to generate TCA cycle intermediates required for cell proliferation, translating into significant reductions in tumor growth and metastatic progression. This therapeutic approach could be considered for future clinical development for OS patients presenting with or at high risk of developing metastasis.
Collapse
Affiliation(s)
- L. Ren
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - V. Ruiz-Rodado
- Metabolomics Section, NeuroOncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - T. Dowdy
- Metabolomics Section, NeuroOncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - S. Huang
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - S. H. Issaq
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - J. Beck
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - H. Wang
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - C. Tran Hoang
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - A. Lita
- Metabolomics Section, NeuroOncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - M. Larion
- Metabolomics Section, NeuroOncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - A. K. LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| |
Collapse
|
29
|
Charan M, Dravid P, Cam M, Setty B, Roberts RD, Houghton PJ, Cam H. Tumor secreted ANGPTL2 facilitates recruitment of neutrophils to the lung to promote lung pre-metastatic niche formation and targeting ANGPTL2 signaling affects metastatic disease. Oncotarget 2020; 11:510-522. [PMID: 32082485 PMCID: PMC7007290 DOI: 10.18632/oncotarget.27433] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/28/2019] [Indexed: 12/28/2022] Open
Abstract
The pre-metastatic niche (PMN) represents an abnormal microenvironment devoid of cancer cells, but favoring tumor growth. Little is known about the mechanisms that generate the PMN or their effects on host cells within metastasis-prone organs. Here, we investigated by using spontaneous metastatic models whether lung epithelial cells are essential for primary tumor induced neutrophil recruitment in lung and subsequently initiating PMN formation in osteosarcoma. We found that serum levels of ANGPTL2 in osteosarcoma patients are significantly higher compared to those in healthy controls and that ANGPTL2 secretion by tumor cells plays an essential role in osteosarcoma metastasis. We determined that tumor-derived ANGPTL2 stimulates lung epithelial cells, which is essential for primary tumor-induced neutrophil recruitment in lung and subsequent pre-metastatic niche formation. Lastly, we identified that a p63 isoform, ΔNp63, drives high level of ANGPTL2 secretion and pharmaceutical inhibition of ANGPTL2 signaling by a non–RGD-based integrin binding peptide (ATN-161) diminished metastatic load in lungs likely due to reduction of the lung pre-metastatic niche formation.
Collapse
Affiliation(s)
- Manish Charan
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Piyush Dravid
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Maren Cam
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Bhuvana Setty
- Department of Hematology & Oncology, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Ryan D Roberts
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Peter J Houghton
- Greehey Children's Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Hakan Cam
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
30
|
Smeester BA, Slipek NJ, Pomeroy EJ, Bomberger HE, Shamsan GA, Peterson JJ, Crosby MR, Draper GM, Becklin KL, Rahrmann EP, McCarthy JB, Odde DJ, Wood DK, Largaespada DA, Moriarity BS. SEMA4C is a novel target to limit osteosarcoma growth, progression, and metastasis. Oncogene 2019; 39:1049-1062. [PMID: 31582836 DOI: 10.1038/s41388-019-1041-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 09/17/2019] [Accepted: 09/23/2019] [Indexed: 02/08/2023]
Abstract
Semaphorins, specifically type IV, are important regulators of axonal guidance and have been increasingly implicated in poor prognoses in a number of different solid cancers. In conjunction with their cognate PLXNB family receptors, type IV members have been increasingly shown to mediate oncogenic functions necessary for tumor development and malignant spread. In this study, we investigated the role of semaphorin 4C (SEMA4C) in osteosarcoma growth, progression, and metastasis. We investigated the expression and localization of SEMA4C in primary osteosarcoma patient tissues and its tumorigenic functions in these malignancies. We demonstrate that overexpression of SEMA4C promotes properties of cellular transformation, while RNAi knockdown of SEMA4C promotes adhesion and reduces cellular proliferation, colony formation, migration, wound healing, tumor growth, and lung metastasis. These phenotypic changes were accompanied by reductions in activated AKT signaling, G1 cell cycle delay, and decreases in expression of mesenchymal marker genes SNAI1, SNAI2, and TWIST1. Lastly, monoclonal antibody blockade of SEMA4C in vitro mirrored that of the genetic studies. Together, our results indicate a multi-dimensional oncogenic role for SEMA4C in metastatic osteosarcoma and more importantly that SEMA4C has actionable clinical potential.
Collapse
Affiliation(s)
- Branden A Smeester
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.,Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Nicholas J Slipek
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.,Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Emily J Pomeroy
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.,Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Heather E Bomberger
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Ghaidan A Shamsan
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.,Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Joseph J Peterson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.,Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Margaret R Crosby
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.,Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Garrett M Draper
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.,Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Kelsie L Becklin
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.,Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Eric P Rahrmann
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - James B McCarthy
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - David J Odde
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.,Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - David K Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - David A Largaespada
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.,Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Branden S Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA. .,Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA. .,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
31
|
Combination Therapy with Disulfiram, Copper, and Doxorubicin for Osteosarcoma: In Vitro Support for a Novel Drug Repurposing Strategy. Sarcoma 2019; 2019:1320201. [PMID: 31379466 PMCID: PMC6657614 DOI: 10.1155/2019/1320201] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 05/19/2019] [Indexed: 12/13/2022] Open
Abstract
Although many cancer cells have significantly higher copper concentrations compared with normal cells and tissues, the role of copper in cancer biology and metastatic disease remains poorly understood. Here, we study the importance of copper in osteosarcoma, which frequently metastasizes to the lungs and is often chemoresistant. K12 and K7M2 are murine OS cells with differing metastatic phenotypes: K7M2 is highly metastatic, whereas K12 is much less so. Intracellular copper levels were determined using atomic absorption. Copper transporters were quantified by qPCR. Cytotoxicity of doxorubicin, disulfiram, and copper(II) chloride was assessed with a cell viability fluorescence stain. Additionally, K7M2 viable cell counts were determined by trypan blue exclusion staining after 72 hours of treatment. Copper levels were found to be significantly higher in K12 OS cells than in K7M2 cells. qPCR showed that K12 cells upregulate the copper influx pump CTR1 and downregulate the copper efflux pump ATP7A compared to K7M2 OS cells. Combination treatment of copper chloride (50 nM) with disulfiram (80 nM) was only cytotoxic to K12 cells. Triple treatment with doxorubicin, disulfiram, and copper displayed potent and durable cytotoxicity of highly metastatic K7M2 cells. We demonstrate here that murine OS cell lines differing in metastatic potential also vary in endogenous copper levels and regulation. Additionally, these differences in copper regulation may contribute to selective cytotoxicity of K12 cells by extremely low doses of copper-potentiated disulfiram. The combination of doxorubicin, disulfiram, and copper should be explored as a therapeutic strategy against OS metastases.
