1
|
Lacinski RA, Dziadowicz SA, Melemai VK, Fitzpatrick B, Pisquiy JJ, Heim T, Lohse I, Schoedel KE, Llosa NJ, Weiss KR, Lindsey BA. Spatial multiplexed immunofluorescence analysis reveals coordinated cellular networks associated with overall survival in metastatic osteosarcoma. Bone Res 2024; 12:55. [PMID: 39333065 PMCID: PMC11436896 DOI: 10.1038/s41413-024-00359-z] [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: 02/05/2024] [Revised: 06/16/2024] [Accepted: 07/18/2024] [Indexed: 09/29/2024] Open
Abstract
Patients diagnosed with advanced osteosarcoma, often in the form of lung metastases, have abysmal five-year overall survival rates. The complexity of the osteosarcoma immune tumor microenvironment has been implicated in clinical trial failures of various immunotherapies. The purpose of this exploratory study was to spatially characterize the immune tumor microenvironment of metastatic osteosarcoma lung specimens. Knowledge of the coordinating cellular networks within these tissues could then lead to improved outcomes when utilizing immunotherapy for treatment of this disease. Importantly, various cell types, interactions, and cellular neighborhoods were associated with five-year survival status. Of note, increases in cellular interactions between T lymphocytes, positive for programmed cell death protein 1, and myeloid-derived suppressor cells were observed in the 5-year deceased cohort. Additionally, cellular neighborhood analysis identified an Immune-Cold Parenchyma cellular neighborhood, also associated with worse 5-year survival. Finally, the Osteosarcoma Spatial Score, which approximates effector immune activity in the immune tumor microenvironment through the spatial proximity of immune and tumor cells, was increased within 5-year survivors, suggesting improved effector signaling in this patient cohort. Ultimately, these data represent a robust spatial multiplexed immunofluorescence analysis of the metastatic osteosarcoma immune tumor microenvironment. Various communication networks, and their association with survival, were described. In the future, identification of these networks may suggest the use of specific, combinatory immunotherapeutic strategies for improved anti-tumor immune responses and outcomes in osteosarcoma.
Collapse
Affiliation(s)
- Ryan A Lacinski
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Cancer Institute, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Sebastian A Dziadowicz
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Vincent K Melemai
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Brody Fitzpatrick
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - John J Pisquiy
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Tanya Heim
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Ines Lohse
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Karen E Schoedel
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Nicolas J Llosa
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Kurt R Weiss
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Brock A Lindsey
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
2
|
Mustokoweni S, Mahyudin F, Setiawati R, Nugrahenny D, Hidayat M, Kalim H, Mintaroem K, Fitri LE, Hogendoorn PCW. Correlation of High-Grade Osteosarcoma Response to Chemotherapy with Enhanced Tissue Immunological Response: Analysis of CD95R, IFN-γ, Catalase, Hsp70, and VEGF. Virchows Arch 2024; 484:925-937. [PMID: 38748263 PMCID: PMC11186924 DOI: 10.1007/s00428-024-03801-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 06/20/2024]
Abstract
High-grade osteosarcoma, a primary malignant bone tumour, is experiencing a global increase in reported incidence with varied prevalence. Despite advances in management, which include surgery and neoadjuvant chemotherapy often an unsatisfactory outcome is found due to poor or heterogeneous response to chemotherapy. Our study delved into chemotherapy responses in osteosarcoma patients and associated molecular expressions, focusing on CD95 receptor (CD95R), interferon (IFN)-γ, catalase, heat-shock protein (Hsp)70, and vascular endothelial growth factor (VEGF). Employing immunohistochemistry and Huvos grading of post-chemo specimens, we analysed formalin-fixed paraffin-embedded (FFPE) osteosarcoma tissue of resected post-chemotherapy specimens from Dr. Soetomo General Academic Hospital in Surabaya, Indonesia (DSGAH), spanning from 2016 to 2020. Results revealed varied responses (poor 40.38%, moderate 48.08%, good 11.54%) and distinct patterns in CD95R, IFN-γ, catalase, Hsp70, and VEGF expression. Significant differences among response groups were observed in CD95R and IFN-γ expression in tumour-infiltrating lymphocytes. The trend of diminishing CD95R expression from poor to good responses, accompanied by an increase in IFN-γ, implied a reduction in the count of viable osteosarcoma cells with the progression of Huvos grading. Catalase expression in osteosarcoma cells was consistently elevated in the poor response group, while Hsp70 expression was highest. VEGF expression in macrophages was significantly higher in the good response group. In conclusion, this study enhances our understanding of immune-chemotherapy interactions in osteosarcoma and identifies potential biomarkers for targeted interventions.
Collapse
Affiliation(s)
- Sjahjenny Mustokoweni
- Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia.
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Airlangga/Dr, Soetomo General Academic Hospital, Mayjen Prof. Dr. Moestopo 6-8, Airlangga, Gubeng, Surabaya, East Java, Indonesia.
| | - Ferdiansyah Mahyudin
- Department of Orthopaedic Surgery and Traumatology, Faculty of Medicine, Universitas Airlangga/Dr, Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Rosy Setiawati
- Department of Radiology, Faculty of Medicine, Universitas Airlangga/Dr, Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Dian Nugrahenny
- Department of Pharmacology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Mohamad Hidayat
- Department of Orthopaedic Surgery and Traumatology, Faculty of Medicine, Universitas Brawijaya/Dr. Saiful Anwar General Hospital, Malang, Indonesia
| | - Handono Kalim
- Department of Internal Medicine, Faculty of Medicine, Universitas Brawijaya/Dr. Saiful Anwar General Hospital, Malang, Indonesia
| | - Karyono Mintaroem
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Loeki Enggar Fitri
- Department of Parasitology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Pancras C W Hogendoorn
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Airlangga/Dr, Soetomo General Academic Hospital, Mayjen Prof. Dr. Moestopo 6-8, Airlangga, Gubeng, Surabaya, East Java, Indonesia.
