1
|
Zhang Y, Gu J, Wang X, Li L, Fu L, Wang D, Wang X, Han X. Opportunities and challenges: mesenchymal stem cells in the treatment of multiple sclerosis. Int J Neurosci 2023; 133:1031-1044. [PMID: 35579409 DOI: 10.1080/00207454.2022.2042690] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 01/08/2022] [Accepted: 02/09/2022] [Indexed: 10/18/2022]
Abstract
Multiple sclerosis (MS) was once considered an untreatable disease. Through years of research, many drugs have been discovered and are widely used for the treatment of MS. However, the current treatment can only alleviate the clinical symptoms of MS and has serious side effects. Mesenchymal stem cells (MSCs) provide neuroprotection by migrating to injured tissues, suppressing inflammation, and fostering neuronal repair. Therefore, MSCs therapy holds great promise for MS treatment. This review aimed to assess the feasibility and safety of use of MSCs in MS treatment as well as its development prospect in clinical treatment by analysing the existing clinical studies.
Collapse
Affiliation(s)
- Yingyu Zhang
- Department of Neurology, China-Japan Union hospital of Jilin University, Changchun city, Jilin, P.R. China
| | - Jiebing Gu
- Department of Neurology, China-Japan Union hospital of Jilin University, Changchun city, Jilin, P.R. China
| | - Xiaoshuang Wang
- Department of Neurology, China-Japan Union hospital of Jilin University, Changchun city, Jilin, P.R. China
| | - Linfang Li
- Department of Neurology, China-Japan Union hospital of Jilin University, Changchun city, Jilin, P.R. China
| | - Lingling Fu
- Department of Neurology, China-Japan Union hospital of Jilin University, Changchun city, Jilin, P.R. China
| | - Di Wang
- Department of Neurology, China-Japan Union hospital of Jilin University, Changchun city, Jilin, P.R. China
| | - Xiuting Wang
- Department of Neurology, China-Japan Union hospital of Jilin University, Changchun city, Jilin, P.R. China
| | - Xuemei Han
- Department of Neurology, China-Japan Union hospital of Jilin University, Changchun city, Jilin, P.R. China
| |
Collapse
|
2
|
Gorman EA, Rynne J, Gardiner HJ, Rostron AJ, Bannard-Smith J, Bentley AM, Brealey D, Campbell C, Curley G, Clarke M, Dushianthan A, Hopkins P, Jackson C, Kefela K, Krasnodembskaya A, Laffey JG, McDowell C, McFarland M, McFerran J, McGuigan P, Perkins GD, Silversides J, Smythe J, Thompson J, Tunnicliffe WS, Welters IDM, Amado-Rodríguez L, Albaiceta G, Williams B, Shankar-Hari M, McAuley DF, O'Kane CM. Repair of Acute Respiratory Distress Syndrome in COVID-19 by Stromal Cells (REALIST-COVID Trial): A Multicenter, Randomized, Controlled Clinical Trial. Am J Respir Crit Care Med 2023; 208:256-269. [PMID: 37154608 DOI: 10.1164/rccm.202302-0297oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023] Open
Abstract
Rationale: Mesenchymal stromal cells (MSCs) may modulate inflammation, promoting repair in coronavirus disease (COVID-19)-related acute respiratory distress syndrome (ARDS). Objectives: We investigated the safety and efficacy of ORBCEL-C (CD362 [cluster of differentiation 362]-enriched, umbilical cord-derived MSCs) in COVID-19-related ARDS. Methods: In this multicenter, randomized, double-blind, allocation-concealed, placebo-controlled trial (NCT03042143), patients with moderate to severe COVID-19-related ARDS were randomized to receive ORBCEL-C (400 million cells) or placebo (Plasma-Lyte 148). The primary safety and efficacy outcomes were the incidence of serious adverse events and oxygenation index at Day 7, respectively. Secondary outcomes included respiratory compliance, driving pressure, PaO2:FiO2 ratio, and Sequential Organ Failure Assessment score. Clinical outcomes relating to duration of ventilation, lengths of ICU and hospital stays, and mortality were collected. Long-term follow-up included diagnosis of interstitial lung disease at 1 year and significant medical events and mortality at 2 years. Transcriptomic analysis was performed on whole blood at Days 0, 4, and 7. Measurements and Main Results: Sixty participants were recruited (final analysis: n = 30 received ORBCEL-C, n = 29 received placebo; 1 participant in the placebo group withdrew consent). Six serious adverse events occurred in the ORBCEL-C group and three in the placebo group (risk ratio, 2.9 [95% confidence interval, 0.6-13.2]; P = 0.25). Day 7 mean (SD) oxygenation index did not differ (ORBCEL-C, 98.3 [57.2] cm H2O/kPa; placebo, 96.6 [67.3] cm H2O/kPa). There were no differences in secondary surrogate outcomes or in mortality at Day 28, Day 90, 1 year, or 2 years. There was no difference in the prevalence of interstitial lung disease at 1 year or significant medical events up to 2 years. ORBCEL-C modulated the peripheral blood transcriptome. Conclusion: ORBCEL-C MSCs were safe in subjects with moderate to severe COVID-19-related ARDS but did not improve surrogates of pulmonary organ dysfunction.
Collapse
Affiliation(s)
- Ellen A Gorman
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Jennifer Rynne
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Hannah J Gardiner
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Anthony J Rostron
- Sunderland Royal Hospital, South Tyneside and Sunderland National Health Service Foundation Trust, Sunderland, United Kingdom
- Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Andrew M Bentley
- Acute Intensive Care Unit, Wythenshawe Hospital, Manchester, United Kingdom
| | - David Brealey
- University College Hospital London, London, United Kingdom
| | | | - Gerard Curley
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mike Clarke
- Northern Ireland Clinical Trials Unit, Belfast, United Kingdom
| | - Ahilanadan Dushianthan
- University Hospital Southampton, Southampton, United Kingdom
- National Institute for Health and Care Research Southampton Biomedical Research Centre, University of Southampton, Southampton, United Kingdom
| | - Phillip Hopkins
- King's Trauma Centre, King's College Hospital, London, United Kingdom
| | - Colette Jackson
- Northern Ireland Clinical Trials Unit, Belfast, United Kingdom
| | - Kallirroi Kefela
- Department of Critical Care, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Anna Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - John G Laffey
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| | - Cliona McDowell
- Northern Ireland Clinical Trials Unit, Belfast, United Kingdom
| | - Margaret McFarland
- Department of Critical Care, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Jamie McFerran
- Northern Ireland Clinical Trials Unit, Belfast, United Kingdom
| | - Peter McGuigan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
- Department of Critical Care, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Gavin D Perkins
- Critical Care Unit, University Hospitals Birmingham, Birmingham, United Kingdom
- Warwick Clinical Trials Unit, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Jonathan Silversides
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
- Department of Critical Care, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Jon Smythe
- National Health Service Blood and Transplant, Oxford, United Kingdom
| | - Jacqui Thompson
- National Health Service Blood and Transplant, Birmingham, United Kingdom
| | | | - Ingeborg D M Welters
- Intensive Care Unit, Royal Liverpool University Hospital, Liverpool, United Kingdom
- Institute of Life Course Medical Sciences, University of Liverpool, Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
| | - Laura Amado-Rodríguez
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Guillermo Albaiceta
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Spain
- Departamento de Biología Funcional, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain; and
| | - Barry Williams
- Independent Patient and Public Representative, Sherborne, United Kingdom
| | - Manu Shankar-Hari
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Daniel F McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
- Department of Critical Care, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Cecilia M O'Kane
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
3
|
Couto PS, Al-Arawe N, Filgueiras IS, Fonseca DLM, Hinterseher I, Catar RA, Chinnadurai R, Bersenev A, Cabral-Marques O, Moll G, Verter F. Systematic review and meta-analysis of cell therapy for COVID-19: global clinical trial landscape, published safety/efficacy outcomes, cell product manufacturing and clinical delivery. Front Immunol 2023; 14:1200180. [PMID: 37415976 PMCID: PMC10321603 DOI: 10.3389/fimmu.2023.1200180] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/24/2023] [Indexed: 07/08/2023] Open
Abstract
During the pandemic of severe respiratory distress syndrome coronavirus 2 (SARS-CoV2), many novel therapeutic modalities to treat Coronavirus 2019 induced disease (COVID-19) were explored. This study summarizes 195 clinical trials of advanced cell therapies targeting COVID-19 that were registered over the two years between January 2020 to December 2021. In addition, this work also analyzed the cell manufacturing and clinical delivery experience of 26 trials that published their outcomes by July 2022. Our demographic analysis found the highest number of cell therapy trials for COVID-19 was in United States, China, and Iran (N=53, 43, and 19, respectively), with the highest number per capita in Israel, Spain, Iran, Australia, and Sweden (N=0.641, 0.232, 0,223, 0.194, and 0.192 trials per million inhabitants). The leading cell types were multipotent mesenchymal stromal/stem cells (MSCs), natural killer (NK) cells, and mononuclear cells (MNCs), accounting for 72%, 9%, and 6% of the studies, respectively. There were 24 published clinical trials that reported on infusions of MSCs. A pooled analysis of these MSC studies found that MSCs provide a relative risk reduction for all-cause COVID-19 mortality of RR=0.63 (95% CI 0.46 to 0.85). This result corroborates previously published smaller meta-analyses, which suggested that MSC therapy demonstrated a clinical benefit for COVID-19 patients. The sources of the MSCs used in these studies and their manufacturing and clinical delivery methods were remarkably heterogeneous, with some predominance of perinatal tissue-derived products. Our results highlight the important role that cell therapy products may play as an adjunct therapy in the management of COVID-19 and its related complications, as well as the importance of controlling key manufacturing parameters to ensure comparability between studies. Thus, we support ongoing calls for a global registry of clinical studies with MSC products that could better link cell product manufacturing and delivery methods to clinical outcomes. Although advanced cell therapies may provide an important adjunct treatment for patients affected by COVID-19 in the near future, preventing pathology through vaccination still remains the best protection to date. We conducted a systematic review and meta-analysis of advanced cell therapy clinical trials as potential novel treatment for COVID-19 (resulting from SARS-CoV-2 coronavirus infection), including analysis of the global clinical trial landscape, published safety/efficacy outcomes (RR/OR), and details on cell product manufacturing and clinical delivery. This study had a 2-year observation interval from start of January 2020 to end of December 2021, including a follow-up period until end of July to identify published outcomes, which covers the most vivid period of clinical trial activity, and is also the longest observation period studied until today. In total, we identified 195 registered advanced cell therapy studies for COVID-19, employing 204 individual cell products. Leading registered trial activity was attributed to the USA, China, and Iran. Through the end of July 2022, 26 clinical trials were published, with 24 out of 26 articles employing intravenous infusions (IV) of mesenchymal stromal/stem cell (MSC) products. Most of the published trials were attributed to China and Iran. The cumulative results from the 24 published studies employing infusions of MSCs indicated an improved survival (RR=0.63 with 95% Confidence Interval 0.46 to 0.85). Our study is the most comprehensive systematic review and meta-analysis on cell therapy trials for COVID-19 conducted to date, clearly identifying the USA, China, and Iran as leading advanced cell therapy trial countries for COVID-19, with further strong contributions from Israel, Spain, Australia and Sweden. Although advanced cell therapies may provide an important adjunct treatment for patients affected by COVID-19 in the future, preventing pathology through vaccination remains the best protection.
Collapse
Affiliation(s)
- Pedro S. Couto
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
- CellTrials.org and Parent’s Guide to Cord Blood Foundation, a non-profit organization headquartered in Brookeville, Brookeville, MD, United States
| | - Nada Al-Arawe
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany
- Vascular Surgery Clinic, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Igor S. Filgueiras
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Dennyson L. M. Fonseca
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of São Paulo (USP), São Paulo, SP, Brazil
| | - Irene Hinterseher
- Vascular Surgery Clinic, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Brandenburg Theodor Fontane, Neuruppin, Germany
- Fakultät der Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburg Technischen Universität (BTU) Cottbus-Senftenberg, Potsdam, Germany
| | - Rusan A. Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany
| | - Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, United States
| | - Alexey Bersenev
- Advanced Cell Therapy (ACT) Laboratory, Yale School of Medicine, New Haven, CT, United States
| | - Otávio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of São Paulo (USP), São Paulo, SP, Brazil
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
- Department of Pharmacy and Postgraduate Program of Health and Science, Federal University of Rio Grande do Norte, Natal, Brazil
- Department of Medicine, Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
- Laboratory of Medical Investigation 29, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Guido Moll
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frances Verter
- CellTrials.org and Parent’s Guide to Cord Blood Foundation, a non-profit organization headquartered in Brookeville, Brookeville, MD, United States
| |
Collapse
|
4
|
Kameishi S, Dunn CM, Oka M, Kim K, Cho YK, Song SU, Grainger DW, Okano T. Rapid and effective preparation of clonal bone marrow-derived mesenchymal stem/stromal cell sheets to reduce renal fibrosis. Sci Rep 2023; 13:4421. [PMID: 36932137 PMCID: PMC10023793 DOI: 10.1038/s41598-023-31437-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 03/11/2023] [Indexed: 03/19/2023] Open
Abstract
Allogeneic "off-the-shelf" mesenchymal stem/stromal cell (MSC) therapy requires scalable, quality-controlled cell manufacturing and distribution systems to provide clinical-grade products using cryogenic cell banking. However, previous studies report impaired cell function associated with administering freeze-thawed MSCs as single cell suspensions, potentially compromising reliable therapeutic efficacy. Using long-term culture-adapted clinical-grade clonal human bone marrow MSCs (cBMSCs) in this study, we engineered cBMSC sheets in 24 h to provide rapid preparation. We then sought to determine the influence of cBMSC freeze-thawing on both in vitro production of pro-regenerative factors and in vivo ability to reduce renal fibrosis in a rat model compared to freshly harvested cBMSCs. Sheets from freeze-thawed cBMSCs sheets exhibited comparable in vitro protein production and gene expression of pro-regenerative factors [e.g., hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), and interleukin 10 (IL-10)] to freshly harvested cBMSC sheets. Additionally, freeze-thawed cBMSC sheets successfully suppressed renal fibrosis in vivo in an established rat ischemia-reperfusion injury model. Despite previous studies reporting that freeze-thawed MSCs exhibit impaired cell functions compared to fresh MSC single cell suspensions, cell sheets engineered from freeze-thawed cBMSCs do not exhibit impaired cell functions, supporting critical steps toward future clinical translation of cBMSC-based kidney disease treatment.
Collapse
Affiliation(s)
- Sumako Kameishi
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA.
- Department of Molecular Pharmaceutics, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, Utah, 84112, USA.
| | - Celia M Dunn
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Masatoshi Oka
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Molecular Pharmaceutics, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, Utah, 84112, USA
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kyungsook Kim
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Molecular Pharmaceutics, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, Utah, 84112, USA
| | | | - Sun U Song
- SCM Lifescience Co., Ltd., Incheon, Republic of Korea
| | - David W Grainger
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Molecular Pharmaceutics, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, Utah, 84112, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA.
