1
|
Zaffagnini M, Boffa A, Andriolo L, Raggi F, Zaffagnini S, Filardo G. Orthobiologic therapies delay the need for hip arthroplasty in patients with avascular necrosis of the femoral head: A systematic review and survival analysis. Knee Surg Sports Traumatol Arthrosc 2024. [PMID: 39543728 DOI: 10.1002/ksa.12532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Abstract
PURPOSE The aim of this systematic review and survival analysis was to quantify the benefits of orthobiologic augmentation therapies for the treatment of avascular necrosis (AVN) of the femoral head and identify the most effective approach to delay the need for total hip arthroplasty (THA). METHODS A systematic review of the literature was performed on PubMed, Scopus, and Cochrane on clinical studies on orthobiologic therapies used alone or as an augmentation to core decompression or other procedures to address hip AVN. A qualitative analysis of the different biological therapies applied was performed. Afterward, the results of these procedures were quantitatively analysed to document their survivorship from THA compared to treatment groups without orthobiologics. Kaplan-Meier analysis was performed for all studies and then by categorising orthobiologics into treatment subgroups. RESULTS A total of 106 studies were included (4505 patients). Different orthobiologic approaches have been evaluated: cell-based therapies including bone marrow aspirate concentrate (BMAC) and bone marrow mesenchymal stromal cells (BM-MSCs), platelet-rich plasma (PRP), or other bioactive molecules applied in the osteonecrotic area or as intra-arterial injections. The survival analysis at 120 months documented a higher (p < 0.0005) cumulative survivorship with orthobiologics (69.4%) compared to controls (48.5%). The superiority was shown specifically for BMAC (p < 0.0005), BM-MSCs (p < 0.0005), intra-arterial (p < 0.0005) and PRP (p = 0.011) approaches, but the direct comparison of these approaches with their controls confirmed benefits only for BMAC (p < 0.0005). CONCLUSION This systematic review and survival analysis demonstrated that orthobiologics have the potential to improve survivorship in patients affected by hip AVN. In particular, the specific analysis of different orthobiologic products supported relevant benefits for BMAC augmentation in terms of survival from the need for THA, while no clear benefits were confirmed for other orthobiologics. LEVEL OF EVIDENCE Level III.
Collapse
Affiliation(s)
- Marco Zaffagnini
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Angelo Boffa
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Luca Andriolo
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Federico Raggi
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Stefano Zaffagnini
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Giuseppe Filardo
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Surgery, EOC, Service of Orthopaedics and Traumatology, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
2
|
Zhang Y, Fan M, Zhang Y. Revolutionizing bone defect healing: the power of mesenchymal stem cells as seeds. Front Bioeng Biotechnol 2024; 12:1421674. [PMID: 39497791 PMCID: PMC11532096 DOI: 10.3389/fbioe.2024.1421674] [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: 04/22/2024] [Accepted: 10/10/2024] [Indexed: 11/07/2024] Open
Abstract
Bone defects can arise from trauma or pathological factors, resulting in compromised bone integrity and the loss or absence of bone tissue. As we are all aware, repairing bone defects is a core problem in bone tissue engineering. While minor bone defects can self-repair if the periosteum remains intact and normal osteogenesis occurs, significant defects or conditions such as congenital osteogenesis imperfecta present substantial challenges to self-healing. As research on mesenchymal stem cell (MSC) advances, new fields of application have emerged; however, their application in orthopedics remains one of the most established and clinically valuable directions. This review aims to provide a comprehensive overview of the research progress regarding MSCs in the treatment of diverse bone defects. MSCs, as multipotent stem cells, offer significant advantages due to their immunomodulatory properties and ability to undergo osteogenic differentiation. The review will encompass the characteristics of MSCs within the osteogenic microenvironment and summarize the research progress of MSCs in different types of bone defects, ranging from their fundamental characteristics and animal studies to clinical applications.
Collapse
Affiliation(s)
- Yueyao Zhang
- Trauma Emergency Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
| | - Mengke Fan
- Trauma Emergency Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
| | - Yingze Zhang
- Trauma Emergency Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
| |
Collapse
|
3
|
Park S, Rahaman KA, Kim YC, Jeon H, Han HS. Fostering tissue engineering and regenerative medicine to treat musculoskeletal disorders in bone and muscle. Bioact Mater 2024; 40:345-365. [PMID: 38978804 PMCID: PMC11228556 DOI: 10.1016/j.bioactmat.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/26/2024] [Accepted: 06/11/2024] [Indexed: 07/10/2024] Open
Abstract
The musculoskeletal system, which is vital for movement, support, and protection, can be impaired by disorders such as osteoporosis, osteoarthritis, and muscular dystrophy. This review focuses on the advances in tissue engineering and regenerative medicine, specifically aimed at alleviating these disorders. It explores the roles of cell therapy, particularly Mesenchymal Stem Cells (MSCs) and Adipose-Derived Stem Cells (ADSCs), biomaterials, and biomolecules/external stimulations in fostering bone and muscle regeneration. The current research underscores the potential of MSCs and ADSCs despite the persistent challenges of cell scarcity, inconsistent outcomes, and safety concerns. Moreover, integrating exogenous materials such as scaffolds and external stimuli like electrical stimulation and growth factors shows promise in enhancing musculoskeletal regeneration. This review emphasizes the need for comprehensive studies and adopting innovative techniques together to refine and advance these multi-therapeutic strategies, ultimately benefiting patients with musculoskeletal disorders.
Collapse
Affiliation(s)
- Soyeon Park
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Khandoker Asiqur Rahaman
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Yu-Chan Kim
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Hojeong Jeon
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Hyung-Seop Han
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| |
Collapse
|
4
|
Yoon SD, Shim BJ, Baek SH, Kim SY. Implantation of Culture-Expanded Bone Marrow Derived Mesenchymal Stromal Cells for Treatment of Osteonecrosis of the Femoral Head. Tissue Eng Regen Med 2024; 21:929-941. [PMID: 38877362 PMCID: PMC11286925 DOI: 10.1007/s13770-024-00647-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: 01/29/2024] [Revised: 04/20/2024] [Accepted: 04/24/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Although core decompression (CD) with stem cell for the treatment of osteonecrosis of the femoral head (ONFH) showed promising results in many reports, the efficacy remains uncertain. We aimed to evaluate the efficacy of CD with culture-expanded autologous bone marrow-derived mesenchymal stem cell (BM-MSC) implantation in early stage ONFH. METHODS A total of 18 patients (22 hips) with ONFH who underwent CD with culture-expanded BM-MSC implantation from September 2013 to July 2020 were retrospectively reviewed. The median age was 35.0 years [interquartile range (IQR), 28.5-42.0], and the median follow-up period was 4.0 years (IQR, 2.0-5.3). The median number of MSCs was 1.06 × 108. To evaluate radiographic and clinical outcomes, Association Research Circulation Osseous (ARCO) classifications, Japanese Investigation Committee classification, combined necrotic angle (CNA) visual analogue scale (VAS) and Harris Hip Score (HHS) were checked at each follow-up. RESULTS The preoperative stage of ONFH was ARCO 2 in 14 hips and ARCO 3a in 8 hips. The ARCO staging was maintained in 7 hips in ARCO 2 and 4 hips in ARCO 3a. The radiographic failure rate of ARCO 2 and 3a was 14.3 and 50%, respectively. Furthermore, CNA decreased to more than 20° in 6 hips (four were ARCO 2 and two were ARCO 3a).There was no significant difference in the VAS and HHS (P = 0.052 and P = 0.535, respectively). Total hip arthroplasty was performed in 4 hips. CONCLUSION CD with culture-expanded autologous BM-MSCs showed promising results for the treatment of early stage ONFH.
