51
|
Shammaa R, El-Kadiry AEH, Abusarah J, Rafei M. Mesenchymal Stem Cells Beyond Regenerative Medicine. Front Cell Dev Biol 2020; 8:72. [PMID: 32133358 PMCID: PMC7040370 DOI: 10.3389/fcell.2020.00072] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are competent suitors of cellular therapy due to their therapeutic impact on tissue degeneration and immune-based pathologies. Additionally, their homing and immunomodulatory properties can be exploited in cancer malignancies to transport pharmacological entities, produce anti-neoplastic agents, or induce anti-tumor immunity. Herein, we create a portfolio for MSC properties, showcasing their distinct multiple therapeutic utilities and successes/challenges thereof in both animal studies and clinical trials. We further highlight the promising potential of MSCs not only in cancer management but also in instigating tumor-specific immunity - i.e., cancer vaccination. Finally, we reflect on the possible reasons impeding the clinical advancement of MSC-based cancer vaccines to assist in contriving novel methodologies from which a therapeutic milestone might emanate.
Collapse
Affiliation(s)
- Riam Shammaa
- Canadian Centre for Regenerative Therapy, Toronto, ON, Canada.,IntelliStem Technologies Inc., Toronto, ON, Canada.,Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada
| | - Abed El-Hakim El-Kadiry
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Montreal, QC, Canada.,Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| | - Jamilah Abusarah
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Moutih Rafei
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.,Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada.,Molecular Biology Program, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
52
|
Fajka-Boja R, Szebeni GJ, Hunyadi-Gulyás É, Puskás LG, Katona RL. Polyploid Adipose Stem Cells Shift the Balance of IGF1/IGFBP2 to Promote the Growth of Breast Cancer. Front Oncol 2020; 10:157. [PMID: 32133294 PMCID: PMC7040181 DOI: 10.3389/fonc.2020.00157] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/29/2020] [Indexed: 12/17/2022] Open
Abstract
Background: The close proximity of adipose tissue and mammary epithelium predispose involvement of adipose cells in breast cancer development. Adipose-tissue stem cells (ASCs) contribute to tumor stroma and promote growth of cancer cells. In our previous study, we have shown that murine ASCs, which undergo polyploidization during their prolonged in vitro culturing, enhanced the proliferation of 4T1 murine breast cancer cells in IGF1 dependent manner. Aims: In the present study, our aim was to clarify the regulation of ASC-derived IGF1. Methods: 4T1 murine breast carcinoma cells were co-transplanted with visceral fat-derived ASCs (vASC) or with the polyploid ASC.B6 cell line into female BALB/c mice and tumor growth and lung metastasis were monitored. The conditioned media of vASCs and ASC.B6 cells were subjected to LC-MS/MS analysis and the production of IGFBP2 was verified by Western blotting. The regulatory effect was examined by adding recombinant IGFBP2 to the co-culture of ASC.B6 and 4T1. Akt/protein kinase B (PKB) activation was detected by Western blotting. Results: Polyploid ASCs promoted the tumor growth and metastasis more potently than vASCs with normal karyotype. vASCs produced the IGF1 regulator IGFBP2, which inhibited proliferation of 4T1 cells. Downregulation of IGFBP2 by polyploidization of ASCs and enhanced secretion of IGF1 allowed survival signaling in 4T1 cells, leading to Akt phosphorylation. Conclusions: Our results implicate that ASCs in the tumor microenvironment actively regulate the growth of breast cancer cells through the IGF/IGFBP system.