Collapse
|
32
|
Qin J, Wang R, Zhao C, Wen J, Dong H, Wang S, Li Y, Zhao Y, Li J, Yang Y, He X, Wang D. Notch signaling regulates osteosarcoma proliferation and migration through Erk phosphorylation. Tissue Cell 2019; 59:51-61. [PMID: 31383289 DOI: 10.1016/j.tice.2019.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/26/2019] [Accepted: 07/01/2019] [Indexed: 12/26/2022]
Abstract
We used a murine spontaneous osteosarcoma cell line with high metastatic potential, the K7M2 cell line to study the role of Notch signaling in the biological manifestations of osteosarcoma, to understand its underlying mechanism in the regulation of cell proliferation and migration, and to improve patient prognosis in cases of osteosarcoma through the discovery of novel therapeutic targets, First, Notch expression in K7M2 was determined by immunostaining, and the γ-secretase inhibitor N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) was used to inhibit proteolytic cleavage of the Notch intracellular domain (NICD), resulting in the inhibition of Notch activation. By using the Sulforhodamine B assay, colony-forming units assay, Brdu and Ki67 staining, and flow cytometry assays of apoptosis and cell cycle stage, DAPT was found to inhibit K7M2 proliferation in a dose-dependent manner. By using wound healing and transwell migration assays, DAPT was found to inhibit K7M2 migration in a dose-dependent manner as well. By using a combination of micro-Raman spectroscopy and K-means clustering analysis, we found that DAPT inhibit a variety of important cell metabolism-related components in most K7M2 cell structures. Then, DAPT was found to inhibit Notch1ICD expression in a concentration-dependent manner, and this expression was directly correlated with Phospho-Erk1/2 (p-Erk) by using Western blotting. To confirm this finding, we used the Notch signaling ligand Jagged1 to activate the Notch signaling pathway, which in turn up-regulated p-Erk, resulting in increased proliferation and migration of K7M2. Using the Erk pathway inhibitor U0126, we showed that p-Erk was downregulated and the proliferation and migration of K7M2 decreased along with it. Finally, we constructed a K7M2 mouse para-tibial tumor model and lung metastatic model. We found DAPT inhibits p-Erk in vivo, effectively controls tumor growth, reduces angiogenesis, reduces metastasis to the lungs, and improves overall survival. In summary, Notch signaling plays an oncogene role and promotes metastasis in osteosarcoma through p-Erk. DAPT effectively inhibits osteosarcoma proliferation and metastasis in vivo and in vitro by inhibiting Erk phosphorylation. Therefore, the inhibition of Notch activation resulted the down-regulation of phosphorylation of Erk pathway can be used as potential therapeutic targets in clinical treatment to improve osteosarcoma prognosis.
Collapse
Affiliation(s)
- Jie Qin
- The Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, PR China
| | - Rui Wang
- The Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, PR China
| | - Chenguang Zhao
- The Department of Rehabilitation, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, PR China
| | - Junxiang Wen
- The Department of Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Hui Dong
- The Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, PR China
| | - Shuang Wang
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi Province, PR China
| | - Yuhuan Li
- The Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, PR China
| | - Yonglin Zhao
- The Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, PR China
| | - Jianjun Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, PR China
| | - Yiting Yang
- The Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, PR China
| | - Xijing He
- The Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, PR China.
| | - Dong Wang
- The Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, PR China.
| |
Collapse
|
33
|
El-Naggar AM, Clarkson PW, Negri GL, Turgu B, Zhang F, Anglesio MS, Sorensen PH. HACE1 is a potential tumor suppressor in osteosarcoma. Cell Death Dis 2019; 10:21. [PMID: 30622235 PMCID: PMC6325116 DOI: 10.1038/s41419-018-1276-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 11/23/2018] [Accepted: 12/06/2018] [Indexed: 12/24/2022]
Abstract
Osteosarcoma is a malignant bone sarcoma characterized by extensive genomic disruption and a propensity for metastatic spread. Osteoid production suggests a close relationship with normal osteoblasts, and the latter are the presumptive cell of origin of this disease. The HACE1 gene, localized to human chromosome 6q21, encodes the HACE1 HECT E3 ligase, a tumor suppressor in diverse tumors that acts in part by targeting the activated form of RAC1 GTPase for proteasomal degradation. Disruption or loss of 6q21 is relatively common in osteosarcomas, and Hace1-/-/Tp53+/- mice frequently develop osteosarcomas, in contrast to Tp53+/- mice, which do not. This suggests an unexplored link between HACE1 loss and osteosarcoma. Here we compared HACE1 expression in normal osteoblasts and osteosarcoma cell lines in vitro by western blotting and quantitative RT-PCR, and in human osteosarcoma specimens by immunohistochemistry. Both HACE1 transcript and protein levels were reduced in osteosarcoma compared to osteoblasts in vitro. Reduced HACE1 expression in osteosarcoma tumors was observed in 76% of cases and associated with high-grade lesions. Further, clonally derived pairs of high and low metastatic osteosarcoma cell lines showed significant downregulation in the high compared to corresponding low metastatic cells. Ectopic expression of HACE1 markedly inhibited anchorage-independent growth and cell motility of HACE1 osteosarcoma cell lines, and was associated with reduced RAC1 activation and decreased reactive oxygen species (ROS). Finally, HACE1 overexpression blocked osteosarcoma xenograft growth and dramatically reduced pulmonary metastases. These findings point to a potential tumor suppressor function for HACE1 in osteosarcoma.
Collapse
Affiliation(s)
- Amal M El-Naggar
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Department of Pathology, Faculty of Medicine, Menoufia University, Menoufia Governorate, Egypt
| | - Paul W Clarkson
- Department of Orthopedics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gian Luca Negri
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Busra Turgu
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Fan Zhang
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael S Anglesio
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Poul H Sorensen
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada. .,Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.
| |
Collapse
|
34
|
Gross AC, Cam H, Phelps DA, Saraf AJ, Bid HK, Cam M, London CA, Winget SA, Arnold MA, Brandolini L, Mo X, Hinckley JM, Houghton PJ, Roberts RD. IL-6 and CXCL8 mediate osteosarcoma-lung interactions critical to metastasis. JCI Insight 2018; 3:99791. [PMID: 30135299 PMCID: PMC6141177 DOI: 10.1172/jci.insight.99791] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/05/2018] [Indexed: 01/01/2023] Open
Abstract
Osteosarcoma (OS), a malignant tumor of bone, kills through aggressive metastatic spread almost exclusively to the lung. Mechanisms driving this tropism for lung tissue remain unknown, though likely invoke specific interactions between tumor cells and other cells within the lung metastatic niche. Aberrant overexpression of ΔNp63 in OS cells directly drives production of IL-6 and CXCL8. All these factors were expressed at higher levels in OS lung metastases than in matched primary tumors from the same patients. Expression in cell lines correlated strongly with lung colonization efficiency in murine xenograft models. Lentivirus-mediated expression endowed poorly metastatic OS cells with increased metastatic capacity. Disruption of IL-6 and CXCL8 signaling using genetic or pharmaceutical inhibitors had minimal effects on tumor cell proliferation in vitro or in vivo, but combination treatment inhibited metastasis across multiple models of metastatic OS. Strong interactions occurred between OS cells and both primary bronchial epithelial cells and bronchial smooth muscle cells that drove feed-forward amplification of IL-6 and CXCL8 production. These results identify IL-6 and CXCL8 as primary mediators of OS lung tropism and suggest pleiotropic, redundant mechanisms by which they might effect metastasis. Combination therapy studies demonstrate proof of concept for targeting these tumor-lung interactions to affect metastatic disease.
