1
|
Liang W, Long H, Zhang H, Bai J, Jiang B, Wang J, Fu L, Ming W, Zhao J, Zeng B. Bone scaffolds-based localized drugs delivery for osteosarcoma: current status and future perspective. Drug Deliv 2024; 31:2391001. [PMID: 39239763 PMCID: PMC11382735 DOI: 10.1080/10717544.2024.2391001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 09/07/2024] Open
Abstract
A common malignant bone neoplasm in teenagers is Osteosarcoma. Chemotherapy, surgical therapy, and radiation therapy together comprise the usual clinical course of treatment for Osteosarcoma. While Osteosarcoma and other bone tumors are typically treated surgically, however, surgical resection frequently fails to completely eradicate tumors, and in turn becomes the primary reason for postoperative recurrence and metastasis, ultimately leading to a high rate of mortality. Patients still require radiation and/or chemotherapy after surgery to stop the spread of the tumor and its metastases, and both treatments have an adverse influence on the body's organ systems. In the postoperative management of osteosarcoma, bone scaffolds can load cargos (growth factors or drugs) and function as drug delivery systems (DDSs). This review describes the different kinds of bone scaffolds that are currently available and highlights key studies that use scaffolds as DDSs for the treatment of osteosarcomas. The discussion also includes difficulties and perspectives regarding the use of scaffold-based DDSs. The study may serve as a source for outlining efficient and secure postoperative osteosarcoma treatment plans.
Collapse
Affiliation(s)
- Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hengguo Long
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hongwei Zhang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Juqin Bai
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Bo Jiang
- Rehabilitation Department, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Jiangwei Wang
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Lifeng Fu
- Department of Orthopedics, Shaoxing City Keqiao District Hospital of Traditional Chinese Medicine, Shaoxing, China
| | - Wenyi Ming
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Jiayi Zhao
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Bin Zeng
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| |
Collapse
|
2
|
Sun C, Li S, Ding J. Biomaterials-Boosted Immunotherapy for Osteosarcoma. Adv Healthc Mater 2024; 13:e2400864. [PMID: 38771618 DOI: 10.1002/adhm.202400864] [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: 03/06/2024] [Revised: 05/05/2024] [Indexed: 05/22/2024]
Abstract
Osteosarcoma (OS) is a primary malignant bone tumor that emanates from mesenchymal cells, commonly found in the epiphyseal end of long bones. The highly recurrent and metastatic nature of OS poses significant challenges to the efficacy of treatment and negatively affects patient prognosis. Currently, available clinical treatment strategies primarily focus on maximizing tumor resection and reducing localized symptoms rather than the complete eradication of malignant tumor cells to achieve ideal outcomes. The biomaterials-boosted immunotherapy for OS is characterized by high effectiveness and a favorable safety profile. This therapeutic approach manipulates the tumor microenvironments at the cellular and molecular levels to impede tumor progression. This review delves into the mechanisms underlying the treatment of OS, emphasizing biomaterials-enhanced tumor immunity. Moreover, it summarizes the immune cell phenotype and tumor microenvironment regulation, along with the ability of immune checkpoint blockade to activate the autoimmune system. Gaining a profound comprehension of biomaterials-boosted OS immunotherapy is imperative to explore more efficacious immunotherapy protocols and treatment options in this setting.
Collapse
Affiliation(s)
- Chao Sun
- Department of Orthopedic Surgery, Orthopedic Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130061, P. R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Shuqiang Li
- Department of Orthopedic Surgery, Orthopedic Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130061, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| |
Collapse
|
3
|
Huang J, Feng Y, Shi Y, Shao W, Li G, Chen G, Li Y, Yang Z, Yao Z. Telomeres and telomerase in Sarcoma disease and therapy. Int J Med Sci 2024; 21:2065-2080. [PMID: 39239547 PMCID: PMC11373546 DOI: 10.7150/ijms.97485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/24/2024] [Indexed: 09/07/2024] Open
Abstract
Sarcoma is a rare tumor derived from the mesenchymal tissue and mainly found in children and adolescents. The outcome for patients with sarcoma is relatively poor compared with that for many other solid malignant tumors. Sarcomas have a highly heterogeneous pathogenesis, histopathology and biological behavior. Dysregulated signaling pathways and various gene mutations are frequently observed in sarcomas. The telomere maintenance mechanism (TMM) has recently been considered as a prognostic factor for patients with sarcomas, and alternative lengthening of telomeres (ALT) positivity has been correlated with poor outcomes in patients with several types of sarcomas. Therefore, telomeres and telomerases may be useful targets for treating sarcomas. This review aims to provide an overview of telomere and telomerase biology in sarcomas.
Collapse
Affiliation(s)
- Jin Huang
- Department of Cancer Research Institute, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Yan Feng
- Department of Cancer Research Institute, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - YangJing Shi
- Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Weilin Shao
- Department of Cancer Research Institute, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Genshan Li
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Gangxian Chen
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Ying Li
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Zuozhang Yang
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Zhihong Yao
- Department of Cancer Research Institute, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| |
Collapse
|
4
|
Karadimas T, Huynh TH, Chose C, Zervoudakis G, Clampitt B, Lapp S, Joyce D, Letson GD, Metts J, Binitie O, Mullinax JE, Lazarides A. Oncolytic Viral Therapy in Osteosarcoma. Viruses 2024; 16:1139. [PMID: 39066301 PMCID: PMC11281467 DOI: 10.3390/v16071139] [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: 07/02/2024] [Revised: 07/13/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
Primary bone malignancies, including osteosarcoma (OS), are rare but aggressive. Current OS treatment, involving surgical resection and chemotherapy, has improved survival for non-metastatic cases but remains ineffective for recurrent or metastatic OS. Oncolytic viral therapy (OVT) is a promising alternative, using naturally occurring or genetically modified viruses to selectively target and lyse cancer cells and induce a robust immune response against remaining OS cells. Various oncolytic viruses (OVs), such as adenovirus, herpes simplex virus, and measles virus, have demonstrated efficacy in preclinical OS models. Combining OVT with other therapeutics, such as chemotherapy or immunotherapy, may further improve outcomes. Despite these advances, challenges in reliability of preclinical models, safety, delivery, and immune response must be addressed to optimize OVT for clinical use. Future research should focus on refining delivery methods, exploring combination treatments, and clinical trials to ensure OVT's efficacy and safety for OS. Overall, OVT represents a novel approach with the potential to drastically improve survival outcomes for patients with OS.
