1
|
Perrier A, Pugnet G, Chaput B, Gandolfi S. Hand surgical and injectable treatments in systemic sclerosis: A systematic review of published cases. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2025:23971983251348059. [PMID: 40538942 PMCID: PMC12174094 DOI: 10.1177/23971983251348059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Accepted: 05/20/2025] [Indexed: 06/22/2025]
Abstract
Introduction Systemic sclerosis is a systemic autoimmune disease that can affect the hands leading to a deterioration in function. Unfortunately, current medical treatments are considered insufficient to improve patients' quality of life. However, innovative surgical and injectable treatments are available. Patients and methods A systematic review of the literature on surgical and injectable treatments available to hand surgeons was carried out according to the PRISMA criteria. All the articles selected were analysed qualitatively and descriptively, and where possible the extracted data were summarised quantitatively. Results and discussion Twenty-nine articles were therefore included, comprising 704 patients and 546 procedures. Nine different treatments were identified. Botulinum toxin injections have shown promising clinical results in the treatment of Raynaud's phenomenon, digital ulcers and pain, but have not been confirmed in randomised placebo-controlled trials. Injections of adipose-derived stromal vascular fraction showed positive results in Raynaud's phenomenon, digital ulcer, pain and function, but not in randomised placebo-controlled trials. Autologous fat grafting has shown significant results in Raynaud Phenomenon (RP) and digital ulcers (DUs) healing and prevention. Peripheral sympathectomy has shown moderate beneficial effects with frequent adverse events, and should be reserved for severe cases. Transluminal angioplasty has shown encouraging results, but studies with a high level of evidence are needed. Finally, symptomatic calcinosis cutis should be treated by resection or carbon dioxide (CO2) laser. A treatment algorithm is provided in the Supplemental Appendix. Conclusion Interventional treatments for scleroderma of the hand have shown an overall trend of efficacy, but some of the beneficial effects have not been found in studies with a high level of evidence. Further well-conducted studies are needed.
Collapse
Affiliation(s)
- Adrien Perrier
- Department of Plastic and Reconstructive Surgery, Hôpital Rangueil, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Gregory Pugnet
- Department of Internal Medicine, Hôpital Rangueil, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Benoit Chaput
- Department of Plastic and Reconstructive Surgery, Hôpital Rangueil, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Silvia Gandolfi
- Department of Plastic and Reconstructive Surgery, Hôpital Rangueil, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| |
Collapse
|
2
|
Kostecka A, Kalamon N, Skoniecka A, Koczkowska M, Skowron PM, Piotrowski A, Pikuła M. Adipose-derived mesenchymal stromal cells in clinical trials: Insights from single-cell studies. Life Sci 2024; 351:122761. [PMID: 38866216 DOI: 10.1016/j.lfs.2024.122761] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/15/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Mesenchymal Stromal Cells (MSCs) offer tremendous potential for the treatment of various diseases and their healing properties have been explored in hundreds of clinical trials. These trails primarily focus on immunological and neurological disorders, as well as regenerative medicine. Adipose tissue is a rich source of mesenchymal stromal cells and methods to obtain and culture adipose-derived MSCs (AD-MSCs) have been well established. Promising results from pre-clinical testing of AD-MSCs activity prompted clinical trials that further led to the approval of AD-MSCs for the treatment of complex perianal fistulas in Crohn's disease and subcutaneous tissue defects. However, AD-MSC heterogeneity along with various manufacturing protocols or different strategies to boost their activity create the need for standardized quality control procedures and safety assessment of the intended cell product. High-resolution transcriptomic methods have been recently gaining attention, as they deliver insight into gene expression profiles of individual cells, helping to deconstruct cellular hierarchy and differentiation trajectories, and to understand cell-cell interactions within tissues. This article presents a comprehensive overview of completed clinical trials evaluating the safety and efficacy of AD-MSC treatment, together with current single-cell studies of human AD-MSC. Furthermore, our work emphasizes the increasing significance of single-cell research in elucidating the mechanisms of cellular action and predicting their therapeutic effects.
Collapse
Affiliation(s)
- Anna Kostecka
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland; 3P - Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
| | - Natalia Kalamon
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland.
| | - Aneta Skoniecka
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, 80-211 Gdańsk, Poland.
| | - Magdalena Koczkowska
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland; 3P - Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland.
| | - Arkadiusz Piotrowski
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland; 3P - Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
| | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, 80-211 Gdańsk, Poland.
| |
Collapse
|
3
|
Lee BW, Kwok SK. Mesenchymal Stem/Stromal Cell-Based Therapies in Systemic Rheumatic Disease: From Challenges to New Approaches for Overcoming Restrictions. Int J Mol Sci 2023; 24:10161. [PMID: 37373308 PMCID: PMC10299481 DOI: 10.3390/ijms241210161] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Systemic rheumatic diseases, such as rheumatoid arthritis, systemic lupus erythematosus, and systemic sclerosis, are chronic autoimmune diseases affecting multiple organs and tissues. Despite recent advances in treatment, patients still experience significant morbidity and disability. Mesenchymal stem/stromal cell (MSC)-based therapy is promising for treating systemic rheumatic diseases due to the regenerative and immunomodulatory properties of MSCs. However, several challenges need to be overcome to use MSCs in clinical practice effectively. These challenges include MSC sourcing, characterization, standardization, safety, and efficacy issues. In this review, we provide an overview of the current state of MSC-based therapies in systemic rheumatic diseases, highlighting the challenges and limitations associated with their use. We also discuss emerging strategies and novel approaches that can help overcome the limitations. Finally, we provide insights into the future directions of MSC-based therapies for systemic rheumatic diseases and their potential clinical applications.
Collapse
Affiliation(s)
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| |
Collapse
|
4
|
Liu YX, Sun JM, Ho CK, Gao Y, Wen DS, Liu YD, Huang L, Zhang YF. Advancements in adipose-derived stem cell therapy for skin fibrosis. World J Stem Cells 2023; 15:342-353. [PMID: 37342214 PMCID: PMC10277960 DOI: 10.4252/wjsc.v15.i5.342] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/30/2023] [Accepted: 04/13/2023] [Indexed: 05/26/2023] Open
Abstract
Pathological scarring and scleroderma, which are the most common conditions of skin fibrosis, pathologically manifest as fibroblast proliferation and extracellular matrix (ECM) hyperplasia. Fibroblast proliferation and ECM hyperplasia lead to fibrotic tissue remodeling, causing an exaggerated and prolonged wound-healing response. The pathogenesis of these diseases has not been fully clarified and is unfortunately accompanied by exceptionally high medical needs and poor treatment effects. Currently, a promising and relatively low-cost treatment has emerged-adipose-derived stem cell (ASC) therapy as a branch of stem cell therapy, including ASCs and their derivatives-purified ASC, stromal vascular fraction, ASC-conditioned medium, ASC exosomes, etc., which are rich in sources and easy to obtain. ASCs have been widely used in therapeutic settings for patients, primarily for the defection of soft tissues, such as breast enhancement and facial contouring. In the field of skin regeneration, ASC therapy has become a hot research topic because it is beneficial for reversing skin fibrosis. The ability of ASCs to control profibrotic factors as well as anti-inflammatory and immunomodulatory actions will be discussed in this review, as well as their new applications in the treatment of skin fibrosis. Although the long-term effect of ASC therapy is still unclear, ASCs have emerged as one of the most promising systemic antifibrotic therapies under development.
Collapse
Affiliation(s)
- Yu-Xin Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Jia-Ming Sun
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Chia-Kang Ho
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Ya Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Dong-Sheng Wen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Yang-Dan Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Lu Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Yi-Fan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| |
Collapse
|
5
|
Adipose Tissue and Adipose-Tissue-Derived Cell Therapies for the Treatment of the Face and Hands of Patients Suffering from Systemic Sclerosis. Biomedicines 2023; 11:biomedicines11020348. [PMID: 36830886 PMCID: PMC9953720 DOI: 10.3390/biomedicines11020348] [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: 12/30/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 01/28/2023] Open
Abstract
Adipose tissue is recognized as a valuable source of cells with angiogenic, immunomodulatory, reparative and antifibrotic properties and emerged as a therapeutic alternative for the regeneration and repair of damaged tissues. The use of adipose-tissue-based therapy is expanding in autoimmune diseases, particularly in Systemic Sclerosis (SSc), a disease in which hands and face are severely affected, leading to disability and a decrease in quality of life. Combining the advantage of an abundant supply of fat tissue and a high abundance of stem/stromal cells, fat grafting and adipose tissue-derived cell-based therapies are attractive therapeutic options in SSc. This review aims to synthesize the evidence to determine the effects of the use of these biological products for face and hands treatment in the context of SSc. This highlights several points: the need to use relevant effectiveness criteria taking into account the clinical heterogeneity of SSc in order to facilitate assessment and comparison of innovative therapies; second, it reveals some impacts of the disease on fat-grafting success; third, an important heterogeneity was noticed regarding the manufacturing of the adipose-derived products and lastly, it shows a lack of robust evidence from controlled trials comparing adipose-derived products with standard care.
Collapse
|
6
|
Xue E, Minniti A, Alexander T, Del Papa N, Greco R, on behalf of The Autoimmune Diseases Working Party (ADWP) of the European Society for Blood and Marrow Transplantation (EBMT). Cellular-Based Therapies in Systemic Sclerosis: From Hematopoietic Stem Cell Transplant to Innovative Approaches. Cells 2022; 11:3346. [PMID: 36359742 PMCID: PMC9658618 DOI: 10.3390/cells11213346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 08/28/2023] Open
Abstract
Systemic sclerosis (SSc) is a systemic disease characterized by autoimmune responses, vasculopathy and tissue fibrosis. The pathogenic mechanisms involve a wide range of cells and soluble factors. The complexity of interactions leads to heterogeneous clinical features in terms of the extent, severity, and rate of progression of skin fibrosis and internal organ involvement. Available disease-modifying drugs have only modest effects on halting disease progression and may be associated with significant side effects. Therefore, cellular therapies have been developed aiming at the restoration of immunologic self-tolerance in order to provide durable remissions or to foster tissue regeneration. Currently, SSc is recommended as the 'standard indication' for autologous hematopoietic stem cell transplantation by the European Society for Blood and Marrow Transplantation. This review provides an overview on cellular therapies in SSc, from pre-clinical models to clinical applications, opening towards more advanced cellular therapies, such as mesenchymal stem cells, regulatory T cells and potentially CAR-T-cell therapies.