- Department of Molecular Pharmaceutics, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, Utah, 84112, USA.
- Institute for Advanced Biomedical Sciences, Tokyo Women's Medical University, Tokyo, Japan.
| |
Collapse
|
5
|
Sadeghi B, Witkamp M, Schefberger D, Arbman A, Ringdén O. Immunomodulation by placenta-derived decidua stromal cells. Role of histocompatibility, accessory cells and freeze-thawing. Cytotherapy 2023; 25:68-75. [PMID: 36333233 DOI: 10.1016/j.jcyt.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 09/15/2022] [Accepted: 10/07/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Human placenta-derived decidua stromal cells (DSCs) are newly introduced stromal cells that have successfully been used in several clinical trials for the treatment of acute inflammatory diseases. Despite published data about DSCs, deeper exploration of mechanisms of action and crosstalk with other immune cells need to be explored. METHODS In mixed lymphocyte culture (MLC), the splenocytes from Balb/c or B6 mice were stimulated using mitogen (concanavalin A), allogeneic (B6 or Balb/c splenocytes) or xenogeneic activation with human peripheral blood mononuclear cells. RESULTS When 10% of the mouse bone marrow-derived-MSC, being autologous, allogeneic or haploidentical (from F1), was added, >95% inhibition was seen. Using human (h)-DSCs, the inhibitory capacity was a median 68% as a xenogeneic immunomodulatory cell when used in mitogen and allogeneic setting in mice MLC. However, when human peripheral blood mononuclear cells were used as stimulator for mouse splenocyte (xenogeneic MLC), hDSC showed a median inhibition of 88%. We explored the presence and function of monocytes in the immunomodulatory function of stromal cells. CD14+ monocyte cells reduced the immunosuppressive effect by hDSC. hDSCs did not show any inhibitory effect on natural killer cell activation and proliferation by interleukin-2. In contrast DSCs increased natural killer proliferation by a median of 58%. Fresh or frozen-thawed hDSCs had similar inhibitory effects on human T-cell proliferation (both allo-stimulation and mitogen stimulation) in vitro. Cell viability at room temperature during 24 h was similar using fresh or freeze-thawed DSCs. CONCLUSIONS To conclude, histocompatibility and CD14+ monocyte cells had an impact on hDSC immunomodulation but frozen-thawed or freshly prepared cells did not.
Collapse
Affiliation(s)
- Behnam Sadeghi
- Translational Cell Therapy Research (TCR), Division of Pediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden.
| | - Myrèse Witkamp
- Translational Cell Therapy Research (TCR), Division of Pediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| | - Dominik Schefberger
- Translational Cell Therapy Research (TCR), Division of Pediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| | - Anna Arbman
- Translational Cell Therapy Research (TCR), Division of Pediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| | - Olle Ringdén
- Translational Cell Therapy Research (TCR), Division of Pediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
6
|
Zhang M, Zhao Y, Wang L, Zheng Y, Yu H, Dong X, He W, Yin Z, Wang Z. Study of the biological characteristics of human umbilical cord mesenchymal stem cells after long-time cryopreservation. Cell Tissue Bank 2022; 23:739-752. [PMID: 35066739 PMCID: PMC9675661 DOI: 10.1007/s10561-021-09973-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/21/2021] [Indexed: 01/22/2023]
Abstract
Human umbilical cord mesenchymal stem cells (hUC-MSCs) have considerable potential in cell therapy. Cryopreservation represents the gold standard in cell storage, but its effect on hUC-MSCs is still not well understood. The aim of this study was to investigate the effect of one year of cryopreservation and thawing on the biological characteristics of hUC-MSCs from the same donors. Fresh hUC-MSCs were cryopreserved in commercial freezing medium (serum-free CellBanker 2) at passage 2. After one year of cryopreservation, the hUC-MSCs were thawed and subcultured to passage 4. The comparison was performed in terms of followings: cell count, viability, morphology, proliferation capacity, differentiation potential and chromosomal stability. The total cell count and viability of hUC-MSCs before and after one year of cryopreservation were 1 × 107 and 96.34% and 0.943 × 107 and 93.81%, respectively. Cryopreserved and fresh hUC-MSCs displayed a similar cell doubling times, expressed the markers CD73, CD90, CD105 and were negative for the markers CD34, CD45, and HLA-DR. Karyotypes were found to be normal after one year of cryopreservation. The trilineage differentiation properties were maintained after cryopreservation. However, when compared to freshly isolated hUC-MSCs from the same donor, cryopreserved hUC-MSCs exhibited decreased expression of osteogenesis- and chondrogenesis-related genes including Runx2, Sox9, and Col1a1, and increased expression of adipogenesis-related genes. These results demonstrated that cryopreservation did not affect cell morphology, surface marker expression, cell viability, proliferative capacity, or chromosomal stability. However, the osteogenic and chondrogenic differentiation capacities of cryopreserved hUC-MSCs were slightly reduced compared with those of fresh cells from the same donor.
Collapse
Affiliation(s)
- Mingqi Zhang
- Department of Stem Cell Center of Precision Medicine Innovation Institute, He University, Hunnan District, No.66 Sishui Street, Shenyang, 110163, China
- Liaoning Key Lab of Ophthalmic Stem Cells, He University, Shenyang, China
- Liaoning Province Ophthalmic Stem Cell Clinical Application Research Center, He Eye Hospital, Shenyang, China
| | - Yan Zhao
- Department of Stem Cell Center of Precision Medicine Innovation Institute, He University, Hunnan District, No.66 Sishui Street, Shenyang, 110163, China
- Liaoning Key Lab of Ophthalmic Stem Cells, He University, Shenyang, China
| | - Le Wang
- Department of Stem Cell Center of Precision Medicine Innovation Institute, He University, Hunnan District, No.66 Sishui Street, Shenyang, 110163, China
- Liaoning Key Lab of Ophthalmic Stem Cells, He University, Shenyang, China
| | - Yuqiang Zheng
- Department of Stem Cell Center of Precision Medicine Innovation Institute, He University, Hunnan District, No.66 Sishui Street, Shenyang, 110163, China
- Liaoning Key Lab of Ophthalmic Stem Cells, He University, Shenyang, China
| | - Hui Yu
- Department of Stem Cell Center of Precision Medicine Innovation Institute, He University, Hunnan District, No.66 Sishui Street, Shenyang, 110163, China
- Liaoning Key Lab of Ophthalmic Stem Cells, He University, Shenyang, China
| | - Xiaoming Dong
- Department of Stem Cell Center of Precision Medicine Innovation Institute, He University, Hunnan District, No.66 Sishui Street, Shenyang, 110163, China
- Liaoning Key Lab of Ophthalmic Stem Cells, He University, Shenyang, China
| | - Wei He
- Department of Stem Cell Center of Precision Medicine Innovation Institute, He University, Hunnan District, No.66 Sishui Street, Shenyang, 110163, China
- Liaoning Key Lab of Ophthalmic Stem Cells, He University, Shenyang, China
- Liaoning Province Ophthalmic Stem Cell Clinical Application Research Center, He Eye Hospital, Shenyang, China
| | - Zhengqin Yin
- Department of Stem Cell Center of Precision Medicine Innovation Institute, He University, Hunnan District, No.66 Sishui Street, Shenyang, 110163, China.
| | - Zhuoshi Wang
- Department of Stem Cell Center of Precision Medicine Innovation Institute, He University, Hunnan District, No.66 Sishui Street, Shenyang, 110163, China.
- Liaoning Key Lab of Ophthalmic Stem Cells, He University, Shenyang, China.
| |
Collapse
|
7
|
Impact of Cryopreservation and Freeze-Thawing on Therapeutic Properties of Mesenchymal Stromal/Stem Cells and Other Common Cellular Therapeutics. CURRENT STEM CELL REPORTS 2022; 8:72-92. [PMID: 35502223 PMCID: PMC9045030 DOI: 10.1007/s40778-022-00212-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2022] [Indexed: 12/19/2022]
Abstract
Purpose of Review Cryopreservation and its associated freezing and thawing procedures–short “freeze-thawing”–are among the final steps in economically viable manufacturing and clinical application of diverse cellular therapeutics. Translation from preclinical proof-of-concept studies to larger clinical trials has indicated that these processes may potentially present an Achilles heel to optimal cell product safety and particularly efficacy in clinical trials and routine use. Recent Findings We review the current state of the literature on how cryopreservation of cellular therapies has evolved and how the application of this technique to different cell types is interlinked with their ability to engraft and function upon transfer in vivo, in particular for hematopoietic stem and progenitor cells (HSPCs), their progeny, and therapeutic cell products derived thereof. We also discuss pros and cons how this may differ for non-hematopoietic mesenchymal stromal/stem cell (MSC) therapeutics. We present different avenues that may be crucial for cell therapy optimization, both, for hematopoietic (e.g., effector, regulatory, and chimeric antigen receptor (CAR)-modified T and NK cell based products) and for non-hematopoietic products, such as MSCs and induced pluripotent stem cells (iPSCs), to achieve optimal viability, recovery, effective cell dose, and functionality of the cryorecovered cells. Summary Targeted research into optimizing the cryopreservation and freeze-thawing routines and the adjunct manufacturing process design may provide crucial advantages to increase both the safety and efficacy of cellular therapeutics in clinical use and to enable effective market deployment strategies to become economically viable and sustainable medicines.
Collapse
|
8
|
Ringdén O, Moll G, Gustafsson B, Sadeghi B. Mesenchymal Stromal Cells for Enhancing Hematopoietic Engraftment and Treatment of Graft-Versus-Host Disease, Hemorrhages and Acute Respiratory Distress Syndrome. Front Immunol 2022; 13:839844. [PMID: 35371003 PMCID: PMC8973075 DOI: 10.3389/fimmu.2022.839844] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/17/2022] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) possess profound immunomodulatory and regenerative properties that are of clinical use in numerous clinical indications with unmet medical need. Common sources of MSCs include among others, bone marrow (BM), fat, umbilical cord, and placenta-derived decidua stromal cells (DSCs). We here summarize our more than 20-years of scientific experience in the clinical use of MSCs and DSCs in different clinical settings. BM-MSCs were first explored to enhance the engraftment of autografts in hematopoietic cell transplantation (HCT) and osteogenesis imperfecta around 30 years ago. In 2004, our group reported the first anti-inflammatory use of BM-MSCs in a child with grade IV acute graft-versus-host disease (GvHD). Subsequent studies have shown that MSCs appear to be more effective in acute than chronic GvHD. Today BM-MSC-therapy is registered for acute GvHD in Japan and for GvHD in children in Canada and New Zeeland. MSCs first home to the lung following intravenous injection and exert strong local and systemic immunomodulatory effects on the host immune system. Thus, they were studied for ameliorating the cytokine storm in acute respiratory distress syndrome (ARDS). Both, MSCs and DSCs were used to treat SARS-CoV-2 coronavirus-induced disease 2019 (COVID-19)-induced ARDS. In addition, they were also used for other novel indications, such as pneumomediastinum, colon perforation, and radiculomyelopathy. MSC and DSCs trigger coagulation and were thus explored to stop hemorrhages. DSCs appear to be more effective for acute GvHD, ARDS, and hemorrhages, but randomized studies are needed to prove superiority. Stromal cell infusion is safe, well tolerated, and only gives rise to a slight fever in a limited number of patients, but no major side effects have been reported in multiple safety studies and metaanalysis. In this review we summarize current evidence from in vitro studies, animal models, and importantly our clinical experience, to support stromal cell therapy in multiple clinical indications. This encloses MSC's effects on the immune system, coagulation, and their safety and efficacy, which are discussed in relation to prominent clinical trials within the field.
Collapse
Affiliation(s)
- Olle Ringdén
- Translational Cell Therapy Research Group, Department of Clinical Sciences, Intervention and Technology (CLNTEC), Division of Pediatrics, Karolinska Institutet, Stockholm, Sweden
| | - Guido Moll
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT) and Berlin-Brandenburg School for Regenerative Therapies (BSRT), Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, All Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Britt Gustafsson
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Behnam Sadeghi
- Translational Cell Therapy Research Group, Department of Clinical Sciences, Intervention and Technology (CLNTEC), Division of Pediatrics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
9
|
Tan L, Liu X, Dou H, Hou Y. Characteristics and regulation of mesenchymal stem cell plasticity by the microenvironment — specific factors involved in the regulation of MSC plasticity. Genes Dis 2022; 9:296-309. [PMID: 35224147 PMCID: PMC8843883 DOI: 10.1016/j.gendis.2020.10.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/05/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs), multipotent stromal cells, have attracted extensive attention in the field of regenerative medicine and cell therapy due to the capacity of self-renewal, multilineage differentiation, and immune regulation. MSCs have different cellular effects in different diseases, and even have markedly different curative effects with different tissue sources, indicating the plasticity of MSCs. The phenotypes, secreted factors, and proliferative, migratory, differentiating, and immunomodulatory effects of MSCs depend on certain mediators present in their microenvironment. Understanding microenvironmental factors and their internal mechanisms in MSC responses may help in subsequent prediction and improvement of clinical benefits. This review highlighted the recent advances in MSC plasticity in the physiological and pathological microenvironment and multiple microenvironmental factors regulating MSC plasticity. It also highlighted some progress in the underlying molecular mechanisms of MSC remodeling in the microenvironment. It might provide references for the improvement in vitro culture of MSCs, clinical application, and in vivo induction.
Collapse
|
10
|
Giri J, Moll G. MSCs in Space: Mesenchymal Stromal Cell Therapeutics as Enabling Technology for Long-Distance Manned Space Travel. CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-022-00207-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Abstract
Purpose of Review
Advancements in space travel, such as space tourism into Earth’s orbit, but also the prospect of long-distance manned space travel to other celestial bodies such as Mars, has generated a clinical need for new enabling technologies to support the long-term well-being of humans during their passage. Here, we will give an outline on the clinical need and practical considerations to MSC therapy as enabling technology for long-distance manned space travel.
Recent Findings
Long-distance space travel entails a threat to the health of astronaut crews due to the low gravity environment and exposure to toxic radiation in space. Multi-organ-system degenerative changes, such as decline in musculoskeletal, hematopoietic, immune system function, and in particular risk of genetic mutations and cancer, are major health concerns. Physical training, pharmacological agents, and protective shielding are among the currently available methods to counteract harmful effects. However, a potential lack of adequate shielding, side effects of pharmacological compounds, and limitations to physical training suggest a need for new countermeasures, to protect space travellers to the best extent. Here, the prospect of cell-based therapy, e.g. mesenchymal stromal/stem cells (MSCs), has been subject to intense research, due to their potent regenerative and immunomodulatory properties. Off-the-shelf MSC therapeutics can be easily maintained in space due to the ambient extremely low-temperature environment, and cryorecovery and even culturing of MSCs under microgravity were shown to be feasible.
Summary
Designing new therapy against harmful radiation is urgent need in space travel. Here we will discuss aspects related to clinical MSC administration to optimize their therapeutic benefit. MSC-based therapy may aid in evolving protective countermeasures for space travellers.