Collapse
Affiliation(s)
- Seong-Dae Yoon
- Department of Orthopedic Surgery, College of Medicine, Kyungpook National University, 130, Dongdeok-Ro, Jung-Gu, Daegu, 41944, South Korea
- Department of Orthopedic Surgery, Kyungpook National University Hospital, 130, Dongdeok-Ro, Jung-Gu, Daegu, 41944, South Korea
| | - Bum-Jin Shim
- Department of Orthopedic Surgery, Yeungnam University Hospital, College of Medicine, Yeungnam University, 170, Hyeonchung-ro, Nam-gu, Daegu, 42415, South Korea
| | - Seung-Hoon Baek
- Department of Orthopedic Surgery, College of Medicine, Kyungpook National University, 130, Dongdeok-Ro, Jung-Gu, Daegu, 41944, South Korea
- Department of Orthopedic Surgery, Kyungpook National University Hospital, 130, Dongdeok-Ro, Jung-Gu, Daegu, 41944, South Korea
| | - Shin-Yoon Kim
- Department of Orthopedic Surgery, College of Medicine, Kyungpook National University, 130, Dongdeok-Ro, Jung-Gu, Daegu, 41944, South Korea.
- Department of Orthopedic Surgery, Kyungpook National University Hospital, 130, Dongdeok-Ro, Jung-Gu, Daegu, 41944, South Korea.
| |
Collapse
|
5
|
Hernigou P, Homma Y, Hernigou J, Flouzat Lachaniette CH, Rouard H, Verrier S. Mesenchymal Stem Cell Therapy for Bone Repair of Human Hip Osteonecrosis with Bilateral Match-Control Evaluation: Impact of Tissue Source, Cell Count, Disease Stage, and Volume Size on 908 Hips. Cells 2024; 13:776. [PMID: 38727312 PMCID: PMC11083454 DOI: 10.3390/cells13090776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
We investigated the impact of mesenchymal stem cell (MSC) therapy on treating bilateral human hip osteonecrosis, analyzing 908 cases. This study assesses factors such as tissue source and cell count, comparing core decompression with various cell therapies. This research emphasizes bone repair according to pre-treatment conditions and the specificities of cell therapy in osteonecrosis repair, indicating a potential for improved bone repair strategies in hips without femoral head collapse. This study utilized a single-center retrospective analysis to investigate the efficacy of cellular approaches in the bone repair of osteonecrosis. It examined the impact on bone repair of tissue source (autologous bone marrow concentrate, allogeneic expanded, autologous expanded), cell quantity (from none in core decompression alone to millions in cell therapy), and osteonecrosis stage and volume. Excluding hips with femoral head collapse, it focused on patients who had bilateral hip osteonecrosis, both pre-operative and post-operative MRIs, and a follow-up of over five years. The analysis divided these patients into seven groups based on match control treatment variations in bilateral hip osteonecrosis, primarily investigating the outcomes between core decompression, washing effect, and different tissue sources of MSCs. Younger patients (<30 years) demonstrated significantly better repair volumes, particularly in stage II lesions, than older counterparts. Additionally, bone repair volume increased with the number of implanted MSCs up to 1,000,000, beyond which no additional benefits were observed. No significant difference was observed in repair outcomes between different sources of MSCs (BMAC, allogenic, or expanded cells). The study also highlighted that a 'washing effect' was beneficial, particularly for larger-volume osteonecrosis when combined with core decompression. Partial bone repair was the more frequent event observed, while total bone repair of osteonecrosis was rare. The volume and stage of osteonecrosis, alongside the number of injected cells, significantly affected treatment outcomes. In summary, this study provides comprehensive insights into the effectiveness and variables influencing the use of mesenchymal stem cells in treating human hip osteonecrosis. It emphasizes the potential of cell therapy while acknowledging the complexity and variability of results based on factors such as age, cell count, and disease stage.
Collapse
Affiliation(s)
- Philippe Hernigou
- Orthopedic Department, University Paris East, Hopital Henri Mondor, 94000 Creteil, France;
| | - Yasuhiro Homma
- Department of Orthopaedics, Faculty of Medicine, Juntendo University, Bunkyo-ku, Tokyo 113-8421, Japan;
| | - Jacques Hernigou
- Department of Orthopaedic Surgery and Traumatology, EpiCURA Baudour Hornu Ath Hospital, 7331 Hainaut, Belgium;
| | | | - Helène Rouard
- Établissement Français du Sang, University Paris East, 94000 Creteil, France;
| | - Sophie Verrier
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland;
| |
Collapse
|
6
|
Wei S, Lu C, Li S, Zhang Q, Cheng R, Pan S, Wu Q, Zhao X, Tian X, Zeng X, Liu Y. Efficacy and safety of mesenchymal stem cell-derived microvesicles in mouse inflammatory arthritis. Int Immunopharmacol 2024; 131:111845. [PMID: 38531171 DOI: 10.1016/j.intimp.2024.111845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
OBJECTIVE To determine the effective and safe intravenous doses of mesenchymal stem cells (MSCs)-derived microvesicles (MVs) and to elucidate the possible causes of death in mice receiving high-dose MVs. METHODS MVs were isolated from human MSCs by gradient centrifugation. Mice with collagen-induced arthritis were treated with different doses of intravenous MVs or MSCs. Arthritis severity, white blood cell count, and serum C-reactive protein levels were measured. To assess the safety profile of MSCs and MVs, mice were treated with different doses of MSCs and MVs, and LD50 was calculated. Mouse lungs and heart were assessed by live fluorescence imaging, histopathological measurements, and immunohistochemistry to explore the possible causes of death. Serum concentrations of cTnT, cTnI, and CK-MB were determined by ELISA. With the H9C2 cardiomyocyte cell line, cellular uptake of MVs was observed using confocal microscopy and cell toxicity was assessed by CCK-8 and flow cytometry. RESULTS Intravenous treatment with MSCs and MVs alleviated inflammatory arthritis, while high doses of MSCs and MVs were lethal. Mice receiving a maximum dose of MSCs (0.1 mL of MSCs at 109/mL) died immediately, while mice receiving a maximum dose of MVs (0.1 mL of MVs at 1012/mL) exhibited tears, drooling, tachycardia, shortness of breath, unbalanced rollover, bouncing, circular crawling, mania, and death. Some mice died after exhibiting convulsions and other symptoms. All mice died shortly after injecting the maximum dose of MSCs. Histologically, mice receiving high doses of MSCs frequently developed pulmonary embolism, while those receiving high doses of MVs died of myocardial infarction. Consistently, the serum levels of cTnT, cTnI, and CK-MB were significantly increased in the MVs-treated group (P < 0.05). The LD50 of intravenous MVs was 1.60 × 1012/kg. Further, MVs could enter the cell. High doses of MVs induced cell apoptosis, though low concentrations of MVs induced cell proliferation. CONCLUSIONS Appropriate dosages of MVs and MSCs are effective treatments for inflammatory arthritis while MVs and MSCs overdose is unsafe by causing cardiopulmonary complications.