Collapse
Affiliation(s)
- Elisabetta Xue
- Hematopoietic and Bone Marrow Transplant Unit, San Raffaele Hospital, 20132 Milan, Italy
| | - Antonina Minniti
- Department of Rheumatology, ASST G. Pini-CTO, 20122 Milan, Italy
| | - Tobias Alexander
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117 Berlin, Germany
| | | | - Raffaella Greco
- Hematopoietic and Bone Marrow Transplant Unit, San Raffaele Hospital, 20132 Milan, Italy
| | | |
Collapse
|
7
|
Khanna D, Caldron P, Martin RW, Kafaja S, Spiera R, Shahouri S, Shah A, Hsu V, Ervin J, Simms R, Domsic RT, Steen V, Hummers LK, Derk C, Mayes M, Chatterjee S, Varga J, Kesten S, Fraser JK, Furst DE. Adipose-Derived Regenerative Cell Transplantation for the Treatment of Hand Dysfunction in Systemic Sclerosis: A Randomized Clinical Trial. Arthritis Rheumatol 2022; 74:1399-1408. [PMID: 35358372 PMCID: PMC9544105 DOI: 10.1002/art.42133] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/08/2022] [Accepted: 03/25/2022] [Indexed: 11/25/2022]
Abstract
Objective Hand dysfunction is common in systemic sclerosis (SSc). We undertook this study to evaluate the capacity of autologous adipose‐derived regenerative cells (ADRCs) to improve hand function in SSc patients. Methods The Scleroderma Treatment with Celution Processed Adipose Derived Regenerative Cells Trial was a prospective, randomized, double‐blind trial of ADRCs, in which ADRCs were obtained from patients with SSc by small‐volume adipose tissue harvest, and the fingers of each patient were injected with ADRCs. The primary end point was change in hand function at 24 and 48 weeks, assessed using the Cochin Hand Function Scale (CHFS). One of the secondary end points included the change in Health Assessment Questionnaire disability index (HAQ DI) at 48 weeks. Separate prespecified analyses were performed for patients with diffuse cutaneous SSc (dcSSc) and those with limited cutaneous SSc (lcSSc). Results Eighty‐eight patients were randomized to receive ADRCs (n = 48 [32 patients with dcSSc and 16 with lcSSc]) or placebo (n = 40 [19 patients with dcSSc and 21 with lcSSc]). Change in hand function according to CHFS score was numerically higher for the ADRC group compared to the placebo group but did not achieve statistical significance (mean ± SD improvement in the CHFS score at 48 weeks 11.0 ± 12.5 versus 8.9 ± 10.5; P = 0.299). For patients with dcSSc, the between‐group difference in the CHFS at 48 weeks was 6.3 points (nominal P = 0.069). For the secondary end point, the dcSSc group exhibited a between‐group difference of 0.17 points in the HAQ DI (nominal P = 0.044) at 48 weeks. Of the ADRC‐treated patients with dcSSc, 52% reported improvement greater than the minimum clinically important difference for both CHFS and HAQ DI compared to 16% in the placebo group (nominal P = 0.016). Small‐volume adipose tissue harvest and ADRC treatment were well tolerated. Conclusion While the primary end point of this trial was not achieved, efficacy trends were observed in patients with dcSSc. Adipose tissue harvest and ADRC injection were demonstrated to be feasible. Further clinical trials of this intervention in the setting of dcSSc are warranted.
Collapse
Affiliation(s)
| | - Paul Caldron
- Arizona Arthritis & Rheumatology Research, Phoenix
| | | | | | | | | | - Ankoor Shah
- Duke University Medical Center, Durham, North Carolina
| | - Vivien Hsu
- Rutgers University, New Brunswick, New Jersey
| | - John Ervin
- Alliance for Multispecialty Research, Kansas City, Missouri
| | - Robert Simms
- Boston University Medical Center, Boston, Massachusetts
| | | | | | | | - Chris Derk
- University of Pennsylvania, Philadelphia
| | | | - Soumya Chatterjee
- Cleveland Clinic Orthopedic & Rheumatologic Institute, Cleveland, Ohio
| | - John Varga
- Northwestern University, Chicago, Illinois, and University of Michigan, Ann Arbor
| | | | | | | |
Collapse
|
8
|
Efficacy and safety of mesenchymal stem cells in the treatment of systemic sclerosis: a systematic review and meta-analysis. Stem Cell Res Ther 2022; 13:118. [PMID: 35313985 PMCID: PMC8935249 DOI: 10.1186/s13287-022-02786-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/17/2022] [Indexed: 11/28/2022] Open
Abstract
Background Systemic sclerosis (SSc) is an autoimmune disease with high morbidity and mortality characterized by fibrosis of the skin and internal organs. Some studies have investigated the use of stem cells to treat SSc. Herein, a systematic review and meta-analysis was conducted to determine the efficacy and safety of mesenchymal stem cells (MSCs) in the treatment of SSc. Methods PubMed, Embase, Cochrane Library, Web of Science, OVID, China National Knowledge Infrastructure and Wanfang databases were searched up to February 1, 2021. Literature screening, data extraction and quality assessment were conducted independently by two researchers in according to the inclusion and exclusion criteria. The discrepancies were resolved by a third researcher. Results A total of 9 studies encompassing 133 SSc patients were included in the study. Compared to the baseline after treatment with MSCs: 1. The modified Rodnan skin score (mRSS) was significantly reduced in patients with SSc (P < 0.00001). 2. MSCs decreased the number of digital ulcer, mouth handicap scale, and visual analog scale of hand pain in SSc patients (P = 0.0007 and P = 0.03, respectively). 3. No statistical differences were detected in Raynaud's condition score and Cochin hand function scale score at 6 months of MSCs therapy (P = 0.5 and P = 0.62). 4. After 12 months of follow-up, MSCs improve carbon monoxide diffusing capacity and forced vital capacity of SSc patients (P < 0.05). 5. Overall, MSCs application was safe; a few cases exhibited swelling at the injection site, diarrhea and arthralgia, which had self-recovery, and no severe adverse events occurred in the included trials. Conclusions MSC therapy improves the degree of skin thickening, lung function, and mouth opening and relieves finger ulcers and pain in patients with SSc without severe adverse events. Thus, MSCs or MSCs combined with plasma and traditional medicine might be an effective and promising treatment of SSc patients. PROSPERO registration number: CRD42020200350
Collapse
|
9
|
Guillo L, Grimaud F, Houser F, Prost C, Jouve E, Philandrianos C, Abellan M, Veran J, Visee C, Beyer-Berjot L, Desjeux A, Dignat-George F, Leone M, Grimaud JC, Sabatier F, Serrero M, Magalon J. Three-year outcome of local injection of autologous stromal vascular fraction cells and microfat in refractory perianal fistulas of Crohn's disease. Stem Cell Res Ther 2022; 13:67. [PMID: 35139888 PMCID: PMC8827195 DOI: 10.1186/s13287-022-02738-x] [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: 05/10/2021] [Accepted: 01/20/2022] [Indexed: 11/10/2022] Open
Abstract
Perianal fistulas in Crohn's disease are frequent and disabling, with a major impact on patients' quality of life. Cell-based therapy using mesenchymal stem cells represents new hope for these patients, but long-term efficacy remains challenging. In a pilot study, including patients with refractory complex perianal fistulas, autologous adipose-derived stromal vascular fraction (ADSVF) combined with microfat achieved combined remission in 60% of cases, with a good safety profile at 1 year. The purpose of this study is to assess whether these results were maintained at longer term. The safety and efficacy data of the ten patients were evaluated retrospectively 3 years after injection on the basis of clinical and radiological data. MRI were analysed according to the MAGNIFI-CD score. No adverse event was attributed to the experimental stem-cell treatment. Combined remission was achieved in 7 patients (70%) and associated with a significant improvement in the MAGNIFI-CD MRI score. In conclusion, the safety and efficacy of ADSVF and microfat injection in Crohn's disease fistulas were maintained at 3 years, demonstrating that this innovative strategy is effective in producing a long-lasting healing effect. The ongoing multicentre randomized placebo-controlled trial (NCT04010526) will be helpful to define the place for this approach in the current therapeutic arsenal.
Collapse
Affiliation(s)
- Lucas Guillo
- Gastroenterology Department, Assistance Publique Hôpitaux de Marseille, University Hospital of Marseille Nord (AP-HM), Chemin des Bourrely, 13015, Marseille, France. .,Cell Therapy Department, INSERM CBT-1409, Assistance Publique Hôpitaux de Marseille, Aix-Marseille University, Marseille, France.
| | - Fanny Grimaud
- Cell Therapy Department, INSERM CBT-1409, Assistance Publique Hôpitaux de Marseille, Aix-Marseille University, Marseille, France
| | - Fanny Houser
- Gastroenterology Department, Assistance Publique Hôpitaux de Marseille, University Hospital of Marseille Nord (AP-HM), Chemin des Bourrely, 13015, Marseille, France
| | - Caroline Prost
- Department of Radiology, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Elisabeth Jouve
- Statistical Department, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Cécile Philandrianos
- Plastic Surgery Department, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Maxime Abellan
- Plastic Surgery Department, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Julie Veran
- Cell Therapy Department, INSERM CBT-1409, Assistance Publique Hôpitaux de Marseille, Aix-Marseille University, Marseille, France
| | - Carine Visee
- Digestive Surgery Department, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Laura Beyer-Berjot
- Digestive Surgery Department, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Ariadne Desjeux
- Gastroenterology Department, Assistance Publique Hôpitaux de Marseille, University Hospital of Marseille Nord (AP-HM), Chemin des Bourrely, 13015, Marseille, France
| | - Françoise Dignat-George
- C2VN, INSERM UMR 1263, Faculté de Pharmacie de Marseille, Aix Marseille University, Marseille, France
| | - Marc Leone
- Department of Anaesthesia and Intensive Care, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Jean-Charles Grimaud
- Gastroenterology Department, Assistance Publique Hôpitaux de Marseille, University Hospital of Marseille Nord (AP-HM), Chemin des Bourrely, 13015, Marseille, France.,Centre d'Investigation Clinique 1409, Assistance Publique Hôpitaux de Marseille, Aix Marseille University, Marseille, France
| | - Florence Sabatier
- Cell Therapy Department, INSERM CBT-1409, Assistance Publique Hôpitaux de Marseille, Aix-Marseille University, Marseille, France.,C2VN, INSERM UMR 1263, Faculté de Pharmacie de Marseille, Aix Marseille University, Marseille, France
| | - Mélanie Serrero
- Gastroenterology Department, Assistance Publique Hôpitaux de Marseille, University Hospital of Marseille Nord (AP-HM), Chemin des Bourrely, 13015, Marseille, France
| | - Jérémy Magalon
- Cell Therapy Department, INSERM CBT-1409, Assistance Publique Hôpitaux de Marseille, Aix-Marseille University, Marseille, France.,C2VN, INSERM UMR 1263, Faculté de Pharmacie de Marseille, Aix Marseille University, Marseille, France
| |
Collapse
|
10
|
Velier M, Daumas A, Simoncini S, Arcani R, Magalon J, Benyamine A, Granel B, Dignat George F, Chabannon C, Sabatier F. Combining systemic and locally applied cellular therapies for the treatment of systemic sclerosis. Bone Marrow Transplant 2022; 57:17-22. [PMID: 34663928 DOI: 10.1038/s41409-021-01492-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/03/2021] [Accepted: 09/29/2021] [Indexed: 02/08/2023]
Abstract
Systemic sclerosis (SSc) is a complex autoimmune disease characterized by a functional and structural alteration of the microvascular network associated with cutaneous and visceral fibrosis lesions. Conventional therapies are based on the use of immunomodulatory molecules and symptomatic management but often prove to be insufficient, particularly for patients suffering from severe and rapidly progressive forms of the disease. In this context, cellular therapy approaches could represent a credible solution with the goal to act on the different components of the disease: the immune system, the vascular system and the extracellular matrix. The purpose of this review is to provide an overview of the cellular therapies available for the management of SSc. The first part will focus on systemically injected therapies, whose primary effect is based on immunomodulatory properties and immune system resetting, including autologous hematopoietic stem cell transplantation and intravenous injection of mesenchymal stem cells. The second part will discuss locally administered regenerative cell therapies, mainly derived from adipose tissue, developed for the management of local complications as hand and face disabilities.