Collapse
|
11
|
Moll G, Ankrum JA, Olson SD, Nolta JA. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:2-13. [PMID: 35641163 PMCID: PMC8895495 DOI: 10.1093/stcltm/szab005] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/27/2021] [Indexed: 11/22/2022] Open
Abstract
The number of mesenchymal stromal/stem cell (MSC) therapeutics and types of clinical applications have greatly diversified during the past decade, including rapid growth of poorly regulated “Stem Cell Clinics” offering diverse “Unproven Stem Cell Interventions.” This product diversification necessitates a critical evaluation of the reliance on the 2006 MSC minimal criteria to not only define MSC identity but characterize MSC suitability for intravascular administration. While high-quality MSC therapeutics have been safely administered intravascularly in well-controlled clinical trials, repeated case reports of mild-to-more-severe adverse events have been reported. These are most commonly related to thromboembolic complications upon infusion of highly procoagulant tissue factor (TF/CD142)-expressing MSC products. As TF/CD142 expression varies widely depending on the source and manufacturing process of the MSC product, additional clinical cell product characterization and guidelines are needed to ensure the safe use of MSC products. To minimize risk to patients receiving MSC therapy, we here propose to supplement the minimal criteria used for characterization of MSCs, to include criteria that assess the suitability of MSC products for intravascular use. If cell products are intended for intravascular delivery, which is true for half of all clinical applications involving MSCs, the effects of MSC on coagulation and hemocompatibility should be assessed and expression of TF/CD142 should be included as a phenotypic safety marker. This adjunct criterion will ensure both the identity of the MSCs as well as the safety of the MSCs has been vetted prior to intravascular delivery of MSC products.
Collapse
Affiliation(s)
- Guido Moll
- BIH Center for Regenerative Therapies (BCRT) and Berlin Brandenburg School of Regenerative Therapies (BSRT), Berlin Institute of Health (BIH) at the Charité—Universitätsmedizin Berlin, corporate member of Freie Universität zu Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Corresponding author: Guido Moll, PhD, BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany.
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering and Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, USA
| | - Scott D Olson
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Jan A Nolta
- Director of the Stem Cell Program, University of California Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
12
|
Kaiser D, Otto NM, McCallion O, Hoffmann H, Zarrinrad G, Stein M, Beier C, Matz I, Herschel M, Hester J, Moll G, Issa F, Reinke P, Roemhild A. Freezing Medium Containing 5% DMSO Enhances the Cell Viability and Recovery Rate After Cryopreservation of Regulatory T Cell Products ex vivo and in vivo. Front Cell Dev Biol 2021; 9:750286. [PMID: 34926446 PMCID: PMC8677839 DOI: 10.3389/fcell.2021.750286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/05/2021] [Indexed: 12/29/2022] Open
Abstract
Cell therapies have significant therapeutic potential in diverse fields including regenerative medicine, transplantation tolerance, and autoimmunity. Within these fields, regulatory T cells (Treg) have been deployed to ameliorate aberrant immune responses with great success. However, translation of the cryopreservation strategies employed for other cell therapy products, such as effector T cell therapies, to Treg therapies has been challenging. The lack of an optimized cryopreservation strategy for Treg products presents a substantial obstacle to their broader application, particularly as administration of fresh cells limits the window available for sterility and functional assessment. In this study, we aimed to develop an optimized cryopreservation strategy for our CD4+CD25+Foxp3+ Treg clinical product. We investigate the effect of synthetic or organic cryoprotectants including different concentrations of DMSO on Treg recovery, viability, phenotype, cytokine production, suppressive capacity, and in vivo survival following GMP-compliant manufacture. We additionally assess the effect of adding the extracellular cryoprotectant polyethylene glycol (PEG), or priming cellular expression of heat shock proteins as strategies to improve viability. We find that cryopreservation in serum-free freezing medium supplemented with 10% human serum albumin and 5% DMSO facilitates improved Treg recovery and functionality and supports a reduced DMSO concentration in Treg cryopreservation protocols. This strategy may be easily incorporated into clinical manufacture protocols for future studies.
Collapse
Affiliation(s)
- Daniel Kaiser
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Natalie Maureen Otto
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Nephrology and Internal Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Oliver McCallion
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Henrike Hoffmann
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ghazaleh Zarrinrad
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Maik Stein
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Carola Beier
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Isabell Matz
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marleen Herschel
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Joanna Hester
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Guido Moll
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Fadi Issa
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Petra Reinke
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Nephrology and Internal Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andy Roemhild
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
13
|
Horie S, Gonzalez H, Brady J, Devaney J, Scully M, O’Toole D, Laffey JG. Fresh and Cryopreserved Human Umbilical-Cord-Derived Mesenchymal Stromal Cells Attenuate Injury and Enhance Resolution and Repair following Ventilation-Induced Lung Injury. Int J Mol Sci 2021; 22:ijms222312842. [PMID: 34884645 PMCID: PMC8657992 DOI: 10.3390/ijms222312842] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Ventilator-induced lung injury (VILI) frequently worsens acute respiratory distress syndrome (ARDS) severity. Human mesenchymal stem/stromal cells (MSCs) offer considerable therapeutic promise, but the key impediments of clinical translation stem from limitations due to cell source and availability, and concerns regarding the loss of efficacy following cryopreservation. These experiments compared the efficacy of umbilical-cord-derived MSCs (UC-MSCs), a readily available and homogenous tissue source, to the previously more widely utilised bone-marrow-derived MSCs (BM-MSCs). We assessed their capacity to limit inflammation, resolve injury and enhance repair in relevant lung mechanical stretch models, and the impact of cryopreservation on therapeutic efficacy. Methods: In series 1, confluent alveolar epithelial layers were subjected to cyclic mechanical stretch (22% equibiaxial strain) and wound injury, and the potential of the secretome from BM- and UC-derived MSCs to attenuate epithelial inflammation and cell death, and enhance wound repair was determined. In series 2, anesthetized rats underwent VILI, and later received, in a randomised manner, 1 × 107 MSCs/kg intravenously, that were: (i) fresh BM-MSCs, (ii) fresh UC-MSCs or (iii) cryopreserved UC-MSCs. Control animals received a vehicle (PBS). The extent of the resolution of inflammation and injury, and repair was measured at 24 h. Results: Conditioned medium from BM-MSCs and UC-MSCs comparably decreased stretch-induced pulmonary epithelial inflammation and cell death. BM-MSCs and UC-MSCs comparably enhanced wound resolution. In animals subjected to VILI, both fresh BM-MSCs and UC-MSCs enhanced injury resolution and repair, while cryopreserved UC-MSCs comparably retained their efficacy. Conclusions: Cryopreserved UC-MSCs can reduce stretch-induced inflammation and cell death, enhance wound resolution, and enhance injury resolution and repair following VILI. Cryopreserved UC-MSCs represent a more abundant, cost-efficient, less variable and equally efficacious source of therapeutic MSC product.
Collapse
Affiliation(s)
- Shahd Horie
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Biomedical Sciences Building, H91 TK33 Galway, Ireland
| | - Hector Gonzalez
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Biomedical Sciences Building, H91 TK33 Galway, Ireland
| | - Jack Brady
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Biomedical Sciences Building, H91 TK33 Galway, Ireland
| | - James Devaney
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Biomedical Sciences Building, H91 TK33 Galway, Ireland
| | - Michael Scully
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Medicine, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland
| | - Daniel O’Toole
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Biomedical Sciences Building, H91 TK33 Galway, Ireland
- Correspondence: (D.O.); (J.G.L.)
| | - John G. Laffey
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Biomedical Sciences Building, H91 TK33 Galway, Ireland
- Medicine, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland
- Department of Anaesthesia, Galway University Hospitals, SAOLTA University Health Group, H91 YR71 Galway, Ireland
- Correspondence: (D.O.); (J.G.L.)
| |
Collapse
|
14
|
Calcat-i-Cervera S, Sanz-Nogués C, O'Brien T. When Origin Matters: Properties of Mesenchymal Stromal Cells From Different Sources for Clinical Translation in Kidney Disease. Front Med (Lausanne) 2021; 8:728496. [PMID: 34616756 PMCID: PMC8488400 DOI: 10.3389/fmed.2021.728496] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
Advanced therapy medicinal products (ATMPs) offer new prospects to improve the treatment of conditions with unmet medical needs. Kidney diseases are a current major health concern with an increasing global prevalence. Chronic renal failure appears after many years of impairment, which opens a temporary window to apply novel therapeutic approaches to delay or halt disease progression. The immunomodulatory, anti-inflammatory, and pro-regenerative properties of mesenchymal stromal cells (MSCs) have sparked interest for their use in cell-based regenerative therapies. Currently, several early-phase clinical trials have been completed and many are ongoing to explore MSC safety and efficacy in a wide range of nephropathies. However, one of the current roadblocks to the clinical translation of MSC therapies relates to the lack of standardization and harmonization of MSC manufacturing protocols, which currently hinders inter-study comparability. Studies have shown that cell culture processing variables can have significant effects on MSC phenotype and functionality, and these are highly variable across laboratories. In addition, heterogeneity within MSC populations is another obstacle. Furthermore, MSCs may be isolated from several sources which adds another variable to the comparative assessment of outcomes. There is now a growing body of literature highlighting unique and distinctive properties of MSCs according to the tissue origin, and that characteristics such as donor, age, sex and underlying medical conditions may alter the therapeutic effect of MSCs. These variables must be taken into consideration when developing a cell therapy product. Having an optimal scale-up strategy for MSC manufacturing is critical for ensuring product quality while minimizing costs and time of production, as well as avoiding potential risks. Ideally, optimal scale-up strategies must be carefully considered and identified during the early stages of development, as making changes later in the bioprocess workflow will require re-optimization and validation, which may have a significant long-term impact on the cost of the therapy. This article provides a summary of important cell culture processing variables to consider in the scale-up of MSC manufacturing as well as giving a comprehensive review of tissue of origin-specific biological characteristics of MSCs and their use in current clinical trials in a range of renal pathologies.
Collapse
Affiliation(s)
| | | | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), CÚRAM, Biomedical Science Building, National University of Ireland, Galway, Ireland
| |
Collapse
|
15
|
Lohmann S, Pool MBF, Rozenberg KM, Keller AK, Moers C, Møldrup U, Møller BK, Lignell SJM, Krag S, Sierra-Parraga JM, Lo Faro ML, Hunter J, Hoogduijn MJ, Baan CC, Leuvenink HGD, Ploeg RJ, Eijken M, Jespersen B. Mesenchymal stromal cell treatment of donor kidneys during ex vivo normothermic machine perfusion: A porcine renal autotransplantation study. Am J Transplant 2021; 21:2348-2359. [PMID: 33382194 DOI: 10.1111/ajt.16473] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 01/25/2023]
Abstract
Normothermic machine perfusion (NMP) of injured kidneys offers the opportunity for interventions to metabolically active organs prior to transplantation. Mesenchymal stromal cells (MSCs) can exert regenerative and anti-inflammatory effects in ischemia-reperfusion injury. The aims of this study were to evaluate the safety and feasibility of MSC treatment of kidneys during NMP using a porcine autotransplantation model, and examine potential MSC treatment-associated kidney improvements up to 14 days posttransplant. After 75 min of kidney warm ischemia, four experimental groups of n = 7 underwent 14 h of oxygenated hypothermic machine perfusion. In three groups this was followed by 240 min of NMP with infusion of vehicle, 10 million porcine, or 10 million human adipose-derived MSCs. All kidneys were autotransplanted after contralateral nephrectomy. MSC treatment did not affect perfusion hemodynamics during NMP or cause adverse effects at reperfusion, with 100% animal survival. MSCs did not affect plasma creatinine, glomerular filtration rate, neutrophil gelatinase-associated lipocalin concentrations or kidney damage assessed by histology during the 14 days, and MSCs retention was demonstrated in renal cortex. Infusing MSCs during ex vivo NMP of porcine kidneys was safe and feasible. Within the short posttransplant follow-up period, no beneficial effects of ex vivo MSC therapy could be demonstrated.
Collapse
Affiliation(s)
- Stine Lohmann
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Merel B F Pool
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Anna K Keller
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Cyril Moers
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ulla Møldrup
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Bjarne K Møller
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Stina J M Lignell
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Søren Krag
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Jesus M Sierra-Parraga
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Maria L Lo Faro
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - James Hunter
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Martin J Hoogduijn
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Henri G D Leuvenink
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rutger J Ploeg
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Marco Eijken
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Bente Jespersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
16
|
Crippa S, Santi L, Berti M, De Ponti G, Bernardo ME. Role of ex vivo Expanded Mesenchymal Stromal Cells in Determining Hematopoietic Stem Cell Transplantation Outcome. Front Cell Dev Biol 2021; 9:663316. [PMID: 34017834 PMCID: PMC8129582 DOI: 10.3389/fcell.2021.663316] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
Overall, the human organism requires the production of ∼1 trillion new blood cells per day. Such goal is achieved via hematopoiesis occurring within the bone marrow (BM) under the tight regulation of hematopoietic stem and progenitor cell (HSPC) homeostasis made by the BM microenvironment. The BM niche is defined by the close interactions of HSPCs and non-hematopoietic cells of different origin, which control the maintenance of HSPCs and orchestrate hematopoiesis in response to the body’s requirements. The activity of the BM niche is regulated by specific signaling pathways in physiological conditions and in case of stress, including the one induced by the HSPC transplantation (HSCT) procedures. HSCT is the curative option for several hematological and non-hematological diseases, despite being associated with early and late complications, mainly due to a low level of HSPC engraftment, impaired hematopoietic recovery, immune-mediated graft rejection, and graft-versus-host disease (GvHD) in case of allogenic transplant. Mesenchymal stromal cells (MSCs) are key elements of the BM niche, regulating HSPC homeostasis by direct contact and secreting several paracrine factors. In this review, we will explore the several mechanisms through which MSCs impact on the supportive activity of the BM niche and regulate HSPC homeostasis. We will further discuss how the growing understanding of such mechanisms have impacted, under a clinical point of view, on the transplantation field. In more recent years, these results have instructed the design of clinical trials to ameliorate the outcome of HSCT, especially in the allogenic setting, and when low doses of HSPCs were available for transplantation.
Collapse
Affiliation(s)
- Stefania Crippa
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ludovica Santi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Margherita Berti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giada De Ponti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Maria Ester Bernardo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy.,University Vita-Salute San Raffaele, Faculty of Medicine, Milan, Italy
| |
Collapse
|
17
|
AlHindi M, Philip MR. Osteogenic differentiation potential and quantification of fresh and cryopreserved dental follicular stem cells-an in vitro analysis. J Stem Cells Regen Med 2021; 17:28-34. [PMID: 34434005 PMCID: PMC8372412 DOI: 10.46582/jsrm.1701004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/23/2021] [Indexed: 11/19/2022]
Abstract
Purpose: To isolate and characterize mesenchymal stem cells of dental follicle from fresh and cryopreserved samples and to test any significant difference in their osteogenic differentiation potential by using digital imaging software. We also investigated whether the cryoprotectant used and its concentration is able to maintain cell count and viability. Methods: Mesenchymal stem cells (MSCs) were isolated from dental follicle of impacted third molars. The osteogenic differentiation potential of dental follicle stem cells was assessed using alizarin red and alkaline phosphatase staining followed by digital imaging quantification of the stains. Results: Dental follicle cells have shown typical characterisation by exhibiting the stem cell stromal markers and hematopoietic markers, but there was variance in the percentage of expression in fresh and cryopreserved samples. There was considerable osteogenic differentiation potential in the fresh sample compared to cryopreserved sample. The cell count and viability were preserved in both samples. Conclusions: The results in the study have shown wide variation of osteogenic differentiation potential in fresh and cryopreserved samples. Also, the cryoprotectant was found to be effective in its purpose at the specified concentration.