Collapse
Affiliation(s)
- Amy C Gross
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Hakan Cam
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, USA.,The Ohio State University James Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Doris A Phelps
- Greehey Children's Cancer Research Institute, San Antonio, Texas, USA
| | - Amanda J Saraf
- The Ohio State University James Comprehensive Cancer Center, Columbus, Ohio, USA.,Division of Pediatric Hematology, Oncology, and BMT, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Hemant K Bid
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, USA.,The Ohio State University James Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Maren Cam
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Cheryl A London
- Department of Veterinary Clinical Sciences and Biosciences, The Ohio State University, Columbus, Ohio, USA.,Cummings School of Veterinary Medicine, Tufts University, Boston, Massachusetts, USA
| | - Sarah A Winget
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Michael A Arnold
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | | | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - John M Hinckley
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, USA.,Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, Ohio, USA
| | - Peter J Houghton
- Greehey Children's Cancer Research Institute, San Antonio, Texas, USA
| | - Ryan D Roberts
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, USA.,The Ohio State University James Comprehensive Cancer Center, Columbus, Ohio, USA.,Division of Pediatric Hematology, Oncology, and BMT, Nationwide Children's Hospital, Columbus, Ohio, USA
| |
Collapse
|
35
|
Gill KS, Fernandes P, Bird B, Soden DM, Forde PF. Combination of electroporation delivered metabolic modulators with low-dose chemotherapy in osteosarcoma. Oncotarget 2018; 9:31473-31489. [PMID: 30140384 PMCID: PMC6101145 DOI: 10.18632/oncotarget.25843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 07/08/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Osteosarcoma accounts for roughly 60% of all malignant bone tumors in children and young adults. The five-year survival rate for localized tumors after surgery and chemotherapy is approximately 70% whilst it drastically reduces to 15-30% in metastatic cases. Metabolic modulation is known to increase sensitivity of cancers to chemotherapy. A novel treatment strategy in Osteosarcoma is needed to battle this devastating malady. RESULTS Electroporation-delivered metabolic modulators were more effective in halting the cell cycle of Osteosarcoma cells and this negatively affects their ability to recover and proliferate, as shown in colony formation assays. Electroporation-delivered metabolic modulators increase the sensitivity of Osteosarcoma cells to chemotherapy and this combination reduces their survivability. CONCLUSION This novel treatment approach highlights the efficacy of electroporation in the delivery of metabolic modulators in Osteosarcoma cells, and increased sensitivity to chemotherapy allowing for a lower dose to be therapeutic. METHODS Metabolic modulations of two Osteosarcoma cell lines were performed with clinically available modulators delivered using electroporation, and its combination with low-dose Cisplatin. The effects of Dicholoroacetic acid, 2-Deoxy-D-glucose and Metformin on cell cycle and recovery of Osteosarcoma cells were assessed. Their sensitivity to chemotherapy was also assessed when treated in combination with electroporation-delivered metabolic modulators.
Collapse
Affiliation(s)
- Kheshwant S. Gill
- Cancer Research at UCC, Western Gateway Building, University College Cork, Cork, Ireland
| | - Philana Fernandes
- Cancer Research at UCC, Western Gateway Building, University College Cork, Cork, Ireland
| | - Brian Bird
- Cancer Research at UCC, Western Gateway Building, University College Cork, Cork, Ireland
- Bons Secours Hospital, Cork, Ireland
| | - Declan M. Soden
- Cancer Research at UCC, Western Gateway Building, University College Cork, Cork, Ireland
| | - Patrick F. Forde
- Cancer Research at UCC, Western Gateway Building, University College Cork, Cork, Ireland
| |
Collapse
|
36
|
Crasto JA, Fourman MS, Morales-Restrepo A, Mahjoub A, Mandell JB, Ramnath K, Tebbets JC, Watters RJ, Weiss KR. Disulfiram reduces metastatic osteosarcoma tumor burden in an immunocompetent Balb/c or-thotopic mouse model. Oncotarget 2018; 9:30163-30172. [PMID: 30046395 PMCID: PMC6059028 DOI: 10.18632/oncotarget.25733] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/14/2018] [Indexed: 12/16/2022] Open
Abstract
Introduction The overall survival rate of patients with osteosarcoma (OS) and pulmonary metastases has remained stagnant at 15–30% for several decades. Disulfiram (DSF) is an FDA-approved aldehyde dehydrogenase inhibitor that reduces the metastatic phenotype of OS cells in vitro. Here we evaluate its in vivo efficacy, as compared to doxorubicin chemotherapy, in a previously-validated orthotopic model of metastatic OS. Results All treatment groups displayed a significantly reduced quantitative OS metastatic burden compared with controls. The metastatic burden of Lo DSF-treated animals was equivalent to the DXR group. Ninety-five percent of control animals displayed evidence of metastatic disease, which was significantly greater than all treatment groups. Discussion Disulfiram treatment resulted in a reduced burden of OS metastatic disease compared with controls. This was statistically-equivalent to doxorubicin. No additive effect was observed between these two therapies. Materials and Methods One-hundred twenty immunocompetent Balb/c mice received proximal tibia paraphyseal injections of 5 × 105 K7M2 murine OS cells. Therapy began three weeks after injection: saline (control), low-dose disulfiram (Lo DSF), high-dose disulfiram (Hi DSF), doxorubicin (DXR), Lo DSF + DXR, and Hi DSF + DXR. Transfemoral amputations were performed at 4 weeks. Quantitative metastatic tumor burden was measured using near-infrared indocyanine green (ICG) angiography.
Collapse
Affiliation(s)
- Jared Anthony Crasto
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mitchell Stephen Fourman
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alejandro Morales-Restrepo
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adel Mahjoub
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan Brendan Mandell
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kavita Ramnath
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Carnegie Mellon University, Pittsburgh, PA, USA
| | - Jessica C Tebbets
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rebecca J Watters
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, PA, USA
| | - Kurt Richard Weiss
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Departments of Anatomic Pathology and General Surgical Oncology, University of Pittsburgh, PA, USA
| |
Collapse
|
37
|
Yohe ME, Gryder BE, Shern JF, Song YK, Chou HC, Sindiri S, Mendoza A, Patidar R, Zhang X, Guha R, Butcher D, Isanogle KA, Robinson CM, Luo X, Chen JQ, Walton A, Awasthi P, Edmondson EF, Difilippantonio S, Wei JS, Zhao K, Ferrer M, Thomas CJ, Khan J. MEK inhibition induces MYOG and remodels super-enhancers in RAS-driven rhabdomyosarcoma. Sci Transl Med 2018; 10:eaan4470. [PMID: 29973406 PMCID: PMC8054766 DOI: 10.1126/scitranslmed.aan4470] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 06/06/2018] [Indexed: 12/22/2022]
Abstract
The RAS isoforms are frequently mutated in many types of human cancers, including PAX3/PAX7 fusion-negative rhabdomyosarcoma. Pediatric RMS arises from skeletal muscle progenitor cells that have failed to differentiate normally. The role of mutant RAS in this differentiation blockade is incompletely understood. We demonstrate that oncogenic RAS, acting through the RAF-MEK [mitogen-activated protein kinase (MAPK) kinase]-ERK (extracellular signal-regulated kinase) MAPK effector pathway, inhibits myogenic differentiation in rhabdomyosarcoma by repressing the expression of the prodifferentiation myogenic transcription factor, MYOG. This repression is mediated by ERK2-dependent promoter-proximal stalling of RNA polymerase II at the MYOG locus. Small-molecule screening with a library of mechanistically defined inhibitors showed that RAS-driven RMS is vulnerable to MEK inhibition. MEK inhibition with trametinib leads to the loss of ERK2 at the MYOG promoter and releases the transcriptional stalling of MYOG expression. MYOG subsequently opens chromatin and establishes super-enhancers at genes required for late myogenic differentiation. Furthermore, trametinib, in combination with an inhibitor of IGF1R, potently decreases rhabdomyosarcoma cell viability and slows tumor growth in xenograft models. Therefore, this combination represents a potential therapeutic for RAS-mutated rhabdomyosarcoma.
Collapse
Affiliation(s)
- Marielle E Yohe
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA.