Collapse
Affiliation(s)
- Thomas Karadimas
- Morsani College of Medicine, University of South Florida Health, Tampa, FL 33602, USA; (T.H.H.); (C.C.); (B.C.); (S.L.)
| | - Thien Huong Huynh
- Morsani College of Medicine, University of South Florida Health, Tampa, FL 33602, USA; (T.H.H.); (C.C.); (B.C.); (S.L.)
| | - Chloe Chose
- Morsani College of Medicine, University of South Florida Health, Tampa, FL 33602, USA; (T.H.H.); (C.C.); (B.C.); (S.L.)
| | - Guston Zervoudakis
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| | - Bryan Clampitt
- Morsani College of Medicine, University of South Florida Health, Tampa, FL 33602, USA; (T.H.H.); (C.C.); (B.C.); (S.L.)
| | - Sean Lapp
- Morsani College of Medicine, University of South Florida Health, Tampa, FL 33602, USA; (T.H.H.); (C.C.); (B.C.); (S.L.)
| | - David Joyce
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| | - George Douglas Letson
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| | - Jonathan Metts
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| | - Odion Binitie
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| | - John E. Mullinax
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| | - Alexander Lazarides
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| |
Collapse
|
5
|
Yuan Z, Zhang Y, Wang X, Wang X, Ren S, He X, Su J, Zheng A, Guo S, Chen Y, Deng S, Wu X, Li M, Du F, Zhao Y, Shen J, Wang Z, Xiao Z. The investigation of oncolytic viruses in the field of cancer therapy. Front Oncol 2024; 14:1423143. [PMID: 39055561 PMCID: PMC11270537 DOI: 10.3389/fonc.2024.1423143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Oncolytic viruses (OVs) have emerged as a potential strategy for tumor treatment due to their ability to selectively replicate in tumor cells, induce apoptosis, and stimulate immune responses. However, the therapeutic efficacy of single OVs is limited by the complexity and immunosuppressive nature of the tumor microenvironment (TME). To overcome these challenges, engineering OVs has become an important research direction. This review focuses on engineering methods and multi-modal combination therapies for OVs aimed at addressing delivery barriers, viral phagocytosis, and antiviral immunity in tumor therapy. The engineering approaches discussed include enhancing in vivo immune response, improving replication efficiency within the tumor cells, enhancing safety profiles, and improving targeting capabilities. In addition, this review describes the potential mechanisms of OVs combined with radiotherapy, chemotherapy, cell therapy and immune checkpoint inhibitors (ICIs), and summarizes the data of ongoing clinical trials. By continuously optimizing engineering strategies and combination therapy programs, we can achieve improved treatment outcomes and quality of life for cancer patients.
Collapse
Affiliation(s)
- Zijun Yuan
- Gulin Traditional Chinese Medicine Hospital, Luzhou, China
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yinping Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Siqi Ren
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xinyu He
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jiahong Su
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Anfu Zheng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Sipeng Guo
- Research And Experiment Center, Sichuan College of Traditional Chinese Medicine, Mianyang, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Zechen Wang
- Gulin Traditional Chinese Medicine Hospital, Luzhou, China
| | - Zhangang Xiao
- Gulin Traditional Chinese Medicine Hospital, Luzhou, China
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
- Department of Pharmacology, School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Mianyang, China
| |
Collapse
|
6
|
Gil-Chinchilla JI, Zapata AG, Moraleda JM, García-Bernal D. Bioengineered Mesenchymal Stem/Stromal Cells in Anti-Cancer Therapy: Current Trends and Future Prospects. Biomolecules 2024; 14:734. [PMID: 39062449 PMCID: PMC11275142 DOI: 10.3390/biom14070734] [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: 05/08/2024] [Revised: 06/11/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are one of the most widely used cell types in advanced therapies due to their therapeutic potential in the regulation of tissue repair and homeostasis, and immune modulation. However, their use in cancer therapy is controversial: they can inhibit cancer cell proliferation, but also potentially promote tumour growth by supporting angiogenesis, modulation of the immune milieu and increasing cancer stem cell invasiveness. This opposite behaviour highlights the need for careful and nuanced use of MSCs in cancer treatment. To optimize their anti-cancer effects, diverse strategies have bioengineered MSCs to enhance their tumour targeting and therapeutic properties or to deliver anti-cancer drugs. In this review, we highlight the advanced uses of MSCs in cancer therapy, particularly as carriers of targeted treatments due to their natural tumour-homing capabilities. We also discuss the potential of MSC-derived extracellular vesicles to improve the efficiency of drug or molecule delivery to cancer cells. Ongoing clinical trials are evaluating the therapeutic potential of these cells and setting the stage for future advances in MSC-based cancer treatment. It is critical to identify the broad and potent applications of bioengineered MSCs in solid tumour targeting and anti-cancer agent delivery to position them as effective therapeutics in the evolving field of cancer therapy.