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
3
|
Yang M, Su Y, Xu K, Zheng H, Yuan Q, Cai Y, Aihaiti Y, Xu P. Ferroptosis-related lncRNAs guiding osteosarcoma prognosis and immune microenvironment. J Orthop Surg Res 2023; 18:787. [PMID: 37858131 PMCID: PMC10588205 DOI: 10.1186/s13018-023-04286-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/14/2023] [Indexed: 10/21/2023] Open
Abstract
OBJECTIVE To investigate the ferroptosis-related long non-coding RNAs (FRLncs) implicated in influencing the prognostic and immune microenvironment in osteosarcoma (OS), and to establish a foundational framework for informing clinical decision making pertaining to OS management. METHODS Transcriptome data and clinical data pertaining to 86 cases of OS, the GSE19276, GSE16088 and GSE33382 datasets, and a list of ferroptosis-related genes (FRGs) were used to establish a risk prognostic model through comprehensive analysis. The identification of OS-related differentially expressed FRGs was achieved through an integrated analysis encompassing the aforementioned 86 OS transcriptome data and the GSE19276, GSE16088 and GSE33382 datasets. Concurrently, OS-related FRLncs were ascertained via co-expression analysis. To establish a risk prognostic model for OS, Univariate Cox regression analysis and Lasso Cox regression analysis were employed. Subsequently, a comprehensive evaluation was conducted, comprising risk curve analysis, survival analysis, receiver operating characteristic curve analysis and independent prognosis analysis. Model validation with distinct clinical subgroups was performed to assess the applicability of the risk prognostic model to diverse patient categories. Moreover, single sample gene set enrichment analysis (ssGSEA) was conducted to investigate variations in immune cell populations and immune functions within the context of the risk prognostic model. Furthermore, an analysis of immune checkpoint differentials yielded insights into immune checkpoint-related genes linked to OS prognosis. Finally, the risk prognosis model was verified by dividing the samples into train group and test group. RESULTS We identified a set of seven FRLncs that exhibit potential as prognostic markers and influence factors of the immune microenvironment in the context of OS. This ensemble encompasses three high-risk FRLncs, denoted as APTR, AC105914.2 and AL139246.5, alongside four low-risk FRLncs, designated as DSCR8, LOH12CR2, AC027307.2 and AC025048.2. Furthermore, our analysis revealed notable down-regulation in the high-risk group across four distinct immune cell types, namely neutrophils, natural killer cells, plasmacytoid dendritic cells and tumor-infiltrating lymphocytes. This down-regulation was also reflected in four key immune functions, antigen-presenting cell (APC)-co-stimulation, checkpoint, cytolytic activity and T cell co-inhibition. Additionally, we identified seven immune checkpoint-associated genes with significant implications for OS prognosis, including CD200R1, HAVCR2, LGALS9, CD27, LAIR1, LAG3 and TNFSF4. CONCLUSION The findings of this study have identified FRLncs capable of influencing OS prognosis and immune microenvironment, as well as immune checkpoint-related genes that are linked to OS prognosis. These discoveries establish a substantive foundation for further investigations into OS survival and offer valuable insights for informing clinical decision making in this context.
Collapse
Affiliation(s)
- Mingyi Yang
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Yani Su
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Ke Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Haishi Zheng
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Qiling Yuan
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Yongsong Cai
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Yirixiati Aihaiti
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
4
|
Hu Y, Mohammad Mirzaei N, Shahriyari L. Bio-Mechanical Model of Osteosarcoma Tumor Microenvironment: A Porous Media Approach. Cancers (Basel) 2022; 14:cancers14246143. [PMID: 36551627 PMCID: PMC9777270 DOI: 10.3390/cancers14246143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma is the most common malignant bone tumor in children and adolescents with a poor prognosis. To describe the progression of osteosarcoma, we expanded a system of data-driven ODE from a previous study into a system of Reaction-Diffusion-Advection (RDA) equations and coupled it with Biot equations of poroelasticity to form a bio-mechanical model. The RDA system includes the spatio-temporal information of the key components of the tumor microenvironment. The Biot equations are comprised of an equation for the solid phase, which governs the movement of the solid tumor, and an equation for the fluid phase, which relates to the motion of cells. The model predicts the total number of cells and cytokines of the tumor microenvironment and simulates the tumor's size growth. We simulated different scenarios using this model to investigate the impact of several biomedical settings on tumors' growth. The results indicate the importance of macrophages in tumors' growth. Particularly, we have observed a high co-localization of macrophages and cancer cells, and the concentration of tumor cells increases as the number of macrophages increases.