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Berkley E Gryder
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Jack F Shern
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Young K Song
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Hsien-Chao Chou
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Sivasish Sindiri
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Arnulfo Mendoza
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Rajesh Patidar
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Rajarashi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Donna Butcher
- Pathology/Histotechnology Laboratory, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21702, USA
| | - Kristine A Isanogle
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Christina M Robinson
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Xiaoling Luo
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jin-Qiu Chen
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Ashley Walton
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Parirokh Awasthi
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Elijah F Edmondson
- Pathology/Histotechnology Laboratory, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21702, USA
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Jun S Wei
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Keji Zhao
- Systems Biology Center, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Javed Khan
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
38
|
Maloney C, Edelman MC, Kallis MP, Soffer SZ, Symons M, Steinberg BM. Intratibial Injection Causes Direct Pulmonary Seeding of Osteosarcoma Cells and Is Not a Spontaneous Model of Metastasis: A Mouse Osteosarcoma Model. Clin Orthop Relat Res 2018; 476:1514-1522. [PMID: 29601385 PMCID: PMC6437576 DOI: 10.1007/s11999.0000000000000291] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/08/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND Although metastasis is the major cause of mortality in patients with osteosarcoma, little is known about how micrometastases progress to gross metastatic disease. Clinically relevant animal models are necessary to facilitate development of new therapies to target indolent pulmonary metastases. Intratibial injection of human and murine osteosarcoma cell lines have been described as orthotopic models that develop spontaneous pulmonary metastasis over time. However, there is variability in reported injection techniques and metastatic efficiency. QUESTIONS/PURPOSES We aimed to characterize a widely used murine model of metastatic osteosarcoma, determine whether it is appropriate to study spontaneous pulmonary metastasis by establishing a reliable volume for intratibial injection, determine the incidence of primary tumor and metastatic formation, determine the kinetics of pulmonary metastatic seeding and outgrowth, and the contribution of the primary tumor to subsequent development of metastasis. METHODS The metastatic mouse osteosarcoma cell line K7M2 was injected into the tibia of mice. The maximum volume that could be injected without leakage was determined using Evan's blue dye (n = 8 mice). Primary tumor formation and metastatic efficiency were determined by measuring the incidence of primary tumor and metastatic formation 4 weeks after intratibial injection (n = 30). The kinetics of metastatic development were determined by performing serial euthanasia at 1, 2, 3, and 4 weeks after injection (n = 24; five to six mice per group). Number of metastatic foci/histologic lung section and metastatic burden/lung section (average surface area of metastatic lesions divided by the total surface area of the lung) was calculated in a blinded fashion. To test the contribution of the primary tumor to subsequent metastases, amputations were performed 30 minutes, 4 hours, or 24 hours after injection (n = 21; five to six mice per group). Mice were euthanized after 4 weeks and metastatic burden calculated as described previously, comparing mice that had undergone amputation with control, nonamputated mice. Differences between groups were calculated using Kruskal-Wallis and one-way analysis of variance. RESULTS The maximum volume of cell suspension that could be injected without leakage was 10 μL. Intratibial injection of tumor cells led to intramedullary tumor formation in 93% of mice by 4 weeks and resulted in detectable pulmonary metastases in 100% of these mice as early as 1 week post-injection. Metastatic burden increased over time (0.88% ± 0.58, week 1; 6.6% ± 5.3, week 2; 16.1% ± 12.5, week 3; and 40.3% ± 14.83, week 4) with a mean difference from week 1 to week 4 of -39.38 (p < 0.001; 95% confidence interval [CI], -57.39 to -21.37), showing pulmonary metastatic growth over time. In contrast, the mean number of metastatic foci did not increase from week 1 to week 4 (36.4 ± 33.6 versus 49.3 ± 26.3, p = 0.18). Amputation of the injected limb at 30 minutes, 4 hours, and 24 hours after injection did not affect pulmonary metastatic burden at 4 weeks, with amputation as early as 30 minutes post-injection resulting in a metastatic burden equivalent to tumor-bearing controls (48.9% ± 6.1% versus 40.9% ± 15.3%, mean difference 7.96, p = 0.819; 95% CI, -33.9 to 18.0). CONCLUSIONS There is immediate seeding of the metastatic site after intratibial injection of the K7M2 osteosarcoma cell line, independent of a primary tumor. This is therefore not a model of spontaneous metastasis. CLINICAL RELEVANCE This model should not be used to study the early components of the metastatic cascade, but rather used as an experimental model of metastasis. Improved understanding of this commonly used model will allow for proper interpretation of existing data and inform the design of future studies exploring the biology of metastasis in osteosarcoma.
Collapse
Affiliation(s)
- Caroline Maloney
- C. Maloney, M. P. Kallis, M. Symons, B. M. Steinberg, The Elmezzi Graduate School of Molecular Medicine, Northwell Health, Manhasset, NY, USA C. Maloney, M. P. Kallis, S. Z. Soffer, M. Symons, B. M. Steinberg, Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA C. Maloney, M. P. Kallis, S. Z. Soffer, Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA M. C. Edelman, Department of Pathology and Laboratory Medicine, Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY, USA
| | | | | | | | | | | |
Collapse
|
39
|
Micrometastatic Drug Screening Platform Shows Heterogeneous Response to MAP Chemotherapy in Osteosarcoma Cell Lines. Clin Orthop Relat Res 2018; 476:1400-1411. [PMID: 29481344 PMCID: PMC6437591 DOI: 10.1007/s11999.0000000000000059] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Approximately 80% of patients with osteosarcoma harbor subclinical pulmonary micrometastases at diagnosis. Conventional chemotherapy includes methotrexate, doxorubicin, and cisplatin (MAP); however, this regimen and thus overall survival (60%-70%) have remained largely unchanged for 30 years. It therefore is necessary to identify novel therapeutics targeting the metastatic progression of osteosarcoma. QUESTIONS/PURPOSES This laboratory study explored application of osteosarcoma spheroids (sarcospheres) for drug screening with the following purposes: (1) to characterize sarcosphere size; (2) to establish accurate measurement of sarcosphere growth; (3) to confirm sarcosphere uniformity; and (4) to apply the platform to evaluate MAP chemotherapy. METHODS Sarcospheres were first characterized to establish accurate measurement of sarcosphere growth and uniform production. The refined platform then was applied to evaluate MAP chemotherapy to validate its use in drug screening. Sarcospheres were generated from highly metastatic human cell lines (143B, MG-63.3, and LM7) by centrifugation to form three-dimensional aggregates modeling micrometastases. Sarcospheres were matured for 24 hours and then incubated with or without drug from Days 0 to 2. Size was assessed by diameter and volume using brightfield microscopy. Growth was measured by volume and resazurin reduction in viable cells. Sarcosphere uniformity was assessed by diameter and resazurin reduction at Day 0 and the Z' factor, a measure of assay suitability for high-throughput screening, was calculated at Day 2. Sarcospheres were treated with individual MAP agents (0 to 1000 μmol/L) to determine concentrations at which 50% of growth from Days 0 to 2 was inhibited (GIC50). Cell lines resistant to MAP in sarcospheres were treated in monolayer for comparison. RESULTS Sarcosphere diameter and growth from Days 0 to 2 were quantitatively dependent on the number of cells seeded and the cell line used. Accurate measurement of growth occurred after resazurin incubation for 6 hours, without EDTA-mediated permeabilization, and was correlated with the number of cells seeded and sarcosphere volume for 143B (Spearman's r: 0.98; p < 0.001), MG-63.3 (0.99; p < 0.001), and LM7 (0.98; p < 0.001). Sarcospheres met established criteria for screening applications as mean Z' factors were greater than 0.5 for all cell lines. Response to MAP therapy was cell line-dependent, because MG-63.3 and LM7 sarcospheres exhibited greater than 2000-fold resistance to methotrexate (GIC50 = 88 ± 36 μmol/L and 174 ± 16 μmol/L, respectively) compared with the 143B cell line (GIC50 = 0.04 ± 0.01 μmol/L; p < 0.001 for MG-63.3 and LM7). MG-63.3 monolayers were more sensitive to methotrexate (GIC50 = 0.01 ± 0.01 μmol/L; p < 0.001) than MG-63.3 sarcospheres, whereas LM7 monolayers remained chemoresistent (GIC50 not reached). CONCLUSIONS This study developed and validated a drug screening platform for progression of osteosarcoma micrometastases. It also highlights heterogeneity among osteosarcoma cell lines. These findings appear to reflect known patient-to-patient heterogeneity and underscore the importance of evaluating multiple tumor models when testing drugs for the treatment of osteosarcoma. CLINICAL RELEVANCE The described approach is a promising starting point for drug screening in osteosarcoma because it is tailored to evaluate micrometastatic disease. A reliable and rapid method to identify novel therapeutics is critical to improve stagnant outcomes for patients with osteosarcoma.