Collapse
Affiliation(s)
- Jesús I. Gil-Chinchilla
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria (IMIB) Pascual Parrilla, Virgen de la Arrixaca University Hospital, University of Murcia, 30120 Murcia, Spain;
| | - Agustín G. Zapata
- Department of Cell Biology, Complutense University, 28040 Madrid, Spain;
| | - Jose M. Moraleda
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria (IMIB) Pascual Parrilla, Virgen de la Arrixaca University Hospital, University of Murcia, 30120 Murcia, Spain;
- Department of Medicine, University of Murcia, 30120 Murcia, Spain
| | - David García-Bernal
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria (IMIB) Pascual Parrilla, Virgen de la Arrixaca University Hospital, University of Murcia, 30120 Murcia, Spain;
- Department of Biochemistry, Molecular Biology and Immunology, University of Murcia, 30120 Murcia, Spain
| |
Collapse
|
7
|
Liang H, Cui M, Tu J, Chen X. Advancements in osteosarcoma management: integrating immune microenvironment insights with immunotherapeutic strategies. Front Cell Dev Biol 2024; 12:1394339. [PMID: 38915446 PMCID: PMC11194413 DOI: 10.3389/fcell.2024.1394339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/29/2024] [Indexed: 06/26/2024] Open
Abstract
Osteosarcoma, a malignant bone tumor predominantly affecting children and adolescents, presents significant therapeutic challenges, particularly in metastatic or recurrent cases. Conventional surgical and chemotherapeutic approaches have achieved partial therapeutic efficacy; however, the prognosis for long-term survival remains bleak. Recent studies have highlighted the imperative for a comprehensive exploration of the osteosarcoma immune microenvironment, focusing on the integration of diverse immunotherapeutic strategies-including immune checkpoint inhibitors, tumor microenvironment modulators, cytokine therapies, tumor antigen-specific interventions, cancer vaccines, cellular therapies, and antibody-based treatments-that are directly pertinent to modulating this intricate microenvironment. By targeting tumor cells, modulating the tumor microenvironment, and activating host immune responses, these innovative approaches have demonstrated substantial potential in enhancing the effectiveness of osteosarcoma treatments. Although most of these novel strategies are still in research or clinical trial phases, they have already demonstrated significant potential for individuals with osteosarcoma, suggesting the possibility of developing new, more personalized and effective treatment options. This review aims to provide a comprehensive overview of the current advancements in osteosarcoma immunotherapy, emphasizing the significance of integrating various immunotherapeutic methods to optimize therapeutic outcomes. Additionally, it underscores the imperative for subsequent research to further investigate the intricate interactions between the tumor microenvironment and the immune system, aiming to devise more effective treatment strategies. The present review comprehensively addresses the landscape of osteosarcoma immunotherapy, delineating crucial scientific concerns and clinical challenges, thereby outlining potential research directions.
Collapse
Affiliation(s)
- Hang Liang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Cui
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Belgiovine C, Mebelli K, Raffaele A, De Cicco M, Rotella J, Pedrazzoli P, Zecca M, Riccipetitoni G, Comoli P. Pediatric Solid Cancers: Dissecting the Tumor Microenvironment to Improve the Results of Clinical Immunotherapy. Int J Mol Sci 2024; 25:3225. [PMID: 38542199 PMCID: PMC10970338 DOI: 10.3390/ijms25063225] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 01/03/2025] Open
Abstract
Despite advances in their diagnosis and treatment, pediatric cancers remain among the leading causes of death in childhood. The development of immunotherapies and other forms of targeted therapies has significantly changed the prognosis of some previously incurable cancers in the adult population. However, so far, the results in pediatric cohorts are disappointing, which is mainly due to differences in tumor biology, including extreme heterogeneity and a generally low tumor mutational burden. A central role in the limited efficacy of immunotherapeutic approaches is played by the peculiar characteristics of the tumor microenvironment (TME) in pediatric cancer, with the scarcity of tumor infiltration by T cells and the abundance of stromal cells endowed with lymphocyte suppressor and tumor-growth-promoting activity. Thus, progress in the treatment of pediatric solid tumors will likely be influenced by the ability to modify the TME while delivering novel, more effective therapeutic agents. In this review, we will describe the TME composition in pediatric solid tumors and illustrate recent advances in treatment for the modulation of immune cells belonging to the TME.
Collapse
Affiliation(s)
- Cristina Belgiovine
- Dipartimento di Scienze Clinico-Chirurgiche, Diagnostiche e Pediatriche, University of Pavia, 27100 Pavia, Italy
- SC Chirurgia Pediatrica, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Kristiana Mebelli
- SC Chirurgia Pediatrica, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Alessandro Raffaele
- SC Chirurgia Pediatrica, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Marica De Cicco
- SSD Cell Factory e Center for Advanced Therapies, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Jessica Rotella
- SC Pediatric Hematology/Oncology, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Paolo Pedrazzoli
- Medical Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
| | - Marco Zecca
- SC Pediatric Hematology/Oncology, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Giovanna Riccipetitoni
- Dipartimento di Scienze Clinico-Chirurgiche, Diagnostiche e Pediatriche, University of Pavia, 27100 Pavia, Italy
- SC Chirurgia Pediatrica, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Patrizia Comoli
- SSD Cell Factory e Center for Advanced Therapies, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
9
|
Wang P, Zhu J, Long Q, Wang Y, Xu H, Tao H, Wu B, Li J, Wu Y, Liu S. LncRNA SATB2-AS1 promotes tumor growth and metastasis and affects the tumor immune microenvironment in osteosarcoma by regulating SATB2. J Bone Oncol 2023; 41:100491. [PMID: 37601080 PMCID: PMC10436287 DOI: 10.1016/j.jbo.2023.100491] [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: 02/05/2023] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 08/22/2023] Open
Abstract