Collapse
|
5
|
Le T, Su S, Kirshtein A, Shahriyari L. Data-Driven Mathematical Model of Osteosarcoma. Cancers (Basel) 2021; 13:cancers13102367. [PMID: 34068946 PMCID: PMC8156666 DOI: 10.3390/cancers13102367] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/10/2021] [Accepted: 05/10/2021] [Indexed: 12/22/2022] Open
Abstract
As the immune system has a significant role in tumor progression, in this paper, we develop a data-driven mathematical model to study the interactions between immune cells and the osteosarcoma microenvironment. Osteosarcoma tumors are divided into three clusters based on their relative abundance of immune cells as estimated from their gene expression profiles. We then analyze the tumor progression and effects of the immune system on cancer growth in each cluster. Cluster 3, which had approximately the same number of naive and M2 macrophages, had the slowest tumor growth, and cluster 2, with the highest population of naive macrophages, had the highest cancer population at the steady states. We also found that the fastest growth of cancer occurred when the anti-tumor immune cells and cytokines, including dendritic cells, helper T cells, cytotoxic cells, and IFN-γ, switched from increasing to decreasing, while the dynamics of regulatory T cells switched from decreasing to increasing. Importantly, the most impactful immune parameters on the number of cancer and total cells were the activation and decay rates of the macrophages and regulatory T cells for all clusters. This work presents the first osteosarcoma progression model, which can be later extended to investigate the effectiveness of various osteosarcoma treatments.
Collapse
Affiliation(s)
- Trang Le
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (T.L.); (S.S.)
| | - Sumeyye Su
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (T.L.); (S.S.)
| | - Arkadz Kirshtein
- Department of Mathematics, Tufts University, Medford, MA 02155, USA;
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (T.L.); (S.S.)
- Correspondence:
| |
Collapse
|
6
|
Le T, Su S, Shahriyari L. Immune classification of osteosarcoma. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:1879-1897. [PMID: 33757216 PMCID: PMC7992873 DOI: 10.3934/mbe.2021098] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Tumor immune microenvironment has been shown to be important in predicting the tumor progression and the outcome of treatments. This work aims to identify different immune patterns in osteosarcoma and their clinical characteristics. We use the latest and best performing deconvolution method, CIBERSORTx, to obtain the relative abundance of 22 immune cells. Then we cluster patients based on their estimated immune abundance and study the characteristics of these clusters, along with the relationship between immune infiltration and outcome of patients. We find that abundance of CD8 T cells, NK cells and M1 Macrophages have a positive association with prognosis, while abundance of γδ T cells, Mast cells, M0 Macrophages and Dendritic cells have a negative association with prognosis. Accordingly, the cluster with the lowest proportion of CD8 T cells, M1 Macrophages and highest proportion of M0 Macrophages has the worst outcome among clusters. By grouping patients with similar immune patterns, we are also able to suggest treatments that are specific to the tumor microenvironment.
Collapse
Affiliation(s)
- Trang Le
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA MA 01003-9305, USA
| | - Sumeyye Su
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA MA 01003-9305, USA
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA MA 01003-9305, USA
| |
Collapse
|
7
|
Prospects for NK Cell Therapy of Sarcoma. Cancers (Basel) 2020; 12:cancers12123719. [PMID: 33322371 PMCID: PMC7763692 DOI: 10.3390/cancers12123719] [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: 11/15/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Sarcomas are a group of aggressive tumors originating from mesenchymal tissues. Patients with advanced disease have poor prognosis due to the ineffectiveness of current treatment protocols. A subset of lymphocytes called natural killer (NK) cells is capable of effective surveillance and clearance of sarcomas, constituting a promising tool for immunotherapeutic treatment. However, sarcomas can cause impairment in NK cell function, associated with enhanced tumor growth and dissemination. In this review, we discuss the molecular mechanisms of sarcoma-mediated suppression of NK cells and their implications for the design of novel NK cell-based immunotherapies against sarcoma. Abstract Natural killer (NK) cells are innate lymphoid cells with potent antitumor activity. One of the most NK cell cytotoxicity-sensitive tumor types is sarcoma, an aggressive mesenchyme-derived neoplasm. While a combination of radical surgery and radio- and chemotherapy can successfully control local disease, patients with advanced sarcomas remain refractory to current treatment regimens, calling for novel therapeutic strategies. There is accumulating evidence for NK cell-mediated immunosurveillance of sarcoma cells during all stages of the disease, highlighting the potential of using NK cells as a therapeutic tool. However, sarcomas display multiple immunoevasion mechanisms that can suppress NK cell function leading to an uncontrolled tumor outgrowth. Here, we review the current evidence for NK cells’ role in immune surveillance of sarcoma during disease initiation, promotion, progression, and metastasis, as well as the molecular mechanisms behind sarcoma-mediated NK cell suppression. Further, we apply this basic understanding of NK–sarcoma crosstalk in order to identify and summarize the most promising candidates for NK cell-based sarcoma immunotherapy.