Collapse
Affiliation(s)
- Shixiong Wei
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuai Fu Yuan, Wang Fu Jing street, Beijing 100730, China; Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - Chenyang Lu
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Sujia Li
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - Qiuping Zhang
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - Ruijuan Cheng
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - ShuYue Pan
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - QiuHong Wu
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - Xueting Zhao
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuai Fu Yuan, Wang Fu Jing street, Beijing 100730, China.
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuai Fu Yuan, Wang Fu Jing street, Beijing 100730, China.
| | - Yi Liu
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China.
| |
Collapse
|
7
|
Kuroda Y, Kawai T, Okuzu Y, Morita Y, Matsuda S. Investigational regenerative medicine for non-traumatic osteonecrosis of the femoral head: a survey of registered clinical trials. Expert Opin Investig Drugs 2024; 33:405-414. [PMID: 38431280 DOI: 10.1080/13543784.2024.2326622] [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: 11/30/2023] [Accepted: 02/28/2024] [Indexed: 03/05/2024]
Abstract
INTRODUCTION Osteonecrosis of the femoral head (ONFH) is a refractory disease requiring joint replacement in young patients. Regenerative therapies have been developed. AREAS COVERED This study surveyed clinical trials on regenerative medicine for ONFH. We extracted clinical trials on non-traumatic ONFH from the websites of five publicly available major registries (EuropeanUnion Clinical Trials Register ([EU-CTR],ClinicalTrials.gov, Chinese ClinicalTrial Registry [ChiCTR], University Hospital Medical InformationNetwork - Clinical Trial Registry [UMIN-CTR] and Australian New Zealand Clinical Trials Registry [ANZCTR]).The trials were classified into six categories based on purpose: surgical treatment, non-drug conservative treatment, conservative drug treatment, therapeutic strategy, diagnosis and pathogenesis, and regenerative therapy.) We extracted 169 clinical trials on ONFH. Of these, 37 were on regenerative medicine, including 29 on cell therapy. Surgical treatment was the most common treatment, followed by regenerative therapy.There were 9 clinical trials registered in the EU-CTR, with 5 on regenerative medicine; 79 trials registered on ClinicalTrials.gov, with 24 on regenerativemedicine; 54 trials registered in the ChiCTR, with 6 on regenerative medicine. EXPERT OPINION The focus of the joint-preserving surgery has shifted to regenerative therapy based on using cell therapy in early-stage ONFH. The global standardisation of regenerative therapy is still ongoing.
Collapse
Affiliation(s)
- Yutaka Kuroda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Toshiyuki Kawai
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Yaichiro Okuzu
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Yugo Morita
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
8
|
Cabrera-Pérez R, Ràfols-Mitjans A, Roig-Molina Á, Beltramone S, Vives J, Batlle-Morera L. Human Wharton's jelly-derived mesenchymal stromal cells promote bone formation in immunodeficient mice when administered into a bone microenvironment. J Transl Med 2023; 21:802. [PMID: 37950242 PMCID: PMC10638709 DOI: 10.1186/s12967-023-04672-9] [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: 08/02/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Wharton's Jelly (WJ) Mesenchymal Stromal Cells (MSC) have emerged as an attractive allogeneic therapy for a number of indications, except for bone-related conditions requiring new tissue formation. This may be explained by the apparent recalcitrance of MSC,WJ to differentiate into the osteogenic lineage in vitro, as opposed to permissive bone marrow (BM)-derived MSCs (MSC,BM) that readily commit to bone cells. Consequently, the actual osteogenic in vivo capacity of MSC,WJ is under discussion. METHODS We investigated how physiological bone environments affect the osteogenic commitment of recalcitrant MSCs in vitro and in vivo. To this end, MSC of BM and WJ origin were co-cultured and induced for synchronous osteogenic differentiation in vitro using transwells. For in vivo experiments, immunodeficient mice were injected intratibially with a single dose of human MSC and bone formation was evaluated after six weeks. RESULTS Co-culture of MSC,BM and MSC,WJ resulted in efficient osteogenesis in both cell types after three weeks. However, MSC,WJ failed to commit to bone cells in the absence of MSC,BM's osteogenic stimuli. In vivo studies showed successful bone formation within the medullar cavity of tibias in 62.5% of mice treated with MSC, WJ. By contrast, new formed trabeculae were only observed in 25% of MSC,BM-treated mice. Immunohistochemical staining of human COXIV revealed the persistence of the infused cells at the site of injection. Additionally, cells of human origin were also identified in the brain, heart, spleen, kidney and gonads in some animals treated with engineered MSC,WJ (eMSC,WJ). Importantly, no macroscopic histopathological alterations, ectopic bone formation or any other adverse events were detected in MSC-treated mice. CONCLUSIONS Our findings demonstrate that in physiological bone microenvironment, osteogenic commitment of MSC,WJ is comparable to that of MSC,BM, and support the use of off-the-shelf allogeneic MSC,WJ products in bone repair and bone regeneration applications.
Collapse
Affiliation(s)
- Raquel Cabrera-Pérez
- Servei de Teràpia Cel·lular i Avançada, Blood and Tissue Bank (BST), 08005, Barcelona, Catalonia, Spain
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR) and Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Catalonia, Spain
| | - Alexis Ràfols-Mitjans
- Centre for Genomic Regulation (CRG), Genomic Regulation, Stem Cells and Cancer Program, The Barcelona Institute of Science and Technology, 08003, Barcelona, Catalonia, Spain
| | - Ángela Roig-Molina
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR) and Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Catalonia, Spain
| | - Silvia Beltramone
- Centre for Genomic Regulation (CRG), Genomic Regulation, Stem Cells and Cancer Program, The Barcelona Institute of Science and Technology, 08003, Barcelona, Catalonia, Spain
| | - Joaquim Vives
- Servei de Teràpia Cel·lular i Avançada, Blood and Tissue Bank (BST), 08005, Barcelona, Catalonia, Spain.
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR) and Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Catalonia, Spain.
- Medicine Department, Universitat Autònoma de Barcelona (UAB), 08193, Barcelona, Catalonia, Spain.
| | - Laura Batlle-Morera
- Centre for Genomic Regulation (CRG), Genomic Regulation, Stem Cells and Cancer Program, The Barcelona Institute of Science and Technology, 08003, Barcelona, Catalonia, Spain.
| |
Collapse
|
9
|
Jakl V, Popp T, Haupt J, Port M, Roesler R, Wiese S, Friemert B, Rojewski MT, Schrezenmeier H. Effect of Expansion Media on Functional Characteristics of Bone Marrow-Derived Mesenchymal Stromal Cells. Cells 2023; 12:2105. [PMID: 37626914 PMCID: PMC10453497 DOI: 10.3390/cells12162105] [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: 07/19/2023] [Revised: 08/07/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The therapeutic efficacy of mesenchymal stromal cells (MSCs) has been shown to rely on their immunomodulatory and regenerative properties. In order to obtain sufficient numbers of cells for clinical applications, MSCs have to be expanded ex vivo. Expansion media with xenogeneic-free (XF) growth-promoting supplements like human platelet lysate (PL) or serum- and xenogeneic-free (SF/XF) formulations have been established as safe and efficient, and both groups provide different beneficial qualities. In this study, MSCs were expanded in XF or SF/XF media as well as in mixtures thereof. MSCs cultured in these media were analyzed for phenotypic and functional properties. MSC expansion was optimal with SF/XF conditions when PL was present. Metabolic patterns, consumption of growth factors, and secretome of MSCs differed depending on the type and concentration of supplement. The lactate per glucose yield increased along with a higher proportion of PL. Many factors in the supernatant of cultured MSCs showed distinct patterns depending on the supplement (e.g., FGF-2, TGFβ, and insulin only in PL-expanded MSC, and leptin, sCD40L PDGF-AA only in SF/XF-expanded MSC). This also resulted in changes in cell characteristics like migratory potential. These findings support current approaches where growth media may be utilized for priming MSCs for specific therapeutic applications.