Collapse
Affiliation(s)
- Mélanie Velier
- C2VN, Aix Marseille Univ, INSERM, INRA, Marseille, France. .,Laboratoire de Culture et Thérapie Cellulaire, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France.
| | - Aurélie Daumas
- C2VN, Aix Marseille Univ, INSERM, INRA, Marseille, France.,Service de Médecine Interne, Gériatrie et Thérapeutique, Hôpital La Timone, AP-HM, Marseille, France
| | | | - Robin Arcani
- C2VN, Aix Marseille Univ, INSERM, INRA, Marseille, France.,Service de Médecine Interne, Gériatrie et Thérapeutique, Hôpital La Timone, AP-HM, Marseille, France
| | - Jérémy Magalon
- C2VN, Aix Marseille Univ, INSERM, INRA, Marseille, France.,Laboratoire de Culture et Thérapie Cellulaire, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France
| | - Audrey Benyamine
- C2VN, Aix Marseille Univ, INSERM, INRA, Marseille, France.,Service de Médecine Interne, Hôpital Nord, pôle MICA, AP-HM, Marseille, France
| | - Brigitte Granel
- C2VN, Aix Marseille Univ, INSERM, INRA, Marseille, France.,Service de Médecine Interne, Hôpital Nord, pôle MICA, AP-HM, Marseille, France
| | | | - Christian Chabannon
- Centre de Thérapie Cellulaire et INSERM CIC BT-1409, Institut Paoli-Calmettes Comprehensive Cancer Center, Marseille, France
| | - Florence Sabatier
- C2VN, Aix Marseille Univ, INSERM, INRA, Marseille, France.,Laboratoire de Culture et Thérapie Cellulaire, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France
| |
Collapse
|
11
|
Hughes M, Allanore Y, El Aoufy K, Denton CP, Khanna D, Krieg T, Matucci-Cerinic M. A Practical Approach to the Management of Digital Ulcers in Patients With Systemic Sclerosis: A Narrative Review. JAMA Dermatol 2021; 157:851-858. [PMID: 34037677 DOI: 10.1001/jamadermatol.2021.1463] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Importance Digital ulcers (DUs) occurring on the fingers in patients with systemic sclerosis (SSc) are associated with substantial pain and disability and are often challenging to treat. However, careful clinical assessment and prompt intervention (wound bed management and systemic pharmacologic treatment) may modify the clinical course. Objectives To provide a practical approach to the assessment and management of SSc-DUs and highlight unmet needs and research priorities. Evidence Review A narrative review of the extant literature was undertaken to provide a broad overview of current knowledge and augmented by expert opinion. Findings Half of the patients with SSc have a history of DUs, and there is a point of prevalence of approximately 10%. Digital ulcers are often very painful and affect all aspects of physical, social, and family life as well as occupation. Digital ulcers are associated with a severe disease course. Systemic sclerosis DUs, particularly those occurring on the fingertips, represent a vascular ischemic complication, although other etiopathogenic factors play an important role. To guide management, a structured clinical approach is required, including DU definition, classification, and categorization. Digital ulcers require a multidisciplinary approach with close cooperation between physicians and specialist nursing and other allied health professionals to guarantee the appropriate treatment and provide patient education. Local wound bed management is necessary for all DUs and is combined with systemic (pharmacologic) treatments. When treating a DU, the clinician should actively review the therapeutic strategy to prevent further DUs, including the level of systemic disease control, and monitor closely for the development of DU complications, including infection and progression to gangrene. Despite a wide available therapeutic armory, a number of unmet needs and challenges remain that that require resolution to optimize DU management. Conclusions and Relevance A practical approach to DU management, including local wound bed management and systemic treatments, is useful. Digital ulcers are of interest to a broad range of dermatologists, rheumatologists, and other physicians providing care for patients with SSc. Careful clinical assessment and prompt intervention can substantially modify the clinical course of DUs in SSc.
Collapse
Affiliation(s)
- Michael Hughes
- Department of Rheumatology, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Yannick Allanore
- Department of Rheumatology, Cochin Hospital, AP-HP, Paris Descartes University, Paris, France
| | - Khadija El Aoufy
- Department of Experimental and Clinical Medicine, University of Florence, Division of Rheumatology AOUC, Florence, Italy
| | - Christopher P Denton
- Centre of Rheumatology, Royal Free Hospital, University College London, London, United Kingdom
| | - Dinesh Khanna
- Scleroderma Program, University of Michigan, Ann Arbor
| | - Thomas Krieg
- Translational Matrix Biology and Department for Dermatology, Medical Faculty, Cologne, Germany
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, University of Florence, Division of Rheumatology AOUC, Florence, Italy
| |
Collapse
|
12
|
Hughes M, Bruni C, Ruaro B, Confalonieri M, Matucci-Cerinic M, Bellando-Randone S. Digital Ulcers in Systemic Sclerosis. Presse Med 2021; 50:104064. [PMID: 33548375 DOI: 10.1016/j.lpm.2021.104064] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/29/2021] [Indexed: 12/28/2022] Open
Abstract
Digital ulcers (DU) are one of the most common complication of Systemic Sclerosis (SSc)-related vasculopathy and represent an important burden for the patients as well as for the society. Still today there is no agreement on the definition, classification and cathegorization of DU even if they are of pivotal importance in clinical practice, for treatment choice and prognostic outcomes, as well as for clinical trials. DU management requires a dedicated multidisciplinary team, that must remain ever vigilant for the development of infective complications and gangrene throughout their disease course, as well as patient education that is crucial to obtain the best compliance to assure the success of the treatment. Currently several drugs are available for DU treatment but in the future, more investigations will be needed to ameliorate the approach and the systemic and local therapies.
Collapse
Affiliation(s)
- Michael Hughes
- Department of Rheumatology, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Cosimo Bruni
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Firenze, & Department of Geriatric Medicine, Division of Rheumatology AOUC, Firenze, Italy
| | - Barbara Ruaro
- Pulmonology Department, University Hospital of Cattinara, University of Trieste, Trieste, Italy
| | - Marco Confalonieri
- Pulmonology Department, University Hospital of Cattinara, University of Trieste, Trieste, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Firenze, & Department of Geriatric Medicine, Division of Rheumatology AOUC, Firenze, Italy
| | - Silvia Bellando-Randone
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Firenze, & Department of Geriatric Medicine, Division of Rheumatology AOUC, Firenze, Italy.
| |
Collapse
|
13
|
Rosa I, Romano E, Fioretto BS, Matucci-Cerinic M, Manetti M. Adipose-derived stem cells: Pathophysiologic implications vs therapeutic potential in systemic sclerosis. World J Stem Cells 2021; 13:30-48. [PMID: 33584978 PMCID: PMC7859990 DOI: 10.4252/wjsc.v13.i1.30] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) residing in the stromal vascular fraction (SVF) of white adipose tissue are recently emerging as an alternative tool for stem cell-based therapy in systemic sclerosis (SSc), a complex connective tissue disorder affecting the skin and internal organs with fibrotic and vascular lesions. Several preclinical and clinical studies have reported promising therapeutic effects of fat grafting and autologous SVF/ADSC-based local treatment for facial and hand cutaneous manifestations of SSc patients. However, currently available data indicate that ADSCs may represent a double-edged sword in SSc, as they may exhibit a pro-fibrotic and anti-adipogenic phenotype, possibly behaving as an additional pathogenic source of pro-fibrotic myofibroblasts through the adipocyte-to-myofibroblast transition process. Thus, in the perspective of a larger employ of SSc-ADSCs for further therapeutic applications, it is important to definitely unravel whether these cells present a comparable phenotype and similar immunosuppressive, anti-inflammatory, anti-fibrotic and pro-angiogenic properties in respect to healthy ADSCs. In light of the dual role that ADSCs seem to play in SSc, this review will provide a summary of the most recent insights into the preclinical and clinical studies employing SVF and ADSCs for the treatment of the disease and, at the same time, will focus on the main findings highlighting the possible involvement of these stem cells in SSc-related fibrosis pathogenesis.
Collapse
Affiliation(s)
- Irene Rosa
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence 50134, Italy
| | - Eloisa Romano
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence 50134, Italy
| | - Bianca Saveria Fioretto
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence 50134, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence 50134, Italy
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence 50134, Italy.
| |
Collapse
|
14
|
Mattei A, Bertrand B, Jouve E, Blaise T, Philandrianos C, Grimaud F, Giraudo L, Aboudou H, Dumoulin C, Arnaud L, Revis J, Galant C, Velier M, Veran J, Dignat-George F, Dessi P, Sabatier F, Magalon J, Giovanni A. Feasibility of First Injection of Autologous Adipose Tissue-Derived Stromal Vascular Fraction in Human Scarred Vocal Folds: A Nonrandomized Controlled Trial. JAMA Otolaryngol Head Neck Surg 2021; 146:355-363. [PMID: 32053141 DOI: 10.1001/jamaoto.2019.4328] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Importance Patients with scarred vocal folds, whether congenitally or after phonosurgery, often exhibit dysphonia that negatively affects daily life and is difficult to treat. The autologous adipose tissue-derived stromal vascular fraction (ADSVF) is a readily accessible source of cells with angiogenic, anti-inflammatory, immunomodulatory, and regenerative properties. Objective To evaluate the feasibility and tolerability of local injections of autologous ADSVF in patients with scarred vocal folds. Design, Setting, and Participants CELLCORDES (Innovative Treatment for Scarred Vocal Cords by Local Injection of Autologous Stromal Vascular Fraction) is a prospective, open-label, single-arm, single-center, nonrandomized controlled trial with a 12-month follow-up and patient enrollment from April 1, 2016, to June 30, 2017. Eight patients with severe dysphonia attributable to vocal fold scarring associated with a congenital malformation or resulting from microsurgical sequelae (voice handicap index score >60 of 120) completed the study. Data analysis was performed from September 1, 2018, to January 1, 2019. Interventions Injection of ADSVF into 1 or 2 vocal folds. Main Outcomes and Measures The primary outcomes were feasibility and the number and severity of adverse events associated with ADSVF-based therapy. The secondary outcomes were changes in vocal assessment, videolaryngostroboscopy, self-evaluation of dysphonia, and quality of life at 1, 6, and 12 months after cell therapy. Results Seven women and 1 man (mean [SD] age, 44.6 [10.4] years) were enrolled in this study. Adverse events associated with liposuction and ADSVF injection occurred; most of them resolved spontaneously. One patient received minor treatment to drain local bruising, and another experienced a minor contour defect at the liposuction site. At 12 months, the voice handicap index score was improved in all patients, with a mean (SD) improvement from baseline of 40.1 (21.5) points. Seven patients (88%) were considered to be responders, defined as improvement by 18 points or more in the voice handicap index score (the minimum clinically important difference). Conclusions and Relevance The findings suggest that autologous ADSVF injection in scarred vocal folds is feasible and tolerable. The findings require confirmation in a randomized clinical trial with a larger population. Trial Registration ClinicalTrials.gov Identifier: NCT02622464.