Collapse
|
8
|
Harris MA, Hons BB, Shekhar TM, Coupland LA, Miles MA, Hawkins CJ. Transient NK Cell Depletion Facilitates Pulmonary Osteosarcoma Metastases After Intravenous Inoculation in Athymic Mice. J Adolesc Young Adult Oncol 2020; 9:667-671. [DOI: 10.1089/jayao.2019.0172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
| | - BBioMed Hons
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia
| | - Tanmay M. Shekhar
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia
| | - Lucy A. Coupland
- The ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Mark A. Miles
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia
| | - Christine J. Hawkins
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia
| |
Collapse
|
9
|
The contribution of immune infiltrates and the local microenvironment in the pathogenesis of osteosarcoma. Cell Immunol 2019; 343:103711. [DOI: 10.1016/j.cellimm.2017.10.011] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/22/2017] [Accepted: 10/26/2017] [Indexed: 12/21/2022]
|
10
|
Westrich JA, Vermeer DW, Silva A, Bonney S, Berger JN, Cicchini L, Greer RO, Song JI, Raben D, Slansky JE, Lee JH, Spanos WC, Pyeon D. CXCL14 suppresses human papillomavirus-associated head and neck cancer through antigen-specific CD8 + T-cell responses by upregulating MHC-I expression. Oncogene 2019; 38:7166-7180. [PMID: 31417179 PMCID: PMC6856418 DOI: 10.1038/s41388-019-0911-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/23/2019] [Accepted: 05/26/2019] [Indexed: 12/30/2022]
Abstract
Evasion of the host immune responses is critical for both persistent human papillomavirus (HPV) infection and associated cancer progression. We have previously shown that expression of the homeostatic chemokine CXCL14 is significantly downregulated by the HPV oncoprotein E7 during cancer progression. Restoration of CXCL14 expression in HPV-positive head and neck cancer (HNC) cells dramatically suppresses tumor growth and increases survival through an immune-dependent mechanism in mice. While CXCL14 recruits natural killer (NK) and T cells to the tumor microenvironment, the mechanism by which CXCL14 mediates tumor suppression through NK and/or T cells remained undefined. Here, we report that CD8+ T cells are required for CXCL14-mediated tumor suppression. Using a CD8+ T cell receptor transgenic model, we show that the CXCL14-mediated antitumor CD8+ T cell responses require antigen specificity. Interestingly, CXCL14 expression restores major histocompatibility complex class I (MHC-I) expression on HPV-positive HNC cells downregulated by HPV, and knockdown of MHC-I expression in HNC cells results in loss of tumor suppression even with CXCL14 expression. These results suggest that CXCL14 enacts antitumor immunity through restoration of MHC-I expression on tumor cells and promoting antigen-specific CD8+ T cell responses to suppress HPV-positive HNC.
Collapse
Affiliation(s)
- Joseph A Westrich
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.,Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Daniel W Vermeer
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD, 57104, USA
| | - Alexa Silva
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Stephanie Bonney
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Jennifer N Berger
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Louis Cicchini
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Robert O Greer
- Departments of Pathology and Dermatology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.,Division of Oral and Maxillofacial Pathology, University of Colorado School of Dental Medicine, Aurora, CO, 80045, USA
| | - John I Song
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - David Raben
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Jill E Slansky
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - John H Lee
- Chan Soon-Shiong Institute for Medicine, El Segundo, CA, 90245, USA
| | - William C Spanos
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD, 57104, USA
| | - Dohun Pyeon
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA. .,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
11
|
Stamatopoulos A, Stamatopoulos T, Gamie Z, Kenanidis E, Ribeiro RDC, Rankin KS, Gerrand C, Dalgarno K, Tsiridis E. Mesenchymal stromal cells for bone sarcoma treatment: Roadmap to clinical practice. J Bone Oncol 2019; 16:100231. [PMID: 30956944 PMCID: PMC6434099 DOI: 10.1016/j.jbo.2019.100231] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 03/14/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Over the past few decades, there has been growing interest in understanding the molecular mechanisms of cancer pathogenesis and progression, as it is still associated with high morbidity and mortality. Current management of large bone sarcomas typically includes the complex therapeutic approach of limb salvage or sacrifice combined with pre- and postoperative multidrug chemotherapy and/or radiotherapy, and is still associated with high recurrence rates. The development of cellular strategies against specific characteristics of tumour cells appears to be promising, as they can target cancer cells selectively. Recently, Mesenchymal Stromal Cells (MSCs) have been the subject of significant research in orthopaedic clinical practice through their use in regenerative medicine. Further research has been directed at the use of MSCs for more personalized bone sarcoma treatments, taking advantage of their wide range of potential biological functions, which can be augmented by using tissue engineering approaches to promote healing of large defects. In this review, we explore the use of MSCs in bone sarcoma treatment, by analyzing MSCs and tumour cell interactions, transduction of MSCs to target sarcoma, and their clinical applications on humans concerning bone regeneration after bone sarcoma extraction.
Collapse
Key Words
- 5-FC, 5-fluorocytosine
- AAT, a1-antitrypsin
- APCs, antigen presenting cells
- ASC, adipose-derived stromal/stem cells
- Abs, antibodies
- Ang1, angiopoietin-1
- BD, bone defect
- BMMSCs, bone marrow-derived mesenchymal stromal cells
- Biology
- Bone
- CAM, cell adhesion molecules
- CCL5, chemokine ligand 5
- CCR2, chemokine receptor 2
- CD, classification determinants
- CD, cytosine deaminase
- CLUAP1, clusterin associated protein 1