Collapse
|
40
|
Wagner F, Holzapfel BM, McGovern JA, Shafiee A, Baldwin JG, Martine LC, Lahr CA, Wunner FM, Friis T, Bas O, Boxberg M, Prodinger PM, Shokoohmand A, Moi D, Mazzieri R, Loessner D, Hutmacher DW. Humanization of bone and bone marrow in an orthotopic site reveals new potential therapeutic targets in osteosarcoma. Biomaterials 2018; 171:230-246. [PMID: 29705656 DOI: 10.1016/j.biomaterials.2018.04.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/11/2018] [Accepted: 04/14/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Existing preclinical murine models often fail to predict effects of anti-cancer drugs. In order to minimize interspecies-differences between murine hosts and human bone tumors of in vivo xenograft platforms, we tissue-engineered a novel orthotopic humanized bone model. METHODS Orthotopic humanized tissue engineered bone constructs (ohTEBC) were fabricated by 3D printing of medical-grade polycaprolactone scaffolds, which were seeded with human osteoblasts and embedded within polyethylene glycol-based hydrogels containing human umbilical vein endothelial cells (HUVECs). Constructs were then implanted at the femur of NOD-scid and NSG mice. NSG mice were then bone marrow transplanted with human CD34 + cells. Human osteosarcoma (OS) growth was induced within the ohTEBCs by direct injection of Luc-SAOS-2 cells. Tissues were harvested for bone matrix and marrow morphology analysis as well as tumor biology investigations. Tumor marker expression was analyzed in the humanized OS and correlated with the expression in 68 OS patients utilizing tissue micro arrays (TMA). RESULTS After harvesting the femurs micro computed tomography and immunohistochemical staining showed an organ, which had all features of human bone. Around the original mouse femur new bone trabeculae have formed surrounded by a bone cortex. Staining for human specific (hs) collagen type-I (hs Col-I) showed human extracellular bone matrix production. The presence of nuclei staining positive for human nuclear mitotic apparatus protein 1 (hs NuMa) proved the osteocytes residing within the bone matrix were of human origin. Flow cytometry verified the presence of human hematopoietic cells. After injection of Luc-SAOS-2 cells a primary tumor and lung metastasis developed. After euthanization histological analysis showed pathognomic features of osteoblastic OS. Furthermore, the tumor utilized the previously implanted HUVECS for angiogenesis. Tumor marker expression was similar to human patients. Moreover, the recently discovered musculoskeletal gene C12orf29 was expressed in the most common subtypes of OS patient samples. CONCLUSION OhTEBCs represent a suitable orthotopic microenvironment for humanized OS growth and offers a new translational direction, as the femur is the most common location of OS. The newly developed and validated preclinical model allows controlled and predictive marker studies of primary bone tumors and other bone malignancies.
Collapse
Affiliation(s)
- Ferdinand Wagner
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia; Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Lindwurmstraße 4, 80337 Munich, Germany; Department of Orthopedics for the University of Regensburg, Asklepios Klinikum Bad Abbach, Kaiser-Karl V.-Allee 3, 93077 Bad Abbach, Germany
| | - Boris M Holzapfel
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia; Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstr. 11, 97074 Wuerzburg, Germany
| | - Jacqui A McGovern
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Abbas Shafiee
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Jeremy G Baldwin
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Laure C Martine
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Christoph A Lahr
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Felix M Wunner
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Thor Friis
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Onur Bas
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Melanie Boxberg
- Institute of Pathology, Klinikum Rechts der Isar, Technical University Munich, Trogerstr. 18, 81675 Munich, Germany
| | - Peter M Prodinger
- Department of Orthopedic Surgery, Klinikum Rechts der Isar, Technical University Munich, Ismaningerstr. 22, 81675 Munich, Germany
| | - Ali Shokoohmand
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Davide Moi
- The University of Queensland, Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Roberta Mazzieri
- The University of Queensland, Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Daniela Loessner
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia; Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Dietmar W Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia; George W Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive Northwest, Atlanta, GA 30332, USA; Institute for Advanced Study, Technical University Munich, Lichtenbergstraße 2a, 85748 Garching, Munich, Germany.
| |
Collapse
|
41
|
Ren L, Hong ES, Mendoza A, Issaq S, Tran Hoang C, Lizardo M, LeBlanc A, Khanna C. Metabolomics uncovers a link between inositol metabolism and osteosarcoma metastasis. Oncotarget 2018; 8:38541-38553. [PMID: 28404949 PMCID: PMC5503552 DOI: 10.18632/oncotarget.15872] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 01/27/2017] [Indexed: 11/25/2022] Open
Abstract
Cancer development and progression are characterized by complex molecular events. The acquisition of these events is primarily believed to result from alterations in gene and protein expression/function. Recent studies have also suggested the role of metabolic alterations, or "metabolic reprogramming," that may similarly contribute to these events. Indeed, our previous investigations in osteosarcoma (OS) identified metabolic changes uniquely linked to metastasis. Based on those findings, here we sought to build a more detailed understanding of the specific alterations in metabolites or metabolic pathways that may be responsible for the observed metastasis-associated metabolic alterations, suggested by gene expression data. This was pursued using a combination of high-throughput liquid- and gas-chromatography-based mass spectrometry (LC/MS and GC/MS) for a global metabolic profiling/subtraction of four pairs of high/low metastatic OS cell lines. By comparing the identity and level of the metabolites between high/low metastatic cells, several metabolic pathways were identified to be differentially activated, such as arginine, glutathione, inositol and fatty acid metabolic pathways. To further interrogate these results, we investigated the effects of inositol pathway dysregulation, through the exposure of metastatic OS cells to IP6 (inositol hexaphosphate). Although IP6 exposures had modest to minimal effects on cell proliferation, we observed reduced cellular glycolysis, down-regulation of PI3K/Akt signaling and suppression of OS metastatic progression. Collectively these data supported further investigation of metabolic sensitivities as anti-metastatic strategies in a clinical setting as well as investigation of altered metabolomics associated with metastatic progression.
