1
|
De la Corte-Rodriguez H, Rodriguez-Merchan EC, Alvarez-Roman MT, Gomez-Cardero P, Jimenez-Yuste V. Ultrasound-guided joint procedures in hemophilia: technique, indications and tips. Expert Rev Hematol 2024; 17:419-430. [PMID: 39008070 DOI: 10.1080/17474086.2024.2380477] [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: 03/21/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
INTRODUCTION The therapeutic approach to pain in hemophilia should be multimodal. Intra-articular injections are a good option when joint lesions do not respond to hematological treatment or rehabilitation and orthopedic surgery is not yet indicated. Performing these procedures under ultrasound guidance has been shown to improve their accuracy and efficacy. AREAS COVERED This article provides a practical overview of the most frequently employed ultrasound-guided intra-articular procedures on the joints of people with hemophilia. The article describes the key elements for performing the technique on the elbow, knee and ankle as the most affected joints. The particularities of the most frequent indications, arthrocentesis, synoviorthesis and analgesic injections with various products are detailed. EXPERT OPINION Current hematological treatments have made it possible to incorporate new therapeutic tools for pain relief for people with hemophilia, including ultrasound-guided joint procedures, which offer excellent results.
Collapse
|
2
|
Théron A, Maumus M, Biron-Andreani C, Sirvent N, Jorgensen C, Noël D. What is the rationale for mesenchymal stromal cells based therapies in the management of hemophilic arthropathies? Osteoarthritis Cartilage 2024; 32:634-642. [PMID: 38160743 DOI: 10.1016/j.joca.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/21/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024]
Abstract
Hemophilia A and B are rare X-linked genetic bleeding disorders due to a complete or partial deficiency in the coagulation factors VIII or IX, respectively. The main treatment for hemophilia is prophylactic and based on coagulation factor replacement therapies. These treatments have significantly reduced bleeding and improved the patients' quality of life. Nevertheless, repeated joint bleedings (hemarthroses), even subclinical hemarthroses, can lead to hemophilic arthropathy (HA). This disabling condition is characterized by chronic pain due to synovial inflammation, cartilage and bone destruction requiring ultimately joint replacement. HA resembles to rheumatoid arthritis because of synovitis but HA is considered as having similarities with osteoarthritis as illustrated by the migration of immune cells, production of inflammatory cytokines, synovial hypertrophy and cartilage damage. Various drugs have been evaluated for the management of HA with limited success. The objective of the review is to discuss new therapeutic approaches with a special focus on the studies that have investigated the potential of using mesenchymal stromal cells (MSCs) in the management of HA. A systematic review of the literature has been made. Most of the studies have focused on the interest of MSCs for the delivery of missing factors VIII or IX but in some studies, more insight on the effect of MSC injection on synovial inflammation or cartilage structure were provided and put in perspective for possible clinical applications.
Collapse
Affiliation(s)
- Alexandre Théron
- IRMB, University of Montpellier, INSERM, Montpellier, France; Resources and Competence Center for Hereditary Hemorrhagic Diseases, CHU Montpellier, Montpellier, France; Department of Pediatric Oncology and Hematology, CHU Montpellier, Montpellier, France
| | - Marie Maumus
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Christine Biron-Andreani
- Resources and Competence Center for Hereditary Hemorrhagic Diseases, CHU Montpellier, Montpellier, France
| | - Nicolas Sirvent
- Department of Pediatric Oncology and Hematology, CHU Montpellier, Montpellier, France
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, Montpellier, France; Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France; Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, Montpellier, France.
| |
Collapse
|
3
|
Théron A, Maumus M, Bony-Garayt C, Sirvent N, Biron-Andreani C, Jorgensen C, Noël D. Mesenchymal Stromal Cells Prevent Blood-induced Degeneration of Chondrocytes in a New Model of Murine Hemarthrosis. Hemasphere 2023; 7:e924. [PMID: 37388924 PMCID: PMC10306440 DOI: 10.1097/hs9.0000000000000924] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023] Open
Abstract
Hemophilia is a rare congenital bleeding disorder caused by deficiency in coagulation factors VIII or IX, which is treated with prophylactic clotting factor concentrates. Nevertheless despite prophylaxis, spontaneous joint bleedings or hemarthroses still occur. The recurrent hemarthroses lead to progressive degradation of the joints and severe hemophilic arthropathy (HA) in patients with moderate and even mild forms of the disease. In absence of disease modifying treatment to stop or even delay HA progression, we aimed at evaluating the therapeutic potential of mesenchymal stromal cells (MSCs)-based therapy. We first developed a relevant and reproducible in vitro model of hemarthrosis relying on blood exposure of primary murine chondrocytes. We found that 30% whole blood for 4 days allowed to induce the characteristic features of hemarthrosis including low survival of chondrocytes, apoptosis induction, and dysregulation of chondrocyte markers in favor of a catabolic and inflammatory phenotype. We then evaluated the potential therapeutic effects of MSCs in this model using different conditions of coculture. Addition of MSCs improved the survival of chondrocytes when added either during the resolution or the acute phases of hemarthrosis and exerted a chondroprotective effect by enhancing the expression of anabolic markers, and reducing the expression of catabolic and inflammatory markers. We here provide the first proof-of-concept that MSCs may exert a therapeutic effect on chondrocytes under hemarthrosis conditions using a relevant in vitro model, thereby confirming a potential therapeutic interest for patients with recurrent joint bleedings.