Collapse
Affiliation(s)
- Alexia Mattei
- Department of Oto-Rhino-Laryngology and Head and Neck Surgery, Assistance Publique-Hôpitaux de Marseille, La Conception University Hospital, Marseille, France.,Laboratoire Parole et Langage, Centre National de la Recherche Scientifique, Aix Marseille University, Aix-en-Provence, France
| | - Baptiste Bertrand
- Department of Plastic and Reconstructive Surgery, Assistance Publique-Hôpitaux de Marseille, La Conception University Hospital, Marseille, France
| | - Elisabeth Jouve
- Assistance Publique-Hôpitaux de Marseille, Institut National de la Sante et de la Recherche Medicale, Institut de Neurosciences des Systèmes, Service de Pharmacologie Clinique et Pharmacovigilance, Centre d'Investigation Clinique Centre de Pharmacologie Clinique et d'Évaluation Thérapeutique, Aix Marseille University, Marseille, France
| | - Théo Blaise
- Assistance Publique-Hôpitaux de Marseille, Institut National de la Sante et de la Recherche Medicale, Institut de Neurosciences des Systèmes, Service de Pharmacologie Clinique et Pharmacovigilance, Centre d'Investigation Clinique Centre de Pharmacologie Clinique et d'Évaluation Thérapeutique, Aix Marseille University, Marseille, France
| | - Cécile Philandrianos
- Department of Plastic and Reconstructive Surgery, Assistance Publique-Hôpitaux de Marseille, La Conception University Hospital, Marseille, France
| | - Fanny Grimaud
- Cell Therapy Department, Assistance Publique-Hôpitaux de Marseille, Institut National de la Sante et de la Recherche Medicale, La Conception University Hospital, Aix Marseille University, Marseille, France
| | - Laurent Giraudo
- Cell Therapy Department, Assistance Publique-Hôpitaux de Marseille, Institut National de la Sante et de la Recherche Medicale, La Conception University Hospital, Aix Marseille University, Marseille, France
| | - Houssein Aboudou
- Cell Therapy Department, Assistance Publique-Hôpitaux de Marseille, Institut National de la Sante et de la Recherche Medicale, La Conception University Hospital, Aix Marseille University, Marseille, France
| | - Chloé Dumoulin
- Cell Therapy Department, Assistance Publique-Hôpitaux de Marseille, Institut National de la Sante et de la Recherche Medicale, La Conception University Hospital, Aix Marseille University, Marseille, France
| | - Laurent Arnaud
- Cell Therapy Department, Assistance Publique-Hôpitaux de Marseille, Institut National de la Sante et de la Recherche Medicale, La Conception University Hospital, Aix Marseille University, Marseille, France
| | - Joana Revis
- Department of Oto-Rhino-Laryngology and Head and Neck Surgery, Assistance Publique-Hôpitaux de Marseille, La Conception University Hospital, Marseille, France.,Laboratoire Parole et Langage, Centre National de la Recherche Scientifique, Aix Marseille University, Aix-en-Provence, France
| | - Camille Galant
- Department of Oto-Rhino-Laryngology and Head and Neck Surgery, Assistance Publique-Hôpitaux de Marseille, La Conception University Hospital, Marseille, France.,Laboratoire Parole et Langage, Centre National de la Recherche Scientifique, Aix Marseille University, Aix-en-Provence, France
| | - Mélanie Velier
- Cell Therapy Department, Assistance Publique-Hôpitaux de Marseille, Institut National de la Sante et de la Recherche Medicale, La Conception University Hospital, Aix Marseille University, Marseille, France.,Institut National de la Sante et de la Recherche Medicale and Institut National de la Recherche Agronomique, Aix Marseille University, Centre Recherche en CardioVasculaire et Nutrition, Marseille, France
| | - Julie Veran
- Cell Therapy Department, Assistance Publique-Hôpitaux de Marseille, Institut National de la Sante et de la Recherche Medicale, La Conception University Hospital, Aix Marseille University, Marseille, France
| | - Françoise Dignat-George
- Cell Therapy Department, Assistance Publique-Hôpitaux de Marseille, Institut National de la Sante et de la Recherche Medicale, La Conception University Hospital, Aix Marseille University, Marseille, France.,Institut National de la Sante et de la Recherche Medicale and Institut National de la Recherche Agronomique, Aix Marseille University, Centre Recherche en CardioVasculaire et Nutrition, Marseille, France
| | - Patrick Dessi
- Department of Oto-Rhino-Laryngology and Head and Neck Surgery, Assistance Publique-Hôpitaux de Marseille, La Conception University Hospital, Marseille, France.,French National Centre for Scientific Research, Centre National de la Recherche Scientifique, Etablissement Français du Sang, Anthropologie bio-culturelle, Droit, Ethique et Santé, Aix Marseille University, Marseille, France
| | - Florence Sabatier
- Cell Therapy Department, Assistance Publique-Hôpitaux de Marseille, Institut National de la Sante et de la Recherche Medicale, La Conception University Hospital, Aix Marseille University, Marseille, France.,Institut National de la Sante et de la Recherche Medicale and Institut National de la Recherche Agronomique, Aix Marseille University, Centre Recherche en CardioVasculaire et Nutrition, Marseille, France
| | - Jérémy Magalon
- Cell Therapy Department, Assistance Publique-Hôpitaux de Marseille, Institut National de la Sante et de la Recherche Medicale, La Conception University Hospital, Aix Marseille University, Marseille, France.,Institut National de la Sante et de la Recherche Medicale and Institut National de la Recherche Agronomique, Aix Marseille University, Centre Recherche en CardioVasculaire et Nutrition, Marseille, France
| | - Antoine Giovanni
- Department of Oto-Rhino-Laryngology and Head and Neck Surgery, Assistance Publique-Hôpitaux de Marseille, La Conception University Hospital, Marseille, France.,Laboratoire Parole et Langage, Centre National de la Recherche Scientifique, Aix Marseille University, Aix-en-Provence, France
| |
Collapse
|
15
|
Herrick AL, Shukla R, Watson REB. Frontiers in translational systemic sclerosis research: A focus on the unmet 'cutaneous' clinical needs (Viewpoint). Exp Dermatol 2020; 29:1144-1153. [DOI: 10.1111/exd.14179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 08/06/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Ariane L. Herrick
- Division of Musculoskeletal and Dermatological Sciences Salford Royal NHS Foundation Trust Manchester Academic Health Science Centre The University of Manchester Manchester UK
| | | | - Rachel E. B. Watson
- Division of Musculoskeletal and Dermatological Sciences Salford Royal NHS Foundation Trust Manchester Academic Health Science Centre The University of Manchester Manchester UK
| |
Collapse
|
16
|
Abedi M, Alavi-Moghadam S, Payab M, Goodarzi P, Mohamadi-jahani F, Sayahpour FA, Larijani B, Arjmand B. Mesenchymal stem cell as a novel approach to systemic sclerosis; current status and future perspectives. CELL REGENERATION (LONDON, ENGLAND) 2020; 9:20. [PMID: 33258056 PMCID: PMC7704834 DOI: 10.1186/s13619-020-00058-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022]
Abstract
Systemic sclerosis is a rare chronic autoimmune disease with extensive microvascular injury, damage of endothelial cells, activation of immune responses, and progression of tissue fibrosis in the skin and various internal organs. According to epidemiological data, women's populations are more susceptible to systemic sclerosis than men. Until now, various therapeutic options are employed to manage the symptoms of the disease. Since stem cell-based treatments have developed as a novel approach to rescue from several autoimmune diseases, it seems that stem cells, especially mesenchymal stem cells as a powerful regenerative tool can also be advantageous for systemic sclerosis treatment via their remarkable properties including immunomodulatory and anti-fibrotic effects. Accordingly, we discuss the contemporary status and future perspectives of mesenchymal stem cell transplantation for systemic sclerosis.
Collapse
Affiliation(s)
- Mina Abedi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Mohamadi-jahani
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Forough Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Abstract
Systemic sclerosis (SSc) is a rare autoimmune disorder with a high mortality rate. There are still many unknowns concerning the pathophysiology of this disease, due to its clinical heterogeneity. Since there is still no curative treatment, researchers focus on finding novel methods to help the patients. One of the valid options is cellular therapy, and mesenchymal stem cells (MSCs)-based therapy yields great expectations. These cells possess especially valuable attributes regarding key points of SSc. Nevertheless, the effectiveness and safety of this therapy must undergo a rigorous process of verification. In preclinical trials, animal models proved to be a valuable source of scientific knowledge regarding SSc. Because of that, it has been possible to test autologous or allogeneic MSCs from various sources in many clinical trials. A lot of aspects still have to be determined to assess their potential in the management of SSc, probably in association with other therapies.
Collapse
|
18
|
Fallet B, Walker UA. Current immunosuppressive and antifibrotic therapies of systemic sclerosis and emerging therapeutic strategies. Expert Rev Clin Pharmacol 2020; 13:1203-1218. [PMID: 33008265 DOI: 10.1080/17512433.2020.1832466] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Systemic sclerosis (SSc) is a rare, difficult to treat disease with profound effects on quality of life and high mortality. Complex and incompletely understood pathophysiologic processes and greatly heterogeneous clinical presentations and outcomes have hampered drug development. AREAS COVERED This review summarizes the currently available immunosuppressive and antifibrotic therapies and discusses novel approaches for the treatment of SSc. We reviewed the literature using the MEDLINE and ClinicalTrial.gov databases between May and September 2020. EXPERT OPINION Available immunosuppressive and antifibrotic drugs only modestly impact the course of the disease. Most drugs are currently only investigated in the subset of patients with early diffuse cutaneous SSc. In this patient population, hematopoietic stem-cell transplantation is currently the only treatment that has demonstrated reversal of lung involvement, enhanced quality of life and reduced long-term mortality, but carries the risk of short-term treatment-related mortality. A great need to provide better therapeutic options to patients exists also for those patients who have limited cutaneous skin involvement. A better understanding of SSc pathophysiology has enabled the identification of numerous new therapeutic targets. The progress made in the design of clinical trials and outcome parameters will likely result in the improvement of effective management options.
Collapse
Affiliation(s)
- Bénédict Fallet
- Department of Rheumatology, University Hospital Basel , Basel, Switzerland
| | - Ulrich A Walker
- Department of Rheumatology, University Hospital Basel , Basel, Switzerland
| |
Collapse
|
19
|
Pignatti M, Spinella A, Cocchiara E, Boscaini G, Lusetti IL, Citriniti G, Lumetti F, Setti G, Dominici M, Salvarani C, De Santis G, Giuggioli D. Autologous Fat Grafting for the Oral and Digital Complications of Systemic Sclerosis: Results of a Prospective Study. Aesthetic Plast Surg 2020; 44:1820-1832. [PMID: 32632623 DOI: 10.1007/s00266-020-01848-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/14/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Systemic sclerosis is a connective tissue disease. Skin involvement of the mouth and hand may compromise function and quality of life. Autologous fat grafting has been described as a specific treatment of these clinical features. We report the results of our prospective study designed to treat and prevent skin complications in systemic sclerosis. MATERIALS AND METHODS We treated 25 patients with mouth and/or hand involvement (microstomia, xerostomia, skin sclerosis, Raynaud's phenomenon and long-lasting digital ulcers) with autologous fat grafting, according to the Coleman's technique, around the mouth and/or at the base of each finger. The surgical procedures were repeated in each patient every 6 months for a total of two or three times. Clinical data were collected before the first surgery and again 6 months after each surgical procedure. Pain, skin thickness, saliva production and disability were assessed with validated tests. RESULTS Overall we performed 63 autologous fat grafting sessions (either on the mouth, on the hands or on both anatomical areas). Results at 6 moths after the last session included improvement of xerostomia evaluated with a sialogram, reduction of the skin tension around the mouth and, in the hands, reduction of the Raynaud phenomenon as well as skin thickness. Pain was reduced while the perception of disability improved. Digital ulcers healed completely in 8/9 patients. CONCLUSIONS Our results confirm the efficacy and safety of autologous fat grafting for the treatment of skin complications and digital ulcers due to systemic sclerosis. In addition, the patients' subjective well-being improved. Level of evidence V This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
|
20
|
Park Y, Lee YJ, Koh JH, Lee J, Min HK, Kim MY, Kim KJ, Lee SJ, Rhie JW, Kim WU, Park SH, Moon SH, Kwok SK. Clinical Efficacy and Safety of Injection of Stromal Vascular Fraction Derived from Autologous Adipose Tissues in Systemic Sclerosis Patients with Hand Disability: A Proof-Of-Concept Trial. J Clin Med 2020; 9:jcm9093023. [PMID: 32961802 PMCID: PMC7565930 DOI: 10.3390/jcm9093023] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/22/2022] Open
Abstract
Background: Stromal vascular fraction (SVF) has recently emerged as a potential therapeutic modality, due to its multipotent cellular components in tissue regeneration. Systemic sclerosis (SSc) is a progressive autoimmune disease that results in hand disability by skin fibrosis and microangiopathies. We performed an open-label study to investigate the efficacy and safety of SVF injection in SSc patients (Clinical Trial number: NCT03060551). Methods: We gathered 20 SSc patients with hand disability, planning for a 24-week follow-up period. SVF was extracted from autologous adipose tissues, processed by the closed system kit, and injected into each finger of SSc patients. We observed various efficacy and safety profiles at each follow-up visit. Results: Among the 20 initially enrolled patients, eighteen received SVF injection, and were completely followed-up for the whole study period. Patients received 3.61 × 106 mesenchymal stem cells into each finger on average. Skin fibrosis, hand edema, and quality of life were significantly improved, and 31.6% of active ulcers were healed at 24 weeks after injections. Semiquantitative results of nailfold capillary microscopy were ameliorated. There was no single serious adverse event related to the procedure. Conclusions: Injection of SVF derived from autologous adipose tissues is tolerable, and shows clinical efficacy in SSc patients.
Collapse
Affiliation(s)
- Youngjae Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.P.); (J.L.); (W.-U.K.); (S.-H.P.)
| | - Yoon Jae Lee
- Department of Plastic Surgery, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea;
| | - Jung Hee Koh
- Division of Rheumatology, Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Bucheon 14647, Korea;
| | - Jennifer Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.P.); (J.L.); (W.-U.K.); (S.-H.P.)
| | - Hong-Ki Min
- Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Centre, Konkuk University School of Medicine, Seoul 05030, Korea;
| | - Moon Young Kim
- Division of Rheumatology, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Incheon 21431, Korea;
| | - Ki Joo Kim
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (K.J.K.); (S.J.L.)
| | - Su Jin Lee
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (K.J.K.); (S.J.L.)
| | - Jong Won Rhie
- Department of Plastic Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Wan-Uk Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.P.); (J.L.); (W.-U.K.); (S.-H.P.)