Collapse
Affiliation(s)
- Maryam AlHindi
- Department of Maxillofacial Surgery, College of Dentistry, King Saud University, Riyadh, PO Box11545, KSA
| | - Manju Roby Philip
- Department of Maxillofacial Surgery, College of Dentistry, King Saud University, Riyadh, PO Box11545, KSA
| |
Collapse
|
18
|
Chen J, Zheng CX, Jin Y, Hu CH. Mesenchymal stromal cell-mediated immune regulation: A promising remedy in the therapy of type 2 diabetes mellitus. STEM CELLS (DAYTON, OHIO) 2021; 39:838-852. [PMID: 33621403 DOI: 10.1002/stem.3357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/03/2021] [Indexed: 11/09/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a major threat to global public health, with increasing prevalence as well as high morbidity and mortality, to which immune dysfunction has been recognized as a crucial contributor. Mesenchymal stromal cells (MSCs), obtained from various sources and possessing potent immunomodulatory abilities, have displayed great therapeutic potential for T2DM. Interestingly, the immunomodulatory capabilities of MSCs are endowed and plastic. Among the multiple mechanisms involved in MSC-mediated immune regulation, the paracrine effects of MSCs have attracted much attention. Of note, extracellular vesicles (EVs), an important component of MSC secretome, have emerged as pivotal mediators of their immunoregulatory effects. Particularly, the necrobiology of MSCs, especially apoptosis, has recently been revealed to affect their immunomodulatory functions in vivo. In specific, a variety of preclinical studies have demonstrated the beneficial effects of MSCs on improving islet function and ameliorating insulin resistance. More importantly, clinical trials have further uncovered the therapeutic potential of MSCs for T2DM. In this review, we outline current knowledge regarding the plasticity and underlying mechanisms of MSC-mediated immune modulation, focusing on the paracrine effects. We also summarize the applications of MSC-based therapies for T2DM in both preclinical studies and clinical trials, with particular emphasis on the modulation of immune system.
Collapse
Affiliation(s)
- Ji Chen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases,Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China.,Department of Oral Implantology, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Chen-Xi Zheng
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases,Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Yan Jin
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases,Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Cheng-Hu Hu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases,Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, People's Republic of China
| |
Collapse
|
19
|
Herzig MC, Christy BA, Montgomery RK, Delavan CP, Jensen KJ, Lovelace SE, Cantu C, Salgado CL, Cap AP, Bynum JA. Interactions of human mesenchymal stromal cells with peripheral blood mononuclear cells in a Mitogenic proliferation assay. J Immunol Methods 2021; 492:113000. [PMID: 33609532 DOI: 10.1016/j.jim.2021.113000] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/05/2020] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Immunomodulation by mesenchymal stromal cells (MSCs) is a potentially important therapeutic modality. MSCs suppress peripheral blood mononuclear cell (PBMC) proliferation in vitro, suggesting a mechanism for suppressing inflammatory responses in vivo. This study details the interactions of PBMCs and MSCs. METHODS Pooled human PBMCs and MSCs were co-cultured at different MSC:PBMC ratios and harvested from 0 to 120 h, with and without phytohaemagglutin A (PHA) stimulation. Proliferation of adherent MSCs and non-adherent PBMCs was assessed by quantitation of ATP levels. PBMC surface marker expression was analyzed by flow cytometry. Indoleamine 2,3-dioxygenase (IDO) activity was determined by kynurenine assay and IDO mRNA by RT-PCR. Cytokine release was measured by ELISA. Immunofluorescent microscopy detected MSC, PBMC, monocyte (CD14+) and apoptotic events. RESULTS PBMC proliferation in response to PHA gave a robust ATP signal by 72 h, which was suppressed by co-culture with densely plated MSCs. Very low level MSC seeding densities relative to PBMC number reproducibly stimulated PBMC proliferation. The CD4+/CD3+ population significantly decreased over time while the CD8+/CD3+ population significantly increased. No change in CD4+/CD8+ ratio is seen with high density MSC co-culture; very low density MSCs augment the changes seen in PHA stimulated PBMCs alone. IDO activity in MSCs co-cultured with PBMCs correlated with PBMC suppression. MSCs increased the secretion of IL-10 and IL-6 from stimulated co-cultures and decreased TNF-α secretion. In stimulated co-culture, low density MSCs decreased in number; fluorescence immunomicroscopy detected association of PBMC with MSC and phosphatidyl serine externalization in both cell populations. CONCLUSIONS A bidirectional interaction between MSCs and PBMCs occurs during co-culture. High numbers of MSCs inhibit PHA-stimulated PBMC proliferation and the PBMC response to stimulation; low numbers of MSCs augment these responses. Low density MSCs are susceptible to attrition, apparently by PBMC-induced apoptosis. These results may have direct application when considering therapeutic dosing of patients; low MSC doses may have unintended detrimental consequences.
Collapse
Affiliation(s)
- Maryanne C Herzig
- Blood and Coagulation Research, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States of America.
| | - Barbara A Christy
- Blood and Coagulation Research, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States of America; Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX, United States of America.
| | - Robbie K Montgomery
- Blood and Coagulation Research, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States of America.
| | - Christopher P Delavan
- Blood and Coagulation Research, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States of America.
| | - Katherine J Jensen
- Blood and Coagulation Research, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States of America.
| | - Sarah E Lovelace
- Blood and Coagulation Research, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States of America.
| | - Carolina Cantu
- Blood and Coagulation Research, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States of America.
| | - Christi L Salgado
- Blood and Coagulation Research, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States of America.
| | - Andrew P Cap
- Blood and Coagulation Research, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States of America; Department of Surgery, UT Health San Antonio, San Antonio, TX, United States of America.
| | - James A Bynum
- Blood and Coagulation Research, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States of America.
| |
Collapse
|
20
|
Bahsoun S, Coopman K, Akam EC. Quantitative assessment of the impact of cryopreservation on human bone marrow-derived mesenchymal stem cells: up to 24 h post-thaw and beyond. Stem Cell Res Ther 2020; 11:540. [PMID: 33317625 PMCID: PMC7734731 DOI: 10.1186/s13287-020-02054-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The effects of cryopreservation on human bone marrow-derived mesenchymal stem cells (hBM-MSCs) are still ill-defined. In this study, a quantitative approach was adopted to measure several post-thaw cell attributes in order to provide an accurate reflection of the freezing and thawing impact. METHODS Fresh and cryopreserved passage-matched cells from three different donors were discretely analysed and compared for their viability, apoptosis level, phenotypic marker expression, metabolic activity, adhesion potential, proliferation rate, colony-forming unit ability (CFUF) and differentiation potentials. RESULTS The results of this study show that cryopreservation reduces cell viability, increases apoptosis level and impairs hBM-MSC metabolic activity and adhesion potential in the first 4 h after thawing. At 24 h post-thaw, cell viability recovered, and apoptosis level dropped but metabolic activity and adhesion potential remained lower than fresh cells. This suggests that a 24-h period is not enough for a full recovery. Beyond 24 h post-thaw, the observed effects are variable for the three cell lines. While no difference is observed in the pre- and post-cryopreservation proliferation rate, cryopreservation reduced the CFUF ability of two of the cell lines and variably affected the adipogenic and osteogenic differentiation potentials of the three cell lines. CONCLUSION The data collected in this study clearly show that fresh and cryopreserved hBM-MSCs are different, and these differences will inevitably introduce variabilities to the product and process development and subsequently imply financial losses. In order to avoid product divergence pre- and post-cryopreservation, effective strategies to mitigate freezing effects must be developed and implemented.
Collapse
Affiliation(s)
- Soukaina Bahsoun
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK.
| | - Karen Coopman
- Centre for Biological Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| | - Elizabeth C Akam
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| |
Collapse
|
21
|
Pietrosi G, Fernández-Iglesias A, Pampalone M, Ortega-Ribera M, Lozano JJ, García-Calderó H, Abad-Jordà L, Conaldi PG, Parolini O, Vizzini G, Luca A, Bosch J, Gracia-Sancho J. Human amniotic stem cells improve hepatic microvascular dysfunction and portal hypertension in cirrhotic rats. Liver Int 2020; 40:2500-2514. [PMID: 32996708 DOI: 10.1111/liv.14610] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Portal hypertension is the main consequence of cirrhosis, responsible for the complications defining clinical decompensation. The only cure for decompensated cirrhosis is liver transplantation, but it is a limited resource and opens the possibility of regenerative therapy. We investigated the potential of primary human amniotic membrane-derived mesenchymal stromal (hAMSCs) and epithelial (hAECs) stem cells for the treatment of portal hypertension and decompensated cirrhosis. METHODS In vitro: Primary liver sinusoidal endothelial cells (LSECs) and hepatic stellate cells (HSCs) from cirrhotic rats (chronic CCl4 inhalation) were co-cultured with hAMSCs, hAECs or vehicle for 24 hours, and their RNA profile was analysed. In vivo: CCl4-cirrhotic rats received 4x106 hAMSCs, 4x106 hAECs, or vehicle (NaCl 0.9%) (intraperitoneal). At 2-weeks we analysed: a) portal pressure (PP) and hepatic microvascular function; b) LSECs and HSCs phenotype; c) hepatic fibrosis and inflammation. RESULTS In vitro experiments revealed sinusoidal cell phenotype amelioration when co-cultured with stem cells. Cirrhotic rats receiving stem cells, particularly hAMSCs, had significantly lower PP than vehicle-treated animals, together with improved liver microcirculatory function. This hemodynamic amelioration was associated with improvement in LSECs capillarization and HSCs de-activation, though hepatic collagen was not reduced. Rats that received amnion derived stem cells had markedly reduced hepatic inflammation and oxidative stress. Finally, liver function tests significantly improved in rats receiving hAMSCs. CONCLUSIONS This preclinical study shows that infusion of human amniotic stem cells effectively decreases PP by ameliorating liver microcirculation, suggesting that it may represent a new treatment option for advanced cirrhosis with portal hypertension.
Collapse
Affiliation(s)
- Giada Pietrosi
- Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione, IRCCS-ISMETT, Palermo, Italy
| | - Anabel Fernández-Iglesias
- Liver Vascular Biology Research Group, Hepatic Hemodynamic Lab. IDIBAPS-Hospital Clínic, CIBEREHD, Barcelona, Spain
| | | | - Martí Ortega-Ribera
- Liver Vascular Biology Research Group, Hepatic Hemodynamic Lab. IDIBAPS-Hospital Clínic, CIBEREHD, Barcelona, Spain
| | - Juan J Lozano
- Liver Vascular Biology Research Group, Hepatic Hemodynamic Lab. IDIBAPS-Hospital Clínic, CIBEREHD, Barcelona, Spain
| | - Héctor García-Calderó
- Liver Vascular Biology Research Group, Hepatic Hemodynamic Lab. IDIBAPS-Hospital Clínic, CIBEREHD, Barcelona, Spain
| | - Laia Abad-Jordà
- Liver Vascular Biology Research Group, Hepatic Hemodynamic Lab. IDIBAPS-Hospital Clínic, CIBEREHD, Barcelona, Spain
| | - Pier G Conaldi
- Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione, IRCCS-ISMETT, Palermo, Italy
| | - Ornella Parolini
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli, Rome, Italy.,Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Vizzini
- Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione, IRCCS-ISMETT, Palermo, Italy
| | - Angelo Luca
- Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione, IRCCS-ISMETT, Palermo, Italy
| | - Jaime Bosch
- Liver Vascular Biology Research Group, Hepatic Hemodynamic Lab. IDIBAPS-Hospital Clínic, CIBEREHD, Barcelona, Spain.,Hepatology, Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Hepatic Hemodynamic Lab. IDIBAPS-Hospital Clínic, CIBEREHD, Barcelona, Spain.,Hepatology, Department for Biomedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
22
|
Chinnadurai R, Forsberg MH, Kink JA, Hematti P, Capitini CM. Use of MSCs and MSC-educated macrophages to mitigate hematopoietic acute radiation syndrome. CURRENT STEM CELL REPORTS 2020; 6:77-85. [PMID: 32944493 DOI: 10.1007/s40778-020-00176-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Purpose of Review Innovative and minimally toxic treatment approaches are sorely needed for the prevention and treatment of hematopoietic acute radiation syndrome (H-ARS). Cell therapies have been increasingly studied for their potential use as countermeasures for accidental and intentional ionizing radiation exposures which can lead to fatal ARS. Mesenchymal stem/stromal cells (MSCs) are a cell therapy that have shown promising results in preclinical studies of ARS, and are being developed in clinical trials specifically for H-ARS. MSCs, MSC-educated macrophages (MEMs) and MSC-exosome educated macrophages (EEMs) all have the potential to be used as adoptive cell therapies for H-ARS. Here we review how MSCs have been reported to mitigate inflammation from radiation injury while also stimulating hematopoiesis during ARS. Recent findings We discuss emerging work with immune cell subsets educated by MSCs, including MEMs and EEMs, in promoting hematopoiesis in xenogeneic models of ARS. We also discuss the first placental-derived MSC product to enter phase I trials, PLX-R18, and the challenges faced by bringing MSC and other cell therapies into the clinic for treating ARS. Summary Although MSCs, MEMs and EEMs are potential cell therapy candidates in promoting hematopoietic HRS, challenges persist in translational clinical development of these products to the clinic. Whether any of these cellular therapies will be sufficient as stand-alone therapies to mitigate H-ARS or if they will be a bridging therapy that insures survival until a curative allogeneic hematopoietic stem cell transplant can be performed are the key questions that will have to be answered.