Collapse
Affiliation(s)
- Alexandre Théron
- IRMB, University of Montpellier, INSERM, Montpellier, France
- Resources and Competence Center for hereditary hemorrhagic diseases, CHU Montpellier, France
- Department of Pediatric Oncology and Hematology, CHU Montpellier, France
| | - Marie Maumus
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | | | - Nicolas Sirvent
- Department of Pediatric Oncology and Hematology, CHU Montpellier, France
| | | | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, Montpellier, France
- Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France
- Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, France
| |
Collapse
|
4
|
Kawamura K, Tanaka Y, Nakasone H, Ishihara Y, Kako S, Kobayashi S, Tanaka Y, Ohmori T, Uchimaru K, Okamoto S, Mineno J, Shiku H, Nishimura S, Kanda Y. Development of a Unique T Cell Receptor Gene-Transferred Tax-Redirected T Cell Immunotherapy for Adult T Cell Leukemia. Biol Blood Marrow Transplant 2020; 26:1377-1385. [PMID: 32311478 DOI: 10.1016/j.bbmt.2020.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 03/24/2020] [Accepted: 04/05/2020] [Indexed: 11/28/2022]
Abstract
Adult T cell leukemia/lymphoma (ATL) is an aggressive peripheral T cell neoplasm caused by infection with human T cell lymphotropic virus type-1 (HTLV-1). Its prognosis remains extremely poor. Tax, the most important regulatory protein for HTLV-1, is associated with the aggressive proliferation of host cells and is also a major target antigen for CD8+ cytotoxic T cells (CTLs). Based on our previous findings that Tax-specific CTLs with a T cell receptor (TCR) containing a unique amino-acid sequence motif exhibit strong HLA-A*24:02-restricted, Tax301-309-specific activity against HTLV-1, we aimed to develop a Tax-redirected T cell immunotherapy for ATL. TCR-ɑ/β genes were cloned from a previously established CTL clone and transduced into peripheral blood mononuclear cells (PBMCs) of healthy volunteers using a retroviral siTCR vector. Then the cytotoxic efficacy against HTLV-1-infected T cells or primary ATL cells was assessed both in vitro and in vivo. The redirected CTLs (Tax-siCTLs) produced a large amount of cytokines and showed strong killing activity against ATL/HTLV-1-infected T cells in vitro, although they did not have universal activity against ATL cells. Next, in a xenograft mouse model using an HTLV-1-infected T cell line (MT-2), in all mice treated with Tax-siCTLs, the tumor rapidly diminished and finally disappeared without normal tissue damage, although all mice that were untreated or treated with non-gene-modified PBMCs died because of tumor progression. Our findings confirm that Tax-siCTLs can exert strong anti-ATL/HTLV-1 effects without a significant reaction against normal cells and have the potential to be a novel immunotherapy for ATL patients.
Collapse
Affiliation(s)
- Koji Kawamura
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Yukie Tanaka
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan; Division of Molecular Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Research Core, Institute of Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hideki Nakasone
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Yuko Ishihara
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Shinichi Kako
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Seiichiro Kobayashi
- Division of Molecular Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuetsu Tanaka
- Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Tsukasa Ohmori
- Department of Biochemistry, Jichi Medical University, Tochigi, Japan
| | - Kaoru Uchimaru
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | | | | | - Hiroshi Shiku
- Department for Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Satoshi Nishimura
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan; Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology, Saitama, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan; Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan.