- CSPG4, Chondroitin sulfate proteoglycan 4
- CX3CL1, chemokine (C-X3-C motif) ligand 1
- CXCL12/CXCR4, C-X-C chemokine ligand 12/ C-X-C chemokine receptor 4
- CXCL12/CXCR7, C-X-C chemokine ligand 12/ C-X-C chemokine receptor 7
- CXCR4, chemokine receptor type 4
- Cell
- DBM, Demineralized Bone Marrow
- DKK1, dickkopf-related protein 1
- ECM, extracellular matrix
- EMT, epithelial-mesenchymal transition
- FGF-2, fibroblast growth factors-2
- FGF-7, fibroblast growth factors-7
- GD2, disialoganglioside 2
- HER2, human epidermal growth factor receptor 2
- HGF, hepatocyte growth factor
- HMGB1/RACE, high mobility group box-1 protein/ receptor for advanced glycation end-products
- IDO, indoleamine 2,3-dioxygenase
- IFN-α, interferon alpha
- IFN-β, interferon beta
- IFN-γ, interferon gamma
- IGF-1R, insulin-like growth factor 1 receptor
- IL-10, interleukin-10
- IL-12, interleukin-12
- IL-18, interleukin-18
- IL-1b, interleukin-1b
- IL-21, interleukin-21
- IL-2a, interleukin-2a
- IL-6, interleukin-6
- IL-8, interleukin-8
- IL11RA, Interleukin 11 Receptor Subunit Alpha
- MAGE, melanoma antigen gene
- MCP-1, monocyte chemoattractant protein-1
- MMP-2, matrix metalloproteinase-2
- MMP2/9, matrix metalloproteinase-2/9
- MRP, multidrug resistance protein
- MSCs, mesenchymal stem/stromal cells
- Mesenchymal
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- OPG, osteoprotegerin
- Orthopaedic
- PBS, phosphate-buffered saline
- PDGF, platelet-derived growth factor
- PDX, patient derived xenograft
- PEDF, pigment epithelium-derived factor
- PGE2, prostaglandin E2
- PI3K/Akt, phosphoinositide 3-kinase/protein kinase B
- PTX, paclitaxel
- RANK, receptor activator of nuclear factor kappa-B
- RANKL, receptor activator of nuclear factor kappa-B ligand
- RBCs, red blood cells
- RES, reticuloendothelial system
- RNA, ribonucleic acid
- Regeneration
- SC, stem cells
- SCF, stem cells factor
- SDF-1, stromal cell-derived factor 1
- STAT-3, signal transducer and activator of transcription 3
- Sarcoma
- Stromal
- TAAs, tumour-associated antigens
- TCR, T cell receptor
- TGF-b, transforming growth factor beta
- TGF-b1, transforming growth factor beta 1
- TNF, tumour necrosis factor
- TNF-a, tumour necrosis factor alpha
- TRAIL, tumour necrosis factor related apoptosis-inducing ligand
- Tissue
- VEGF, vascular endothelial growth factor
- VEGFR, vascular endothelial growth factor receptor
- WBCs, white blood cell
- hMSCs, human mesenchymal stromal cells
- rh-TRAIL, recombinant human tumour necrosis factor related apoptosis-inducing ligand
Collapse
Affiliation(s)
- Alexandros Stamatopoulos
- Academic Orthopaedic Unit, Papageorgiou General Hospital, Aristotle University Medical School, West Ring Road of Thessaloniki, Pavlos Melas Area, N. Efkarpia, 56403 Thessaloniki, Greece
- Center of Orthopaedics and Regenerative Medicine (C.O.RE.), Center for Interdisciplinary Research and Innovation (C.I.R.I.), Aristotle University Thessaloniki, Greece
| | - Theodosios Stamatopoulos
- Academic Orthopaedic Unit, Papageorgiou General Hospital, Aristotle University Medical School, West Ring Road of Thessaloniki, Pavlos Melas Area, N. Efkarpia, 56403 Thessaloniki, Greece
- Center of Orthopaedics and Regenerative Medicine (C.O.RE.), Center for Interdisciplinary Research and Innovation (C.I.R.I.), Aristotle University Thessaloniki, Greece
| | - Zakareya Gamie
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Eustathios Kenanidis
- Academic Orthopaedic Unit, Papageorgiou General Hospital, Aristotle University Medical School, West Ring Road of Thessaloniki, Pavlos Melas Area, N. Efkarpia, 56403 Thessaloniki, Greece
- Center of Orthopaedics and Regenerative Medicine (C.O.RE.), Center for Interdisciplinary Research and Innovation (C.I.R.I.), Aristotle University Thessaloniki, Greece
| | - Ricardo Da Conceicao Ribeiro
- School of Mechanical and Systems Engineering, Stephenson Building, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | - Kenneth Samora Rankin
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Craig Gerrand
- Royal National Orthopaedic Hospital, Brockley Hill, Stanmore, HA7 4LP, UK
| | - Kenneth Dalgarno
- School of Mechanical and Systems Engineering, Stephenson Building, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | - Eleftherios Tsiridis
- Academic Orthopaedic Unit, Papageorgiou General Hospital, Aristotle University Medical School, West Ring Road of Thessaloniki, Pavlos Melas Area, N. Efkarpia, 56403 Thessaloniki, Greece
- Center of Orthopaedics and Regenerative Medicine (C.O.RE.), Center for Interdisciplinary Research and Innovation (C.I.R.I.), Aristotle University Thessaloniki, Greece
| |
Collapse
|
12
|
Wang Z, Wang Z, Li B, Wang S, Chen T, Ye Z. Innate Immune Cells: A Potential and Promising Cell Population for Treating Osteosarcoma. Front Immunol 2019; 10:1114. [PMID: 31156651 PMCID: PMC6531991 DOI: 10.3389/fimmu.2019.01114] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 05/01/2019] [Indexed: 12/13/2022] Open
Abstract
Advanced, recurrent, or metastasized osteosarcomas remain challenging to cure or even alleviate. Therefore, the development of novel therapeutic strategies is urgently needed. Cancer immunotherapy has greatly improved in recent years, with options including adoptive cellular therapy, vaccination, and checkpoint inhibitors. As such, immunotherapy is becoming a potential strategy for the treatment of osteosarcoma. Innate immunocytes, the first line of defense in the immune system and the bridge to adaptive immunity, are one of the vital effector cell subpopulations in cancer immunotherapy. Innate immune cell-based therapy has shown potent antitumor activity against hematologic malignancies and some solid tumors, including osteosarcoma. Importantly, some immune checkpoints are expressed on both innate and adaptive immune cells, modulating their functions in tumor immunity. Therefore, blocking or activating immune checkpoint-mediated downstream signaling pathways can improve the therapeutic effects of innate immune cell-based therapy. In this review, we summarize the current status and future prospects of innate immune cell-based therapy for the treatment of osteosarcoma, with a focus on the potential synergistic effects of combination therapy involving innate immunotherapy and immune checkpoint inhibitors/oncolytic viruses.