Collapse
Affiliation(s)
- Ling Ren
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Ellen S Hong
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Arnulfo Mendoza
- Molecular Oncology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Sameer Issaq
- Molecular Oncology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Christine Tran Hoang
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Michael Lizardo
- Molecular Oncology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Amy LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Chand Khanna
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.,Dr. Khanna is currently with Ethos Veterinary Health, Woburn MA and Ethos Discovery, Washington DC, USA
| |
Collapse
|
42
|
Grisez BT, Ray JJ, Bostian PA, Markel JE, Lindsey BA. Highly metastatic K7M2 cell line: A novel murine model capable of in vivo imaging via luciferase vector transfection. J Orthop Res 2018; 36:10.1002/jor.23868. [PMID: 29427436 PMCID: PMC6086764 DOI: 10.1002/jor.23868] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 01/24/2018] [Indexed: 02/04/2023]
Abstract
Osteosarcoma is rare and little improvement in survival rates has occurred in the last 25 years despite modern chemotherapeutic treatment. Bioluminescent cell lines for the modeling of osteosarcoma have shown success in tracking metastases in vivo, but commonly use adenoviral vectors to transfect the native cell line with bioluminescent reporters. The purpose of this study was to develop an orthotopic model for metastatic osteosarcoma capable of in vivo monitoring of metastatic and primary tumor burden in an immunocompetent mouse and compare that model to its wild type pathogenesis. K7M2 cells were transfected using a plasmid vector and were stable after 12 weeks. Thirty-four female BALB/c mice aged 4-5 weeks underwent orthotopic implantation of either wild type (n = 12) or transfected (n = 22) K7M2 cells in the proximal tibia. Mice were monitored for tumor growth and weekly In Vivo Imaging System (IVIS) imaging was performed to monitor for pulmonary metastasis. Although tumors developed sooner in the wild type group, no significant differences were seen compared to Transfected Group 1 in rate of inoculation, growth rates after first detection, metastatic rate, and time between inoculation and death. This study establishes a new murine model for metastatic osteosarcoma using the K7M2-wt cell line transfected with a non-viral plasmid luciferase vector. The benefits of this preclinical model include an intact immune system and orthotopically driven metastatic disease; this model appears comparable to its wild type counterpart. In the future, the model may be used to examine promising immunomodulatory therapies using bioluminescence in vivo. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Brian T Grisez
- Department of Orthopaedics, West Virginia University, PO Box 9196, Morgantown, West Virginia 26506-9196
| | - Justin J Ray
- Department of Orthopaedics, West Virginia University, PO Box 9196, Morgantown, West Virginia 26506-9196
| | - Phillip A Bostian
- Department of Orthopaedics, West Virginia University, PO Box 9196, Morgantown, West Virginia 26506-9196
| | - Justin E Markel
- Department of Orthopaedics, West Virginia University, PO Box 9196, Morgantown, West Virginia 26506-9196
| | - Brock A Lindsey
- Department of Orthopaedics, West Virginia University, PO Box 9196, Morgantown, West Virginia 26506-9196
| |
Collapse
|
43
|
Morrow JJ, Bayles I, Funnell APW, Miller TE, Saiakhova A, Lizardo MM, Bartels CF, Kapteijn MY, Hung S, Mendoza A, Dhillon G, Chee DR, Myers JT, Allen F, Gambarotti M, Righi A, DiFeo A, Rubin BP, Huang AY, Meltzer PS, Helman LJ, Picci P, Versteeg H, Stamatoyannopolus J, Khanna C, Scacheri PC. Positively selected enhancer elements endow osteosarcoma cells with metastatic competence. Nat Med 2018; 24:176-185. [PMID: 29334376 PMCID: PMC5803371 DOI: 10.1038/nm.4475] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 12/18/2017] [Indexed: 12/13/2022]
Abstract
Metastasis results from a complex set of traits acquired by tumor cells, distinct from those necessary for tumorigenesis. Here, we investigate the contribution of enhancer elements to the metastatic phenotype of osteosarcoma. Through epigenomic profiling, we identify substantial differences in enhancer activity between primary and metastatic human tumors and between near isogenic pairs of highly lung metastatic and nonmetastatic osteosarcoma cell lines. We term these regions metastatic variant enhancer loci (Met-VELs). Met-VELs drive coordinated waves of gene expression during metastatic colonization of the lung. Met-VELs cluster nonrandomly in the genome, indicating that activity of these enhancers and expression of their associated gene targets are positively selected. As evidence of this causal association, osteosarcoma lung metastasis is inhibited by global interruptions of Met-VEL-associated gene expression via pharmacologic BET inhibition, by knockdown of AP-1 transcription factors that occupy Met-VELs, and by knockdown or functional inhibition of individual genes activated by Met-VELs, such as that encoding coagulation factor III/tissue factor (F3). We further show that genetic deletion of a single Met-VEL at the F3 locus blocks metastatic cell outgrowth in the lung. These findings indicate that Met-VELs and the genes they regulate play a functional role in metastasis and may be suitable targets for antimetastatic therapies.
Collapse
Affiliation(s)
- James J. Morrow
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ian Bayles
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Tyler E. Miller
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alina Saiakhova
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Michael M. Lizardo
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, 20892 USA
| | - Cynthia F. Bartels
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Maaike Y. Kapteijn
- Thrombosis and Hemostasis Division, Department of Internal Medicine, LUMC, Leiden, Netherlands
| | - Stevephen Hung
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Arnulfo Mendoza
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, 20892 USA
| | - Gursimran Dhillon
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Daniel R. Chee
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA
| | - Jay T. Myers
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Frederick Allen
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Marco Gambarotti
- Research Laboratory, Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136, Bologna, Italy
| | - Alberto Righi
- Research Laboratory, Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136, Bologna, Italy
| | - Analisa DiFeo
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Brian P. Rubin
- Departments of Anatomic Pathology and Molecular Genetics, Cleveland Clinic, Lerner Research Institute and Taussig Cancer Center, Cleveland, OH 44195, USA
| | - Alex Y. Huang
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Paul S. Meltzer
- Genetics Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, 20892 USA
| | - Lee J. Helman
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, 20892 USA
| | - Piero Picci
- Research Laboratory, Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136, Bologna, Italy
| | - Henri Versteeg
- Thrombosis and Hemostasis Division, Department of Internal Medicine, LUMC, Leiden, Netherlands
| | | | - Chand Khanna
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, 20892 USA
| | - Peter C. Scacheri
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
- Research Laboratory, Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136, Bologna, Italy
| |
Collapse
|
44
|
Alizadeh E, Lyons SM, Castle JM, Prasad A. Measuring systematic changes in invasive cancer cell shape using Zernike moments. Integr Biol (Camb) 2017; 8:1183-1193. [PMID: 27735002 DOI: 10.1039/c6ib00100a] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
We study the shape characteristics of osteosarcoma cancer cell lines on surfaces of differing hydrophobicity using Zernike moments to represent cell shape. We compare the shape characteristics of four invasive cell lines with a corresponding less-invasive parental line on three substrates. Cell shapes of each pair of cell lines are quite close and display overlapping characteristics. To quantitatively study shape changes in high-dimensional parameter space we project down to principal component space and define a vector that summarizes average shape differences. Using this vector we find that three of the four pairs of cell lines show similar changes in shape, while the fourth pair shows a very different pattern of changes. We find that shape differences are sufficient to enable a neural network to classify cells accurately as belonging to the highly invasive or the less invasive phenotype. The patterns of shape changes were also reproducible for repetitions of the experiment. We also find that shape changes on different substrates show similarities between the eight cells studied, but the differences were typically not enough to permit classification. Our paper strongly suggests that shape may provide a means to read out the phenotypic state of some cell types, and shape analysis can be usefully performed using a Zernike moment representation.