| |
Collapse
|
5
|
Zhang F, Yan X, Li M, Hua B, Xiao X, Monahan PE, Sun J. Exploring the Potential Feasibility of Intra-Articular Adeno-Associated Virus-Mediated Gene Therapy for Hemophilia Arthropathy. Hum Gene Ther 2020; 31:448-458. [PMID: 32079420 DOI: 10.1089/hum.2019.355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Hemophilia arthropathy (HA) represents the majority of morbidity in severe hemophilia patients, especially in resource-limited countries. Adeno-associated virus (AAV)-mediated gene therapy is showing promise for managing hemophilia. However, patients with neutralizing antibodies (NAbs) against AAV, and inhibitors to clotting factors, are excluded from such therapy. This study explored the feasibility of AAV-mediated local gene therapy for HA. Factor VIII knockout (FVIII-/-) mice, with or without a FVIII inhibitor, were subjected to hemarthrosis induction and treated with either intravenous (IV) or intraarticular (IA) recombinant human factor VIII (rhFVIII). To investigate whether rhFVIII carried the risk to develop a FVIII inhibitor, FVIII-/- mice were treated with three doses of IV or IA rhFVIII and inhibitor development was measured. In patients with established HA requiring synovial fluid aspiration, plasma, and synovial fluid were collected and measured for anti-AAV capsid IgG (serotypes 1-9 and 843) and NAbs for AAV843. IA rhFVIII provided better protection from synovitis compared with IV rhFVIII, with or without the FVIII inhibitor. While IV rhFVIII led to all FVIII-/- mice developing an FVIII inhibitor (n = 31, median 4.9 Bethesda units [BU]/mL), only 50% of the mice developed a FVIII inhibitor by IA administration, and at a lower titer (median 0.55 BU/mL). In hemophilia patients, total anti-AAV IgG was lowest for AAV4 and AAV5, both in plasma and synovial fluid. Anti-AAV IgGs in synovial fluid for most samples were lower or similar to the plasma levels. These results show that direct IA rhFVIII administration yields better protection against bleeding-induced joint damage, even in the presence of an inhibitor antibody. IA rhFVIII delivery carried a lower risk of FVIII inhibitor formation compared with IV FVIII. The anti-AAV antibody level in synovial fluid was similar or lower than the plasma level, supporting the feasibility of local gene therapy for managing HA.
Collapse
Affiliation(s)
- Feixu Zhang
- School of Bioengineering, East China University of Science and Technology, Shanghai, China.,School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xiaobo Yan
- Department of Hematology, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Min Li
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Baolai Hua
- Department of Hematology, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Xiao Xiao
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Paul E Monahan
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina.,Harold R. Roberts Comprehensive Hemophilia Diagnosis and Treatment Center, University of North Carolina, Chapel Hill, North Carolina.,Spark Therapeutics, Philadelphia, Pennsylvania
| | - Junjiang Sun
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina.,Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
6
|
Rose M, Gao K, Cortez-Toledo E, Agu E, Hyllen AA, Conroy K, Pan G, Nolta JA, Wang A, Zhou P. Endothelial cells derived from patients' induced pluripotent stem cells for sustained factor VIII delivery and the treatment of hemophilia A. Stem Cells Transl Med 2020; 9:686-696. [PMID: 32162786 PMCID: PMC7214661 DOI: 10.1002/sctm.19-0261] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/29/2020] [Indexed: 12/12/2022] Open
Abstract
Hemophilia A (HA) is a bleeding disorder characterized by spontaneous and prolonged hemorrhage. The disease is caused by mutations in the coagulation factor 8 gene (F8) leading to factor VIII (FVIII) deficiency. Since FVIII is primarily produced in endothelial cells (ECs) in a non‐diseased human being, ECs hold great potential for development as a cell therapy for HA. We showed that HA patient‐specific induced pluripotent stem cells (HA‐iPSCs) could provide a renewable supply of ECs. The HA‐iPSC‐derived ECs were transduced with lentiviral vectors to stably express the functional B domain deleted F8 gene, the luciferase gene, and the enhanced green fluorescent protein gene (GFP). When transplanted intramuscularly into neonatal and adult immune deficient mice, the HA‐iPSC‐derived ECs were retained in the animals for at least 10‐16 weeks and maintained their expression of FVIII, GFP, and the endothelial marker CD31, as demonstrated by bioluminescence imaging and immunostaining, respectively. When transplanted into HA mice, these transduced HA‐iPSC‐derived ECs significantly reduced blood loss in a tail‐clip bleeding test and produced therapeutic plasma levels (11.2%‐369.2%) of FVIII. Thus, our studies provide proof‐of‐concept that HA‐iPSC‐derived ECs can serve as a factory to deliver FVIII for the treatment of HA not only in adults but also in newborns.