Collapse
Affiliation(s)
- Zenan Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Zhan Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Binghao Li
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Shengdong Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Tao Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Zhaoming Ye
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| |
Collapse
|
13
|
Yang X, Zhang W, Xu P. NK cell and macrophages confer prognosis and reflect immune status in osteosarcoma. J Cell Biochem 2019; 120:8792-8797. [PMID: 30556159 DOI: 10.1002/jcb.28167] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/08/2018] [Indexed: 01/24/2023]
Abstract
Osteosarcoma (OS) is a common primary malignant bone tumor in young adolescents. About 30% of patients with OS have a recurrence, and the overall survival after OS recurrence is not good. In this study, we aimed to analyze and identify factors that influence prognosis after OS relapse. We retrieved the Gene Expression Omnibus data set and collected a series of transcriptome data with clinical information, including microRNA (miRNA) and messenger RNA (mRNA) expression profiles in recurrent OS. Upon comparison of the dysregulated genes of survival status in the recurrent OS samples, it was found that there were 268 differential expressed (DE) mRNAs and six DE miRNAs. These genes are related to pathways in cancer. We also predicted the interaction networks of these DE mRNAs and miRNAs. Further, we applied cibersort to estimate the proportion of immune cell types and we discovered that natural killer cells and macrophages have different abundance between good prognosis and poor prognosis. Our study indicates that for recurrent OS samples, there are several differences between these two groups, including gene expression and the status of immune activation. The immunity status is a candidate signature for disease prediction, prevention, and therapy choices.
Collapse
Affiliation(s)
- Xianliang Yang
- Department of Orthopedic, The First People's Hospital of Wenling, The Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, Zhejiang, China
| | - Wenbin Zhang
- Department of Orthopedic, The First People's Hospital of Wenling, The Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, Zhejiang, China
| | - Panfeng Xu
- Department of Orthopedic, The First People's Hospital of Wenling, The Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, Zhejiang, China
| |
Collapse
|
14
|
|
15
|
Otoukesh B, Boddouhi B, Moghtadaei M, Kaghazian P, Kaghazian M. Novel molecular insights and new therapeutic strategies in osteosarcoma. Cancer Cell Int 2018; 18:158. [PMID: 30349420 PMCID: PMC6192346 DOI: 10.1186/s12935-018-0654-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/01/2018] [Indexed: 12/18/2022] Open
Abstract
Osteosarcoma (OS) is one of the most prevalent malignant cancers with lower survival and poor overall prognosis mainly in children and adolescents. Identifying the molecular mechanisms and OS stem cells (OSCs) as new concepts involved in disease pathogenesis and progression may potentially lead to new therapeutic targets. Therefore, therapeutic targeting of OSCs can be one of the most important and effective strategies for the treatment of OS. This review describes the new molecular targets of OS as well as novel therapeutic approaches in the design of future investigations and treatment.
Collapse
Affiliation(s)
- Babak Otoukesh
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, 1445613131 Iran
| | - Bahram Boddouhi
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, 1445613131 Iran
| | - Mehdi Moghtadaei
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, 1445613131 Iran
| | - Peyman Kaghazian
- Department of Orthopedic and Traumatology, Universitätsklinikum Bonn, Bonn, Germany
| | - Maria Kaghazian
- Department of Biology, Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
16
|
Wang S, Li H, Ye C, Lin P, Li B, Zhang W, Sun L, Wang Z, Xue D, Teng W, Zhou X, Lin N, Ye Z. Valproic Acid Combined with Zoledronate Enhance γδ T Cell-Mediated Cytotoxicity against Osteosarcoma Cells via the Accumulation of Mevalonate Pathway Intermediates. Front Immunol 2018. [PMID: 29535738 PMCID: PMC5835048 DOI: 10.3389/fimmu.2018.00377] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The long-term survival of osteosarcoma has remained unchanged in the last several decades. Immunotherapy is proved to be a promising therapeutic strategy against osteosarcoma, especially for those with metastasis. Our previous study explored the sensibilization of zoledronate (ZOL) in γδ T cell-mediated cytotoxicity against osteosarcoma, but we have not yet elucidated the specific mechanism. Besides, high concentration is required to achieve these effects, whereas plasma ZOL concentration declines rapidly in the circulation. Valproic acid (VPA), a histone deacetylase inhibitor commonly used as the antiepileptic drug, has attracted much attention due to its synergistic antitumor efficacy with chemotherapy or immunotherapy. Here, we demonstrated that VPA combined with ZOL revealed the synergistic effect in enhancing antitumor efficacy of γδ T cells against osteosarcoma cells. This enhancement was mainly TCR-mediated and largely dependent on granule exocytose pathway. Of note, our findings indicated that ZOL sensitized osteosarcoma cells to γδ T cells by increasing the accumulation of the mevalonate pathway intermediates, which could be facilitated by VPA. We also found that this combination had similar effects on primary osteosarcoma cells. All the results suggested that VPA combined with ZOL could reduce the dose required to achieve a significant antitumor effect of γδ T cells, promoting it to be a novel therapy against osteosarcoma.