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.P.); (J.L.); (W.-U.K.); (S.-H.P.)
| | - Suk-Ho Moon
- Department of Plastic Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
- Correspondence: (S.-H.M.); (S.-K.K.)
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.P.); (J.L.); (W.-U.K.); (S.-H.P.)
- Correspondence: (S.-H.M.); (S.-K.K.)
| |
Collapse
|
21
|
Araujo DB, Dantas JR, Silva KR, Souto DL, Pereira MDFC, Moreira JP, Luiz RR, Claudio-Da-Silva CS, Gabbay MAL, Dib SA, Couri CEB, Maiolino A, Rebelatto CLK, Daga DR, Senegaglia AC, Brofman PRS, Baptista LS, Oliveira JEP, Zajdenverg L, Rodacki M. Allogenic Adipose Tissue-Derived Stromal/Stem Cells and Vitamin D Supplementation in Patients With Recent-Onset Type 1 Diabetes Mellitus: A 3-Month Follow-Up Pilot Study. Front Immunol 2020; 11:993. [PMID: 32582156 PMCID: PMC7280537 DOI: 10.3389/fimmu.2020.00993] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 04/27/2020] [Indexed: 01/06/2023] Open
Abstract
Objective: To evaluate the short term safety and potential therapeutic effect of allogenic adipose tissue-derived stromal/stem cells (ASCs) + cholecalciferol in patients with recent-onset T1D. Methods: Prospective, phase II, open trial, pilot study in which patients with recent onset T1D received ASCs (1 × 106 cells/kg) and cholecalciferol 2000 UI/day for 3 months (group 1) and were compared to controls with standard insulin therapy (group 2). Adverse events, C-peptide (CP), insulin dose, HbA1c, time in range (TIR), glucose variability (continuous glucose monitoring) and frequency of CD4+FoxP3+ T-cells (flow cytometry) were evaluated at baseline (T0) and after 3 months (T3). Results: 13 patients were included (8: group 1; 5: group 2). Their mean age and disease duration were 26.7 ± 6.1 years and 2.9 ± 1.05 months. Adverse events were transient headache (n = 8), mild local reactions (n = 7), tachycardia (n = 4), abdominal cramps (n = 1), thrombophlebitis (n = 4), mild floaters (n = 2), central retinal vein occlusion (n = 1, complete resolution). At T3, group 1 had lower insulin requirement (0.22 ± 0.17 vs. 0.61±0.26IU/Kg; p = 0.01) and HbA1c (6.47 ± 0.86 vs. 7.48 ± 0.52%; p = 0.03) than group 2. In group 1, 2 patients became insulin free (for 4 and 8 weeks) and all were in honeymoon at T3 (vs. none in group 2; p = 0.01). CP variations did not differ between groups (−4.6 ± 29.1% vs. +2.3 ± 59.65%; p = 0.83). Conclusions: Allogenic ASCs + cholecalciferol without immunosuppression was associated with stability of CP and unanticipated mild transient adverse events in patients with recent onset T1D. ClinicalTrials.gov registration: NCT03920397.
Collapse
Affiliation(s)
- Debora B Araujo
- Federal University of Rio de Janeiro, Nutrology and Diabetes Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Joana R Dantas
- Federal University of Rio de Janeiro, Nutrology and Diabetes Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Karina R Silva
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Rio de Janeiro, Brazil
| | - Débora L Souto
- Nutrology and Diabetes Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Jessica P Moreira
- Biostatistics Department, Institute of Public Health Studies, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ronir R Luiz
- Biostatistics Department, Institute of Public Health Studies, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Monica A L Gabbay
- Department of Stem Cell Therapy, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Sergio A Dib
- Department of Stem Cell Therapy, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | | | - Angelo Maiolino
- Hematology Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carmen L K Rebelatto
- Core Cell Technology, Pontifical Catholic University of Paraná, Curitiba, Brazil
| | - Debora R Daga
- Core Cell Technology, Pontifical Catholic University of Paraná, Curitiba, Brazil
| | | | - Paulo R S Brofman
- Surgical Clinic D at University of Sao Paulo, Core Cell Technology, Pontifical Catholic University of Paraná, Curitiba, Brazil
| | - Leandra Santos Baptista
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Rio de Janeiro, Brazil.,Multidisciplinary Center for Biological Research (Numpex-Bio), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - José E P Oliveira
- Nutrology and Diabetes Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lenita Zajdenverg
- Nutrology and Diabetes Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Melanie Rodacki
- Nutrology and Diabetes Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Manetti M. Could autologous adipose-derived stromal vascular fraction turn out an unwanted source of profibrotic myofibroblasts in systemic sclerosis? Ann Rheum Dis 2020; 79:e55. [PMID: 30867149 DOI: 10.1136/annrheumdis-2019-215288] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/02/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence I-50134, Italy
| |
Collapse
|
23
|
Magalon J, Velier M, Simoncini S, Dignat-George F, Granel B, Paul P, Sabatier F. Response to: 'Adipose stromal vascular fraction and regenerative therapy in SSc: response to the article by Magalon et al' by De Benedetto et al. Ann Rheum Dis 2020; 79:e54. [PMID: 30787004 DOI: 10.1136/annrheumdis-2019-215132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/05/2019] [Accepted: 02/05/2019] [Indexed: 01/02/2023]
Affiliation(s)
- Jérémy Magalon
- Cell Therapy Department, Hôpital de la Conception, Marseille, France
- Aix Marseille University, Marseille, France
| | - Mélanie Velier
- Cell Therapy Department, Hôpital de la Conception, Marseille, France
- Aix Marseille University, Marseille, France
| | | | | | - Brigitte Granel
- Aix Marseille University, Marseille, France
- Internal Medicine Department, Hôpital Nord, Marseille, France
| | - Pascale Paul
- Cell Therapy Department, Hôpital de la Conception, Marseille, France
- Aix Marseille University, Marseille, France
| | - Florence Sabatier
- Cell Therapy Department, Hôpital de la Conception, Marseille, France
- Aix Marseille University, Marseille, France
| |
Collapse
|
24
|
Rockel JS, Rabani R, Viswanathan S. Anti-fibrotic mechanisms of exogenously-expanded mesenchymal stromal cells for fibrotic diseases. Semin Cell Dev Biol 2019; 101:87-103. [PMID: 31757583 DOI: 10.1016/j.semcdb.2019.10.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/11/2019] [Accepted: 10/30/2019] [Indexed: 12/17/2022]
Abstract
Most chronic diseases involving inflammation have a fibrotic component that involves remodeling and excess accumulation of extracellular matrix components. Left unchecked, fibrosis leads to organ failure and death. Mesenchymal stromal cells (MSCs) are emerging as a potent cell-based therapy for a wide spectrum of fibrotic conditions due to their immunomodulatory, anti-inflammatory and anti-fibrotic properties. This review provides an overview of known mechanisms by which MSCs mediate their anti-fibrotic actions and in relation to animal models of pulmonary, liver, renal and cardiac fibrosis. Recent MSC clinical trials results in liver, lung, skin, kidney and hearts are discussed and next steps for future MSC-based therapies including pre-activated or genetically-modified cells, or extracellular vesicles are also considered.
Collapse
Affiliation(s)
- Jason S Rockel
- Arthritis Program, University Health Network, Toronto, ON, Canada; Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.
| | - Razieh Rabani
- Arthritis Program, University Health Network, Toronto, ON, Canada; Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Sowmya Viswanathan
- Arthritis Program, University Health Network, Toronto, ON, Canada; Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Division of Hematology, Department of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
25
|
VELIER M, SIMONCINI S, ABELLAN M, FRANCOIS P, EAP S, LAGRANGE A, BERTRAND B, DAUMAS A, GRANEL B, DELORME B, DIGNAT GEORGE F, MAGALON J, SABATIER F. Adipose-Derived Stem Cells from Systemic Sclerosis Patients Maintain Pro-Angiogenic and Antifibrotic Paracrine Effects In Vitro. J Clin Med 2019; 8:E1979. [PMID: 31739569 PMCID: PMC6912239 DOI: 10.3390/jcm8111979] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/09/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023] Open
Abstract
Innovative therapies based on autologous adipose-derived stem/stromal cells (ASC) are currently being evaluated for treatment of systemic sclerosis (SSc). Although paracrine angiogenic and antifibrotic effects are considered the predominant mechanisms of ASC therapeutic potential, the impact of SSc on ASC paracrine functions remains controversial. In this study, phenotype, senescence, differentiation potential, and molecular profile were determined in ASC from SSc patients (SSc-ASC) (n = 7) and healthy donors (HD-ASC) (n = 7). ASC were co-cultured in indirect models with dermal fibroblasts (DF) from SSc patients or endothelial cells to assess their pro-angiogenic and antifibrotic paracrine effects. The angiogenic activity of endothelial cells was measured in vitro using tube formation and spheroid assays. DF collagen and alpha smooth muscle actin (αSMA) content were quantified after five days of co-culture with ASC. Differentiation capacity, senescence, and mRNA profiles did not differ significantly between SSc-ASC and HD-ASC. SSc-ASC retained the ability to stimulate angiogenesis through paracrine mechanisms; however, functional assays revealed reduced potential compared to HD-ASC. DF fibrosis markers were significantly decreased after co-culture with SSc-ASC. Together, these results indicate that SSc effects do not significantly compromise the angiogenic and the antifibrotic paracrine properties of ASC, thereby supporting further development of ASC-based autologous therapies for SSc treatment.
Collapse
Affiliation(s)
- Mélanie VELIER
- Aix Marseille University, INSERM, INRA, C2VN, 13005 Marseille, France
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 13005 Marseille, France
| | | | - Maxime ABELLAN
- Plastic Surgery Department, Hôpital de la Conception, AP-HM, 13005 Marseille, France
| | - Pauline FRANCOIS
- Aix Marseille University, INSERM, INRA, C2VN, 13005 Marseille, France
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 13005 Marseille, France
| | - Sandy EAP
- R&D Department, Macopharma, 59420 Mouvaux, France
| | | | - Baptiste BERTRAND
- Plastic Surgery Department, Hôpital de la Conception, AP-HM, 13005 Marseille, France
| | - Aurélie DAUMAS
- Aix Marseille University, INSERM, INRA, C2VN, 13005 Marseille, France
- Internal Medicine Department, Hôpital Nord & Hôpital de la Timone, AP-HM, 13005 Marseille, France
| | - Brigitte GRANEL
- Aix Marseille University, INSERM, INRA, C2VN, 13005 Marseille, France
- Internal Medicine Department, Hôpital Nord & Hôpital de la Timone, AP-HM, 13005 Marseille, France
| | | | | | - Jérémy MAGALON
- Aix Marseille University, INSERM, INRA, C2VN, 13005 Marseille, France
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 13005 Marseille, France
| | - Florence SABATIER
- Aix Marseille University, INSERM, INRA, C2VN, 13005 Marseille, France
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 13005 Marseille, France
| |
Collapse
|
26
|
Manetti M, Romano E, Rosa I, Fioretto BS, Praino E, Guiducci S, Iannone F, Ibba-Manneschi L, Matucci-Cerinic M. Systemic Sclerosis Serum Steers the Differentiation of Adipose-Derived Stem Cells Toward Profibrotic Myofibroblasts: Pathophysiologic Implications. J Clin Med 2019; 8:E1256. [PMID: 31430950 PMCID: PMC6723717 DOI: 10.3390/jcm8081256] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/06/2019] [Accepted: 08/16/2019] [Indexed: 12/16/2022] Open
Abstract
Systemic sclerosis (SSc; scleroderma) is characterized by life-threatening progressive multiorgan fibrosis orchestrated by profibrotic myofibroblasts originating from different sources. Because recent data demonstrated that the majority of myofibroblasts in a murine scleroderma model arise from adipocytic progenitors through the adipocyte-myofibroblast transition process, we sought to determine whether the SSc microenvironment may affect the differentiation potential of adipose-derived stem cells (ADSC). Normal human ADSC from three donors were treated with serum from SSc patients (n = 6), serum from healthy individuals (n = 6), or recombinant human transforming growth factor-β1 (TGFβ1) as positive control of myofibroblastic phenotype induction. ADSC were subjected to in vitro adipogenic differentiation for up to 21 days in the presence of different stimuli followed by lipid content quantification. In selected experiments, adipocytic and mesenchymal/myofibroblast marker gene and protein expression levels were assessed by Real-Time PCR, immunoblotting and immunofluorescence after administration of different stimuli for 72 and 96 h, respectively. Cell contractile phenotype was assayed by collagen gel contraction assay. Likewise stimulation with TGFβ1, SSc serum was able to significantly inhibit the adipocyte differentiation of ADSC as testified by a strong decrease in red-colored lipid droplets after 21 days of adipogenic induction. Treatment of ADSC either with SSc serum or TGFβ1 resulted in the acquisition of a myofibroblast-like phenotype characterized by a reduced expression of the adipocytic markers perilipin and adiponectin, a significant upregulation of the mesenchymal/myofibroblast markers α-SMA+ stress fibers, S100A4 and type I collagen, and an ability to effectively contract collagen gels. In SSc, the pathologic environment may favor the differentiation of ADSC into profibrotic and contractile myofibroblast-like cells. These findings strengthen the notion that the generation of myofibroblasts from ADSC may be relevant in SSc pathophysiology potentially representing a new target for the prevention/treatment of multiorgan fibrosis.