Collapse
Affiliation(s)
- Melanie Rose
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California
| | - Kewa Gao
- Department of Surgery, University of California Davis Medical Center, Sacramento, California.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California
| | - Elizabeth Cortez-Toledo
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California
| | - Emmanuel Agu
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California
| | - Alicia A Hyllen
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California
| | - Kelsey Conroy
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California
| | - Guangjin Pan
- Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jan A Nolta
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California.,University of California Davis Gene Therapy Center, Sacramento, California
| | - Aijun Wang
- Department of Surgery, University of California Davis Medical Center, Sacramento, California.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California.,Department of Biomedical Engineering, University of California Davis, Davis, California
| | - Ping Zhou
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California.,University of California Davis Gene Therapy Center, Sacramento, California
| |
Collapse
|
7
|
Shomali N, Gharibi T, Vahedi G, Mohammed RN, Mohammadi H, Salimifard S, Marofi F. Mesenchymal stem cells as carrier of the therapeutic agent in the gene therapy of blood disorders. J Cell Physiol 2019; 235:4120-4134. [PMID: 31691976 DOI: 10.1002/jcp.29324] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 09/30/2019] [Indexed: 12/16/2022]
Abstract
Nonhematopoietic stem cells as a delivery platform of therapeutic useful genes have attracted widespread attention in recent years, owing to gained a long lifespan, easy separation, high proliferation, and high transfection capacity. Mesenchymal stem/stromal cells (MSCs) are the choice of the cells for gene and cell therapy due to high self-renewal capacity, high migration rate to the site of the tumor, and with immune suppressive and anti-inflammatory properties. Hence, it has a high potential of safety genetic modification of MSCs for antitumor gene expression and has paved the way for the clinical application of these cells to target the therapy of cancers and other diseases. The aim of gene therapy is targeted treatment of cancers and diseases through recovery, change, or enhancement cell performance to the sustained secretion of useful therapeutic proteins and induction expression of the functional gene in intended tissue. Recent developments in the vectors designing leading to the increase and durability of expression and improvement of the safety of the vectors that overcome a lot of problems, such as durability of expression and the host immune response. Nowadays, gene therapy approach is used by MSCs as a delivery vehicle in the preclinical and the clinical trials for the secretion of erythropoietin, recombinant antibodies, coagulation factors, cytokines, as well as angiogenic inhibitors in many blood disorders like anemia, hemophilia, and malignancies. In this study, we critically discuss the status of gene therapy by MSCs as a delivery vehicle for the treatment of blood disorders. Finally, the results of clinical trial studies are assessed, highlighting promising advantages of this emerging technology in the clinical setting.
Collapse
Affiliation(s)
- Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ghasem Vahedi
- Department of Immunology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Rebar N Mohammed
- Bone Marrow Transplant Center, Hiwa Cancer Hospital, Suleimanyah, Iraq.,Department of Microbiology, College of Veterinary Medicine, University of Sulaimani, Suleimanyah, Iraq
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Sevda Salimifard
- Department of Hematology and Blood Transfusion, School of Allied Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Buccheri E, Avola M, Vitale N, Pavone P, Vecchio M. Haemophilic arthropathy: A narrative review on the use of intra-articular drugs for arthritis. Haemophilia 2019; 25:919-927. [PMID: 31639263 DOI: 10.1111/hae.13857] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/16/2019] [Accepted: 09/18/2019] [Indexed: 09/15/2023]
Abstract
INTRODUCTION Intra-articular injections of various drugs are commonly used in patients with degenerative osteoarthritis and also in haemophilic patients. Haemophilic arthropathy is a particular type of secondary osteoarthritis (OA), but the degeneration of strong synovial, cartilaginous and subchondral constituents is provoked by the direct action of iron and blood in the joint. AIM OF THE STUDY The aim of this study is to review the literature regarding the use of various intra-articular drugs in joints affected by haemophilic arthropathy. METHODS We reviewed the data from the literature; the search was performed on three medical electronic databases (PubMed, Cochrane Library and Scopus Web of Science) by three authors (B. E., A. M. and V. N.) from 3 December 2018 till 15 December 2018. The search string was as follows: (hyaluronic acid OR viscosupplementation OR platelet-rich plasma OR corticosteroid OR mesenchymal stem cells) AND (haemophilia OR haemophilic arthropathy OR haemophilic arthritis). RESULTS Once the research was performed, a total of 300 articles were identified. 47 selected articles were analysed by the reviewers, and the eligibility of the study inclusion was assessed independently. Twelve papers were included based on clear fulfilment of the inclusion criteria. Thirty-five articles were excluded for the following reasons: no full text or accessible data for 14 of them, 15 involved surgery or rehabilitation therapy as the primary topic and 6 were systematic reviews (the main topics were beyond the haemophilic arthropathy). CONCLUSION Although the degree of scientific evidence of the publications on intra-articular injections of various drugs (hyaluronic acid, corticosteriods, PRP and MSCs) in haemophilia is very low, it seems that intra-articular injections of hyaluronic acid can relieve joint pain for months and can be repeated every 6-12 months, which is why they can be recommended. Corticosteroid injections seem to relieve joint pain for a few weeks, but their routine use is not recommended in haemophilia. The efficacy of PRP and MSCs in haemophilic arthropathy is pending confirmation, which is why they are not currently recommended.