Collapse
Affiliation(s)
- Elaheh Alizadeh
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA.
| | - Samanthe Merrick Lyons
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Jordan Marie Castle
- Department of Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Ashok Prasad
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA. and School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
45
|
Gianferante DM, Mirabello L, Savage SA. Germline and somatic genetics of osteosarcoma - connecting aetiology, biology and therapy. Nat Rev Endocrinol 2017; 13:480-491. [PMID: 28338660 DOI: 10.1038/nrendo.2017.16] [Citation(s) in RCA: 315] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clinical outcomes and treatment modalities for osteosarcoma, the most common primary cancer of bone, have changed very little over the past 30 years. The peak incidence of osteosarcoma occurs during the adolescent growth spurt, which suggests that bone growth and pubertal hormones are important in the aetiology of the disease. Tall stature, high birth weight and certain inherited cancer predisposition syndromes are well-described risk factors for osteosarcoma. Common genetic variants are also associated with osteosarcoma. The somatic genome of osteosarcoma is highly aneuploid, exhibits extensive intratumoural heterogeneity and has a higher mutation rate than most other paediatric cancers. Complex pathways related to bone growth and development and tumorigenesis are also important in osteosarcoma biology. In this Review, we discuss the contributions of germline and somatic genetics, tumour biology and animal models in improving our understanding of osteosarcoma aetiology, and their potential to identify novel therapeutic targets and thus improve the lives of patients with osteosarcoma.
Collapse
Affiliation(s)
- D Matthew Gianferante
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, Maryland 20892, USA
| | - Lisa Mirabello
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, Maryland 20892, USA
| | - Sharon A Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, Maryland 20892, USA
| |
Collapse
|
46
|
Robl B, Botter SM, Boro A, Meier D, Neri D, Fuchs B. Evaluation of F8-TNF-α in Models of Early and Progressive Metastatic Osteosarcoma. Transl Oncol 2017; 10:419-430. [PMID: 28448958 PMCID: PMC5406547 DOI: 10.1016/j.tranon.2017.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/16/2017] [Accepted: 02/16/2017] [Indexed: 12/17/2022] Open
Abstract
The targeted delivery of tumor necrosis factor-α (TNF-α) with antibodies specific to splice isoforms of fibronectin [e.g., F8-TNF, specific to the extra-domain A (EDA) domain of fibronectin] has already shown efficacy against experimental sarcomas but has not yet been investigated in orthotopic sarcomas. Here, we investigated F8-TNF in a syngeneic K7 M2-derived orthotopic model of osteosarcoma as a treatment against pulmonary metastases, the most frequent cause of osteosarcoma-related death. Immunofluorescence on human osteosarcoma tissue confirmed the presence of EDA in primary tumors (PTs) as well as metastases. In mice, the efficacy of F8-TNF against PTs and early pulmonary metastases was evaluated. Intratibial PT growth was not affected by F8-TNF, yet early micrometastases were reduced possibly due to an F8-TNF-dependent attraction of pulmonary CD4+, CD8+, and natural killer cells. Furthermore, immunofluorescence revealed stronger expression of EDA in early pulmonary metastases compared with PT tissue. To study progressing pulmonary metastases, a hind limb amputation model was established, and the efficacy of F8-TNF, alone or combined with doxorubicin, was investigated. Despite the presence of EDA in metastases, no inhibition of progressive metastatic growth was detected. No significant differences in numbers of CD4+ or CD8+ cells or F4/80+ and Ly6G+ myeloid-derived cells were observed, although a strong association between metastatic growth and presence of pulmonary Ly6G+ myeloid-derived cells was detected. In summary, these findings demonstrate the potential of F8-TNF in activating the immune system and reducing early metastatic growth yet suggest a lack of efficacy of F8-TNF alone or combined with doxorubicin against progressing osteosarcoma metastases.
Collapse
Affiliation(s)
- Bernhard Robl
- Laboratory for Orthopedic Research, Department of Orthopedics, Balgrist University Hospital, Zurich, Switzerland.
| | - Sander Martijn Botter
- Laboratory for Orthopedic Research, Department of Orthopedics, Balgrist University Hospital, Zurich, Switzerland.
| | - Aleksandar Boro
- Laboratory for Orthopedic Research, Department of Orthopedics, Balgrist University Hospital, Zurich, Switzerland.
| | - Daniela Meier
- Laboratory for Orthopedic Research, Department of Orthopedics, Balgrist University Hospital, Zurich, Switzerland.
| | - Dario Neri
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland.
| | - Bruno Fuchs
- Laboratory for Orthopedic Research, Department of Orthopedics, Balgrist University Hospital, Zurich, Switzerland.
| |
Collapse
|
47
|
Botham R, Roth HS, Book AP, Roady PJ, Fan TM, Hergenrother PJ. Small-Molecule Procaspase-3 Activation Sensitizes Cancer to Treatment with Diverse Chemotherapeutics. ACS CENTRAL SCIENCE 2016; 2:545-59. [PMID: 27610416 PMCID: PMC4999974 DOI: 10.1021/acscentsci.6b00165] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Indexed: 05/08/2023]
Abstract
Conventional chemotherapeutics remain essential treatments for most cancers, but their combination with other anticancer drugs (including targeted therapeutics) is often complicated by unpredictable synergies and multiplicative toxicities. As cytotoxic anticancer chemotherapeutics generally function through induction of apoptosis, we hypothesized that a molecularly targeted small molecule capable of facilitating a central and defining step in the apoptotic cascade, the activation of procaspase-3 to caspase-3, would broadly and predictably enhance activity of cytotoxic drugs. Here we show that procaspase-activating compound 1 (PAC-1) enhances cancer cell death induced by 15 different FDA-approved chemotherapeutics, across many cancer types and chemotherapeutic targets. In particular, the promising combination of PAC-1 and doxorubicin induces a synergistic reduction in tumor burden and enhances survival in murine tumor models of osteosarcoma and lymphoma. This PAC-1/doxorubicin combination was evaluated in 10 pet dogs with naturally occurring metastatic osteosarcoma or lymphoma, eliciting a biologic response in 3 of 6 osteosarcoma patients and 4 of 4 lymphoma patients. Importantly, in both mice and dogs, coadministration of PAC-1 with doxorubicin resulted in no additional toxicity. On the basis of the mode of action of PAC-1 and the high expression of procaspase-3 in many cancers, these results suggest the combination of PAC-1 with cytotoxic anticancer drugs as a potent and general strategy to enhance therapeutic response.