Our previous report has identified a lncRNA SATB2-AS1, which was significantly up-regulated in osteosarcoma tissue and promotes the proliferation of osteosarcoma cells in vitro. However, the mechanisms of SATB2-AS1 regulating the growth and metastasis of osteosarcoma cells in vivo and its role in the prognosis of osteosarcoma patients are still unclear. In this study, the transcriptome sequencing data of 87 patients with osteosarcoma from the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database and 7 patients from our clinical center (GZFPH) was used to evaluate the importance of SATB2-AS1 and SATB2 on the prognosis. The effect of SATB2-AS1 on the growth and metastasis of osteosarcoma cells in vivo was verified by a mouse tumor model. The potential mechanisms of SATB2-AS1 regulating SATB2 were further explored by dual-luciferase reporter gene assay, RNA pull-down assay, and bioinformatics analysis. The results suggested that increased co-expression of SATB2-AS1 and SATB2 was significantly associated with poor overall survival (OS) and relapse-free survival (RFS), and was a biomarker for risk stratification in patients with osteosarcoma. Mechanistically, SATB2-AS1 promotes tumor growth and lung metastasis by regulating SATB2 in vivo. SATB2-AS1 directly binds to POU3F1 for mediating SATB2 expression in MNNG/HOS cells. In addition, SATB2-AS1 and SATB2 might be potential immunomodulators for negatively affecting immune cell infiltration by the IL-17 signaling pathway. In summary, SATB2-AS1 promoted tumor cell growth and lung metastasis by activating SATB2, thereby associated with poor prognosis in patients with osteosarcoma, which indicated that SATB2-AS1 and SATB2 might be novel biomarkers for risk stratification and promising therapeutic targets for osteosarcoma.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Oncology, the Second Affiliated Hospital, and School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong 510180, PR China
- Department of Oncology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China
- Guangzhou First People’s Hospital, Guangzhou, Guangdong 510180, PR China
| | - Jianwei Zhu
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China
- Guangzhou First People’s Hospital, Guangzhou, Guangdong 510180, PR China
| | - Qingqin Long
- Department of Oncology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China
- Guangzhou First People’s Hospital, Guangzhou, Guangdong 510180, PR China
| | - Yan Wang
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China
- Guangzhou First People’s Hospital, Guangzhou, Guangdong 510180, PR China
| | - Huihua Xu
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China
- Guangzhou First People’s Hospital, Guangzhou, Guangdong 510180, PR China
| | - Huimin Tao
- Department of Oncology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China
- Guangzhou First People’s Hospital, Guangzhou, Guangdong 510180, PR China
| | - Biwen Wu
- Department of Oncology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China
- Guangzhou First People’s Hospital, Guangzhou, Guangdong 510180, PR China
| | - Jiajun Li
- Department of Oncology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China
- Guangzhou First People’s Hospital, Guangzhou, Guangdong 510180, PR China
| | - Yong Wu
- Department of Oncology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China
- Guangzhou First People’s Hospital, Guangzhou, Guangdong 510180, PR China
| | - Sihong Liu
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, PR China
- Guangzhou First People’s Hospital, Guangzhou, Guangdong 510180, PR China
| |
Collapse
|
10
|
Todosenko N, Khlusov I, Yurova K, Khaziakhmatova O, Litvinova L. Signal Pathways and microRNAs in Osteosarcoma Growth and the Dual Role of Mesenchymal Stem Cells in Oncogenesis. Int J Mol Sci 2023; 24:ijms24108993. [PMID: 37240338 DOI: 10.3390/ijms24108993] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
The major challenges in Osteosarcoma (OS) therapy are its heterogeneity and drug resistance. The development of new therapeutic approaches to overcome the major growth mechanisms of OS is urgently needed. The search for specific molecular targets and promising innovative approaches in OS therapy, including drug delivery methods, is an urgent problem. Modern regenerative medicine focuses on harnessing the potential of mesenchymal stem cells (MSCs) because they have low immunogenicity. MSCs are important cells that have received considerable attention in cancer research. Currently, new cell-based methods for using MSCs in medicine are being actively investigated and tested, especially as carriers for chemotherapeutics, nanoparticles, and photosensitizers. However, despite the inexhaustible regenerative potential and known anticancer properties of MSCs, they may trigger the development and progression of bone tumors. A better understanding of the complex cellular and molecular mechanisms of OS pathogenesis is essential to identify novel molecular effectors involved in oncogenesis. The current review focuses on signaling pathways and miRNAs involved in the development of OS and describes the role of MSCs in oncogenesis and their potential for antitumor cell-based therapy.
Collapse
Affiliation(s)
- Natalia Todosenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Igor Khlusov
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
- Laboratory of Cellular and Microfluidic Technologies, Siberian State Medical University, 2, Moskovskii Trakt, 634050 Tomsk, Russia
| | - Kristina Yurova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Olga Khaziakhmatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Larisa Litvinova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
- Laboratory of Cellular and Microfluidic Technologies, Siberian State Medical University, 2, Moskovskii Trakt, 634050 Tomsk, Russia
| |
Collapse
|
11
|
Morales-Molina A, Rodriguez-Milla MÁ, Gambera S, Cejalvo T, de Andrés B, Gaspar ML, García-Castro J. Toll-like Receptor Signaling-deficient Cells Enhance Antitumor Activity of Cell-based Immunotherapy by Increasing Tumor Homing. CANCER RESEARCH COMMUNICATIONS 2023; 3:347-360. [PMID: 36875156 PMCID: PMC9976589 DOI: 10.1158/2767-9764.crc-22-0365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/29/2022] [Accepted: 02/06/2023] [Indexed: 02/11/2023]
Abstract
Cancer immunotherapy aims to activate the immune system. Some immunotherapeutic agents can be loaded in carrier cells for delivering to the tumors. However, a challenge with cell-based therapies is the selection of the appropriate cells to produce effective clinical outcomes. We hypothesize that therapies based on cells presenting a natural low proinflammatory profile ("silent cells") in the peripheral blood would result in better antitumor responses by increasing their homing to the tumor site. We studied our hypothesis in an immunotherapy model consisting of mesenchymal stromal cells (MSCs) carrying oncolytic adenoviruses for the treatment of immunocompetent mice. Toll-like receptor signaling-deficient cells (TLR4, TLR9, or MyD88 knockout) were used as "silent cells," while regular MSCs were used as control. Although in vitro migration was similar in regular and knockout carrier cells, in vivo tumor homing of silent cells was significantly higher after systemic administration. This better homing to the tumor site was highly related to the mild immune response triggered by these silent cells in peripheral blood. As a result, the use of silent cells significantly improved the antitumor efficacy of the treatment in comparison with the use of regular MSCs. While cancer immunotherapies generally aim to boost local immune responses in the tumor microenvironment, low systemic inflammation after systemic administration of the treatment may indeed enhance their tumor homing and improve the overall antitumor effect. These findings highlight the importance of selecting appropriate donor cells as therapeutic carriers in cell-based therapies for cancer treatment. Significance Cells carrying drugs, virus, or other antitumor agents are commonly used for the treatment of cancer. This research shows that silent cells are excellent carriers for immunotherapies, improving tumor homing and enhancing the antitumor effect.