Collapse
Affiliation(s)
- Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
| | - Eloisa Romano
- Division of Rheumatology and Scleroderma Unit, AOUC, Department of Geriatric Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Division of Rheumatology and Scleroderma Unit, AOUC, Department of Geriatric Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Bianca Saveria Fioretto
- Division of Rheumatology and Scleroderma Unit, AOUC, Department of Geriatric Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Emanuela Praino
- Rheumatology Unit, Department of Emergency and Organ Transplantation, University of Bari, 70121 Bari, Italy
| | - Serena Guiducci
- Division of Rheumatology and Scleroderma Unit, AOUC, Department of Geriatric Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Florenzo Iannone
- Rheumatology Unit, Department of Emergency and Organ Transplantation, University of Bari, 70121 Bari, Italy
| | - Lidia Ibba-Manneschi
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Marco Matucci-Cerinic
- Division of Rheumatology and Scleroderma Unit, AOUC, Department of Geriatric Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| |
Collapse
|
27
|
Durchschein F, Moazedi-Fuerst F, Kielhauser S, Lackner A, Wiedner M, Koch H, Justich I, Eherer A. Helpful, albeit hazardous! Esophageal stem-cell injection in systemic sclerosis. Ther Adv Musculoskelet Dis 2019; 11:1759720X19863618. [PMID: 31360239 PMCID: PMC6637824 DOI: 10.1177/1759720x19863618] [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: 12/20/2018] [Accepted: 06/22/2019] [Indexed: 11/17/2022] Open
Abstract
Background Over 90% of patients with systemic sclerosis suffer from gastroesophageal reflux. Esophageal motility disturbances are associated with a reduced life quality and may force interstitial lung disease progression. We wanted to determine whether we can improve gastroesophageal reflux in these patients by esophageal stem-cell injection. Methods We performed a pilot study including eights patients with systemic sclerosis and symptomatic gastroesophageal reflux. Sampling of adipose tissue was performed by an experienced plastic surgeon under local anesthesia. The collected fat was injected into the submucosa of the distal esophagus, each time 1 ml in all four quadrants starting 2, 4 and 6 cm proximal to the Z line (ending up to a total volume of 12 ml). Before the intervention, 3, 6 and finally 12 months after the procedure, patients answered the Gastroesophageal Reflux Disease Health-Related Quality of Life Questionnaire (GERD HRQL) and a high-resolution manometry was performed to quantify changes in motility function. Results All patients showed an improvement in the GERD HRQL score after the stem-cell injection and a lower dosage of proton-pump inhibitors. The manometric findings showed no change throughout the time. A serious adverse event occurred, as one patient developed multiple cerebellar embolic infarcts. Conclusion Because of the favorable effect in all patients, a safe route for esophageal fat injection needs to be developed.
Collapse
Affiliation(s)
| | - Florentine Moazedi-Fuerst
- Division of Rheumatology, Department of Internal Medicine, Medical University Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Sonja Kielhauser
- Division of Rheumatology, Medical University Graz, Graz, Austria
| | - Angelika Lackner
- Division of Rheumatology, Medical University Graz, Graz, Austria
| | - Maria Wiedner
- Division of Plastic, Aesthetic and Reconstructive Surgery, Medical University of Graz, Austria
| | - Horst Koch
- Division of Plastic, Aesthetic and Reconstructive Surgery, Medical University of Graz, Austria
| | - Ivo Justich
- Division of Plastic, Aesthetic and Reconstructive Surgery, Medical University of Graz, Austria
| | - Andreas Eherer
- Division of Gastroenterology and Hepatology, Medical University Graz, Graz, Austria
| |
Collapse
|
28
|
Herrick AL. Raynaud's phenomenon. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2019; 4:89-101. [PMID: 35382391 PMCID: PMC8922643 DOI: 10.1177/2397198319826467] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 11/27/2018] [Indexed: 09/05/2023]
Abstract
Raynaud's phenomenon can be either primary (idiopathic) or secondary to underlying disease including systemic sclerosis. Primary Raynaud's phenomenon is very common, affecting approximately 3%-5% of the general population. Although much rarer, systemic sclerosis-related Raynaud's phenomenon can be particularly severe, progressing to digital ulceration in approximately 50% of patients. Raynaud's phenomenon can have a major impact on quality of life. This review has a focus on the systemic sclerosis-related Raynaud's phenomenon (which is the most researched form of Raynaud's phenomenon and probably the most challenging to treat) and on recent advances. Epidemiology (including transition from 'isolated' to systemic sclerosis-related Raynaud's phenomenon), pathogenesis, diagnosis and assessment are discussed, followed by the treatment of both 'uncomplicated' and 'complicated' Raynaud's phenomena (i.e. Raynaud's phenomenon which has progressed to digital ulceration and/or critical ischaemia). Finally, some of the major challenges for the next 5-10 years are highlighted.
Collapse
Affiliation(s)
- Ariane L Herrick
- Centre for Musculoskeletal Research, The University of Manchester, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester, UK
| |
Collapse
|
29
|
Magalon J, Velier M, Simoncini S, François P, Bertrand B, Daumas A, Benyamine A, Boissier R, Arnaud L, Lyonnet L, Fernandez S, Dignat-George F, Casanova D, Guillet B, Granel B, Paul P, Sabatier F. Molecular profile and proangiogenic activity of the adipose-derived stromal vascular fraction used as an autologous innovative medicinal product in patients with systemic sclerosis. Ann Rheum Dis 2019; 78:391-398. [PMID: 30612118 DOI: 10.1136/annrheumdis-2018-214218] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/31/2018] [Accepted: 11/16/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The autologous stromal vascular fraction (SVF) from adipose tissue is an alternative to cultured adipose-derived stem cells for use in regenerative medicine and represents a promising therapy for vasculopathy and hand disability in systemic sclerosis (SSc). However, the bioactivity of autologous SVF is not documented in this disease context. This study aimed to compare the molecular and functional profiles of the SVF-based medicinal product obtained from SSc and healthy subjects. METHODS Good manufacturing practice (GMP)-grade SVF from 24 patients with SSc and 12 healthy donors (HD) was analysed by flow cytometry to compare the distribution of the CD45- and CD45+ haematopoietic cell subsets. The ability of SVF to form a vascular network was assessed using Matrigel in vivo assay. The transcriptomic and secretory profiles of the SSc-SVF were assessed by RNA sequencing and multiplex analysis, respectively, and were compared with the HD-SVF. RESULTS The distribution of the leucocyte, endothelial, stromal, pericyte and transitional cell subsets was similar for SSc-SVF and HD-SVF. SSc-SVF retained its vasculogenic capacity, but the density of neovessels formed in SVF-loaded Matrigel implanted in nude mice was slightly decreased compared with HD-SVF. SSc-SVF displayed a differential molecular signature reflecting deregulation of angiogenesis, endothelial activation and fibrosis. CONCLUSIONS Our study provides the first evidence that SSc does not compromise the vascular repair capacity of SVF, supporting its use as an innovative autologous biotherapy. The characterisation of the specific SSc-SVF molecular profile provides new perspectives for delineating markers of the potency of SVF and its targets for the treatment of SSc.
Collapse
Affiliation(s)
- Jérémy Magalon
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
| | - Mélanie Velier
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
| | | | - Pauline François
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
| | - Baptiste Bertrand
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
- Plastic Surgery Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Aurélie Daumas
- Internal Medicine Department, Hôpital Nord & Hôpital de la Timone, AP-HM, Marseille, France
| | - Audrey Benyamine
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
- Internal Medicine Department, Hôpital Nord & Hôpital de la Timone, AP-HM, Marseille, France
| | - Romain Boissier
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
- Urology Surgery Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Laurent Arnaud
- Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Luc Lyonnet
- Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | | | - Françoise Dignat-George
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
- Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Dominique Casanova
- Plastic Surgery Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Benjamin Guillet
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
- CERIMED, Aix-Marseille University, AP-HM, Marseille, France
| | - Brigitte Granel
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
- Internal Medicine Department, Hôpital Nord & Hôpital de la Timone, AP-HM, Marseille, France
| | - Pascale Paul
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
| | - Florence Sabatier
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France
- INSERM, INRA, C2VN, Aix-Marseille University, Marseille, France
| |
Collapse
|
30
|
Bertheuil N, Chaput B, Ménard C, Varin A, Laloze J, Watier E, Tarte K. Adipose mesenchymal stromal cells: Definition, immunomodulatory properties, mechanical isolation and interest for plastic surgery. ANN CHIR PLAST ESTH 2019; 64:1-10. [DOI: 10.1016/j.anplas.2018.07.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 07/13/2018] [Indexed: 12/14/2022]
|
31
|
Rozier P, Maria A, Goulabchand R, Jorgensen C, Guilpain P, Noël D. Mesenchymal Stem Cells in Systemic Sclerosis: Allogenic or Autologous Approaches for Therapeutic Use? Front Immunol 2018; 9:2938. [PMID: 30619298 PMCID: PMC6302042 DOI: 10.3389/fimmu.2018.02938] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/29/2018] [Indexed: 12/18/2022] Open
Abstract
Systemic sclerosis (SSc) is a rare autoimmune disease, which is potentially lethal. The physiopathology of the disease is still incompletely elucidated although the role of fibroblasts, endothelial cells (ECs), immune cells. and the environment (i.e., oxidative stress) has been demonstrated. This is an intractable disease with an urgent need to provide better therapeutic options to patients. Mesenchymal stem cells (MSCs) represent a promising therapeutic approach thanks to the number of trophic and pleiotropic properties they exert. Among these, MSCs display anti-fibrotic, angiogenic, and immunomodulatory capacities that might be of interest in the treatment of SSc by acting on different processes that are dysregulated in the disease. In the recent years, the therapeutic effectiveness of MSCs has been demonstrated in different preclinical animal models and is being investigated in phase I clinical trials. Both allogenic and autologous transplantation of MSCs isolated from bone marrow or adipose tissue is being evaluated. The rationale for using allogenic MSCs in SSc, as well as in other autoimmune diseases, is based on the possibility that autologous MSCs might be altered in these diseases. In SSc, reports from the literature are controversial. Nevertheless, the role of the oxidative environment and of the crosstalk with neighboring cells (fibroblasts and ECs) on the functional properties of MSCs has been reported. Here, we review the preclinical and clinical data reporting the interest of MSC-based treatment in SSc and question the use of autologous or allogeneic MSCs in perspective of clinical applications.