Collapse
Affiliation(s)
- Rachel
C. Botham
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Howard S. Roth
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Alison P. Book
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Patrick J. Roady
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Timothy M. Fan
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Paul J. Hergenrother
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
- E-mail:
| |
Collapse
|
48
|
Ren L, Mendoza A, Zhu J, Briggs JW, Halsey C, Hong ES, Burkett SS, Morrow J, Lizardo MM, Osborne T, Li SQ, Luu HH, Meltzer P, Khanna C. Characterization of the metastatic phenotype of a panel of established osteosarcoma cells. Oncotarget 2016; 6:29469-81. [PMID: 26320182 PMCID: PMC4745740 DOI: 10.18632/oncotarget.5177] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 09/25/2015] [Indexed: 11/25/2022] Open
Abstract
Osteosarcoma (OS) is the most common bone tumor in pediatric patients. Metastasis is a major cause of mortality and morbidity. The rarity of this disease coupled with the challenges of drug development for metastatic cancers have slowed the delivery of improvements in long-term outcomes for these patients. In this study, we collected 18 OS cell lines, confirmed their expression of bone markers and complex karyotypes, and characterized their in vivo tumorgenicity and metastatic potential. Since prior reports included conflicting descriptions of the metastatic and in vivo phenotypes of these models, there was a need for a comparative assessment of metastatic phenotypes using identical procedures in the hands of a single investigative group. We expect that this single characterization will accelerate the study of this metastatic cancer. Using these models we evaluated the expression of six previously reported metastasis-related OS genes. Ezrin was the only gene consistently differentially expressed in all the pairs of high/low metatstatic OS cells. We then used a subtractive gene expression approach of the high and low human metastatic cells to identify novel genes that may be involved in OS metastasis. PHLDA1 (pleckstrin homology-like domain, family A) was identified as one of the genes more highly expressed in the high metastatic compared to low metastatic cells. Knocking down PHLDA1 with siRNA or shRNA resulted in down regulation of the activities of MAPKs (ERK1/2), c-Jun N-terminal kinases (JNK), and p38 mitogen-activated protein kinases (MAPKs). Reducing the expression of PHLDA1 also delayed OS metastasis progression in mouse xenograft models.
Collapse
Affiliation(s)
- Ling Ren
- Molecular Oncology Section - Metastasis Biology Group, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Arnulfo Mendoza
- Molecular Oncology Section - Metastasis Biology Group, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Jack Zhu
- Genetic Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Joseph W Briggs
- Molecular Oncology Section - Metastasis Biology Group, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Charles Halsey
- Molecular Pathology Unit, Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Ellen S Hong
- Molecular Oncology Section - Metastasis Biology Group, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Sandra S Burkett
- Comparative Molecular Cytogenetics Core Facility, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - James Morrow
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michael M Lizardo
- Molecular Oncology Section - Metastasis Biology Group, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Tanasa Osborne
- National Institute of Environmental Health, Research Triangle Park, North Carolina, USA
| | - Samuel Q Li
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Hue H Luu
- Department of Orthopedic Surgery & Rehabilitation Medicine, University of Chicago, Medicine & Biological Sciences, Chicago, USA
| | - Paul Meltzer
- Genetic Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Chand Khanna
- Molecular Oncology Section - Metastasis Biology Group, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
49
|
Morrow JJ, Mendoza A, Koyen A, Lizardo MM, Ren L, Waybright TJ, Hansen RJ, Gustafson DL, Zhou M, Fan TM, Scacheri PC, Khanna C. mTOR Inhibition Mitigates Enhanced mRNA Translation Associated with the Metastatic Phenotype of Osteosarcoma Cells In Vivo. Clin Cancer Res 2016; 22:6129-6141. [PMID: 27342399 DOI: 10.1158/1078-0432.ccr-16-0326] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 05/20/2016] [Accepted: 06/13/2016] [Indexed: 12/20/2022]
Abstract
PURPOSE To successfully metastasize, tumor cells must respond appropriately to biological stressors encountered during metastatic progression. We sought to test the hypothesis that enhanced efficiency of mRNA translation during periods of metastatic stress is required for metastatic competence of osteosarcoma and that this metastasis-specific adaptation is amenable to therapeutic intervention. EXPERIMENTAL DESIGN We employ novel reporter and proteomic systems that enable tracking of mRNA translation efficiency and output in metastatic osteosarcoma cells as they colonize the lungs. We test the potential to target mRNA translation as an antimetastatic therapeutic strategy through pharmacokinetic studies and preclinical assessment of the prototypic mTOR inhibitor, rapamycin, across multiple models of metastasis. RESULTS Metastatic osteosarcoma cells translate mRNA more efficiently than nonmetastatic cells during critical stressful periods of metastatic colonization of the lung. Rapamycin inhibits translational output during periods of metastatic stress, mitigates lung colonization, and prolongs survival. mTOR-inhibiting exposures of rapamycin are achievable in mice using treatment schedules that correspond to human doses well below the MTDs defined in human patients, and as such are very likely to be tolerated over long exposures alone and in combination with other agents. CONCLUSIONS Metastatic competence of osteosarcoma cells is dependent on efficient mRNA translation during stressful periods of metastatic progression, and the mTOR inhibitor, rapamycin, can mitigate this translation and inhibit metastasis in vivo Our data suggest that mTOR pathway inhibitors should be reconsidered in the clinic using rationally designed dosing schedules and clinical metrics related to metastatic progression. Clin Cancer Res; 22(24); 6129-41. ©2016 AACR.
Collapse
Affiliation(s)
- James J Morrow
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio.,Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Arnulfo Mendoza
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Allyson Koyen
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Michael M Lizardo
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Ling Ren
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Timothy J Waybright
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Ryan J Hansen
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado.,Pharmacology Shared Resource, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado
| | - Daniel L Gustafson
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado.,Pharmacology Shared Resource, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado
| | - Ming Zhou
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois, Urbana, Illinois
| | - Peter C Scacheri
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Chand Khanna
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland.
| |
Collapse
|
50
|
Xue YL, Meng XQ, Ma LJ, Yuan Z. Plumbagin exhibits an anti-proliferative effect in human osteosarcoma cells by downregulating FHL2 and interfering with Wnt/β-catenin signalling. Oncol Lett 2016; 12:1095-1100. [PMID: 27446400 DOI: 10.3892/ol.2016.4725] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 03/10/2016] [Indexed: 11/05/2022] Open
Abstract
Plumbagin, a naphthoquinone constituent of Plumbago zeylanica L. (Plumbaginaceae) is widely used in traditional Chinese medicine as an antifungal, antibacterial and anti-inflammatory agent. Plumbagin is known to exhibit proapoptotic, antiangiogenic and antimetastatic effects in cancer cells. The transcriptional co-factor four and a half LIM domains 2 (FHL2) is a multifunctional adaptor protein that is involved in the regulation of gene expression, signal transduction and cell proliferation and differentiation, and also acts as a tumor suppressor or oncoprotein depending on the tissue microenvironment. The present study investigated the effect of plumbagin on FHL2 expression, Wnt/β-catenin signalling and its anti-proliferative activity in various human osteosarcoma cell lines, including SaOS2, MG63, HOS and U2OS. The cells were exposed to plumbagin and the expression of FHL2 was evaluated using western blot analysis. Furthermore, the anti-proliferative effect of plumbagin was evaluated using a 3-(4,5 dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. In addition, since FHL2 is involved in Wnt/β-catenin signaling, the effect of plumbagin on β-catenin and its primary target genes, including v-myc avian myelocytomatosis viral oncogene homolog (c-Myc) and WNT1 inducible signaling pathway protein-1 (WISP-1), was evaluated using western blot analysis. It was observed that plumbagin suppressed the expression of FHL2 and exhibited significant anti-proliferative activity in osteosarcoma cells. It also attenuated Wnt/β-catenin signalling by downregulating β-catenin and its target genes, including c-Myc and WISP-1. In conclusion, plumbagin demonstrated anti-proliferative activity in osteosarcoma cells by downregulating FHL2 and interfering with Wnt/β-catenin signalling.
Collapse
Affiliation(s)
- Yuan-Liang Xue
- Department of Orthopedics of Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| | - Xiang-Qi Meng
- Department of Orthopedics, Suzhou Hospital of Traditional Chinese Medicine, Suzhou, Jiangsu 215000, P.R. China
| | - Long-Jun Ma
- Department of Orthopedics, People's Hospital of Yangxin, Binzhou, Shandong 251800, P.R. China
| | - Zhen Yuan
- Department of Orthopedics, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| |
Collapse
|