Collapse
Affiliation(s)
- Alvaro Morales-Molina
- Cellular Biotechnology Unit, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Miguel Ángel Rodriguez-Milla
- Cellular Biotechnology Unit, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Stefano Gambera
- Cellular Biotechnology Unit, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Teresa Cejalvo
- Cellular Biotechnology Unit, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Biological Products, Advanced Therapies and Biotechnology, Department of Medicines for Human Use, AEMPS, Madrid, Spain
| | - Belén de Andrés
- Immunology Laboratory, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Spain
| | - María-Luisa Gaspar
- Immunology Laboratory, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Spain
| | - Javier García-Castro
- Cellular Biotechnology Unit, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
12
|
Antuamwine BB, Bosnjakovic R, Hofmann-Vega F, Wang X, Theodosiou T, Iliopoulos I, Brandau S. N1 versus N2 and PMN-MDSC: A critical appraisal of current concepts on tumor-associated neutrophils and new directions for human oncology. Immunol Rev 2022; 314:250-279. [PMID: 36504274 DOI: 10.1111/imr.13176] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Research on tumor-associated neutrophils (TAN) currently surges because of the well-documented strong clinical relevance of tumor-infiltrating neutrophils. This relevance is illustrated by strong correlations between high frequencies of intratumoral neutrophils and poor outcome in the majority of human cancers. Recent high-dimensional analysis of murine neutrophils provides evidence for unexpected plasticity of neutrophils in murine models of cancer and other inflammatory non-malignant diseases. New analysis tools enable deeper insight into the process of neutrophil differentiation and maturation. These technological and scientific developments led to the description of an ever-increasing number of distinct transcriptional states and associated phenotypes in murine models of disease and more recently also in humans. At present, functional validation of these different transcriptional states and potential phenotypes in cancer is lacking. Current functional concepts on neutrophils in cancer rely mainly on the myeloid-derived suppressor cell (MDSC) concept and the dichotomous and simple N1-N2 paradigm. In this manuscript, we review the historic development of those concepts, critically evaluate these concepts against the background of our own work and provide suggestions for a refinement of current concepts in order to facilitate the transition of TAN research from experimental insight to clinical translation.
Collapse
Affiliation(s)
- Benedict Boateng Antuamwine
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Rebeka Bosnjakovic
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Francisca Hofmann-Vega
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Xi Wang
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Theodosios Theodosiou
- Department of Basic Sciences, School of Medicine, University of Crete, Heraklion, Greece
| | - Ioannis Iliopoulos
- Department of Basic Sciences, School of Medicine, University of Crete, Heraklion, Greece
| | - Sven Brandau
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany.,German Cancer Consortium, Partner Site Essen-Düsseldorf, Essen, Germany
| |
Collapse
|
13
|
Herrador-Cañete G, Zalacain M, Labiano S, Laspidea V, Puigdelloses M, Marrodan L, Garcia-Moure M, Gonzalez-Huarriz M, Marco-Sanz J, Ausejo-Mauleon I, de la Nava D, Hernández-Osuna R, Martínez-García J, Silva-Pilipich N, Gurucega E, Patiño-García A, Hernández-Alcoceba R, Smerdou C, Alonso MM. Galectin-3 inhibition boosts the therapeutic efficacy of Semliki Forest virus in pediatric osteosarcoma. Mol Ther Oncolytics 2022; 26:246-264. [PMID: 35949950 PMCID: PMC9345771 DOI: 10.1016/j.omto.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/07/2022] [Indexed: 12/04/2022] Open
Abstract
The outcomes of metastatic and nonresponder pediatric osteosarcoma patients are very poor and have not improved in the last 30 years. These tumors harbor a highly immunosuppressive environment, making existing immunotherapies ineffective. Here, we evaluated the use of Semliki Forest virus (SFV) vectors expressing galectin-3 (Gal3) inhibitors as therapeutic tools, since both the inhibition of Gal3, which is involved in immunosuppression and metastasis, and virotherapy based on SFV have been demonstrated to reduce tumor progression in different tumor models. In vitro, inhibitors based on the Gal3 amino-terminal domain alone (Gal3-N) or fused to a Gal3 peptide inhibitor (Gal3-N-C12) were able to block the binding of Gal3 to the surface of activated T cells. In vivo, SFV expressing Gal3-N-C12 induced strong antitumor responses in orthotopic K7M2 and MOS-J osteosarcoma tumors, leading to complete regressions in 47% and 30% of mice, respectively. Pulmonary metastases were also reduced in K7M2 tumor-bearing mice after treatment with SFV-Gal3-N-C12. Both the antitumor and antimetastatic responses were dependent on modulation of the immune system, primarily including an increase in tumor-infiltrating lymphocytes and a reduction in the immunosuppressive environment inside tumors. Our results demonstrated that SFV-Gal3-N-C12 could constitute a potential therapeutic agent for osteosarcoma patients expressing Gal3.