Collapse
Affiliation(s)
| | - Marianna Avola
- Graduate School of Physical Medicine and Rehabilitation, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Nicolò Vitale
- Graduate School of Physical Medicine and Rehabilitation, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Piero Pavone
- University-Hospital "Policlinico-Vittorio Emanuele, " University of Catania, Catania, Italy
| | - Michele Vecchio
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
- Rehabilitation Unit, "AOU Policlinico Vittorio Emanuele", Catania, Italy
| |
Collapse
|
9
|
He P, Zhang F, Zhong C, Li M, Zheng J, Hua B, Sun J. Timely and large dose of clotting factor IX provides better joint wound healing after hemarthrosis in hemophilia B mice. Int J Hematol 2019; 110:59-68. [PMID: 31006077 DOI: 10.1007/s12185-019-02639-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/27/2019] [Accepted: 03/19/2019] [Indexed: 12/15/2022]
Abstract
Bleeding into the joints represents the major morbidity of severe hemophilia and predisposes it to hemophilic arthropathy (HA). In a reproducible hemarthrosis mouse model, we found distinct changes in thrombin activity in joint tissue homogenate following exposure of the joint to blood in wide type (WT) and hemophilic B mice. Specifically, at early time points (4 h and 24 h) after hemarthrosis, thrombin activity in WT mice quickly peaked at 4 h, and returned to baseline after 1 week. In hemophilia B mice, there was no/minimal thrombin activity in joint tissues at 4 h and 24 h, whereas at 72 h and thereafter, thrombin activity kept rising, and persisted at a higher level. Nevertheless, prothrombin had not decreased in both WT and hemophilia. The pattern was also confirmed by Western blotting and immunostaining. To optimize the protection against development of HA, we tested different treatment regimens by administration of clotting factor IX into hemophilia B mouse after hemarthrosis induction, including a total of 600 IU/kg FIX within the first 24 h or the whole 2-week period. We concluded that timely (in the first 24 h) and sufficient hemostasis correction is critical for a better protection against the development of hemophilic arthropathy.
Collapse
Affiliation(s)
- Ping He
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Feixu Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Chen Zhong
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Min Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jing Zheng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Baolai Hua
- Department of Hematology, Clinical Medical College, Yangzhou University, Yangzhou, China.
| | - Junjiang Sun
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China.
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, USA.
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
10
|
Borsotti C, Follenzi A. New technologies in gene therapy for inducing immune tolerance in hemophilia A. Expert Rev Clin Immunol 2018; 14:1013-1019. [PMID: 30345839 DOI: 10.1080/1744666x.2018.1539667] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Conventional hemophilia treatment is based on repeated infusion of the missing clotting factor. This therapy is lifelong, expensive and can result in the formation of neutralizing antibodies, thus causing failure of the treatment and requiring higher doses of the replacement drug. Areas covered: Gene and cell therapies offer the advantage of providing a definitive and long-lasting correction of the mutated gene, promoting its physiological expression and preventing neutralizing antibody development. This review focuses on the most recent approaches that have been shown to prevent and even eradicate immune response toward the replaced factor. Expert commentary: Despite the encouraging data demonstrated by ongoing clinical trials and pre-clinical studies, more extensive investigations are necessary to establish the long-term safety and efficacy of gene therapy treatments in maintaining immune tolerance.