Collapse
Affiliation(s)
- Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA
| | - Matthew H Forsberg
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - John A Kink
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Christian M Capitini
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
23
|
Non-toxic freezing media to retain the stem cell reserves in adipose tissues. Cryobiology 2020; 96:137-144. [PMID: 32687840 DOI: 10.1016/j.cryobiol.2020.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 07/01/2020] [Accepted: 07/16/2020] [Indexed: 01/10/2023]
Abstract
Subcutaneous adipose tissue is a rich source of stromal vascular fraction (SVF) and adipose-derived stromal/stem cells (ASCs) that are inherently multipotent and exhibit regenerative properties. In current practice, lipoaspirate specimens harvested from liposuction surgeries are routinely discarded as a biohazard waste due to a lack of simple, cost effective, and validated cryopreservation protocols. The aim of this study is to develop a xenoprotein-free cryoprotective agent cocktail that will allow for short-term (up to 6 months) preservation of lipoaspirate tissues suitable for fat grafting and/or stromal/stem cell isolation when stored at achievable temperatures (-20 °C or -80 °C). Lipoaspirates donated by three consenting healthy donors undergoing elective cosmetic liposuction surgeries were suspended in five freezing media (FM1: 10% DMSO and 35% BSA; FM2: 2% DMSO and 43% BSA; FM3: 10% DMSO and 35% lipoaspirate saline; FM4: 2% DMSO and 6% HSA; and FM5: 40% lipoaspirate saline and 10% PVP) all suspended in 1X DMEM/F12 and frozen using commercially available freezers (-20 °C or -80 °C) and stored at least for a 1 month. After 1 month of freezing storage, SVF cells and ASCs were isolated from the frozen-thawed lipoaspirates by digestion with collagenase type I. Cell viability was evaluated by fluorescence microscopy after staining with acridine orange and ethidium bromide. The SVF isolated from lipoaspirates frozen at -80 °C retained comparable cell viability with the tested freezing media (FM2, FM3, FM4) comparable with the conventional DMSO and animal serum media (FM1), whereas the FM5 media resulted in lower viability. In contrast, tissues frozen and stored at -20 °C did not yield live SVF cells after thawing and collagenase digestion. The surface marker expression (CD90, CD29, CD34, CD146, CD31, and CD45) of ASCs from frozen lipoaspirates at -80 °C in different cryoprotectant media were also evaluated and no significant differences were found between the groups. The adipogenic and osteogenic differentiation potential were studied by histochemical staining and gene expression by qRT-PCR. Oil Red O staining for adipogenesis revealed that the CPA media FM1, FM4 and FM5 displayed robust differentiation. Alizarin Red S staining for osteogenesis revealed that FM1 and FM4 media displayed superior differentiation in comparison to other tested media. Measurement of adipogenic and osteogenic gene expression by qRT-PCR provided similar outcomes and indicated that FM4 CPA media comparable with FM1 for adipogenesis and osteogenesis.
Collapse
|
24
|
Ntege EH, Sunami H, Shimizu Y. Advances in regenerative therapy: A review of the literature and future directions. Regen Ther 2020; 14:136-153. [PMID: 32110683 PMCID: PMC7033303 DOI: 10.1016/j.reth.2020.01.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/14/2020] [Accepted: 01/26/2020] [Indexed: 12/14/2022] Open
Abstract
There is enormous global anticipation for stem cell-based therapies that are safe and effective. Numerous pre-clinical studies present encouraging results on the therapeutic potential of different cell types including tissue derived stem cells. Emerging evidences in different fields of research suggest several cell types are safe, whereas their therapeutic application and effectiveness remain challenged. Multiple factors that influence treatment outcomes are proposed including immunocompatibility and potency, owing to variations in tissue origin, ex-vivo methodologies for preparation and handling of the cells. This communication gives an overview of literature data on the different types of cells that are potentially promising for regenerative therapy. As a case in point, the recent trends in research and development of the mesenchymal stem cells (MSCs) for cell therapy are considered in detail. MSCs can be isolated from a variety of tissues and organs in the human body including bone marrow, adipose, synovium, and perinatal tissues. However, MSC products from the different tissue sources exhibit unique or varied levels of regenerative abilities. The review finally focuses on adipose tissue-derived MSCs (ASCs), with the unique properties such as easier accessibility and abundance, excellent proliferation and differentiation capacities, low immunogenicity, immunomodulatory and many other trophic properties. The suitability and application of the ASCs, and strategies to improve the innate regenerative capacities of stem cells in general are highlighted among others.
Collapse
Affiliation(s)
- Edward H. Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Japan
- Research Center for Regenerative Medicine, School of Medicine, University of the Ryukyus, Japan
| | - Hiroshi Sunami
- Research Center for Regenerative Medicine, School of Medicine, University of the Ryukyus, Japan
| | - Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Japan
| |
Collapse
|
25
|
Fu X, Xu B, Jiang J, Du X, Yu X, Yan Y, Li S, Inglis BM, Ma H, Wang H, Pei X, Si W. Effects of cryopreservation and long-term culture on biological characteristics and proteomic profiles of human umbilical cord-derived mesenchymal stem cells. Clin Proteomics 2020; 17:15. [PMID: 32489333 PMCID: PMC7247169 DOI: 10.1186/s12014-020-09279-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 05/15/2020] [Indexed: 12/14/2022] Open
Abstract
Background Human umbilical cord-derived MSCs (hUC-MSCs) have been identified as promising seeding cells in tissue engineering and clinical applications of regenerative medicine due to their advantages of simple acquisition procedure and the capability to come from a young tissue donor over the other MSCs sources. In clinical applications, large scale production is required and optimal cryopreservation and culture conditions are essential to autologous and allogeneic transplantation in the future. However, the influence of cryopreserved post-thaw and long-term culture on hUC-MSCs remains unknown, especially in terms of specific protein expression. Therefore, biological characteristics and proteomic profiles of hUC-MSCs after cryopreserving and long-term culturing were investigated. Methods Firstly, hUC-MSCs were isolated from human umbilical cord tissues and identified through morphology, surface markers and tri-lineage differentiation potential at passage 3, and then the biological characteristics and proteomic profiles were detected and compared after cryopreserving and long-term culturing at passage 4 and continuously cultured to passage 10 with detection occurring here as well. The proteomic profiles were tested by using the isobaric tags for relative and absolute quantification (iTRAQ) labeling technique and differential protein were confirmed by mass spectrometry. Results The results showed no significant differences in phenotypes including morphology, surface marker and tri-lineage differentiation potential but have obvious changes in translation level, which is involved in metabolism, cell cycle and other pathways. Conclusion This suggests that protein expression may be used as an indicator of hUC-MSCs security testing before applying in clinical settings, and it is also expected to provide the foundation or standardization guide of hUC-MSCs applications in regenerative medicine.
Collapse
Affiliation(s)
- Xufeng Fu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004 China.,Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500 China
| | - Bo Xu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004 China
| | - Jiang Jiang
- Department of Obstetrics, The First People's Hospital of Yunnan Province, Kunming, 650032 China
| | - Xing Du
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004 China
| | - Xiaoli Yu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004 China
| | - Yaping Yan
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500 China
| | - Shanshan Li
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500 China
| | - Briauna Marie Inglis
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500 China
| | - Huiming Ma
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004 China
| | - Hongyan Wang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004 China
| | - Xiuying Pei
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004 China
| | - Wei Si
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500 China
| |
Collapse
|
26
|
Mello TG, Rosado-de-Castro PH, Campos RMP, Vasques JF, Rangel-Junior WS, Mattos RSDARD, Puig-Pijuan T, Foerster BU, Gutfilen B, Souza SAL, Boltze J, Paiva FF, Mendez-Otero R, Pimentel-Coelho PM. Intravenous Human Umbilical Cord-Derived Mesenchymal Stromal Cell Administration in Models of Moderate and Severe Intracerebral Hemorrhage. Stem Cells Dev 2020; 29:586-598. [PMID: 32160799 DOI: 10.1089/scd.2019.0176] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is as a life-threatening condition that can occur in young adults, often causing long-term disability. Recent preclinical data suggest mesenchymal stromal cell (MSC)-based therapies as promising options to minimize brain damage after ICH. However, therapeutic evidence and mechanistic insights are still limited, particularly when compared with other disorders such as ischemic stroke. Herein, we employed a model of collagenase-induced ICH in young adult rats to investigate the potential therapeutic effects of an intravenous injection of human umbilical cord Wharton's jelly-derived MSCs (hUC-MSCs). Two doses of collagenase were used to cause moderate or severe hemorrhages. Magnetic resonance imaging showed that animals treated with hUC-MSCs after moderate ICH had smaller residual hematoma volumes than vehicle-treated rats, whereas the cell therapy failed to decrease the hematoma volume in animals with a severe ICH. Functional assessments (rotarod and elevated body swing tests) were performed for up to 21 days after ICH. Enduring neurological impairments were seen only in animals subjected to severe ICH, but the cell therapy did not induce statistically significant improvements in the functional recovery. The biodistribution of Technetium-99m-labeled hUC-MSCs was also evaluated, showing that most cells were found in organs such as the spleen and lungs 24 h after transplantation. Nevertheless, it was possible to detect a weak signal in the brain, which was higher in the ipsilateral hemisphere of rats subjected to a severe ICH. These data indicate that hUC-MSCs have moderately beneficial effects in cases of less severe brain hemorrhages in rats by decreasing the residual hematoma volume, and that optimization of the therapy is still necessary.
Collapse
Affiliation(s)
- Tanira Giara Mello
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto de Engenharia Nuclear, Comissão Nacional de Energia Nuclear, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | - Paulo Henrique Rosado-de-Castro
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil.,Departamento de Radiologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Juliana Ferreira Vasques
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | | | | | - Teresa Puig-Pijuan
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | - Bernd Uwe Foerster
- Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, Brazil
| | - Bianca Gutfilen
- Departamento de Radiologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sergio Augusto Lopes Souza
- Departamento de Radiologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | | | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | - Pedro Moreno Pimentel-Coelho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| |
Collapse
|
27
|
Sadatpoor SO, Salehi Z, Rahban D, Salimi A. Manipulated Mesenchymal Stem Cells Applications in Neurodegenerative Diseases. Int J Stem Cells 2020; 13:24-45. [PMID: 32114741 PMCID: PMC7119211 DOI: 10.15283/ijsc19031] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/07/2019] [Accepted: 04/13/2019] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells that have multilinear differentiation and self-renewal abilities. These cells are immune-privileged as they express no or low level of class-II major histocompatibility complex (MHC-II) and other costimulatory molecules. Having neuroprotective and regenerative properties, MSCs can be used to ameliorate several intractable neurodegenerative disorders by affecting both innate and adaptive immune systems. Several manipulations like pretreating MSCs with different conditions or agents, and using molecules derived from MSCs or genetically manipulating them, are the common and practical ways that can be used to strengthen MSCs survival and potency. Improved MSCs can have significantly enhanced impacts on diseases compared to MSCs not manipulated. In this review, we describe some of the most important manipulations that have been exerted on MSCs to improve their therapeutic functions and their applications in ameliorating three prevalent neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and Huntington's disease.
Collapse
Affiliation(s)
- Seyyed omid Sadatpoor
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Immunology Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Dariush Rahban
- Department of Nanomedicine, School of Advanced Medical Technologies, Tehran University of Medical Science, Tehran, Iran
| | - Ali Salimi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Moll G, Hoogduijn MJ, Ankrum JA. Editorial: Safety, Efficacy and Mechanisms of Action of Mesenchymal Stem Cell Therapies. Front Immunol 2020; 11:243. [PMID: 32133010 PMCID: PMC7040069 DOI: 10.3389/fimmu.2020.00243] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/30/2020] [Indexed: 12/19/2022] Open
Affiliation(s)
- Guido Moll
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany
| | - Martin J Hoogduijn
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
29
|
Thompson M, Mei SH, Wolfe D, Champagne J, Fergusson D, Stewart DJ, Sullivan KJ, Doxtator E, Lalu M, English SW, Granton J, Hutton B, Marshall J, Maybee A, Walley KR, Santos CD, Winston B, McIntyre L. Cell therapy with intravascular administration of mesenchymal stromal cells continues to appear safe: An updated systematic review and meta-analysis. EClinicalMedicine 2020; 19:100249. [PMID: 31989101 PMCID: PMC6970160 DOI: 10.1016/j.eclinm.2019.100249] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/04/2019] [Accepted: 12/17/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Characterization of the mesenchymal stromal cell (MSC) safety profile is important as this novel therapy continues to be evaluated in clinical trials for various inflammatory conditions. Due to an increase in published randomized controlled trials (RCTs) from 2012-2019, we performed an updated systematic review to further characterize the MSC safety profile. METHODS MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials and Web of Science (to May 2018) were searched. RCTs that compared intravascular delivery of MSCs to controls in adult populations were included. Pre-specified adverse events were grouped according to: (1) immediate, (2) infection, (3) thrombotic/embolic, and (4) longer-term events (mortality, malignancy). Adverse events were pooled and meta-analyzed by fitting inverse-variance binary random effects models. Primary and secondary clinical efficacy endpoints were summarized descriptively. FINDINGS 7473 citations were reviewed and 55 studies met inclusion criteria (n = 2696 patients). MSCs as compared to controls were associated with an increased risk of fever (Relative Risk (RR) = 2·48, 95% Confidence Interval (CI) = 1·27-4·86; I2 = 0%), but not non-fever acute infusional toxicity, infection, thrombotic/embolic events, death, or malignancy (RR = 1·16, 0·99, 1·14, 0·78, 0·93; 95% CI = 0·70-1·91, 0·81-1·21, 0·67-1·95, 0·65-0·94, 0·60-1·45; I2 = 0%, 0%, 0%, 0%, 0%). No included trials were ended prematurely due to safety concerns. INTERPRETATIONS MSC therapy continues to exhibit a favourable safety profile. Future trials should continue to strengthen study rigor, reporting of MSC characterization, and adverse events. FUNDING Stem Cell Network, Ontario Institute for Regenerative Medicine and Ontario Research Fund.
Collapse
Affiliation(s)
- Mary Thompson
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Shirley H.J. Mei
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Dianna Wolfe
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Josée Champagne
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Dean Fergusson
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Duncan J. Stewart
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Katrina J. Sullivan
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Emily Doxtator
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Manoj Lalu
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Shane W. English
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - John Granton
- Department of Medicine (Critical Care), University of Toronto, Toronto, Ontario, Canada
| | - Brian Hutton
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - John Marshall
- Department of Surgery (Critical Care), University of Toronto, Toronto, Ontario, Canada
| | - Alies Maybee
- Patient Advisors Network, Toronto, Ontario, Canada
| | - Keith R. Walley
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Claudia Dos Santos
- Department of Surgery (Critical Care), University of Toronto, Toronto, Ontario, Canada
| | - Brent Winston
- Department of Critical Care, Medicine, and Biochemistry and Microbiology, University of Calgary, Calgary, Alberta, Canada
| | - Lauralyn McIntyre
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
30
|
Lee OJ, Luk F, Korevaar SS, Koch TG, Baan CC, Merino A, Hoogduijn MJ. The Importance of Dosing, Timing, and (in)Activation of Adipose Tissue-Derived Mesenchymal Stromal Cells on Their Immunomodulatory Effects. Stem Cells Dev 2020; 29:38-48. [DOI: 10.1089/scd.2019.0225] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Olivia J. Lee
- Section of Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Franka Luk
- Section of Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sander S. Korevaar
- Section of Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Thomas G. Koch
- Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Carla C. Baan
- Section of Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ana Merino
- Section of Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Martin J. Hoogduijn
- Section of Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
31
|
Guadix JA, López-Beas J, Clares B, Soriano-Ruiz JL, Zugaza JL, Gálvez-Martín P. Principal Criteria for Evaluating the Quality, Safety and Efficacy of hMSC-Based Products in Clinical Practice: Current Approaches and Challenges. Pharmaceutics 2019; 11:pharmaceutics11110552. [PMID: 31652984 PMCID: PMC6921040 DOI: 10.3390/pharmaceutics11110552] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 12/20/2022] Open
Abstract
Human Mesenchymal Stem Cells (hMSCs) play an important role as new therapeutic alternatives in advanced therapies and regenerative medicine thanks to their regenerative and immunomodulatory properties, and ability to migrate to the exact area of injury. These properties have made hMSCs one of the more promising cellular active substances at present, particularly in terms of the development of new and innovative hMSC-based products. Currently, numerous clinical trials are being conducted to evaluate the therapeutic activity of hMSC-based products on specific targets. Given the rapidly growing number of hMSC clinical trials in recent years and the complexity of these products due to their cellular component characteristics and medicinal product status, there is a greater need to define more stringent, specific, and harmonized requirements to characterize the quality of the hMSCs and enhance the analysis of their safety and efficacy in final products to be administered to patients. These requirements should be implemented throughout the manufacturing process to guarantee the function and integrity of hMSCs and to ensure that the hMSC-based final product consistently meets its specifications across batches. This paper describes the principal phases involved in the design of the manufacturing process and updates the specific technical requirements needed to address the appropriate clinical use of hMSC-based products. The challenges and limitations to evaluating the safety, efficacy, and quality of hMSCs have been also reviewed and discussed.