Collapse
Affiliation(s)
- Viktoria Jakl
- Institute for Transfusion Medicine, University Hospital Ulm, 89081 Ulm, Germany; (V.J.)
| | - Tanja Popp
- Bundeswehr Institute of Radiobiology, 80937 Munich, Germany (J.H.); (M.P.)
| | - Julian Haupt
- Bundeswehr Institute of Radiobiology, 80937 Munich, Germany (J.H.); (M.P.)
- Clinic for Trauma Surgery and Orthopedics, Army Hospital Ulm, 89081 Ulm, Germany
| | - Matthias Port
- Bundeswehr Institute of Radiobiology, 80937 Munich, Germany (J.H.); (M.P.)
| | - Reinhild Roesler
- Core Unit of Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081 Ulm, Germany; (R.R.); (S.W.)
| | - Sebastian Wiese
- Core Unit of Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081 Ulm, Germany; (R.R.); (S.W.)
| | - Benedikt Friemert
- Clinic for Trauma Surgery and Orthopedics, Army Hospital Ulm, 89081 Ulm, Germany
| | - Markus T. Rojewski
- Institute for Transfusion Medicine, University Hospital Ulm, 89081 Ulm, Germany; (V.J.)
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, 89081 Ulm, Germany
| | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, University Hospital Ulm, 89081 Ulm, Germany; (V.J.)
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, 89081 Ulm, Germany
| |
Collapse
|
10
|
Xu H, Wang W, Liu X, Huang W, Zhu C, Xu Y, Yang H, Bai J, Geng D. Targeting strategies for bone diseases: signaling pathways and clinical studies. Signal Transduct Target Ther 2023; 8:202. [PMID: 37198232 DOI: 10.1038/s41392-023-01467-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/02/2023] [Accepted: 04/19/2023] [Indexed: 05/19/2023] Open
Abstract
Since the proposal of Paul Ehrlich's magic bullet concept over 100 years ago, tremendous advances have occurred in targeted therapy. From the initial selective antibody, antitoxin to targeted drug delivery that emerged in the past decades, more precise therapeutic efficacy is realized in specific pathological sites of clinical diseases. As a highly pyknotic mineralized tissue with lessened blood flow, bone is characterized by a complex remodeling and homeostatic regulation mechanism, which makes drug therapy for skeletal diseases more challenging than other tissues. Bone-targeted therapy has been considered a promising therapeutic approach for handling such drawbacks. With the deepening understanding of bone biology, improvements in some established bone-targeted drugs and novel therapeutic targets for drugs and deliveries have emerged on the horizon. In this review, we provide a panoramic summary of recent advances in therapeutic strategies based on bone targeting. We highlight targeting strategies based on bone structure and remodeling biology. For bone-targeted therapeutic agents, in addition to improvements of the classic denosumab, romosozumab, and PTH1R ligands, potential regulation of the remodeling process targeting other key membrane expressions, cellular crosstalk, and gene expression, of all bone cells has been exploited. For bone-targeted drug delivery, different delivery strategies targeting bone matrix, bone marrow, and specific bone cells are summarized with a comparison between different targeting ligands. Ultimately, this review will summarize recent advances in the clinical translation of bone-targeted therapies and provide a perspective on the challenges for the application of bone-targeted therapy in the clinic and future trends in this area.
Collapse
Affiliation(s)
- Hao Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Wentao Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Xin Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Chen Zhu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
11
|
Blanco JF, Garcia-Garcia FJ, Villarón EM, da Casa C, Fidalgo H, López-Parra M, Santos JA, Sánchez-Guijo F. Long-Term Results of a Phase I/II Clinical Trial of Autologous Mesenchymal Stem Cell Therapy for Femoral Head Osteonecrosis. J Clin Med 2023; 12:jcm12062117. [PMID: 36983120 PMCID: PMC10051457 DOI: 10.3390/jcm12062117] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/08/2023] [Accepted: 03/01/2023] [Indexed: 03/30/2023] Open
Abstract
(1) Background: Osteonecrosis of the femoral head (ONFH) is characterized by impaired vascularization with ischemia resulting in bone cell death, leading to the deterioration of the hip joint. Mesenchymal stem/stromal cells (MSCs) are an attractive potential therapeutic approach in this setting. The aim of this study is to evaluate the clinical improvement in terms of pain and quality of life, as well as the safety of the procedure during the follow-up of patients. (2) Methods: A Phase I-II Open-Label Non-Randomized Prospective clinical trial was conducted. Eight patients with idiopathic ONFH and stage < IIC in the ARCO classification were included. Four weeks before therapy, 40 mL of autologous bone marrow was obtained, and MSCs were expanded under Good-Manufacturing-Practice (GMP) standards. Study medication consisted of a suspension of autologous BM-derived MSCs (suspended in a solution of 5-10 mL of saline and 5% human albumin) in a single dose of 0.5-1 × 106 cells/kg of the patient, administered intraosseously with a trocar and under radioscopic control. Per-protocol monitoring of patients included a postoperative period of 12 months, with a clinical and radiological assessment that included the visual analog scale (VAS), the Harris scale, the SF-36, and the radiological evolution of both hips. In addition, all patients were further followed up for eight years to assess the need for long-term total hip replacement (THR) surgery. (3) Results: Median age of patients included was 48.38 ± 7.38 years, and all patients were men. Autologous MSCs were expanded in all cases. There were no adverse effects related to cell administration. Regarding efficacy, both VAS and ODI scores improved after surgery. Radiologically, 12.5% of patients improved at the end of follow-up, whereas 50% improved clinically. No adverse effects related to the procedure were recorded, and none of the patients needed THR surgery within the first year after MSC therapy. (4) Conclusions: The use of autologous MSCs for patients with ONFH disease is feasible, safe in the long term, and potentially effective.