Collapse
Affiliation(s)
- Chiara Borsotti
- a Department of Health Sciences , Università del Piemonte Orientale , Novara , Italy
| | - Antonia Follenzi
- a Department of Health Sciences , Università del Piemonte Orientale , Novara , Italy
| |
Collapse
|
11
|
Advances in gene therapy for hemophilia: basis, current status, and future perspectives. Int J Hematol 2018; 111:31-41. [DOI: 10.1007/s12185-018-2513-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 02/08/2023]
|
12
|
Ohmori T, Mizukami H, Katakai Y, Kawai S, Nakamura H, Inoue M, Shu T, Sugimoto H, Sakata Y. Safety of intra-articular transplantation of lentivirally transduced mesenchymal stromal cells for haemophilic arthropathy in a non-human primate. Int J Hematol 2018; 108:239-245. [PMID: 29737459 DOI: 10.1007/s12185-018-2465-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 12/01/2022]
Abstract
Joint bleeding and resultant arthropathy are major determinants of quality of life in haemophilia patients. We previously developed a mesenchymal stromal cell (MSC)-based treatment approach for haemophilic arthropathy in a mouse model of haemophilia A. Here, we evaluated the long-term safety of intra-articular injection of lentivirally transduced autologous MSCs in non-human primates. Autologous bone-marrow-derived MSCs transduced with a lentiviral vector expressing coagulation factor VIII (FVIII) were injected into the left knee joint of cynomolgus monkeys. We first conducted codon optimization to increase FVIII production in the cells. Lentiviral transduction of autologous MSCs resulted in a significant increase of FVIII in the culture supernatant before transplantation. We did not find any tumour generation around the knee structure at 11-16 months after injection by magnetic resonance imaging. The proviral sequence of the simian immunodeficiency virus lentiviral vector was not detected in the heart, lungs, spleen, liver, testis, or bone marrow by real-time quantitative PCR. We confirmed the long-term safety of intra-articular injection of transduced MSCs in a non-human primate. The procedure may be an attractive therapeutic approach for joint diseases in haemophilia patients.
Collapse
Affiliation(s)
- Tsukasa Ohmori
- Department of Biochemistry, Jichi Medical University School of Medicine, 3111-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Yuko Katakai
- The Corporation for Production and Research of Laboratory Primates, 1-16-2 Sakura, Tsukuba, Ibaraki, 305-0003, Japan
| | - Sho Kawai
- Department of Radiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hitoyasu Nakamura
- Department of Radiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Makoto Inoue
- ID Pharma Inc., 6 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Tsugumine Shu
- ID Pharma Inc., 6 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Hideharu Sugimoto
- Department of Radiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Yoichi Sakata
- Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| |
Collapse
|
13
|
Sokal EM, Lombard CA, Roelants V, Najimi M, Varma S, Sargiacomo C, Ravau J, Mazza G, Jamar F, Versavau J, Jacobs V, Jacquemin M, Eeckhoudt S, Lambert C, Stéphenne X, Smets F, Hermans C. Biodistribution of Liver-Derived Mesenchymal Stem Cells After Peripheral Injection in a Hemophilia A Patient. Transplantation 2017; 101:1845-1851. [PMID: 28738402 DOI: 10.1097/tp.0000000000001773] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND With the exception of liver transplantation, there is no cure for hemophilia, which is currently managed by preemptive replacement therapy. Liver-derived stem cells are in clinical development for inborn and acquired liver diseases and could represent a curative treatment for hemophilia A. The liver is a major factor VIII (FVIII) synthesis site, and mesenchymal stem cells have been shown to control joint bleeding in animal models of hemophilia. Adult-derived human liver stem cells (ADHLSCs) have mesenchymal characteristics and have been shown able to engraft in and repopulate both animal and human livers. Thus, the objectives were to evaluate the potency of ADHLSCs to control bleeding in a hemophilia A patient and assess the biodistribution of the cells after intravenous injection. METHODS A patient suffering from hemophilia A was injected with repeated doses of ADHLSCs via a peripheral vein (35 million In-oxine-labeled cells, followed by 125 million cells the next day, and 3 infusions of 250 million cells every 2 weeks thereafter; total infusion period, 50 days). RESULTS After cell therapy, we found a temporary (15 weeks) decrease in the patient's FVIII requirements and severe bleeding complications, despite a lack of increase in circulating FVIII. The cells were safely administered to the patient via a peripheral vein. Biodistribution analysis revealed an initial temporary entrapment of the cells in the lungs, followed by homing to the liver and to a joint afflicted with hemarthrosis. CONCLUSION These results suggest the potential use of ADHLSCs in the treatment of hemophilia A.