Collapse
Affiliation(s)
- Juan Antonio Guadix
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Instituto Malagueño de Biomedicina (IBIMA), Campus de Teatinos s/n, Málaga E-29071, Spain.
- BIONAND, Centro Andaluz de Nanomedicina y Biotecnología (Junta de Andalucía, Universidad de Málaga), c/ Severo Ochoa nº25, Campanillas, Málaga E-29590, Spain.
| | - Javier López-Beas
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Seville 41092, Spain.
| | - Beatriz Clares
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Granada E-18071, Spain.
| | - José Luis Soriano-Ruiz
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Granada E-18071, Spain.
| | - José Luis Zugaza
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, Leioa E-48940, Spain.
- Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, building 205, Zamudio E-48170, Spain.
- IKERBASQUE, Basque Foundation for Science, María Díaz de Haro 3, Bilbao E-48013, Spain.
| | - Patricia Gálvez-Martín
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Granada E-18071, Spain.
- R&D Human Health, Bioibérica S.A.U., Barcelona E-08029, Spain.
| |
Collapse
|
32
|
Czekaj P, Król M, Limanówka Ł, Michalik M, Lorek K, Gramignoli R. Assessment of animal experimental models of toxic liver injury in the context of their potential application as preclinical models for cell therapy. Eur J Pharmacol 2019; 861:172597. [PMID: 31408648 DOI: 10.1016/j.ejphar.2019.172597] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/04/2019] [Accepted: 08/08/2019] [Indexed: 02/06/2023]
Abstract
Preclinical animal models allow to study development and progression of several diseases, including liver disorders. These studies, for ethical reasons and medical limits, are impossible to carry out in human patients. At the same time, such experimental models constitute an important source of knowledge on pathomechanisms for drug- and virus-induced hepatotoxicity, both acute and chronic. Carbon tetrachloride, D-Galactosamine, and retrorsine are xenobiotics that can be used in immunocompetent animal models of hepatotoxicity, where chemical-intoxicated livers present histological features representative of human viruses-related infection. A prolonged derangement into liver architecture and functions commonly lead to cirrhosis, eventually resulting in hepatocellular carcinoma. In human, orthotopic liver transplantation commonly resolve most the problems related to cirrhosis. However, the shortage of donors does not allow all the patients in the waiting list to receive an organ on time. A promising alternative treatment for acute and chronic liver disease has been advised in liver cell transplantation, but the limited availability of hepatocytes for clinical approaches, in addition to the immunosuppressant regiment required to sustain cellular long-term engraftment have been encouraging the use of alternative cell sources. A recent effective source of stem cells have been recently identified in the human amnion membrane. Human amnion epithelial cells (hAEC) have been preclinically tested and proven sufficient to rescue immunocompetent rodents lethally intoxicated with drugs. The adoption of therapeutic procedures based on hAEC transplant in immunocompetent recipients affected by liver diseases, as well as patients with immune-related disorders, may constitute a successful new alternative therapy in regenerative medicine.
Collapse
Affiliation(s)
- Piotr Czekaj
- Department of Cytophysiology, Chair of Histology and Embryology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland, Medyków 18 str., 40-752, Katowice, Poland.
| | - Mateusz Król
- Students Scientific Society, Department of Cytophysiology, Chair of Histology and Embryology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland, Medyków 18 str., 40-752, Katowice, Poland.
| | - Łukasz Limanówka
- Students Scientific Society, Department of Cytophysiology, Chair of Histology and Embryology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland, Medyków 18 str., 40-752, Katowice, Poland
| | - Marcin Michalik
- Students Scientific Society, Department of Cytophysiology, Chair of Histology and Embryology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland, Medyków 18 str., 40-752, Katowice, Poland
| | - Katarzyna Lorek
- Students Scientific Society, Department of Cytophysiology, Chair of Histology and Embryology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland, Medyków 18 str., 40-752, Katowice, Poland
| | - Roberto Gramignoli
- Department of Laboratory Medicine (LABMED), H5, Division of Pathology, Karolinska Institutet, Alfred Nobels Allé 8, 14152, Huddinge, Sweden.
| |
Collapse
|
33
|
Therapeutic Effects of Intravenous Injection of Fresh and Frozen Thawed HO-1-Overexpressed Ad-MSCs in Dogs with Acute Spinal Cord Injury. Stem Cells Int 2019; 2019:8537541. [PMID: 31481975 PMCID: PMC6701425 DOI: 10.1155/2019/8537541] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/27/2019] [Indexed: 12/31/2022] Open
Abstract
Owing to the antioxidant and anti-inflammatory functions of hemeoxygenase-1 (HO-1), HO-1-expressing canine adipose-derived mesenchymal stem cells (Ad-MSCs) could be efficacious in treating spinal cord injury (SCI). Further, frozen thawed HO-1 Ad-MSCs could be instantly available as an emergency treatment for SCI. We compared the effects of intravenous treatment with freshly cultured HO-1 Ad-MSCs (HO-1 MSCs), only green fluorescent protein-expressing Ad-MSCs (GFP MSCs), and frozen thawed HO-1 Ad-MSCs (FT-HO-1 MSCs) in dogs with acute SCI. For four weeks, dogs were evaluated for improvement in hind limb locomotion using a canine Basso Beattie Bresnahan (cBBB) score. Upon completion of the study, injured spinal cord segments were harvested and used for western blot and histopathological analyses. All cell types had migrated to the injured spinal cord segment. The group that received HO-1 MSCs showed significant improvement in the cBBB score within four weeks. This group also showed significantly higher expression of NF-M and reduced astrogliosis. There was reduced expression of proinflammatory cytokines (IL6, TNF-α, and IL-1β) and increased expression of anti-inflammatory markers (IL-10, HO-1) in the HO-1 MSC group. Histopathological assessment revealed decreased fibrosis at the epicenter of the lesion and increased myelination in the HO-1 MSC group. Together, these data suggest that HO-1 MSCs could improve hind limb function by increasing the anti-inflammatory reaction, leading to neural sparing. Further, we found similar results between GFP MSCs and FT-HO-1 MSCs, which suggest that FT-HO-1 MSCs could be used as an emergency treatment for SCI.
Collapse
|
34
|
Oja S, Kaartinen T, Ahti M, Korhonen M, Laitinen A, Nystedt J. The Utilization of Freezing Steps in Mesenchymal Stromal Cell (MSC) Manufacturing: Potential Impact on Quality and Cell Functionality Attributes. Front Immunol 2019; 10:1627. [PMID: 31379832 PMCID: PMC6646664 DOI: 10.3389/fimmu.2019.01627] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/01/2019] [Indexed: 01/20/2023] Open
Abstract
Some recent reports suggest that cryopreserved and thawed mesenchymal stromal cells (MSCs) may have impaired functional properties as compared to freshly harvested MSCs from continuous cultures. A cryopreservation step in the manufacturing process brings important benefits, since it enables immediate off-the-shelf access to the products and a completion of all quality testing before batch release and administration to the patient. Cryopreservation is also inevitable in MSC banking strategies. In this study, we present the results from the MSC stability testing program of our in-house manufactured clinical-grade allogeneic bone marrow-derived MSC product that is expanded in platelet lysate and frozen in passage 2. The current manufacturing protocol contains only one freezing step and the frozen MSC product is thawed bed-side at the clinic. We can conclude superior viability and cell recovery of the frozen and thawed MSC product utilizing the validated freezing and thawing protocols we have developed. The MSC phenotype and differentiation potential was generally found to be unaltered after thawing, but the thawed cells exhibited a 50% reduced, but not completely abolished, performance in an in vitro immunosuppression assay. The in vitro immunosuppression assay results should, however, be interpreted with caution, since the chosen assay mainly measures one specific immunosuppressive mechanism of MSCs to suppress T-cell proliferation. Since at least two freezing steps are usually necessary in MSC banking strategies, we went on to investigate the impact of repeated freezing on MSC quality attributes. We can conclude that two freezing steps with a preceding cell culture phase of at least one passage before freezing is feasible and does not substantially affect basic cell manufacturing parameters or quality attributes of the final frozen and thawed product. Our results suggest, however, that an exhaustive number of freezing steps (≥4) may induce earlier senescence. In conclusion, our results support the utilization of frozen MSC products and MSC banking strategies, but emphasize the need of always performing detailed studies on also the cryopreserved MSC counterpart and to carefully report the cryopreservation and thawing protocols.
Collapse
Affiliation(s)
- Sofia Oja
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Tanja Kaartinen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Marja Ahti
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Matti Korhonen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Anita Laitinen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Johanna Nystedt
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| |
Collapse
|
35
|
Hoogduijn MJ, Lombardo E. Mesenchymal Stromal Cells Anno 2019: Dawn of the Therapeutic Era? Concise Review. Stem Cells Transl Med 2019; 8:1126-1134. [PMID: 31282113 PMCID: PMC6811696 DOI: 10.1002/sctm.19-0073] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/17/2019] [Indexed: 12/11/2022] Open
Abstract
2018 was the year of the first marketing authorization of an allogeneic stem cell therapy by the European Medicines Agency. The authorization concerns the use of allogeneic adipose tissue-derived mesenchymal stromal cells (MSCs) for treatment of complex perianal fistulas in Crohn's disease. This is a breakthrough in the field of MSC therapy. The last few years have, furthermore, seen some breakthroughs in the investigations into the mechanisms of action of MSC therapy. Although the therapeutic effects of MSCs have largely been attributed to their secretion of immunomodulatory and regenerative factors, it has now become clear that some of the effects are mediated through host phagocytic cells that clear administered MSCs and in the process adapt an immunoregulatory and regeneration supporting function. The increased interest in therapeutic use of MSCs and the ongoing elucidation of the mechanisms of action of MSCs are promising indicators that 2019 may be the dawn of the therapeutic era of MSCs and that there will be revived interest in research to more efficient, practical, and sustainable MSC-based therapies. Stem Cells Translational Medicine 2019;8:1126-1134.
Collapse
Affiliation(s)
- Martin J Hoogduijn
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
36
|
Sierra Parraga JM, Rozenberg K, Eijken M, Leuvenink HG, Hunter J, Merino A, Moers C, Møller BK, Ploeg RJ, Baan CC, Jespersen B, Hoogduijn MJ. Effects of Normothermic Machine Perfusion Conditions on Mesenchymal Stromal Cells. Front Immunol 2019; 10:765. [PMID: 31024574 PMCID: PMC6469476 DOI: 10.3389/fimmu.2019.00765] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/22/2019] [Indexed: 12/16/2022] Open
Abstract
Ex-situ normothermic machine perfusion (NMP) of transplant kidneys allows assessment of kidney quality and targeted intervention to initiate repair processes prior to transplantation. Mesenchymal stromal cells (MSC) have been shown to possess the capacity to stimulate kidney repair. Therefore, the combination of NMP and MSC therapy offers potential to repair transplant kidneys. It is however unknown how NMP conditions affect MSC. In this study the effect of NMP perfusion fluid on survival, metabolism and function of thawed cryopreserved human (h)MSC and porcine (p)MSC in suspension conditions was studied. Suspension conditions reduced the viability of pMSC by 40% in both perfusion fluid and culture medium. Viability of hMSC was reduced by suspension conditions by 15% in perfusion fluid, whilst no differences were found in survival in culture medium. Under adherent conditions, survival of the cells was not affected by perfusion fluid. The perfusion fluid did not affect survival of fresh MSC in suspension compared to the control culture medium. The freeze-thawing process impaired the survival of hMSC; 95% survival of fresh hMSC compared to 70% survival of thawed hMSC. Moreover, thawed MSC showed increased levels of reactive oxygen species, which indicates elevated levels of oxidative stress, and reduced mitochondrial activity, which implies reduced metabolism. The adherence of pMSC and hMSC to endothelial cells was reduced after the thawing process, effect which was particularly profound in in the perfusion fluid. To summarize, we observed that conditions required for machine perfusion are influencing the behavior of MSC. The freeze-thawing process reduces survival and metabolism and increases oxidative stress, and diminishes their ability to adhere to endothelial cells. In addition, we found that hMSC and pMSC behaved differently, which has to be taken into consideration when translating results from animal experiments to clinical studies.
Collapse
Affiliation(s)
- Jesus M Sierra Parraga
- Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Kaithlyn Rozenberg
- Nuffield Department of Surgical Sciences and Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Marco Eijken
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark.,Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Henri G Leuvenink
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, Groningen, Netherlands
| | - James Hunter
- Nuffield Department of Surgical Sciences and Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Ana Merino
- Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Cyril Moers
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, Groningen, Netherlands
| | - Bjarne K Møller
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Rutger J Ploeg
- Nuffield Department of Surgical Sciences and Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Carla C Baan
- Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Bente Jespersen
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Martin J Hoogduijn
- Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
37
|
Rojewski MT, Lotfi R, Gjerde C, Mustafa K, Veronesi E, Ahmed AB, Wiesneth M, Körper S, Sensebé L, Layrolle P, Hellem S, Schrezenmeier H. Translation of a standardized manufacturing protocol for mesenchymal stromal cells: A systematic comparison of validation and manufacturing data. Cytotherapy 2019; 21:468-482. [PMID: 30926359 DOI: 10.1016/j.jcyt.2019.03.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/22/2018] [Accepted: 03/02/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Many data are available on expansion protocols for mesenchymal stromal cells (MSCs) for both experimental settings and manufacturing for clinical trials. However, there is a lack of information on translation of established protocols for Good Manufacturing Practice (GMP) from validation to manufacturing for clinical application. We present the validation and translation of a standardized pre-clinical protocol for isolation and expansion of MSCs for a clinical trial for reconstitution of alveolar bone. METHODS Key parameters of 22 large-scale expansions of MSCs from bone marrow (BM) for validation were compared with 11 expansions manufactured for the clinical trial "Jaw bone reconstruction using a combination of autologous mesenchymal stromal cells and biomaterial prior to dental implant placement (MAXILLO1)" aimed at reconstruction of alveolar bone. RESULTS Despite variations of the starting material, the robust protocol led to stable performance characteristics of expanded MSCs. Manufacturing of the autologous advanced therapy medicinal product MAXILLO-1-MSC was possible, requiring 21 days for each product. Transport of BM aspirates and MSCs within 24 h was guaranteed. MSCs fulfilled quality criteria requested by the national competent authority. In one case, the delivered MSCs developed a mosaic in chromosomal finding, showing no abnormality in differentiation capacity, growth behavior or surface marker expression during long-term culture. The proportion of cells with the mosaic decreased in long-term culture and cells stopped growth after 38.4 population doublings. CONCLUSIONS Clinical use of freshly prepared MSCs, manufactured according to a standardized and validated protocol, is feasible for bone regeneration, even if there was a long local distance between manufacturing center and clinical site. Several parameters, such as colony forming units fibroblasts (CFU-F), percentage of CD34+ cells, cell count of mononuclear cells (MNCs) and white blood cells (WBCs), of the BM may serve as a predictive tool for the yield of MSCs and may help to avoid unnecessary costs for MSC manufacturing due to insufficient cell expansion rates.