Collapse
Affiliation(s)
- Juan F Blanco
- Orthopaedic Surgery and Traumatology Department, University Hospital of Salamanca, 37007 Salamanca, Spain
- Biomedical Research Institute of Salamanca (IBSAL), 37007 Salamanca, Spain
- Regenerative Medicine and Cell Therapy Network Center of Castilla y Leon, Gerencia Regional de Salud, 47011 Valladolid, Spain
- Health Outcomes-Oriented Cooperative Research Networks in Advanced Therapies (RICORS TERAV), Instituto de Salud Carlos III, 28220 Madrid, Spain
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Francisco J Garcia-Garcia
- Orthopaedic Surgery and Traumatology Department, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Eva M Villarón
- Biomedical Research Institute of Salamanca (IBSAL), 37007 Salamanca, Spain
- Regenerative Medicine and Cell Therapy Network Center of Castilla y Leon, Gerencia Regional de Salud, 47011 Valladolid, Spain
- Health Outcomes-Oriented Cooperative Research Networks in Advanced Therapies (RICORS TERAV), Instituto de Salud Carlos III, 28220 Madrid, Spain
- Cell Therapy Unit, Hematology Department, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Carmen da Casa
- Biomedical Research Institute of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Helena Fidalgo
- Biomedical Research Institute of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Miriam López-Parra
- Biomedical Research Institute of Salamanca (IBSAL), 37007 Salamanca, Spain
- Regenerative Medicine and Cell Therapy Network Center of Castilla y Leon, Gerencia Regional de Salud, 47011 Valladolid, Spain
- Health Outcomes-Oriented Cooperative Research Networks in Advanced Therapies (RICORS TERAV), Instituto de Salud Carlos III, 28220 Madrid, Spain
- Cell Therapy Unit, Hematology Department, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - José A Santos
- Biomedical Research Institute of Salamanca (IBSAL), 37007 Salamanca, Spain
- Radiology Department, University Hospital of Salamanca, 37007 Salamanca, Spain
- Department of Biomedical Sciences and Diagnostics, University of Salamanca, 37007 Salamanca, Spain
| | - Fermín Sánchez-Guijo
- Biomedical Research Institute of Salamanca (IBSAL), 37007 Salamanca, Spain
- Regenerative Medicine and Cell Therapy Network Center of Castilla y Leon, Gerencia Regional de Salud, 47011 Valladolid, Spain
- Health Outcomes-Oriented Cooperative Research Networks in Advanced Therapies (RICORS TERAV), Instituto de Salud Carlos III, 28220 Madrid, Spain
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Cell Therapy Unit, Hematology Department, University Hospital of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
12
|
Jakl V, Ehmele M, Winkelmann M, Ehrenberg S, Eiseler T, Friemert B, Rojewski MT, Schrezenmeier H. A novel approach for large-scale manufacturing of small extracellular vesicles from bone marrow-derived mesenchymal stromal cells using a hollow fiber bioreactor. Front Bioeng Biotechnol 2023; 11:1107055. [PMID: 36761296 PMCID: PMC9904364 DOI: 10.3389/fbioe.2023.1107055] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/09/2023] [Indexed: 01/25/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are promising therapeutic candidates in a variety of diseases due to having immunomodulatory and pro-regenerative properties. In recent years, MSC-derived small extracellular vesicles (sEVs) have attracted increasing interest as a possible alternative to conventional cell therapy. However, translational processes of sEVs for clinical applications are still impeded by inconsistencies regarding isolation procedures and culture conditions. We systematically compared different methods for sEV isolation from conditioned media of ex vivo expanded bone marrow-derived MSCs and demonstrated considerable variability of quantity, purity, and characteristics of sEV preparations obtained by these methods. The combination of cross flow filtration with ultracentrifugation for sEV isolation resulted in sEVs with similar properties as compared to isolation by differential centrifugation combined with ultracentrifugation, the latter is still considered as gold standard for sEV isolation. In contrast, sEV isolation by a combination of precipitation with polyethylene glycol and ultracentrifugation as well as cross flow filtration and size exclusion chromatography resulted in sEVs with different characteristics, as shown by surface antigen expression patterns. The MSC culture requires a growth-promoting supplement, such as platelet lysate, which contains sEVs itself. We demonstrated that MSC culture with EV-depleted platelet lysate does not alter MSC characteristics, and conditioned media of such MSC cultures provide sEV preparations enriched for MSC-derived sEVs. The results from the systematic stepwise evaluation of various aspects were combined with culture of MSCs in a hollow fiber bioreactor. This resulted in a strategy using cross flow filtration with subsequent ultracentrifugation for sEV isolation. In conclusion, this workflow provides a semi-automated, efficient, large-scale-applicable, and good manufacturing practice (GMP)-grade approach for the generation of sEVs for clinical use. The use of EV-depleted platelet lysate is an option to further increase the purity of MSC-derived sEVs.
Collapse
Affiliation(s)
- Viktoria Jakl
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany
| | - Melanie Ehmele
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, Ulm, Germany
| | - Martina Winkelmann
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany
| | - Simon Ehrenberg
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany
| | - Tim Eiseler
- Clinic of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Benedikt Friemert
- Clinic for Trauma Surgery and Orthopedics, Army Hospital Ulm, Ulm, Germany
| | - Markus Thomas Rojewski
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, Ulm, Germany
| | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, Ulm, Germany
| |
Collapse
|
13
|
Burns JS, Kassem M. Identifying Biomarkers for Osteogenic Potency Assay Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1420:39-58. [PMID: 37258783 DOI: 10.1007/978-3-031-30040-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
There has been extensive exploration of how cells may serve as advanced therapy medicinal products to treat skeletal pathologies. Osteoblast progenitors responsible for production of extracellular matrix that is subsequently mineralized during bone formation have been characterised as a rare bone marrow subpopulation of cell culture plastic adherent cells. Conveniently, they proliferate to form single-cell derived colonies of fibroblastoid cells, termed colony forming unit fibroblasts that can subsequently differentiate to aggregates resembling small areas of cartilage or bone. However, donor heterogeneity and loss of osteogenic differentiation capacity during extended cell culture have made the discovery of reliable potency assay biomarkers difficult. Nonetheless, functional osteoblast models derived from telomerised human bone marrow stromal cells have allowed extensive comparative analysis of gene expression, microRNA, morphological phenotypes and secreted proteins. This chapter highlights numerous insights into the molecular mechanisms underpinning osteogenic differentiation of multipotent stromal cells and bone formation, discussing aspects involved in the choice of useful biomarkers for functional attributes that can be quantitively measured in osteogenic potency assays.
Collapse
Affiliation(s)
- Jorge S Burns
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy.
| | - Moustapha Kassem
- University Hospital of Odense, University of Southern Denmark, Odense, Denmark
- Danish Stem Cell Center, University of Copenhagen, Copenhagen, Denmark
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Gómez-Barrena E, Padilla-Eguiluz NG, López-Marfil M, Ruiz de la Reina R. Volume and location of bone regeneration after autologous expanded mesenchymal stromal cells in hip osteonecrosis : a pilot study. Bone Joint Res 2022; 11:881-889. [PMID: 36464628 PMCID: PMC9792875 DOI: 10.1302/2046-3758.1112.bjr-2022-0152.r1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIMS Successful cell therapy in hip osteonecrosis (ON) may help to avoid ON progression or total hip arthroplasty (THA), but the achieved bone regeneration is unclear. The aim of this study was to evaluate amount and location of bone regeneration obtained after surgical injection of expanded autologous mesenchymal stromal cells from the bone marrow (BM-hMSCs). METHODS A total of 20 patients with small and medium-size symptomatic stage II femoral head ON treated with 140 million BM-hMSCs through percutaneous forage in the EudraCT 2012-002010-39 clinical trial were retrospectively evaluated through preoperative and postoperative (three and 12 months) MRI. Then, 3D reconstruction of the original lesion and the observed postoperative residual damage after bone regeneration were analyzed and compared per group based on treatment efficacy. RESULTS The mean preoperative lesion volume was 18.7% (SD 10.2%) of the femoral head. This reduced to 11.6% (SD 7.5%) after three months (p = 0.015) and 3.7% (SD 3%) after one year (p < 0.001). Bone regeneration in healed cases represented a mean 81.2% (SD 13.8%) of the initial lesion volume at one year. Non-healed cases (n = 1 stage progression; n = 3 THAs) still showed bone regeneration but this did not effectively decrease the ON volume. A lesion size under mean 10% (SD 6%) of the femoral head at three months predicted no ON stage progression at one year. Regeneration in the lateral femoral head (C2 under Japanese Investigation Committee (JCI) classification) and in the central and posterior regions of the head was predominant in cases without ON progression. CONCLUSION Bone regeneration was observed in osteonecrotic femoral heads three months after expanded autologous BM-hMSC injection, and the volume and location of regeneration indicated the success of the therapy.Cite this article: Bone Joint Res 2022;11(12):881-889.