Collapse
Affiliation(s)
- Etienne M Sokal
- 1 Université Catholique de Louvain, Cliniques Universitaires St Luc, Service de Gastroentérologie & Hépatologie Pédiatrique, Brussels, Belgium.2 Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Brussels, Belgium.3 Centre de Thérapie Cellulaire et Tissulaire, Cliniques Universitaires St Luc, Brussels, Belgium.4 Service de Médecine Nucléaire, Cliniques Universitaires St Luc, Brussels, Belgium.5 Division of Medicine, Institute for Liver and Digestive Health, Royal Free Hospital, University College of London, London, United Kingdom.6 Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Belgium.7 Service d'Hématologie, Cliniques Universitaires St Luc, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Composition and Function of the Interstitial Fluid. Protein Sci 2016. [DOI: 10.1201/9781315374307-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
15
|
Rodriguez-Merchan EC. Intra-articular injections of mesenchymal stem cells (MSCs) as a treatment for hemophilic arthropathy. Expert Rev Hematol 2016; 9:737-41. [DOI: 10.1080/17474086.2016.1203780] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
16
|
Sokal EM, Lombard C, Mazza G. Mesenchymal stem cell treatment for hemophilia: a review of current knowledge. J Thromb Haemost 2015; 13 Suppl 1:S161-6. [PMID: 26149017 DOI: 10.1111/jth.12933] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Hemophilia remains a non-curative disease, and patients are constrained to undergo repeated injections of clotting factors. In contrast, the sustained production of endogenous factors VIII (FVIII) or IX (FIX) by the patient's own cells could represent a curative treatment. Gene therapy has thus provided new hope for these patients. However, the issues surrounding the durability of expression and immune responses against gene transfer vectors remain. Cell therapy, involving stem cells expanded in vitro, can provide de novo protein synthesis and, if implanted successfully, could induce a steady-state production of low quantities of factors, which may keep the patient above the level required to prevent spontaneous bleeding. Liver-derived stem cells are already being assessed in clinical trials for inborn errors of metabolism and, in view of their capacity to produce FVIII and FIX in cell culture, they are now also being considered for clinical application in hemophilia patients.
Collapse
Affiliation(s)
- E M Sokal
- Service de gastroentérologie et hépatologie pédiatrique, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Cliniques St Luc, Brussels, Belgium
| | - C Lombard
- Université Catholique de Louvain, Brussels, Belgium
| | - G Mazza
- Division of Medicine, Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London, UK
| |
Collapse
|
17
|
Ohmori T, Mizukami H, Ozawa K, Sakata Y, Nishimura S. New approaches to gene and cell therapy for hemophilia. J Thromb Haemost 2015; 13 Suppl 1:S133-42. [PMID: 26149014 DOI: 10.1111/jth.12926] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hemophilia is considered suitable for gene therapy because it is caused by a single gene abnormality, and therapeutic coagulation factor levels may vary across a broad range. Recent success of hemophilia B gene therapy with an adeno-associated virus (AAV) vector in a clinical trial showed the real prospect that, through gene therapy, a cure for hemophilia may become a reality. However, AAV-mediated gene therapy is not applicable to patients with hemophilia A at present, and neutralizing antibodies against AAV reduce the efficacy of AAV-mediated strategies. Because patients that benefit from AAV treatment (hemophilia B without neutralizing antibodies) are estimated to represent only 15% of total patients with hemophilia, the development of basic technologies for hemophilia A and those that result in higher therapeutic effects are critical. In this review, we present an outline of gene therapy methods for hemophilia, including the transition of technical developments thus far and our novel techniques.
Collapse
Affiliation(s)
- T Ohmori
- Research Division of Cell and Molecular Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - H Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - K Ozawa
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Y Sakata
- Research Division of Cell and Molecular Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - S Nishimura
- Research Division of Cell and Molecular Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan
- Translational Systems Biology and Medicine Initiative, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
Ravanbod R, Torkaman G, Mophid M, Mohammadali F. Experimental study on the role of intra-articular injection of MSCs on cartilage regeneration in haemophilia. Haemophilia 2015; 21:693-701. [DOI: 10.1111/hae.12659] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2015] [Indexed: 12/12/2022]
Affiliation(s)
- R. Ravanbod
- Department of Physical Therapy; Biomechanical Research Laboratory; Tarbiat Modares University; Tehran Iran
| | - G. Torkaman
- Department of Physical Therapy; Biomechanical Research Laboratory; Tarbiat Modares University; Tehran Iran
| | - M. Mophid
- Department of Histology; Baquiyatallah University of Medical Sciences; Tehran Iran
| | - F. Mohammadali
- Department of Hematology; Tarbiat Modares University; Tehran Iran
| |
Collapse
|
19
|
Shim G, Lee S, Han J, Kim G, Jin H, Miao W, Yi TG, Cho YK, Song SU, Oh YK. Pharmacokinetics and in vivo fate of intra-articularly transplanted human bone marrow-derived clonal mesenchymal stem cells. Stem Cells Dev 2015; 24:1124-32. [PMID: 25519508 DOI: 10.1089/scd.2014.0240] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In this study, we report the pharmacokinetics and in vivo fate of intra-articularly transplanted human mesenchymal stem cells (MSCs) in comparison with those of intravenously administered cells. Bone marrow-derived human clonal mesenchymal stem cells (hcMSCs) were transplanted to nude mice through intravenous or intra-articular routes. The numbers of hcMSCs in blood and tissue samples were measured by the quantitative real-time-polymerase chain reaction (qPCR) with human Alu (hAlu) as a detection marker. Following intra-articular transplantation, the blood levels of hcMSCs peaked 8 h postdose and gradually diminished, showing a 95-fold higher mean residence time than hcMSCs delivered through the intravenous route. Unlike intravenously administered hcMSCs, intra-articularly injected hcMSCs were mainly retained at injection joint sites where their levels 8 h postdose were 116-fold higher than those in muscle tissues. Regardless of injection routes, biodistribution patterns did not significantly differ between normal and osteoarthritis-induced mice. Quantitative analysis using hAlu-specific qPCR revealed that hcMSC levels in joint tissues were significantly higher than those in muscle tissues 120 days postdose. These dramatic differences in kinetic behavior and fate of intra-articularly transplanted hcMSCs compared with intravenously administered hcMSCs may provide insights useful for the development of human MSCs for arthritis therapeutics.