Collapse
Affiliation(s)
- Shengdong Wang
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Hengyuan Li
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Chenyi Ye
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Peng Lin
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Binghao Li
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Wei Zhang
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Lingling Sun
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Zhan Wang
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Deting Xue
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Wangsiyuan Teng
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Xingzhi Zhou
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Nong Lin
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Zhaoming Ye
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
Abstract
BACKGROUND Early lymphocyte recovery following chemotherapy has been associated with improved outcome in many cancers, including in one small study of osteosarcoma patients. MATERIALS AND METHODS To confirm this finding, we retrospectively reviewed data from 53 patients with newly diagnosed osteosarcoma who had blood counts on day 14 (±1 d) following the first cycle of cisplatin and doxorubicin. RESULTS The median absolute lymphocyte count (ALC) 14 days after starting the first cycle of chemotherapy (ALC-14) was 1990 cells/μL (range: 600 to 6470). For 32 patients with an ALC-14≥1800 cells/μL, the 5-year progression-free survival (PFS) was 69%, compared with 33% for patients with an ALC-14 of <1800 cell/μL (P=0.036). In multivariable analysis of factors including age, sex, metastatic disease, and favorable histologic response to induction chemotherapy, ALC-14 was significantly associated with PFS (P=0.0081) and overall survival (P=0.0131). The use of ALC-14 appears to further stratify PFS and overall survival among patients when grouped by histologic response. CONCLUSIONS We confirmed that early lymphocyte recovery was associated with outcome in pediatric osteosarcoma. Although presumably reflecting immune-mediated tumor control, the precise mechanism for this is unclear. Further study of peripheral blood lymphocyte subpopulations in prospectively treated patients is underway.
Collapse
|
18
|
Heymann MF, Brown HK, Heymann D. Drugs in early clinical development for the treatment of osteosarcoma. Expert Opin Investig Drugs 2016; 25:1265-1280. [DOI: 10.1080/13543784.2016.1237503] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Marie-Françoise Heymann
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
- INSERM, UMR 957, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Equipe Ligue 2012, Faculty of Medicine, University of Nantes, Nantes, France
- Nantes University Hospital, Nantes, France
- European Associated Laboratory, Sarcoma Research Unit, Medical School, INSERM-University of Sheffield, Sheffield, UK
| | - Hannah K. Brown
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
- European Associated Laboratory, Sarcoma Research Unit, Medical School, INSERM-University of Sheffield, Sheffield, UK
| | - Dominique Heymann
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
- INSERM, UMR 957, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Equipe Ligue 2012, Faculty of Medicine, University of Nantes, Nantes, France
- Nantes University Hospital, Nantes, France
- European Associated Laboratory, Sarcoma Research Unit, Medical School, INSERM-University of Sheffield, Sheffield, UK
| |
Collapse
|
19
|
Osteosarcoma: Cells-of-Origin, Cancer Stem Cells, and Targeted Therapies. Stem Cells Int 2016; 2016:3631764. [PMID: 27366153 PMCID: PMC4913005 DOI: 10.1155/2016/3631764] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/10/2016] [Indexed: 12/25/2022] Open
Abstract
Osteosarcoma (OS) is the most common type of primary solid tumor that develops in bone. Although standard chemotherapy has significantly improved long-term survival over the past few decades, the outcome for those patients with metastatic or recurrent OS remains dismally poor and, therefore, novel agents and treatment regimens are urgently required. A hypothesis to explain the resistance of OS to chemotherapy is the existence of drug resistant CSCs with progenitor properties that are responsible of tumor relapses and metastasis. These subpopulations of CSCs commonly emerge during tumor evolution from the cell-of-origin, which are the normal cells that acquire the first cancer-promoting mutations to initiate tumor formation. In OS, several cell types along the osteogenic lineage have been proposed as cell-of-origin. Both the cell-of-origin and their derived CSC subpopulations are highly influenced by environmental and epigenetic factors and, therefore, targeting the OS-CSC environment and niche is the rationale for many recently postulated therapies. Likewise, some strategies for targeting CSC-associated signaling pathways have already been tested in both preclinical and clinical settings. This review recapitulates current OS cell-of-origin models, the properties of the OS-CSC and its niche, and potential new therapies able to target OS-CSCs.
Collapse
|
20
|
Regan D, Dow S. Manipulation of Innate Immunity for Cancer Therapy in Dogs. Vet Sci 2015; 2:423-439. [PMID: 29061951 PMCID: PMC5644648 DOI: 10.3390/vetsci2040423] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/20/2015] [Accepted: 11/23/2015] [Indexed: 12/23/2022] Open
Abstract
Over the last one to two decades, the field of cancer immunotherapy has rapidly progressed from early preclinical studies to a successful clinical reality and fourth major pillar of human cancer therapy. While current excitement in the field of immunotherapy is being driven by several major breakthroughs including immune checkpoint inhibitors and adoptive cell therapies, these advances stem from a foundation of pivotal studies demonstrating the immune systems role in tumor control and eradication. The following will be a succinct review on veterinary cancer immunotherapy as it pertains to manipulation of the innate immune system to control tumor growth and metastasis. In addition, we will provide an update on recent progress in our understanding of the innate immune system in veterinary tumor immunology, and how these gains may lead to novel therapies for the treatment of cancer in companion animals.
Collapse
Affiliation(s)
- Daniel Regan
- Flint Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80525, USA.
- The Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80525, USA.
| | - Steven Dow
- Flint Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80525, USA.