Collapse
Affiliation(s)
- Enrique Gómez-Barrena
- Department of Orthopedic Surgery and Traumatology, La Paz Hospital, IdiPaz, Universidad Autónoma de Madrid, Madrid, Spain, Enrique Gómez-Barrena. E-mail:
| | - Norma-Griset Padilla-Eguiluz
- Department of Orthopedic Surgery and Traumatology, La Paz Hospital, IdiPaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta López-Marfil
- Biomedical Engineering Master program, Universidad Politécnica de Madrid, Madrid, Spain
| | | | | |
Collapse
|
15
|
Mastrolia I, Giorgini A, Murgia A, Loschi P, Petrachi T, Rasini V, Pinelli M, Pinto V, Lolli F, Chiavelli C, Grisendi G, Baschieri MC, Santis GD, Catani F, Dominici M, Veronesi E. Autologous Marrow Mesenchymal Stem Cell Driving Bone Regeneration in a Rabbit Model of Femoral Head Osteonecrosis. Pharmaceutics 2022; 14:pharmaceutics14102127. [PMID: 36297562 PMCID: PMC9610232 DOI: 10.3390/pharmaceutics14102127] [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: 07/28/2022] [Revised: 09/21/2022] [Accepted: 10/03/2022] [Indexed: 11/07/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a progressive degenerative disease that ultimately requires a total hip replacement. Mesenchymal stromal/stem cells (MSCs), particularly the ones isolated from bone marrow (BM), could be promising tools to restore bone tissue in ONFH. Here, we established a rabbit model to mimic the pathogenic features of human ONFH and to challenge an autologous MSC-based treatment. ON has been originally induced by the synergic combination of surgery and steroid administration. Autologous BM-MSCs were then implanted in the FH, aiming to restore the damaged tissue. Histological analyses confirmed bone formation in the BM-MSC treated rabbit femurs but not in the controls. In addition, the model also allowed investigations on BM-MSCs isolated before (ON-BM-MSCs) and after (ON+BM-MSCs) ON induction to dissect the impact of ON damage on MSC behavior in an affected microenvironment, accounting for those clinical approaches foreseeing MSCs generally isolated from affected patients. BM-MSCs, isolated before and after ON induction, revealed similar growth rates, immunophenotypic profiles, and differentiation abilities regardless of the ON. Our data support the use of ON+BM-MSCs as a promising autologous therapeutic tool to treat ON, paving the way for a more consolidated use into the clinical settings.
Collapse
Affiliation(s)
- Ilenia Mastrolia
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
- Correspondence:
| | - Andrea Giorgini
- Division of Orthopedics, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Alba Murgia
- Technopole of Mirandola TPM, Mirandola, 41037 Modena, Italy
| | | | | | - Valeria Rasini
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Massimo Pinelli
- Division of Plastic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Valentina Pinto
- Division of Plastic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Francesca Lolli
- Division of Plastic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Chiara Chiavelli
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Giulia Grisendi
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Maria Cristina Baschieri
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Giorgio De Santis
- Division of Plastic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Fabio Catani
- Division of Orthopedics, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
- Technopole of Mirandola TPM, Mirandola, 41037 Modena, Italy
| | - Elena Veronesi
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
- Technopole of Mirandola TPM, Mirandola, 41037 Modena, Italy
| |
Collapse
|
16
|
Castillo Mercado JS, Rojas Lievano J, Zaldivar B, Barajas C, Fierro G, González JC. Atraumatic osteonecrosis of the humeral head: pathophysiology and current concepts of evaluation and treatment. JSES REVIEWS, REPORTS, AND TECHNIQUES 2022; 2:277-284. [PMID: 37588865 PMCID: PMC10426613 DOI: 10.1016/j.xrrt.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
The humeral head is considered the second most common site for osteonecrosis to occur after the femoral head. As seen in the femoral head, the circulatory implications characteristic of this condition are attributable to the interaction between a genetic predisposition and the exposure to certain risk factors. There is no consensus regarding the pathogenesis of osteonecrosis, yet the final common pathway results in disrupted blood supply, increased intraosseous pressure, and bone death. Disease staging using radiography and magnetic resonance imaging is predictive of disease progression and can help the orthopedic surgeon to guide treatment. Although there is a myriad of treatment modalities, there is a lack of high-quality evidence to conclude what is the most appropriate treatment option for each stage of humeral head osteonecrosis. Nonoperative treatment is the preferred option in early-stage disease, and it may prevent disease progression. Nonetheless, in some cases, disease progression occurs despite nonoperative measures, and surgical treatment is required. The purpose of this article is to provide an updated review of the available evidence on risk factors, diagnosis, and treatment of atraumatic humeral head osteonecrosis.
Collapse
Affiliation(s)
| | - Jorge Rojas Lievano
- Department of Orthopedics and Traumatology, Hospital Universitario Fundacion Santa Fe de Bogota, Bogota, Colombia
| | - Brandon Zaldivar
- Department of Orthopedics and Traumatology, Hospital Universitario Fundacion Santa Fe de Bogota, Bogota, Colombia
| | - Camilo Barajas
- Department of Orthopedics and Traumatology, Hospital Universitario Fundacion Santa Fe de Bogota, Bogota, Colombia
| | - Guido Fierro
- Department of Orthopedics and Traumatology, Hospital Universitario Fundacion Santa Fe de Bogota, Bogota, Colombia
- Facultad de Medicina, Universidad de los Andes, Bogota, Colombia
| | - Juan Carlos González
- Department of Orthopedics and Traumatology, Hospital Universitario Fundacion Santa Fe de Bogota, Bogota, Colombia
- Facultad de Medicina, Universidad de los Andes, Bogota, Colombia
| |
Collapse
|
17
|
Capelli C, Frigerio S, Lisini D, Nava S, Gaipa G, Belotti D, Cabiati B, Budelli S, Lazzari L, Bagnarino J, Tanzi M, Comoli P, Perico N, Introna M, Golay J. A comprehensive report of long-term stability data for a range ATMPs: A need to develop guidelines for safe and harmonized stability studies. Cytotherapy 2022; 24:544-556. [PMID: 35177338 DOI: 10.1016/j.jcyt.2021.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND AIMS Advanced therapy medicinal products (ATMPs) are novel drugs based on genes, cells or tissues developed to treat many different diseases. Stability studies of each new ATMP need to be performed to define its shelf life and guarantee efficacy and safety upon infusion, and these are presently based on guidelines originally drafted for standard pharmaceutical drugs, which have properties and are stored in conditions quite different from cell products. The aim of this report is to provide evidence-based information for stability studies on ATMPs that will facilitate the interlaboratory harmonization of practices in this area. METHODS We have collected and analyzed the results of stability studies on 19 different cell-based experimental ATMPs, produced by five authorized cell factories forming the Lombardy "Plagencell network" for use in 36 approved phase I/II clinical trials; most were cryopreserved and stored in liquid nitrogen vapors for 1 to 13 years. RESULTS The cell attributes collected in stability studies included cell viability, immunophenotype and potency assays, in particular immunosuppression, cytotoxicity, cytokine release and proliferation/differentiation capacity. Microbiological attributes including sterility, endotoxin levels and mycoplasma contamination were also analyzed. All drug products (DPs), cryopreserved in various excipients containing 10% DMSO and in different primary containers, were very stable long term at <-150°C and did not show any tendency for diminished viability or efficacy for up to 13.5 years. CONCLUSIONS Our data indicate that new guidelines for stability studies, specific for ATMPs and based on risk analyses, should be drafted to harmonize practices, significantly reduce the costs of stability studies without diminishing safety. Some specific suggestions are presented in the discussion.