Collapse
Affiliation(s)
- Guillermo Herrador-Cañete
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Gene Therapy and Regulation of Gene Expression Program, Cima Universidad de Navarra, Pamplona 31008, Spain
| | - Marta Zalacain
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Sara Labiano
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Virginia Laspidea
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Montserrat Puigdelloses
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Lucía Marrodan
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Marc Garcia-Moure
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Marisol Gonzalez-Huarriz
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Javier Marco-Sanz
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Iker Ausejo-Mauleon
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Daniel de la Nava
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Reyes Hernández-Osuna
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Javier Martínez-García
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Gene Therapy and Regulation of Gene Expression Program, Cima Universidad de Navarra, Pamplona 31008, Spain
| | - Noelia Silva-Pilipich
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Gene Therapy and Regulation of Gene Expression Program, Cima Universidad de Navarra, Pamplona 31008, Spain
| | - Elisabeth Gurucega
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Bioinformatics Platform, Cima Universidad de Navarra, Pamplona 31008, Spain
| | - Ana Patiño-García
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Rubén Hernández-Alcoceba
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Gene Therapy and Regulation of Gene Expression Program, Cima Universidad de Navarra, Pamplona 31008, Spain
| | - Cristian Smerdou
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Gene Therapy and Regulation of Gene Expression Program, Cima Universidad de Navarra, Pamplona 31008, Spain
| | - Marta M Alonso
- Health Research Institute of Navarra (IdiSNA), Pamplona 31008, Spain.,Solid Tumor Program, Cima Universidad de Navarra, Pamplona 31008, Spain.,Department of Pediatrics, Clínica Universidad de Navarra, Pamplona 31008, Spain
| |
Collapse
|
14
|
Wilson-Robles HM, Bygott T, Kelly TK, Miller TM, Miller P, Matsushita M, Terrell J, Bougoussa M, Butera T. Evaluation of plasma nucleosome concentrations in dogs with a variety of common cancers and in healthy dogs. BMC Vet Res 2022; 18:329. [PMID: 36045415 PMCID: PMC9429572 DOI: 10.1186/s12917-022-03429-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 08/19/2022] [Indexed: 01/02/2023] Open
Abstract
Background Cell free DNA, in the form of nucleosomes, is released into circulation during apoptosis and necrosis in a variety of diseases. They are small fragments of chromosomes that are composed of DNA wrapped around a histone core made of four duplicate histone proteins forming an octamer. The nucleosome compartment is a relatively uninvestigated area of circulating tumor biomarkers in dogs. The objectives of this study were to quantify and better characterize nucleosome concentrations in 528 dogs with various common malignancies and compare them to 134 healthy dogs. Results The sensitivity of increased circulating nucleosome concentrations for the detection of cancer in all dogs was 49.8% with a specificity of 97% with an area under the curve of 68.74%. The top 4 malignancies detected by the test included lymphoma, hemangiosarcoma, histiocytic sarcoma and malignant melanoma. The malignancies least likely to be detected were soft tissue sarcomas, osteosarcoma and mast cell tumors. Conclusions A variety of tumor types may cause increased nucleosome concentrations in dogs. Tumors of hematopoietic origin are most likely to cause elevations and local tumors such as soft tissue sarcomas are least likely to cause elevations in plasma nucleosome concentrations.
Collapse
|
15
|
Zhu T, Han J, Yang L, Cai Z, Sun W, Hua Y, Xu J. Immune Microenvironment in Osteosarcoma: Components, Therapeutic Strategies and Clinical Applications. Front Immunol 2022; 13:907550. [PMID: 35720360 PMCID: PMC9198725 DOI: 10.3389/fimmu.2022.907550] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/26/2022] [Indexed: 12/21/2022] Open
Abstract
Osteosarcoma is a primary malignant tumor that tends to threaten children and adolescents, and the 5-year event-free survival rate has not improved significantly in the past three decades, bringing grief and economic burden to patients and society. To date, the genetic background and oncogenesis mechanisms of osteosarcoma remain unclear, impeding further research. The tumor immune microenvironment has become a recent research hot spot, providing novel but valuable insight into tumor heterogeneity and multifaceted mechanisms of tumor progression and metastasis. However, the immune microenvironment in osteosarcoma has been vigorously discussed, and the landscape of immune and non-immune component infiltration has been intensively investigated. Here, we summarize the current knowledge of the classification, features, and functions of the main infiltrating cells, complement system, and exosomes in the osteosarcoma immune microenvironment. In each section, we also highlight the complex crosstalk network among them and the corresponding potential therapeutic strategies and clinical applications to deepen our understanding of osteosarcoma and provide a reference for imminent effective therapies with reduced adverse effects.