- The Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80525, USA.
| |
Collapse
|
21
|
Koehl U, Kalberer C, Spanholtz J, Lee DA, Miller JS, Cooley S, Lowdell M, Uharek L, Klingemann H, Curti A, Leung W, Alici E. Advances in clinical NK cell studies: Donor selection, manufacturing and quality control. Oncoimmunology 2015; 5:e1115178. [PMID: 27141397 PMCID: PMC4839369 DOI: 10.1080/2162402x.2015.1115178] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/24/2015] [Accepted: 10/27/2015] [Indexed: 11/13/2022] Open
Abstract
Natural killer (NK) cells are increasingly used in clinical studies in order to treat patients with various malignancies. The following review summarizes platform lectures and 2013–2015 consortium meetings on manufacturing and clinical use of NK cells in Europe and United States. A broad overview of recent pre-clinical and clinical results in NK cell therapies is provided based on unstimulated, cytokine-activated, as well as genetically engineered NK cells using chimeric antigen receptors (CAR). Differences in donor selection, manufacturing and quality control of NK cells for cancer immunotherapies are described and basic recommendations are outlined for harmonization in future NK cell studies.
Collapse
Affiliation(s)
- U Koehl
- Institute of Cellular Therapeutics, IFB-Tx, Hannover Medical School , Hannover, Germany
| | - C Kalberer
- Diagnostic Hematology, University Hospital Basel , Basel, Switzerland
| | - J Spanholtz
- Glycostem Therapeutics , Oss, the Netherlands
| | - D A Lee
- University of Texas MD Anderson Cancer Center, Pediatrics , Houston, TX, USA
| | - J S Miller
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota , Minneapolis, MN, USA
| | - S Cooley
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota , Minneapolis, MN, USA
| | - M Lowdell
- Department of Hematology, Royal Free Hospital, UCL Medical School , London, UK
| | - L Uharek
- Hematology and Oncology, Benjamin Franklin faculty of Charité , Berlin, Germany
| | - H Klingemann
- NantKwest Inc., Research & Development , Cambridge, MA, USA
| | - A Curti
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. and A. Seràgnoli", Berlin, University of Bologna , Italy
| | - W Leung
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital , Memphis, TN, USA
| | - E Alici
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm Sweden; Cell therapies institute, Nova Southeastern University, Fort Lauderdale, FL, USA; Hematology Center, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| |
Collapse
|
22
|
Roberts SS, Chou AJ, Cheung NKV. Immunotherapy of Childhood Sarcomas. Front Oncol 2015; 5:181. [PMID: 26301204 PMCID: PMC4528283 DOI: 10.3389/fonc.2015.00181] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/23/2015] [Indexed: 12/29/2022] Open
Abstract
Pediatric sarcomas are a heterogeneous group of malignant tumors of bone and soft tissue origin. Although more than 100 different histologic subtypes have been described, the majority of pediatric cases belong to the Ewing’s family of tumors, rhabdomyosarcoma and osteosarcoma. Most patients that present with localized stage are curable with surgery and/or chemotherapy; however, those with metastatic disease at diagnosis or those who experience a relapse continue to have a very poor prognosis. New therapies for these patients are urgently needed. Immunotherapy is an established treatment modality for both liquid and solid tumors, and in pediatrics, most notably for neuroblastoma and osteosarcoma. In the past, immunomodulatory agents such as interferon, interleukin-2, and liposomal-muramyl tripeptide phosphatidyl-ethanolamine have been tried, with some activity seen in subsets of patients; additionally, various cancer vaccines have been studied with possible benefit. Monoclonal antibody therapies against tumor antigens such as disialoganglioside GD2 or immune checkpoint targets such as CTLA-4 and PD-1 are being actively explored in pediatric sarcomas. Building on the success of adoptive T cell therapy for EBV-related lymphoma, strategies to redirect T cells using chimeric antigen receptors and bispecific antibodies are rapidly evolving with potential for the treatment of sarcomas. This review will focus on recent preclinical and clinical developments in targeted agents for pediatric sarcomas with emphasis on the immunobiology of immune checkpoints, immunoediting, tumor microenvironment, antibody engineering, cell engineering, and tumor vaccines. The future integration of antibody-based and cell-based therapies into an overall treatment strategy of sarcoma will be discussed.
Collapse
Affiliation(s)
- Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center , New York, NY , USA
| | - Alexander J Chou
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center , New York, NY , USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center , New York, NY , USA
| |
Collapse
|
23
|
Hattinger CM, Fanelli M, Tavanti E, Vella S, Ferrari S, Picci P, Serra M. Advances in emerging drugs for osteosarcoma. Expert Opin Emerg Drugs 2015; 20:495-514. [PMID: 26021401 DOI: 10.1517/14728214.2015.1051965] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Osteosarcoma (OS), the most common primary malignant bone tumor, is currently treated with pre- and postoperative chemotherapy in association with the surgical removal of the tumor. Conventional treatments allow to cure about 60 - 65% of patients with primary tumors and only 20 - 25% of patients with recurrent disease. New treatment approaches and drugs are therefore highly warranted to improve prognosis. AREAS COVERED This review focuses on the therapeutic approaches that are under development or clinical evaluation in OS. Information was obtained from different and continuously updated data bases, as well as from literature searches, in which particular relevance was given to reports and reviews on new targeted therapies under clinical investigation in high-grade OS. EXPERT OPINION OS is a heterogeneous tumor, with a great variability in treatment response between patients. It is therefore unlikely that a single therapeutic tool will be uniformly successful for all OS patients. This claims for the validation of new treatment approaches together with biologic/(pharmaco)genetic markers, which may select the most appropriate subgroup of patients for each treatment approach. Since some promising novel agents and treatment strategies are currently tested in Phase I/II/III clinical trials, we may hope that new therapies with superior efficacy and safety profiles will be identified in the next few years.
Collapse
|