Collapse
Affiliation(s)
- Chiara Capelli
- Center of Cellular Therapy "G. Lanzani", ASST Papa Giovanni XXIII, Bergamo, Italy; Fondazione per la Ricerca Ospedale di Bergamo, Bergamo, Italy
| | - Simona Frigerio
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniela Lisini
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sara Nava
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Gaipa
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Daniela Belotti
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Benedetta Cabiati
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Silvia Budelli
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Lorenza Lazzari
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Jessica Bagnarino
- UOSD Cell Factory, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Matteo Tanzi
- UOSD Cell Factory, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Patrizia Comoli
- UOSD Cell Factory, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Norberto Perico
- Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Istituto Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Martino Introna
- Center of Cellular Therapy "G. Lanzani", ASST Papa Giovanni XXIII, Bergamo, Italy.
| | - Josée Golay
- Center of Cellular Therapy "G. Lanzani", ASST Papa Giovanni XXIII, Bergamo, Italy; Fondazione per la Ricerca Ospedale di Bergamo, Bergamo, Italy
| |
Collapse
|
18
|
Ohori-Morita Y, Niibe K, Limraksasin P, Nattasit P, Miao X, Yamada M, Mabuchi Y, Matsuzaki Y, Egusa H. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:434-449. [PMID: 35267026 PMCID: PMC9052431 DOI: 10.1093/stcltm/szab030] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 12/02/2021] [Indexed: 11/14/2022] Open
Affiliation(s)
- Yumi Ohori-Morita
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Kunimichi Niibe
- Corresponding authors: Kunimichi Niibe, DDS, PhD, Associate Professor, Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai-city, Miyagi 980-8575, Japan. Tel: +81-22-717-8363; Fax: +81-22-717-8367;
| | - Phoonsuk Limraksasin
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Praphawi Nattasit
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Xinchao Miao
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Masahiro Yamada
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Yo Mabuchi
- Department of Biochemistry and Biophysics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yumi Matsuzaki
- Department of Life Science, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Hiroshi Egusa
- Hiroshi Egusa, DDS, PhD, Director, Center for Advanced Stem Cell and Regenerative Research, Professor and Chair, Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai-city 980-8575, Japan. Tel: +81-22-717-8363; Fax: +81-22-717-8367;
| |
Collapse
|
19
|
Mebarki M, Iglicki N, Marigny C, Abadie C, Nicolet C, Churlaud G, Maheux C, Boucher H, Monsel A, Menasché P, Larghero J, Faivre L, Cras A. Development of a human umbilical cord-derived mesenchymal stromal cell-based advanced therapy medicinal product to treat immune and/or inflammatory diseases. Stem Cell Res Ther 2021; 12:571. [PMID: 34774107 PMCID: PMC8590372 DOI: 10.1186/s13287-021-02637-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Background Umbilical cord-derived mesenchymal stromal cells (UC-MSCs) revealed their key role in immune regulation, offering promising therapeutic perspectives for immune and inflammatory diseases. We aimed to develop a production process of an UC-MSC-based product and then to characterize UC-MSC properties and immunomodulatory activities in vitro, related to their clinical use and finally, to transfer this technology to a good manufacturing practice (GMP) compliant facility, to manufacture an advanced therapy medicinal product (ATMP). Methods Fifteen human umbilical cords (UCs) were collected to develop the production process. Three batches of UC-MSCs from a single donor were characterized at basal state and after in vitro pro-inflammatory stimulation by interferon-γ (IFNγ) and tumor necrosis factor-α (TNFα). Proliferation, immunophenotype, activation markers’ expression and the inhibition of T cell proliferation were assessed. Finally, this technology was transferred to a GMP-compliant facility to manufacture an UC-MSC-based ATMP, from a single donor, using the explant method followed by the establishment of master and work cell stocks. Results Twelve UCs were processed successfully allowing to isolate UC-MSCs with doubling time and population doubling remaining stable until passage 4. CD90, CD105, CD73, CD44, CD29, CD166 expression was positive; CD14, CD45, CD31, HLA-DR, CD40, CD80 and CD86 expression was negative, while CD146 and HLA-ABC expression was heterogeneous. Cell morphology, proliferation and immunophenotype were not modified by inflammatory treatment. Indoleamine 2,3-dioxygenase (IDO) expression was significantly induced by IFNγ and IFNγ + TNFα versus non-treated cells. Intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) expression was induced significantly after priming. T cell proliferation was significantly decreased in the presence of UC-MSCs in a dose-dependent manner. This inhibitory effect was improved by IFNγ or IFNγ + TNFα, at UC-MSCs:PBMC ratio 1:10 and 1:30, whereas only IFNγ allowed to decrease significantly T cell proliferation at ratio 1:100. The manufacturing process of the UC-MSC-based ATMP was qualified and authorized by the French regulatory agency for clinical use (NCT04333368). Conclusion This work allowed to develop an investigational UC-MSC-based ATMP authorized for clinical use. Our results showed that an inflammatory environment preserves the biological properties of UC-MSCs with an improvement of their immunomodulatory functions. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02637-7.
Collapse
Affiliation(s)
- Miryam Mebarki
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France. .,INSERM U976, Université de Paris, 75010, Paris, France. .,Faculté de Pharmacie, Université de Paris, 75006, Paris, France.
| | | | - Céline Marigny
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France
| | - Camille Abadie
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France
| | - Claire Nicolet
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France
| | - Guillaume Churlaud
- AP-HP, Hôpital Saint-Louis, Centre MEARY de Thérapie Cellulaire Et Génique, 75010, Paris, France
| | - Camille Maheux
- AP-HP, Hôpital Saint-Louis, Centre MEARY de Thérapie Cellulaire Et Génique, 75010, Paris, France
| | - Hélène Boucher
- AP-HP, Hôpital Saint-Louis, Centre MEARY de Thérapie Cellulaire Et Génique, 75010, Paris, France
| | - Antoine Monsel
- Unité de Soins Intensifs Et Département de Biothérapies, inflammation et immunopathologie, AP-HP, Hôpital La Pitié-Salpêtrière, 75013, Paris, France.,INSERM UMR-S 959, Université Sorbonne, 75012, Paris, France
| | - Philippe Menasché
- Département de Chirurgie Cardiovasculaire, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - Jérôme Larghero
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France.,INSERM U976, Université de Paris, 75010, Paris, France.,AP-HP, Hôpital Saint-Louis, Centre MEARY de Thérapie Cellulaire Et Génique, 75010, Paris, France
| | - Lionel Faivre
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France.,INSERM U976, Université de Paris, 75010, Paris, France
| | - Audrey Cras
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France. .,INSERM UMR1140, Université de Paris, 75006, Paris, France. .,Faculté de Pharmacie, Université de Paris, 75006, Paris, France.