Collapse
Affiliation(s)
- Tianyi Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Jing Han
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Liu Yang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Zhengdong Cai
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Wei Sun
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Yingqi Hua
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Jing Xu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| |
Collapse
|
16
|
Maneta E, Fultang L, Taylor J, Pugh M, Jenkinson W, Anderson G, Coomarasamy A, Kilby MD, Lissauer DM, Mussai F, De Santo C. G-CSF induces CD15 + CD14 + cells from granulocytes early in the physiological environment of pregnancy and the cancer immunosuppressive microenvironment. Clin Transl Immunology 2022; 11:e1395. [PMID: 35602884 PMCID: PMC9114661 DOI: 10.1002/cti2.1395] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/21/2022] [Accepted: 05/03/2022] [Indexed: 01/04/2023] Open
Abstract
Objectives Recombinant granulocyte colony-stimulating factor (G-CSF) is frequently administered to patients with cancer to enhance granulocyte recovery post-chemotherapy. Clinical trials have also used G-CSF to modulate myeloid cell function in pregnancy and inflammatory diseases. Although the contribution of G-CSF to expanding normal granulocytes is well known, the effect of this cytokine on the phenotype and function of immunosuppressive granulocytic cells remains unclear. Here, we investigate the impact of physiological and iatrogenic G-CSF on an as yet undescribed granulocyte phenotype and ensuing outcome on T cells in the settings of cancer and pregnancy. Methods Granulocytes from patients treated with recombinant G-CSF, patients with late-stage cancer and women enrolled on a trial of recombinant G-CSF were phenotyped by flow cytometry. The ability and mechanism of polarised granulocytes to suppress T-cell proliferation were assessed by cell proliferation assays, flow cytometry and ELISA. Results We observed that G-CSF leads to a significant upregulation of CD14 expression on CD15+ granulocytes. These CD15+CD14+ cells are identified in the blood of patients with patients undergoing neutrophil mobilisation with recombinant G-CSF, and physiologically in women early in pregnancy or in those treated as a part of a clinical trial. Immunohistochemistry of tumor tissue or placental tissue identified the expression of G-CSF. The G-CSF upregulates the release of reactive oxygen species (ROS) in CD15+CD14+ cells leading to the suppression of T-cell proliferation. Conclusions G-CSF induces a population of ROS+ immunosuppressive CD15+CD14+ granulocytes. Strategies for how recombinant G-CSF can be scheduled to reduce effects on T-cell therapies should be developed in future clinical studies.
Collapse
Affiliation(s)
- Ebtehag Maneta
- Institute of Immunology and Immunotherapy University of Birmingham Birmingham UK
| | - Livingstone Fultang
- Institute of Immunology and Immunotherapy University of Birmingham Birmingham UK
| | - Jemma Taylor
- Institute of Immunology and Immunotherapy University of Birmingham Birmingham UK
| | - Matthew Pugh
- Institute of Immunology and Immunotherapy University of Birmingham Birmingham UK
| | - William Jenkinson
- Institute of Immunology and Immunotherapy University of Birmingham Birmingham UK
| | - Graham Anderson
- Institute of Immunology and Immunotherapy University of Birmingham Birmingham UK
| | - Arri Coomarasamy
- Tommy's National Centre for Miscarriage Research Institute of Metabolism and Systems Research University of Birmingham Birmingham UK
| | - Mark D Kilby
- Institute of Metabolism and Systems Research University of Birmingham Birmingham UK.,Fetal Medicine Centre Birmingham Women's & Children's Foundation Trust Edgbaston, Birmingham UK
| | - David M Lissauer
- Institute of Life Course and Medical Sciences University of Liverpool Liverpool UK.,Malawi-Liverpool-Wellcome Trust Blantyre Malawi
| | - Francis Mussai
- Institute of Immunology and Immunotherapy University of Birmingham Birmingham UK
| | - Carmela De Santo
- Institute of Immunology and Immunotherapy University of Birmingham Birmingham UK
| |
Collapse
|
17
|
de la Nava D, Selvi KM, Alonso MM. Immunovirotherapy for Pediatric Solid Tumors: A Promising Treatment That is Becoming a Reality. Front Immunol 2022; 13:866892. [PMID: 35493490 PMCID: PMC9043602 DOI: 10.3389/fimmu.2022.866892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy has seen tremendous strides in the last decade, acquiring a prominent position at the forefront of cancer treatment since it has been proven to be efficacious for a wide variety of tumors. Nevertheless, while immunotherapy has changed the paradigm of adult tumor treatment, this progress has not yet been translated to the pediatric solid tumor population. For this reason, alternative curative therapies are urgently needed for the most aggressive pediatric tumors. In recent years, oncolytic virotherapy has consolidated as a feasible strategy for cancer treatment, not only for its tumor-specific effects and safety profile but also for its capacity to trigger an antitumor immune response. This review will summarize the current status of immunovirotherapy to treat cancer, focusing on pediatric solid malignancies. We will revisit previous basic, translational, and clinical research and discuss advances in overcoming the existing barriers and limitations to translate this promising therapeutic as an every-day cancer treatment for the pediatric and young adult populations.
Collapse
Affiliation(s)
- Daniel de la Nava
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
- Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Kadir Mert Selvi
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
- Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Marta M. Alonso
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
- Programs in Solid Tumors and Neuroscience, Foundation for the Applied Medical Research, Pamplona, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
18
|
Hu H, Deng X, Song Q, Yang W, Zhang Y, Liu W, Wang S, Liang Z, Xing X, Zhu J, Zhang J, Shao Z, Wang B, Zhang Y. Mitochondria-targeted accumulation of oxygen-irrelevant free radicals for enhanced synergistic low-temperature photothermal and thermodynamic therapy. J Nanobiotechnology 2021; 19:390. [PMID: 34823543 PMCID: PMC8620660 DOI: 10.1186/s12951-021-01142-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/13/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Although lower temperature (< 45 °C) photothermal therapy (LPTT) have attracted enormous attention in cancer therapy, the therapeutic effect is still unsatisfying when applying LPTT alone. Therefore, combining with other therapies is urgently needed to improve the therapeutic effect of LPTT. Recently reported oxygen-irrelevant free radicals based thermodynamic therapy (TDT) exhibit promising potential for hypoxic tumor treatment. However, overexpression of glutathione (GSH) in cancer cells would potently scavenge the free radicals before their arrival to the specific site and dramatically diminish the therapeutic efficacy. METHODS AND RESULTS In this work, a core-shell nanoplatform with an appropriate size composed of arginine-glycine-aspartate (RGD) functioned polydopamine (PDA) as a shell and a triphenylphosphonium (TPP) modified hollow mesoporous manganese dioxide (H-mMnO2) as a core was designed and fabricated for the first time. This nanostructure endows a size-controllable hollow cavity mMnO2 and thickness-tunable PDA layers, which effectively prevented the pre-matured release of encapsulated azo initiator 2,2'-azobis[2-(2-imidazolin-2-yl) propane] dihydrochloride (AIBI) and revealed pH/NIR dual-responsive release performance. With the mitochondria-targeting ability of TPP, the smart nanocomposites (AIBI@H-mMnO2-TPP@PDA-RGD, AHTPR) could efficiently induce mitochondrial associated apoptosis in cancer cells at relatively low temperatures (< 45 °C) via selectively releasing oxygen-irrelevant free radicals in mitochondria and facilitating the depletion of intracellular GSH, exhibiting the advantages of mitochondria-targeted LPTT/TDT. More importantly, remarkable inhibition of tumor growth was observed in a subcutaneous xenograft model of osteosarcoma (OS) with negligible side effects. CONCLUSIONS The synergistic therapy efficacy was confirmed by effectively inducing cancer cell death in vitro and completely eradicating the tumors in vivo. Additionally, the excellent biosafety and biocompatibility of the nanoplatforms were confirmed both in vitro and in vivo. Taken together, the current study provides a novel paradigm toward oxygen-independent free-radical-based cancer therapy, especially for the treatment of hypoxic solid tumors.