Collapse
Affiliation(s)
- Markus Thomas Rojewski
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg - Hessia, Ulm, Germany.
| | - Ramin Lotfi
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany; Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg - Hessia, Ulm, Germany
| | - Cecilie Gjerde
- Institute of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Kamal Mustafa
- Institute of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Elena Veronesi
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Aymen B Ahmed
- Section for Haematology, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Markus Wiesneth
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg - Hessia, Ulm, Germany
| | - Sixten Körper
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg - Hessia, Ulm, Germany
| | - Luc Sensebé
- UMR5273 Centre national de la recherche scientifique (CNRS), UPS, Établissement francais du sang (EFS)-INSERM U1031, STROMAlab, Toulouse, France; Établissement francais du sang (EFS) Pyrénées-Méditeranée, Toulouse, France
| | - Pierre Layrolle
- Inserm U957, Laboratory for Pathophysiology of Bone Resorption, Faculty of Medicine, University of Nantes, Nantes, France
| | - Sølve Hellem
- Institute of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany; Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg - Hessia, Ulm, Germany
| |
Collapse
|
38
|
Moll G, Ankrum JA, Kamhieh-Milz J, Bieback K, Ringdén O, Volk HD, Geissler S, Reinke P. Intravascular Mesenchymal Stromal/Stem Cell Therapy Product Diversification: Time for New Clinical Guidelines. Trends Mol Med 2019; 25:149-163. [PMID: 30711482 DOI: 10.1016/j.molmed.2018.12.006] [Citation(s) in RCA: 267] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/04/2018] [Accepted: 12/17/2018] [Indexed: 12/13/2022]
Abstract
Intravascular infusion is the most popular route for therapeutic multipotent mesenchymal stromal/stem cell (MSC) delivery in hundreds of clinical trials. Meta-analysis has demonstrated that bone marrow MSC infusion is safe. It is not clear if this also applies to diverse new cell products derived from other sources, such as adipose and perinatal tissues. Different MSC products display varying levels of highly procoagulant tissue factor (TF) and may adversely trigger the instant blood-mediated inflammatory reaction (IBMIR). Suitable strategies for assessing and controlling hemocompatibility and optimized cell delivery are crucial for the development of safer and more effective MSC therapies.
Collapse
Affiliation(s)
- Guido Moll
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany.
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, USA
| | - Julian Kamhieh-Milz
- Department of Transfusion Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Olle Ringdén
- Translational Cell Therapy Research (TCR), Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Hans-Dieter Volk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Institute of Medical Immunology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Berlin Center for Advanced Therapies (BECAT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Equal contribution senior authorship
| | - Sven Geissler
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Julius Wolff Institute (JWI), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Equal contribution senior authorship
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Berlin Center for Advanced Therapies (BECAT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Equal contribution senior authorship
| |
Collapse
|
39
|
Salmenkari H, Laitinen A, Forsgård RA, Holappa M, Lindén J, Pasanen L, Korhonen M, Korpela R, Nystedt J. The use of unlicensed bone marrow-derived platelet lysate-expanded mesenchymal stromal cells in colitis: a pre-clinical study. Cytotherapy 2019; 21:175-188. [PMID: 30611671 DOI: 10.1016/j.jcyt.2018.11.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/04/2018] [Accepted: 11/13/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) are a promising candidate for treatment of inflammatory disorders, but their efficacy in human inflammatory bowel diseases (IBDs) has been inconsistent. Comparing the results from various pre-clinical and clinical IBD studies is also challenging due to a large variation in study designs. METHODS In this comparative pre-clinical study, we compared two administration routes and investigated the safety and feasibility of both fresh and cryopreserved platelet-lysate-expanded human bone marrow-derived MSCs without additional licensing in a dextran sodium sulfate (DSS) colitis mouse model both in the acute and regenerative phases of colitis. Body weight, macroscopic score for inflammation and colonic interleukin (IL)-1β and tumor necrosis factor (TNF)α concentrations were determined in both phases of colitis. Additionally, histopathology was assessed and Il-1β and Agtr1a messenger RNA (mRNA) levels and angiotensin-converting enzyme (ACE) protein levels were measured in the colon in the regenerative phase of colitis. RESULTS Intravenously administered MSCs exhibited modest anti-inflammatory capacity in the acute phase of colitis by reducing IL-1β protein levels in the inflamed colon. There were no clear improvements in mice treated with fresh or cryopreserved unlicensed MSCs according to weight monitoring results, histopathology and macroscopic score results. Pro-inflammatory ACE protein expression and shedding were reduced by cryopreserved MSCs in the colon. CONCLUSIONS In conclusion, we observed a good safety profile for bone marrow-derived platelet lysate-expanded MSCs in a mouse pre-clinical colitis model, but the therapeutic effect of MSCs prepared without additional licensing (i.e. such as MSCs are administered in graft-versus-host disease) was modest in the chosen in vivo model system and limited to biochemical improvements in cytokines without a clear benefit in histopathology or body weight development.
Collapse
Affiliation(s)
- Hanne Salmenkari
- Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anita Laitinen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Richard A Forsgård
- Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mervi Holappa
- Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jere Lindén
- Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Lauri Pasanen
- Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Matti Korhonen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Riitta Korpela
- Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Johanna Nystedt
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland.
| |
Collapse
|
40
|
Clinical Application of Stem/Stromal Cells in COPD. STEM CELL-BASED THERAPY FOR LUNG DISEASE 2019. [PMCID: PMC7121219 DOI: 10.1007/978-3-030-29403-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive life-threatening disease that is significantly increasing in prevalence and is predicted to become the third leading cause of death worldwide by 2030. At present, there are no true curative treatments that can stop the progression of the disease, and new therapeutic strategies are desperately needed. Advances in cell-based therapies provide a platform for the development of new therapeutic approaches in severe lung diseases such as COPD. At present, a lot of focus is on mesenchymal stem (stromal) cell (MSC)-based therapies, mainly due to their immunomodulatory properties. Despite increasing number of preclinical studies demonstrating that systemic MSC administration can prevent or treat experimental COPD and emphysema, clinical studies have not been able to reproduce the preclinical results and to date no efficacy or significantly improved lung function or quality of life has been observed in COPD patients. Importantly, the completed appropriately conducted clinical trials uniformly demonstrate that MSC treatment in COPD patients is well tolerated and no toxicities have been observed. All clinical trials performed so far, have been phase I/II studies, underpowered for the detection of potential efficacy. There are several challenges ahead for this field such as standardized isolation and culture procedures to obtain a cell product with high quality and reproducibility, administration strategies, improvement of methods to measure outcomes, and development of potency assays. Moreover, COPD is a complex pathology with a diverse spectrum of clinical phenotypes, and therefore it is essential to develop methods to select the subpopulation of patients that is most likely to potentially respond to MSC administration. In this chapter, we will discuss the current state of the art of MSC-based cell therapy for COPD and the hurdles that need to be overcome.
Collapse
|
41
|
Chinnadurai R, Rajan D, Qayed M, Arafat D, Garcia M, Liu Y, Kugathasan S, Anderson LJ, Gibson G, Galipeau J. Potency Analysis of Mesenchymal Stromal Cells Using a Combinatorial Assay Matrix Approach. Cell Rep 2018; 22:2504-2517. [PMID: 29490284 PMCID: PMC5855117 DOI: 10.1016/j.celrep.2018.02.013] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 09/11/2017] [Accepted: 02/02/2018] [Indexed: 02/07/2023] Open
Abstract
Assays that can characterize MSC immune potency need to be identified for use in advanced clinical trials. MSCs possess a number of putative regenerative and immunomodulatory properties, and an assay matrix approach may best capture involved effector pathways. We have tested two assay systems to measure the potency of MSCs derived from human subjects: MSC secretome analysis and a quantitative RNA-based array for genes specific to immunomodulatory and homing properties of MSCs. Secretome analysis identified a unique cytokine signature that is upregulated by MSCs or downregulated in responder PBMCs and correlated with T cell suppression. Use of interferon-γ as a surrogate for the action of activated PBMCs on MSCs served as an alternative for the use of human PBMCs as responder cells in a potency assay. Our approach and results define and simplify the multifunctional or matrix responses of MSCs and may serve as a platform for robust potency analysis.
Collapse
Affiliation(s)
- Raghavan Chinnadurai
- Department of Medicine, University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin - Madison, Madison, WI 53705, USA
| | - Devi Rajan
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| | - Muna Qayed
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| | - Dalia Arafat
- School of Biology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | | - Yifei Liu
- Department of Statistics, University of Wisconsin - Madison, Madison, WI 53706, USA
| | - Subra Kugathasan
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| | - Larry J Anderson
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| | - Greg Gibson
- School of Biology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Jacques Galipeau
- Department of Medicine, University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin - Madison, Madison, WI 53705, USA.
| |
Collapse
|
42
|
Packthongsuk K, Rathbun T, Troyer D, Davis DL. Porcine Wharton's jelly cells distribute throughout the body after intraperitoneal injection. Stem Cell Res Ther 2018; 9:38. [PMID: 29444715 PMCID: PMC5813394 DOI: 10.1186/s13287-018-0775-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 01/08/2018] [Accepted: 01/15/2018] [Indexed: 12/12/2022] Open
Abstract
Background Wharton's jelly cells (WJCs) have multiple differentiation potentials and are easily harvested in large numbers. WJCs are well tolerated in allogeneic environments and there is a growing list of their therapeutic effects. Most therapies require administering large numbers of cells and this is generally accomplished by intravenous injection. Here, we studied the locations of porcine WJCs in immune-competent, allogeneic hosts after intraperitoneal (IP) injection. Methods Male porcine WJCs were administered to female neonatal piglets by IP injection. The location of transplanted cells was examined at 6 h, 24 h, and 7 days after administration using confocal microscopy and polymerase chain reaction (PCR). Transplanted cells were also retrieved from the intestines of recipients and were cultured. Previously transplanted cells were identified by fluorescence in-situ hybridization (FISH) using a Y-chromosome probe. Results Allogeneic cells were identified in the small and large intestine, stomach, liver, spleen, diaphragm, omentum, kidney, pancreas, mesenteric lymph nodes, heart, lungs, uterus, bladder, and skeletal muscle. Male cells (SRY positive) were found in cultures of cells harvested from the intestinal mucosa 1 week after administration of male porcine WJCs. Conclusions Our results show that porcine WJCs distribute widely to the organs in immunocompetent allogeneic hosts after IP administration. They may distribute through the lymphatics initially, and a prominent site of incorporation is the mucosa of the gastrointestinal tract. In that location they could function in the niche of endogenous stem cells and provide secretory products to cells in the tissue damaged by intestinal disease. Electronic supplementary material The online version of this article (10.1186/s13287-018-0775-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kreeson Packthongsuk
- National Institute of Animal Health (NIAH) 50/2 Kasetklang, Pahonyothin Rd., Jatujak, Ladyao, Bangkok, 10900, Thailand
| | - Theresa Rathbun
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, 66506, KS, USA
| | - Deryl Troyer
- Department of Anatomy and Physiology, Kansas State University, Manhattan, 66506, KS, USA
| | - Duane L Davis
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, 66506, KS, USA.
| |
Collapse
|
43
|
Zanata F, Shaik S, Devireddy RV, Wu X, Ferreira LM, Gimble JM. Cryopreserved Adipose Tissue-Derived Stromal/Stem Cells: Potential for Applications in Clinic and Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 951:137-146. [PMID: 27837560 DOI: 10.1007/978-3-319-45457-3_11] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adipose-Derived Stromal/Stem Cells (ASC) have considerable potential for regenerative medicine due to their abilities to proliferate, differentiate into multiple cell lineages, high cell yield, relative ease of acquisition, and almost no ethical concerns since they are derived from adult tissue. Storage of ASC by cryopreservation has been well described that maintains high cell yield and viability, stable immunophenotype, and robust differentiation potential post-thaw. This ability is crucial for banking research and for clinical therapeutic purposes that avoid the morbidity related to repetitive liposuction tissue harvests. ASC secrete various biomolecules such as cytokines which are reported to have immunomodulatory properties and therapeutic potential to reverse symptoms of multiple degenerative diseases/disorders. Nevertheless, safety regarding the use of these cells clinically is still under investigation. This chapter focuses on the different aspects of cryopreserved ASC and the methods to evaluate their functionality for future clinical use.
Collapse
Affiliation(s)
- Fabiana Zanata
- Federal University of Sao Paulo, Sao Paulo, SP, Brazil
- Center for Stem Cell Research & Regenerative Medicine, Tulane University, New Orleans, LA, USA
| | - Shahensha Shaik
- Department of Mechanical Engineering, Louisiana State University, Baton Rouge, LA, USA
| | - Ram V Devireddy
- Department of Mechanical Engineering, Louisiana State University, Baton Rouge, LA, USA
| | - Xiying Wu
- La Cell LLC, New Orleans BioInnovation Center, Suite 304, 1441 Canal Street, New Orleans, LA, 70112, USA
| | | | - Jeffrey M Gimble
- Center for Stem Cell Research & Regenerative Medicine, Tulane University, New Orleans, LA, USA.