| |
Collapse
|
20
|
El-Jawhari JJ, Ganguly P, Jones E, Giannoudis PV. Bone Marrow Multipotent Mesenchymal Stromal Cells as Autologous Therapy for Osteonecrosis: Effects of Age and Underlying Causes. Bioengineering (Basel) 2021; 8:69. [PMID: 34067727 PMCID: PMC8156020 DOI: 10.3390/bioengineering8050069] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/29/2021] [Accepted: 05/13/2021] [Indexed: 12/21/2022] Open
Abstract
Bone marrow (BM) is a reliable source of multipotent mesenchymal stromal cells (MSCs), which have been successfully used for treating osteonecrosis. Considering the functional advantages of BM-MSCs as bone and cartilage reparatory cells and supporting angiogenesis, several donor-related factors are also essential to consider when autologous BM-MSCs are used for such regenerative therapies. Aging is one of several factors contributing to the donor-related variability and found to be associated with a reduction of BM-MSC numbers. However, even within the same age group, other factors affecting MSC quantity and function remain incompletely understood. For patients with osteonecrosis, several underlying factors have been linked to the decrease of the proliferation of BM-MSCs as well as the impairment of their differentiation, migration, angiogenesis-support and immunoregulatory functions. This review discusses the quality and quantity of BM-MSCs in relation to the etiological conditions of osteonecrosis such as sickle cell disease, Gaucher disease, alcohol, corticosteroids, Systemic Lupus Erythematosus, diabetes, chronic renal disease and chemotherapy. A clear understanding of the regenerative potential of BM-MSCs is essential to optimize the cellular therapy of osteonecrosis and other bone damage conditions.
Collapse
Affiliation(s)
- Jehan J El-Jawhari
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK
- Clinical Pathology Department, Mansoura University, Mansoura 35516, Egypt
| | - Payal Ganguly
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, UK; (P.G.); (E.J.); (P.V.G.)
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, UK; (P.G.); (E.J.); (P.V.G.)
| | - Peter V Giannoudis
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, UK; (P.G.); (E.J.); (P.V.G.)
- Academic Department of Trauma and Orthopedic, School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
21
|
Hernigou P, Auregan JC, Dubory A, Flouzat Lachaniette CH, Rouard H. Ankle osteonecrosis in fifty-one children and adolescent's leukemia survivors: a prospective randomized study on percutaneous mesenchymal stem cells treatment. INTERNATIONAL ORTHOPAEDICS 2021; 45:2383-2393. [PMID: 33893522 DOI: 10.1007/s00264-021-05051-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/16/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Corticoid treatment associated with haematologic treatments can lead to ankle osteonecrosis in children's survivors of acute leukemia (ALL). Based on the efficiency of mesenchymal stem cells (MSCs) in hip osteonecrosis, we performed an evaluation of this treatment in 51 children and adolescents who had symptomatic ankle osteonecrosis after therapy for haematologic cancer. MATERIAL AND METHODS The 51 patients had a total of 79 osteonecrosis sites on MRI, with 29 talus sites, 18 metaphyseal tibia sites, 12 epiphyseal tibia sites, eight calcaneus sites, six fibula sites, four navicular sites, and two cuboid sites. In this prospective randomized trial, 37 ankles were addressed for cell therapy, 37 others for core decompression alone, and 20 were considered as a control group without treatment. We analyzed the outcome of this treatment osteonecrosis, the number and characteristics of bone marrow mesenchymal cells (MSCs) that could be transplanted, and the risks of tumorigenesis in these patients with haematologic cancers. The patients were operated on over a period of ten years from 2000 to 2010 and were monitored through December 31, 2019. RESULTS Despite a normal systemic blood cells count, MSCs in the iliac crest (counted as CFU-F) were in low number (1021 MSCs/mL; range 314-3015) and were of host origin after even allogeneic bone marrow transplantation. Better clinical outcomes (pain, foot and ankle deformity) and osteonecrosis repair on MRI with absence of collapse were obtained in ankles that received cell therapy as compared with those with core decompression alone or those without initial surgery. No tumour was found on MRI at the sites of injection and this study found no increased risk of recurrence or of new cancer in this population after an average follow-up of 15 years. CONCLUSIONS These results suggest that autologous MSCs can improve the quality of life of leukemia survivors with ankle osteonecrosis.
Collapse
Affiliation(s)
- Philippe Hernigou
- Orthopedic Department, Henri Mondor Hospital, University Paris East, Paris, France.
| | - Jean Charles Auregan
- Orthopedic Department, Antoine Beclère Hospital, University Paris West, Paris, France
| | - Arnaud Dubory
- Orthopedic Department, Henri Mondor Hospital, University Paris East, Paris, France
| | | | - Hélène Rouard
- Etablissement Français du Sang, Henri Mondor Hospital, University Paris East, Paris, France
| |
Collapse
|
22
|
Implantation of autologous Expanded Mesenchymal Stromal Cells in Hip Osteonecrosis through Percutaneous Forage: Evaluation of the Operative Technique. J Clin Med 2021; 10:jcm10040743. [PMID: 33673388 PMCID: PMC7918570 DOI: 10.3390/jcm10040743] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 11/17/2022] Open
Abstract
Bone forage to treat early osteonecrosis of the femoral head (ONFH) has evolved as the channel to percutaneously deliver cell therapy into the femoral head. However, its efficacy is variable and the drivers towards higher efficacy are currently unknown. The aim of this study was to evaluate the forage technique and correlate it with the efficacy to heal ONFH in a multicentric, multinational clinical trial to implant autologous mesenchymal stromal cells expanded from bone marrow (BM-hMSCs). Methods: In the context of EudraCT 2012-002010-39, patients with small and medium-sized (mean volume = 13.3%, range: 5.4 to 32.2) ONFH stage II (Ficat, ARCO, Steinberg) C1 and C2 (Japanese Investigation Committee (JIC)) were treated with percutaneous forage and implantation of 140 million BM-hMSCs in a standardized manner. Postoperative hip radiographs (AP—anteroposterior and lateral), and MRI sections (coronal and transverse) were retrospectively evaluated in 22 patients to assess the femoral head drilling orientation in both planes, and its relation to the necrotic area. Results: Treatment efficacy was similar in C1 and C2 (coronal plane) and in anterior to posterior (transverse plane) osteonecrotic lesions. The drill crossed the sclerotic rim in all cases. The forage was placed slightly valgus, at 139.3 ± 8.4 grades (range, 125.5–159.3) with higher dispersion (f = 2.6; p = 0.034) than the anatomical cervicodiaphyseal angle. Bonferroni’s correlation between both angles was 0.50 (p = 0.028). More failures were seen with a varus drill positioning, aiming at the central area of the femoral head, outside the weight-bearing area (WBA) (p = 0.049). In the transverse plane, the anterior positioning of the drill did not result in better outcomes (p = 0.477). Conclusion: The forage drilling to deliver cells should be positioned within the WBA in the coronal plane, avoiding varus positioning, and central to anterior in the transverse plane. The efficacy of delivered MSCs to regenerate bone in ONFH could be influenced by the drilling direction. Standardization of this surgical technique is desirable.
Collapse
|