Collapse
Affiliation(s)
- Hongzhi Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051 China
- NHC Key Laboratory of Intelligent Orthopeadic Equipment, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei China
| | - Xiangtian Deng
- School of Medicine, Nankai University, Tianjin, 300071 China
| | - Qingcheng Song
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051 China
- NHC Key Laboratory of Intelligent Orthopeadic Equipment, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei China
| | - Wenbo Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Yiran Zhang
- School of Medicine, Nankai University, Tianjin, 300071 China
| | - Weijian Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051 China
- NHC Key Laboratory of Intelligent Orthopeadic Equipment, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei China
| | - Shangyu Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Zihui Liang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-Constructed by the Province and Ministry, Hubei University, Wuhan, 430062 China
| | - Xin Xing
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051 China
- NHC Key Laboratory of Intelligent Orthopeadic Equipment, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei China
| | - Jian Zhu
- School of Medicine, Nankai University, Tianjin, 300071 China
| | - Junzhe Zhang
- School of Medicine, Nankai University, Tianjin, 300071 China
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051 China
- NHC Key Laboratory of Intelligent Orthopeadic Equipment, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Baichuan Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Yingze Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051 China
- NHC Key Laboratory of Intelligent Orthopeadic Equipment, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei China
| |
Collapse
|
19
|
Mouchemore KA, Anderson RL. Immunomodulatory effects of G-CSF in cancer: Therapeutic implications. Semin Immunol 2021; 54:101512. [PMID: 34763974 DOI: 10.1016/j.smim.2021.101512] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/23/2021] [Indexed: 01/04/2023]
Abstract
Numerous preclinical studies have reported a pro-tumour role for granulocyte colony-stimulating factor (G-CSF) that is predominantly mediated by neutrophils and MDSCs, the major G-CSF receptor expressing populations. In the presence of G-CSF (either tumour-derived or exogenous) these myeloid populations commonly exhibit a T cell suppressive phenotype. However, the direct effects of this cytokine on other immune lineages, such as T and NK cells, are not as well established. Herein we discuss the most recent data relating to the effect of G-CSF on the major immune populations, exclusively in the context of cancer. Recent publications have drawn attention to the other tumour-promoting effects of G-CSF on myeloid cells, including NETosis, promotion of cancer stemness and skewed differentiation of bone marrow progenitors towards myelopoiesis. Although G-CSF is safely and commonly used as a supportive therapy to prevent or treat chemotherapy-associated neutropenia in cancer patients, we also discuss the potential impacts of G-CSF on other anti-cancer treatments. Importantly, considerations for immune checkpoint blockade are highlighted, as many publications report a T cell suppressive effect of G-CSF that may diminish the effectiveness of this immunotherapy.
Collapse
Affiliation(s)
- Kellie A Mouchemore
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Robin L Anderson
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
20
|
Sarhadi VK, Daddali R, Seppänen-Kaijansinkko R. Mesenchymal Stem Cells and Extracellular Vesicles in Osteosarcoma Pathogenesis and Therapy. Int J Mol Sci 2021; 22:11035. [PMID: 34681692 PMCID: PMC8537935 DOI: 10.3390/ijms222011035] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/01/2021] [Accepted: 10/09/2021] [Indexed: 12/29/2022] Open
Abstract
Osteosarcoma (OS) is an aggressive bone tumor that mainly affects children and adolescents. OS has a strong tendency to relapse and metastasize, resulting in poor prognosis and survival. The high heterogeneity and genetic complexity of OS make it challenging to identify new therapeutic targets. Mesenchymal stem cells (MSCs) are multipotent stem cells that can differentiate into adipocytes, osteoblasts, or chondroblasts. OS is thought to originate at some stage in the differentiation process of MSC to pre-osteoblast or from osteoblast precursors. MSCs contribute to OS progression by interacting with tumor cells via paracrine signaling and affect tumor cell proliferation, invasion, angiogenesis, immune response, and metastasis. Extracellular vesicles (EVs), secreted by OS cells and MSCs in the tumor microenvironment, are crucial mediators of intercellular communication, driving OS progression by transferring miRNAs/RNA and proteins to other cells. MSC-derived EVs have both pro-tumor and anti-tumor effects on OS progression. MSC-EVs can be also engineered to deliver anti-tumor cargo to the tumor site, which offers potential applications in MSC-EV-based OS treatment. In this review, we highlight the role of MSCs in OS, with a focus on EV-mediated communication between OS cells and MSCs and their role in OS pathogenesis and therapy.
Collapse
|