- La Cell LLC, New Orleans BioInnovation Center, Suite 304, 1441 Canal Street, New Orleans, LA, 70112, USA.
| |
Collapse
|
44
|
Cryopreserved or Fresh Mesenchymal Stromal Cells: Only a Matter of Taste or Key to Unleash the Full Clinical Potential of MSC Therapy? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 951:77-98. [PMID: 27837556 DOI: 10.1007/978-3-319-45457-3_7] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) harbor great therapeutic potential for numerous diseases. From early clinical trials, success and failure analysis, bench-to-bedside and back-to-bench approaches, there has been a great gain in knowledge, still leaving a number of questions to be answered regarding optimal manufacturing and quality of MSCs for clinical application. For treatment of many acute indications, cryobanking may remain a prerequisite, but great uncertainty exists considering the therapeutic value of freshly thawed (thawed) and continuously cultured (fresh) MSCs. The field has seen an explosion of new literature lately, outlining the relevance of the topic. MSCs appear to have compromised immunomodulatory activity directly after thawing for clinical application. This may provide a possible explanation for failure of early clinical trials. It is not clear if and how quickly MSCs recover their full therapeutic activity, and if the "cryo stun effect" is relevant for clinical success. Here, we will share our latest insights into the relevance of these observations for clinical practice that will be discussed in the context of the published literature. We argue that the differences of fresh and thawed MSCs are limited but significant. A key issue in evaluating potency differences is the time point of analysis after thawing. To date, prospective double-blinded randomized clinical studies to evaluate potency of both products are lacking, although recent progress was made with preclinical assessment. We suggest refocusing therapeutic MSC development on potency and safety assays with close resemblance of the clinical reality.
Collapse
|
45
|
Baygan A, Aronsson-Kurttila W, Moretti G, Tibert B, Dahllöf G, Klingspor L, Gustafsson B, Khoein B, Moll G, Hausmann C, Svahn BM, Westgren M, Remberger M, Sadeghi B, Ringden O. Safety and Side Effects of Using Placenta-Derived Decidual Stromal Cells for Graft-versus-Host Disease and Hemorrhagic Cystitis. Front Immunol 2017; 8:795. [PMID: 28744284 PMCID: PMC5504152 DOI: 10.3389/fimmu.2017.00795] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/22/2017] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are increasingly used in regenerate medicine. Placenta-derived decidual stromal cells (DSCs) are a novel therapy for acute graft-versus-host-disease (GVHD) and hemorrhagic cystitis (HC) after allogeneic hematopoietic stem cell transplantation (HSCT). DSCs are more immunosuppressive than MSCs. We assessed adverse events and safety using DSCs among 44 treated patients and 40 controls. The median dose of infused cells was 1.5 (range 0.9–2.9) × 106 DSCs/kg. The patients were given 2 (1–5) doses, with a total of 82 infusions. Monitoring ended 3 months after the last DSC infusion. Three patients had transient reactions during DSC infusion. Laboratory values, hemorrhages, and transfusions were similar in the two groups. The frequency of leukemic relapse (2/2, DSC/controls) and invasive fungal infections (6/6) were the same in the two groups. Causes of death were those seen in HSCT patients: infections (5/3), respiratory failure (1/1), circulatory failure (3/1), thromboembolism (1/0), multiorgan failure (0/1), and GVHD and others (2/7). One-year survival for the DSC patients with GVHD was 67%, which was significantly better than achieved previously at our center. One-year survival was 90% in the DSC-treated HC group. DSC infusions appear safe. Randomized studies are required to prove efficacy.
Collapse
Affiliation(s)
- Arjang Baygan
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Wictor Aronsson-Kurttila
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Gianluca Moretti
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Babylonia Tibert
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Göran Dahllöf
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lena Klingspor
- Department of Microbiology, Uppsala University Hospital, Uppsala, Sweden
| | - Britt Gustafsson
- Department of Pediatrics, Uppsala University Hospital, Uppsala, Sweden
| | - Bita Khoein
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Guido Moll
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden.,Charité Universitätsmedizin, Berlin, Germany
| | - Charlotta Hausmann
- Center for Allogeneic Stem Cell Transplantation, Department of Pathology/Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Britt-Marie Svahn
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Westgren
- Department of Obstetrics and Gynecology, Karolinska University Hospital, Stockholm, Sweden
| | - Mats Remberger
- Center for Allogeneic Stem Cell Transplantation, Department of Pathology/Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Behnam Sadeghi
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Olle Ringden
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
46
|
Chabot D, Tremblay T, Paré I, Bazin R, Loubaki L. Transient warming events occurring after freezing impairs umbilical cord-derived mesenchymal stromal cells functionality. Cytotherapy 2017; 19:978-989. [PMID: 28606762 DOI: 10.1016/j.jcyt.2017.04.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/28/2017] [Accepted: 04/27/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) have shown promising results for the treatment of refractory acute graft-versus-host disease. While safety of MSC infusion has been demonstrated, the use of cryopreserved MSCs in clinical trials has raised concerns regarding the retention of their functional activity. This has led to the recommendation by experts in the field to use freshly harvested MSCs, even though this approach is much less practical from a logistic point of view. In the present study, we revisited the impact of cryopreservation on MSC functionality and addressed the possibility that warming events on frozen cells rather than cryopreservation per se could impact MSC functionality. METHODS Following controlled-rate freezing to -130°C, umbilical cord-derived MSCs were left at room temperature (RT) for 2-10 min or on dry ice for 10 min, before being transferred into liquid nitrogen (LqN2). MSCs of each group were subsequently tested (viability, functionality and cellular damage) and compared with their freshly harvested counterparts. RESULTS We demonstrated that freshly harvested MSCs as well as cryopreserved MSCs that were left on dry ice following step-down freezing have comparable viability, functionality and integrity. In contrast, cryopreserved MSCs that were left at RT before being transferred into LqN2 were functionally impaired and showed cellular damage upon thawing even though they exhibited high viability. DISCUSSION Warming events after freezing and not cryopreservation per se significantly impair MSC functionality, indicating that cryopreserved MSCs can be an advantageous alternative to freshly harvested cells for therapeutic purposes.
Collapse
Affiliation(s)
- Dominique Chabot
- Department of Research and Development, Héma-Québec, Québec City, QC, Canada; Department of Biochemistry, Microbiology and Bioinformatics, Laval University, Québec City, QC, Canada
| | - Tony Tremblay
- Department of Research and Development, Héma-Québec, Québec City, QC, Canada
| | - Isabelle Paré
- Department of Research and Development, Héma-Québec, Québec City, QC, Canada
| | - Renée Bazin
- Department of Research and Development, Héma-Québec, Québec City, QC, Canada; Department of Biochemistry, Microbiology and Bioinformatics, Laval University, Québec City, QC, Canada
| | - Lionel Loubaki
- Department of Research and Development, Héma-Québec, Québec City, QC, Canada; Department of Biochemistry, Microbiology and Bioinformatics, Laval University, Québec City, QC, Canada.
| |
Collapse
|
47
|
Cytokine treatment optimises the immunotherapeutic effects of umbilical cord-derived MSC for treatment of inflammatory liver disease. Stem Cell Res Ther 2017; 8:140. [PMID: 28595619 PMCID: PMC5465593 DOI: 10.1186/s13287-017-0590-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/12/2017] [Accepted: 05/17/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSC) possess immunomodulatory properties and low immunogenicity, both crucial properties for their development into an effective cellular immunotherapy. They have shown benefit in clinical trials targeting liver diseases; however the efficacy of MSC therapy will benefit from improvement of the immunomodulatory and immunogenic properties of MSC. METHODS MSC derived from human umbilical cords (ucMSC) were treated for 3 days in vitro with various inflammatory factors, interleukins, vitamins and serum deprivation. Their immunogenicity and immunomodulatory capacity were examined by gene-expression analysis, surface-marker expressions, IDO activity, PGE2 secretion and inhibition of T cell proliferation and IFNγ production. Furthermore, their activation of NK cell cytotoxicity was investigated via CD107a expression on NK cells. The immunomodulatory capacity, biodistribution and survival of pre-treated ucMSC were investigated in a CCl4-induced liver disease mouse model. In addition, capacity of pre-treated MSC to ameliorate liver inflammation was examined in an ex vivo liver inflammation co-culture model. RESULTS IFN-γ and a multiple cytokine cocktail (MC) consisting of IFN-γ, TGFβ and retinoic acid upregulated the expression of immunomodulatory factor PD-L1 and IDO activity. Subsequently, both treatments enhanced the capacity of ucMSC to inhibit CD4 and CD8 T cell proliferation and IFN-γ production. The susceptibility of ucMSC for NK cell lysis was decreased by IFN-β, TGFβ and MC treatment. In vivo, no immunomodulation was observed by the ucMSC. Four hours after intravenous infusion in mice with CCl4-induced inflammatory liver injury, the majority of ucMSC were trapped in the lungs. Rapid clearance of ucMSC(VitB6), ucMSC(Starv + VitB6) and ucMSC(MC) and altered bio-distribution of ucMSC(TGFβ) compared to untreated ucMSC was observed. In the ex vivo co-culture system with inflammatory liver slices ucMSC(MC) showed significantly enhanced modulatory capacity compared to untreated ucMSC. CONCLUSIONS The present study demonstrates the responsiveness of ucMSC to in vitro optimisation treatment. The observed improvements in immunomodulatory capacity as well as immunogenicity after MC treatment may improve the efficacy of ucMSC as immunotherapy targeted towards liver inflammation.
Collapse
|
48
|
Shaik S, Hayes D, Gimble J, Devireddy R. Inducing Heat Shock Proteins Enhances the Stemness of Frozen-Thawed Adipose Tissue-Derived Stem Cells. Stem Cells Dev 2017; 26:608-616. [PMID: 28052723 PMCID: PMC5393415 DOI: 10.1089/scd.2016.0289] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/04/2017] [Indexed: 01/01/2023] Open
Abstract
Extensive research has been performed to determine the effect of freezing protocol and cryopreservation agents on the viability of adipose tissue-derived stromal/stem cells (ASCs) as well as other cells. Unfortunately, the conclusion one may draw after decades of research utilizing fundamentally similar cryopreservation techniques is that a barrier exists, which precludes full recovery. We hypothesize that agents capable of inducing a subset of heat shock proteins (HSPs) and chaperones will reduce the intrinsic barriers to the post-thaw recovery of ASCs. ASCs were exposed to 43°C for 1 h to upregulate HSPs, and the temporal HSP expression profile postheat shock was determined by performing quantitative polymerase chain reaction (PCR) and western blotting assays. The expression levels of HSP70 and HSP32 were found to be maximum at 3 h after the heat shock, whereas HSP90 and HSP27 remain unchanged. The heat shocked ASCs cryopreserved during maximal HSPs expression exhibited increased post-thaw viability than the nonheat shocked samples. Histochemical staining and quantitative reverse transcription-PCR indicated that the ASC differentiation potential was retained. Thus, suggesting that the upregulation of HSPs before a freezing insult is beneficial to ASCs and a potential alternative to the use of harmful cryoprotective agents.
Collapse
Affiliation(s)
- Shahensha Shaik
- Bioengineering Laboratory, Department of Mechanical Engineering, Louisiana State University, Baton Rouge, Louisiana
| | - Daniel Hayes
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania
| | - Jeffrey Gimble
- La Cell LLC and Center for Stem Cell Research and Regenerative Medicine and Departments of Medicine, Structural and Cellular Biology, and Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ram Devireddy
- Bioengineering Laboratory, Department of Mechanical Engineering, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
49
|
Pollock K, Samsonraj RM, Dudakovic A, Thaler R, Stumbras A, McKenna DH, Dosa PI, van Wijnen AJ, Hubel A. Improved Post-Thaw Function and Epigenetic Changes in Mesenchymal Stromal Cells Cryopreserved Using Multicomponent Osmolyte Solutions. Stem Cells Dev 2017; 26:828-842. [PMID: 28178884 DOI: 10.1089/scd.2016.0347] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Current methods for freezing mesenchymal stromal cells (MSCs) result in poor post-thaw function, which limits the clinical utility of these cells. This investigation develops a novel approach to preserve MSCs using combinations of sugars, sugar alcohols, and small-molecule additives. MSCs frozen using these solutions exhibit improved post-thaw attachment and a more normal alignment of the actin cytoskeleton compared to cells exposed to dimethylsulfoxide (DMSO). Osteogenic and chondrogenic differentiation assays show that cells retain their mesenchymal lineage properties. Genomic analysis indicates that the different freezing media evaluated have different effects on the levels of DNA hydroxymethylation, which are a principal epigenetic mark and a key step in the demethylation of CpG doublets. RNA sequencing and quantitative real time-polymerase chain reaction validation demonstrate that transcripts for distinct classes of cytoprotective genes, as well as genes related to extracellular matrix structure and growth factor/receptor signaling are upregulated in experimental freezing solutions compared to DMSO. For example, the osmotic regulator galanin, the antiapoptotic marker B cell lymphoma 2, as well as the cell surface adhesion molecules CD106 (vascular cell adhesion molecule 1) and CD54 (intracellular adhesion molecule 1) are all elevated in DMSO-free solutions. These studies validate the concept that DMSO-free solutions improve post-thaw biological functions and are viable alternatives for freezing MSCs. These novel solutions promote expression of cytoprotective genes, modulate the CpG epigenome, and retain the differentiation ability of MSCs, suggesting that osmolyte-based freezing solutions may provide a new paradigm for therapeutic cell preservation.
Collapse
Affiliation(s)
- Kathryn Pollock
- 1 Department of Biomedical Engineering, University of Minnesota , Minneapolis, Minnesota
| | | | - Amel Dudakovic
- 2 Department of Orthopedic Surgery, Mayo Clinic , Rochester, Minnesota
| | - Roman Thaler
- 2 Department of Orthopedic Surgery, Mayo Clinic , Rochester, Minnesota
| | - Aron Stumbras
- 3 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| | - David H McKenna
- 4 Department of Laboratory Medicine and Pathology, University of Minnesota , Minneapolis, Minnesota
| | - Peter I Dosa
- 5 Institute for Therapeutics Discovery and Development, University of Minnesota , Minneapolis, Minnesota
| | | | - Allison Hubel
- 6 Department of Mechanical Engineering, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
50
|
Immunomodulatory Role of Mesenchymal Stem Cell Therapy in Vascularized Composite Allotransplantation. J Transplant 2016; 2016:6951693. [PMID: 27822384 PMCID: PMC5086388 DOI: 10.1155/2016/6951693] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 09/15/2016] [Indexed: 12/12/2022] Open
Abstract
This review aims to summarize contemporary evidence of the in vitro and in vivo immunomodulatory effects of mesenchymal stem cells (MSCs) in promoting vascularized composite allotransplant (VCA) tolerance. An extensive literature review was performed to identify pertinent articles of merit. Prospective preclinical trials in mammal subjects receiving VCA (or skin allograft) with administration of MSCs were reviewed. Prospective clinical trials with intravascular delivery of MSCs in human populations undergoing solid organ transplant were also identified and reviewed. Sixteen preclinical studies are included. Eleven studies compared MSC monotherapy to no therapy; of these, ten reported improved graft survival, which was statistically significantly prolonged in eight. Eight studies analyzed allograft survival with MSC therapy as an adjunct to proven immunosuppressive regimens. In these studies, daily immunosuppression was transiently delivered and then stopped. In all studies, treatment-free graft survival was statistically significantly prolonged in animals that received MSC therapy. MSCs have been safely administered clinically and their use in renal transplant clinical trials provides evidence that they improve allograft transplant tolerance in clinical practice. There is potential for MSC induction therapy to overcome many of the obstacles to widespread VCA in clinical practice. Preclinical studies are needed before MSC-induced VCA tolerance becomes a clinical reality.
Collapse
|