Collapse
Affiliation(s)
- Pauline Rozier
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Department of Internal Medicine, Multiorganic Diseases, Saint-Eloi Hospital, Montpellier, France
| | - Alexandre Maria
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Department of Internal Medicine, Multiorganic Diseases, Saint-Eloi Hospital, Montpellier, France
| | - Radjiv Goulabchand
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Department of Internal Medicine, Multiorganic Diseases, Saint-Eloi Hospital, Montpellier, France
| | - Christian Jorgensen
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Lapeyronie University Hospital, Montpellier, France
| | - Philippe Guilpain
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Department of Internal Medicine, Multiorganic Diseases, Saint-Eloi Hospital, Montpellier, France
| | - Danièle Noël
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Lapeyronie University Hospital, Montpellier, France
| |
Collapse
|
32
|
Autologous adipose-derived stromal vascular fraction and scarred vocal folds: first clinical case report. Stem Cell Res Ther 2018; 9:202. [PMID: 30053913 PMCID: PMC6062967 DOI: 10.1186/s13287-018-0842-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/24/2018] [Accepted: 03/15/2018] [Indexed: 11/18/2022] Open
|
33
|
Comella K, Parlo M, Daly R, Dominessy K. First-in-man intravenous implantation of stromal vascular fraction in psoriasis: a case study. Int Med Case Rep J 2018; 11:59-64. [PMID: 29606893 PMCID: PMC5868735 DOI: 10.2147/imcrj.s163612] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Stromal vascular fraction (SVF) is a mixture of adipose-derived stem cells/mesenchymal stem cells, endothelial/progenitors, pericytes, fibroblasts, and other cells obtained from fat tissue. A small sample of fat or adipose tissue can be obtained under local anesthesia using a cannula. After an enzymatic digestion and centrifugation, the adipocytes (fat cells) are removed to obtain an SVF. Here, we describe the rationale and, to our knowledge, the first clinical implementation of SVF intravenously in a patient with severe psoriasis. METHODS Adipose tissue (60 mL) was collected under local anesthesia via a mini-lipoaspirate procedure. The SVF was separated from the adipocytes via centrifugation after an enzymatic digestion. The cells were resuspended in normal saline and injected via bolus push intravenous. The subject was monitored over a period of 12 months for safety (adverse events), medication changes, and quality of life parameters. RESULTS The patient did not report any safety concerns and did not experience any severe adverse events. The patient demonstrated a significant decrease in symptoms with a noticeable difference in skin quality appearance. Psoriasis area and severity index score went from 50.4 at baseline to 0.3 at 1 month follow-up. CONCLUSION Overall, the patient reported improved quality of life and willingness to continue treatments. This successful initial case study demonstrates that this may be a feasible treatment plan for patients suffering from psoriasis.
Collapse
|
34
|
Adipose-derived cellular therapies in solid organ and vascularized-composite allotransplantation. Curr Opin Organ Transplant 2018; 22:490-498. [PMID: 28873074 DOI: 10.1097/mot.0000000000000452] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Controlling acute allograft rejection following vascularized composite allotransplantation requires strict adherence to courses of systemic immunosuppression. Discovering new methods to modulate the alloreactive immune response is essential for widespread application of vascularized composite allotransplantation. Here, we discuss how adipose-derived cellular therapies represent novel treatment options for immune modulation and tolerance induction in vascularized composite allotransplantation. RECENT FINDINGS Adipose-derived mesenchymal stromal cells are cultured from autologous or allogeneic adipose tissue and possess immunomodulatory qualities capable of prolonging allograft survival in animal models of vascularized composite allotransplantation. Similar immunosuppressive and immunomodulatory effects have been observed with noncultured adipose stromal-vascular-fraction-derived therapies, albeit publication of in-vivo stromal vascular fraction cell modulation in transplantation models is lacking. However, both stromal vascular fraction and adipose derived mesenchymal stem cell therapies have the potential to effectively modulate acute allograft rejection via recruitment and induction of regulatory immune cells. SUMMARY To date, most reports focus on adipose derived mesenchymal stem cells for immune modulation in transplantation despite their phenotypic plasticity and reliance upon culture expansion. Along with the capacity for immune modulation, the supplemental wound healing and vasculogenic properties of stromal vascular fraction, which are not shared by adipose derived mesenchymal stem cells, hint at the profound therapeutic impact stromal vascular fraction-derived treatments could have on controlling acute allograft rejection and tolerance induction in vascularized composite allotransplantation. Ongoing projects in the next few years will help design the best applications of these well tolerated and effective treatments that should reduce the risk/benefit ratio and allow more patients access to vascularized composite allotransplantation therapy.
Collapse
|
35
|
Wollina U, Koch A, Heinig B, Tchernev G, Lotti T. Cutaneous Microembolism of Fingers and Toes. Open Access Maced J Med Sci 2018; 6:166-169. [PMID: 29484019 PMCID: PMC5816294 DOI: 10.3889/oamjms.2018.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 08/19/2017] [Accepted: 08/20/2017] [Indexed: 01/08/2023] Open
Abstract
A macro vascular embolism is a well-known emergency. In contrast, cutaneous microembolism is a lesser known symptom. However, cutaneous microembolism of fingers and toes is a red flag symptom for vascular emergencies. The underlying cause may involve infectious, immunological, metabolic and physical disorders, coagulation disorders and malignancies. Early recognition can help to live safe.
Collapse
Affiliation(s)
- Uwe Wollina
- Department of Dermatology and Allergology, Städtisches Klinikum Dresden, Friedrichstrasse 41, 01067 Dresden, Germany
| | - André Koch
- Department of Dermatology and Allergology, Städtisches Klinikum Dresden, Friedrichstrasse 41, 01067 Dresden, Germany
| | - Birgit Heinig
- Center for Physical Therapy and Rehabilitative Medicine, Städtisches Klinikum Dresden, Dresden, Germany
| | - Georgi Tchernev
- Department of Dermatology, Venereology and Dermatologic Surgery, Medical Institute of Ministry of Interior, Sofia, Bulgaria.,Onkoderma Policlinic for Dermatology and Dermatologic Surgery, Sofia, Bulgaria
| | | |
Collapse
|
36
|
Scleroderma skin ulcers definition, classification and treatment strategies our experience and review of the literature. Autoimmun Rev 2017; 17:155-164. [PMID: 29196241 DOI: 10.1016/j.autrev.2017.11.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Skin ulcers (SU) are one of the most frequent manifestations of systemic sclerosis (SSc). SSc-SU are very painful, often persistent and recurrent; they may lead to marked impairment of patient's activities and quality of life. Despite their severe impact on the whole SSc patient's management, the proposed definition, classification criteria, and therapeutic strategies of SSc-SU are still controversial. OBJECTIVE The present study aimed to elaborate a comprehensive proposal of definition, classification, and therapeutic strategy of SSc-SU on the basis of our long-term single center experience along with a careful revision of the world literature on the same topic. METHODS A series of 282 SSc patients (254 females and 28 males; 84% with limited and 16% diffuse cutaneous SSc; mean age of 51.5±13.9SD at SSc onset; mean follow-up 5.8±4.6SDyears) enrolled during the last decade at our Rheumatology Unit were retrospectively evaluated with specific attention to SSc-SU. The SSc-SU were classified in 5 subtypes according to prominent pathogenetic mechanism(s) and localization, namely 1. digital ulcers (DU) of the hands or feet, 2. SU on bony prominence, 3. SU on calcinosis, 4. SU of lower limbs, and 5. DU presenting with gangrene. This latter is a very harmful evolution of both DU of the hands and feet needing a differential diagnosis with critical limb ischemia. RESULTS During the follow up period, one or more episodes of SSc-SU were recorded in over half patients (156/282, 55%); skin lesions were often recurrent and difficult-to-heal because of local complications, mainly infections (67.3%), in some cases associated to osteomyelitis (19.2%), gangrene (16%), and/or amputation (11.5%). SSc-SU were significantly associated with lower patients' mean age at the disease onset (p=0.024), male gender (p=0.03), diffuse cutaneous subset (p=0.015), calcinosis (p=0.002), telangiectasia (p=0.008), melanodermia (p<0.001), abnormal PAPs (p=0.036), and/or altered inflammation reactant (CRP, p=0.001). Therapeutic strategy of SSc-SU included both systemic and local pharmacological treatments with particular attention to complicating infections and chronic/procedural pain, as well as a number of non-pharmacological measures. Integrated local treatments were often decisive for the SSc-SU healing; they were mainly based on the wound bed preparation principles that are summarized in the acronym TIME (necrotic Tissue, Infection/Inflammation, Moisture balance, and Epithelization). The updated review of the literature focusing on this challenging issue was analyzed in comparison with our experience. CONCLUSIONS The recent advancement of knowledge and management strategies of SSc-SU achieved during the last years lead to the clear-cut improvement of patients' quality of life and reduced long-term disability.
Collapse
|
37
|
Foubert P, Zafra D, Liu M, Rajoria R, Gutierrez D, Tenenhaus M, Fraser JK. Autologous adipose-derived regenerative cell therapy modulates development of hypertrophic scarring in a red Duroc porcine model. Stem Cell Res Ther 2017; 8:261. [PMID: 29141687 PMCID: PMC5688645 DOI: 10.1186/s13287-017-0704-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 12/18/2022] Open
Abstract
Background Effective prevention and treatment of hypertrophic scars (HTSs), a common consequence of deep-partial thickness injury, remain a significant clinical challenge. Previous studies from our group have shown that autologous adipose-derived regenerative cells (ADRCs) represent a promising approach to improve wound healing and, thereby, impact HTS development. The purpose of this study was to assess the influence of local delivery of ADRCs immediately following deep-partial thickness cutaneous injury on HTS development in the red Duroc (RD) porcine model. Methods Bilateral pairs of deep-partial thickness excisional wounds (2 mm depth; 58 cm2 area) were created using an electric dermatome on RD pigs (n = 12). Autologous ADRCs were isolated from the inguinal fat pad and then sprayed directly onto the wound at a dose of 0.25 × 106 viable cells/cm2. The paired contralateral wound received vehicle control. Wound healing and development of HTS were assessed over 6 months using digital imaging, quantitative measurement of skin hardness and pigmentation, and histology. Results Data showed that ADRC treatment led to reduced scar hyperpigmentation compared to control (p < 0.05). Using the Durometer, at 2 and 6 months post-injury, skin hardness was 10–20% lower in ADRCs-treated wounds compared to control vehicle (p < 0.05). A similar trend was observed with the skin fibrometer. ADRC treatment promoted more normal collagen organization, improvement in the number of rete ridges (p < 0.01), longer elastic fiber length (p < 0.01), and reduced hypervascularity (blood vessel density; p < 0.05). ADRC treatment was associated with modulation of IL-6 expression within the wound/scar with upregulation 2 weeks after injury (wound healing phase) and downregulation at 2 months (early scarring phase) post-treatment compared to control Conclusions These findings support the potential therapeutic value of autologous ADRC administration for reduction of HTS development following deep-partial cutaneous injury. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0704-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Philippe Foubert
- Cytori Therapeutics Inc, 3020 Callan Road, 92121, San Diego, CA, USA.
| | - Diana Zafra
- Cytori Therapeutics Inc, 3020 Callan Road, 92121, San Diego, CA, USA
| | - Mike Liu
- Cytori Therapeutics Inc, 3020 Callan Road, 92121, San Diego, CA, USA
| | | | | | - Mayer Tenenhaus
- UCSD Medical Center, University of California, San Diego, CA, USA
| | - John K Fraser
- Cytori Therapeutics Inc, 3020 Callan Road, 92121, San Diego, CA, USA
| |
Collapse
|
38
|
Comella K, Parlo M, Daly R, Depasquale V, Edgerton E, Mallory P, Schmidt R, Drake WP. Safety Analysis of Autologous Stem Cell Therapy in a Variety of Degenerative Diseases and Injuries Using the Stromal Vascular Fraction. J Clin Med Res 2017; 9:935-942. [PMID: 29038672 PMCID: PMC5633095 DOI: 10.14740/jocmr3187w] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 09/25/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Stem cells from adipose tissue offer a novel therapy for patients with damaged tissue. Stromal vascular fraction (SVF) injected into patients may reduce inflammation, promote healing, and repair damaged/scarred tissue. SVF can be isolated from fat (adipose) tissue in an outpatient procedure. The SVF population includes mesenchymal stem cells (MSCs), pericytes, endothelial/progenitor cells, fibroblasts and growth factors where the adipocyte (fat cell) population has been removed. Here we describe the use of SVF in the clinic for degenerative diseases in orthopedics, neurological conditions and systemic conditions in 676 patients. METHODS This study demonstrated the strong safety profile from a multi-center analysis of SVF injection in treating various diseases. Approximately 60 mL of fat tissue was removed from the abdomen or flanks using a local tumescent liposuction procedure. The fat was separated via centrifuge to isolate the SVF and the cells were delivered intraarticularly, intravenously, intrathecally, or intradiscally directly into the same patient. All subjects were monitored for adverse events. RESULTS The procedure demonstrates exceptional patient safety, and the study underscores the safety of autologous stem cell therapy in general. Few adverse events were reported and were overwhelmingly of mild and transient nature, such as the expected soreness at the site of liposuction and occasional headache. CONCLUSION The three deaths reported were most likely not related to the treatment but instead to the underlying disease. Our study demonstrates a strong safety profile with low complication rates.