Collapse
Affiliation(s)
- Gayong Shim
- 1 Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Hemophilia is an X-linked inherited bleeding disorder consisting of two classifications, hemophilia A and hemophilia B, depending on the underlying mutation. Although the disease is currently treatable with intravenous delivery of replacement recombinant clotting factor, this approach represents a significant cost both monetarily and in terms of quality of life. Gene therapy is an attractive alternative approach to the treatment of hemophilia that would ideally provide life-long correction of clotting activity with a single injection. In this review, we will discuss the multitude of approaches that have been explored for the treatment of both hemophilia A and B, including both in vivo and ex vivo approaches with viral and nonviral delivery vectors.
Collapse
Affiliation(s)
- Geoffrey L Rogers
- University of Florida, Department of Pediatrics, Division of Cellular and Molecular Therapy, Gainesville, FL 32610
| | - Roland W Herzog
- University of Florida, Department of Pediatrics, Division of Cellular and Molecular Therapy, Gainesville, FL 32610
| |
Collapse
|
21
|
Kashiwakura Y, Ohmori T, Mimuro J, Madoiwa S, Inoue M, Hasegawa M, Ozawa K, Sakata Y. Production of functional coagulation factor VIII from iPSCs using a lentiviral vector. Haemophilia 2014; 20:e40-4. [PMID: 24354485 DOI: 10.1111/hae.12311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2013] [Indexed: 12/17/2022]
Abstract
The use of induced pluripotent stem cells (iPSCs) as an autologous cell source has shed new light on cell replacement therapy with respect to the treatment of numerous hereditary disorders. We focused on the use of iPSCs for cell-based therapy of haemophilia. We generated iPSCs from mesenchymal stem cells that had been isolated from C57BL/6 mice. The mouse iPSCs were generated through the induction of four transcription factor genes Oct3/4, Klf-4, Sox-2 and c-Myc. The derived iPSCs released functional coagulation factor VIII (FVIII) following transduction with a simian immunodeficiency virus vector. The subcutaneous transplantation of iPSCs expressing FVIII into nude mice resulted in teratoma formation, and significantly increased plasma levels of FVIII. The plasma concentration of FVIII was at levels appropriate for human therapy at 2-4 weeks post transplantation. Our data suggest that iPSCs could be an attractive and prospective autologous cell source for the production of coagulation factor, and that engineered iPSCs expressing coagulation factor might provide a cell-based therapeutic strategy appropriate for haemophilia.
Collapse
Affiliation(s)
- Y Kashiwakura
- Research Division of Cell and Molecular Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan; Department of Immunology, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Consistently measurable and persistent expression of circulating clotting factor activity, associated with decreased clinical bleeding, has been achieved for the first time in a hemophilia gene therapy trial. This review examines the successes and limitations of this clinical trial for hemophilia B and approaches to advance beyond this milestone. RECENT FINDINGS Although a self-complementary serotype 8 adeno-associated virus (scAAV8) vector approach directed factor IX expression of up to 6% in a human trial, the apparent need to suppress vector dose-dependent immune-mediated liver inflammation in some patients at the highest dose highlighted the next steps to optimize the risk-benefit of hemophilia gene therapy. The approaches being pursued include manufacturing modifications to eliminate contaminating empty vector capsids, the utilization of factor IX and factor VIII modified transgenes to improve secretion or function of the transgene product, and adjunctive pharmacologic and molecular approaches to overcome limitations imposed by naturally occurring antibodies against vectors and by the large size of the factor VIII gene. SUMMARY Preclinical data suggest strategies in development may build upon the first gene therapy success and achieve factor IX correction sufficient to prevent bleeding without toxicity and translate success to hemophilia A gene therapy.
Collapse
|