Collapse
Affiliation(s)
- Kristin Comella
- US Stem Cell, Inc., 13794 NW 4th Street, Suite 212, Sunrise, FL 33325, USA
- Panama College of Cell Science, 3rd Floor, C&H Towers Corner of Great Marlboro and Great George Streets Roseau, 00152, Commonwealth of Dominica
| | - Michelle Parlo
- US Stem Cell Clinic, 12651 Sunrise Blvd, Suite 104, Sunrise, FL 33323, USA
| | - Rosemary Daly
- US Stem Cell Clinic, 12651 Sunrise Blvd, Suite 104, Sunrise, FL 33323, USA
| | - Vincent Depasquale
- NSI Stem Cell, 3118 Gulf to Bay Blvd, Suite 310, Clearwater, FL 33759, USA
| | - Eric Edgerton
- NSI Stem Cell, 3118 Gulf to Bay Blvd, Suite 310, Clearwater, FL 33759, USA
| | - Patrick Mallory
- Mallory Family Wellness, 1548 North Boise Ave., Loveland, CO 80538, USA
| | - Roy Schmidt
- New Life Medical Group, 15 Stonebridge, Jackson, TN 38305, USA
| | - Walter P. Drake
- Panama College of Cell Science, 3rd Floor, C&H Towers Corner of Great Marlboro and Great George Streets Roseau, 00152, Commonwealth of Dominica
| |
Collapse
|
39
|
Tabatabaei Qomi R, Sheykhhasan M. Adipose-derived stromal cell in regenerative medicine: A review. World J Stem Cells 2017; 9:107-117. [PMID: 28928907 PMCID: PMC5583529 DOI: 10.4252/wjsc.v9.i8.107] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/30/2017] [Accepted: 05/18/2017] [Indexed: 02/06/2023] Open
Abstract
The application of appropriate cell origin for utilizing in regenerative medicine is the major issue. Various kinds of stem cells have been used for the tissue engineering and regenerative medicine. Such as, several stromal cells have been employed as treat option for regenerative medicine. For example, human bone marrow-derived stromal cells and adipose-derived stromal cells (ADSCs) are used in cell-based therapy. Data relating to the stem cell therapy and processes associated with ADSC has developed remarkably in the past 10 years. As medical options, both the stromal vascular and ADSC suggests good opportunity as marvelous cell-based therapeutics. The some biological features are the main factors that impact the regenerative activity of ADSCs, including the modulation of the cellular immune system properties and secretion of bioactive proteins such as cytokines, chemokines and growth factors, as well as their intrinsic anti-ulcer and anti-inflammatory potential. A variety of diseases have been treated by ADSCs, and it is not surprising that there has been great interest in the possibility that ADSCs might be used as therapeutic strategy to improve a wider range of diseases. This is especially important when it is remembered that routine therapeutic methods are not completely effective in treat of diseases. Here, it was discuss about applications of ADSC to colitis, liver failure, diabetes mellitus, multiple sclerosis, orthopaedic disorders, hair loss, fertility problems, and salivary gland damage.
Collapse
Affiliation(s)
- Reza Tabatabaei Qomi
- Department of Stem Cell, the Academic Center for Education, Culture and Research, PO Box QOM-3713189934, Qom, Iran
| | - Mohsen Sheykhhasan
- Department of Stem Cell, the Academic Center for Education, Culture and Research, PO Box QOM-3713189934, Qom, Iran
| |
Collapse
|
40
|
Comella K, Blas JAP, Ichim T, Lopez J, Limon J, Moreno RC. Autologous Stromal Vascular Fraction in the Intravenous Treatment of End-Stage Chronic Obstructive Pulmonary Disease: A Phase I Trial of Safety and Tolerability. J Clin Med Res 2017; 9:701-708. [PMID: 28725319 PMCID: PMC5505307 DOI: 10.14740/jocmr3072w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 05/23/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a consistently progressive, ultimately fatal disease for which no treatment exists capable of either reversing or even interrupting its course. It afflicts more than 5% of the population in many countries, and it accordingly represents the third most frequent cause of death in the US, where it accounts for more than 600 billion in health care costs, morbidity, and mortality. Adipose tissue contains within its stromal compartment a high abundance of adipose stem/stromal cells (ASCs), which can be readily separated from the adipocyte population by methods which require less than 2 h of processing time and yield a concentrated cellular preparation termed the stromal vascular fraction (SVF). The SVF contains all cellular elements of fat, excluding adipocytes. Recent clinical studies have begun to explore the feasibility and safety of the local injection or intravascular delivery of SVF or more purified populations of ASCs derived by culture protocols. Several pre-clinical studies have demonstrated a remarkable ability of ASC to nearly fully ameliorate the progress of emphysema due to cigarette smoke exposure as well as other causes. However, no prior clinical studies have evaluated the safety of administration of either ASC or SVF in subjects with COPD. We hypothesized that harvest, isolation, and immediate intravenous infusion of autologous SVF would be feasible and safe in subjects with COPD; and that such an approach, if ultimately determined to be efficacious as well as safe, would provide a highly practical method for treatment of COPD. METHODS In this study, an initial phase I trial evaluating the early and delayed safety of SVF infusion was performed. Twelve subjects were enrolled in the study, in which adipose tissue was harvested using standard liposuction techniques, followed by SVF isolation and intravenous infusion of 150 - 300 million cells. Standardized questionnaires were administered to study feasibility as well as immediate and delayed outcomes and adverse events as primary endpoints. Secondary endpoints included subjective wellness and attitudes towards the procedure, as well as willingness to undergo the procedure a second time. The follow-up time ranged from 3 to 12 months, averaging 12 months. RESULTS Of the 12 subjects, only one experienced an immediate adverse event, related to bruising from the liposuction. No observed pulmonary or cardiac issues were observed as related to the procedure. There were no deaths over the 12-month study period, and none identified in the subsequent telephonic follow-up. Attitudes toward the procedure were predominantly positive, and 92% of the study subjects expressed a desire to undergo the procedure a second time. CONCLUSIONS This study is the first to demonstrate safety of SVF infusion in humans with serious pulmonary disease. Specifically, the use of intravenous infusion as a route to achieve pulmonary cellular targeting did not lead to clinical pulmonary compromise. The intravenous administration of SVF should be further explored as a potentially feasible and safe method for delivery leading to possible therapeutic benefit.
Collapse
Affiliation(s)
- Kristin Comella
- US Stem Cell, Inc., 13794 NW 4th Street, Suite 212, Sunrise, FL 33325, USA
| | - Jesus A. Perez Blas
- University of Baja California, Mexico and Hospital Angeles, Tijuana, Mexico, Regenerative Medicine Institute, Mexico
| | - Tom Ichim
- University of Baja California, Mexico and Hospital Angeles, Tijuana, Mexico, Regenerative Medicine Institute, Mexico
| | - Javier Lopez
- University of Baja California, Mexico and Hospital Angeles, Tijuana, Mexico, Regenerative Medicine Institute, Mexico
| | - Jose Limon
- University of Baja California, Mexico and Hospital Angeles, Tijuana, Mexico, Regenerative Medicine Institute, Mexico
| | - Ruben Corral Moreno
- University of Baja California, Mexico and Hospital Angeles, Tijuana, Mexico, Regenerative Medicine Institute, Mexico
| |
Collapse
|
41
|
DiCarlo AL, Tamarat R, Rios CI, Benderitter M, Czarniecki CW, Allio TC, Macchiarini F, Maidment BW, Jourdain JR. Cellular Therapies for Treatment of Radiation Injury: Report from a NIH/NIAID and IRSN Workshop. Radiat Res 2017; 188:e54-e75. [PMID: 28605260 DOI: 10.1667/rr14810.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In recent years, there has been increasing concern over the possibility of a radiological or nuclear incident occurring somewhere in the world. Intelligence agencies frequently report that terrorist groups and rogue nations are seeking to obtain radiological or nuclear weapons of mass destruction. In addition, there exists the real possibility that safety of nuclear power reactors could be compromised by natural (such as the tsunami and subsequent Fukushima accident in Japan in March, 2011) or accidental (Three Mile Island, 1979 and Chernobyl, 1986) events. Although progress has been made by governments around the world to prepare for these events, including the stockpiling of radiation countermeasures, there are still challenges concerning care of patients injured during a radiation incident. Because the deleterious and pathological effects of radiation are so broad, it is desirable to identify medical countermeasures that can have a beneficial impact on several tissues and organ systems. Cellular therapies have the potential to impact recovery and tissue/organ regeneration for both early and late complications of radiation exposure. These therapies, which could include stem or blood progenitor cells, mesenchymal stromal cells (MSCs) or cells derived from other tissues (e.g., endothelium or placenta), have shown great promise in treating other nonradiation injuries to and diseases of the bone marrow, skin, gastrointestinal tract, brain, lung and heart. To explore the potential use of these therapies in the treatment of victims after acute radiation exposure, the National Institute of Allergy and Infectious Diseases co-sponsored an international workshop in July, 2015 in Paris, France with the Institut de Radioprotection et de Sûreté Nucléaire. The workshop included discussions of data available from testing in preclinical models of radiation injury to different organs, logistics associated with the practical use of cellular therapies for a mass casualty incident, as well as international regulatory requirements for authorizing such drug products to be legally and readily used in such incidents. This report reviews the data presented, as well as key discussion points from the meeting.
Collapse
Affiliation(s)
- Andrea L DiCarlo
- a Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Radia Tamarat
- b Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Fontenay-aux-Roses, France
| | - Carmen I Rios
- a Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Marc Benderitter
- b Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Fontenay-aux-Roses, France
| | | | | | - Francesca Macchiarini
- e Previously -RNCP, DAIT, NIAID, NIH; now National Institute on Aging (NIA), NIH, Bethesda, Maryland
| | | | - Jean-Rene Jourdain
- b Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Fontenay-aux-Roses, France
| |
Collapse
|
42
|
Ilic D, Trento C, Bisioti A. Latest developments in the field of stem cell research and regenerative medicine compiled from publicly available information and press releases from nonacademic institutions 1 October–30 November 2016. Regen Med 2017. [DOI: 10.2217/rme-2016-0172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Dusko Ilic
- Stem Cell Laboratories, Guy's Assisted Conception Unit, Division of Women's Health, Faculty of Science & Medicine, King's College London, London, UK
| | - Cristina Trento
- Department of Haemato-Oncology, Rayne Institute, Faculty of Science & Medicine, King's College London, London, UK
| | - Alexandra Bisioti
- Cellular Therapy from Bench to Market Program, Faculty of Science & Medicine, King's College London, London, UK
| |
Collapse
|