Collapse
Affiliation(s)
- Roberta Fajka-Boja
- Artificial Chromosome and Stem Cell Research Laboratory, Biological Research Centre, Institute of Genetics, Szeged, Hungary
| | - Gábor J Szebeni
- Laboratory of Functional Genomics, Biological Research Centre, Institute of Genetics, Szeged, Hungary.,Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Éva Hunyadi-Gulyás
- Laboratory of Proteomics Research, Biological Research Centre, Institute of Biochemistry, Szeged, Hungary
| | - László G Puskás
- Laboratory of Functional Genomics, Biological Research Centre, Institute of Genetics, Szeged, Hungary.,Avidin Ltd., Szeged, Hungary
| | - Róbert L Katona
- Artificial Chromosome and Stem Cell Research Laboratory, Biological Research Centre, Institute of Genetics, Szeged, Hungary
| |
Collapse
|
53
|
Stem cells out of the bag: characterization of ex vivo expanded mesenchymal stromal cells for possible clinical use. Future Sci OA 2020; 6:FSO449. [PMID: 32140248 PMCID: PMC7050601 DOI: 10.2144/fsoa-2019-0129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aim: Mesenchymal stromal cells (MSC) are a promising tool for cellular therapy and regenerative medicine. One major difficulty in establishing a MSC expansion protocol is the large volume of bone marrow (BM) required. We studied whether cells trapped within a collection bag and filter system could be considered as a source of MSC. Results: From the 20 BM collection bag and filter systems, we recovered an average of 1.68 × 108 mononuclear cells, which is the equivalent to 60 ml of filtered BM. Mononuclear cells were expanded ex vivo to 17 × 106 MSC, with purity shown by a CD44+, CD105+, CD90+ and CD73+ immunophenotype, a reduction of 20% proliferating cells in a mixed lymphocyte reaction and also the ability of adipocyte differentiation. Conclusion: Long-term MSC cultures were established from the usually discarded BM collection bag and filter, maintaining an appropriate phenotype and function, being suitable for both investigation and clinical settings. Mesenchymal stromal cells (MSC) are a promising tool for cellular therapy and regenerative medicine. One major difficulty in obtaining MSC is the large volume of bone marrow (BM) required from a healthy donor. From usually discarded collection bags of BM collected for transplant, we recovered a number of cells equivalent to 60 ml of BM and expanded functional MSC with high purity. We believe that those recovered cells are an alternative to BM for obtaining MSC. The routinely recovery of such cells in reference centers, in a way similar to a public cord-blood bank, could benefit the scientific community, once further research is conducted to confirm results.
Collapse
|
54
|
Averyanov A, Koroleva I, Konoplyannikov M, Revkova V, Lesnyak V, Kalsin V, Danilevskaya O, Nikitin A, Sotnikova A, Kotova S, Baklaushev V. First-in-human high-cumulative-dose stem cell therapy in idiopathic pulmonary fibrosis with rapid lung function decline. Stem Cells Transl Med 2020; 9:6-16. [PMID: 31613055 PMCID: PMC6954714 DOI: 10.1002/sctm.19-0037] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 08/18/2019] [Indexed: 12/17/2022] Open
Abstract
Previous phase I studies demonstrated safety and some beneficial effects of mesenchymal stem cells (MSCs) in patients with mild to moderate idiopathic pulmonary fibrosis (IPF). The aim of our study was to evaluate the safety, tolerability, and efficacy of a high cumulative dose of bone marrow MSCs in patients with rapid progressive course of severe to moderate IPF. Twenty patients with forced ventilation capacity (FVC) ≥40% and diffusing capacity of the lung for carbon monoxide (DLCO) ≥20% with a decline of both >10% over the previous 12 months were randomized into two groups: one group received two intravenous doses of allogeneic MSCs (2 × 108 cells) every 3 months, and the second group received a placebo. A total amount of 1.6 × 109 MSCs had been administered to each patient after the study completion. There were no significant adverse effects after administration of MSCs in any patients. In the group of MSC therapy, we observed significantly better improvement for the 6-minute walk distance in 13 weeks, for DLCO in 26 weeks, and for FVC in 39 weeks compared with placebo. FVC for 12 months in the MSCs therapy group increased by 7.8% from baseline, whereas it declined by 5.9% in the placebo group. We did not find differences between the groups in mortality (two patients died in each group) or any changes in the high-resolution computed tomography fibrosis score. In patients with IPF and a rapid pulmonary function decline, therapy with high doses of allogeneic MSCs is a safe and promising method to reduce disease progression.
Collapse
Affiliation(s)
- Alexander Averyanov
- Federal Research and Clinical Center of Federal Medical‐Biologic AgencyMoscowRussia
- Pulmonology Scientific Research Institute under Federal Medical‐Biologic AgencyMoscowRussia
| | - Irina Koroleva
- Federal Research and Clinical Center of Federal Medical‐Biologic AgencyMoscowRussia
| | | | - Veronika Revkova
- Federal Research and Clinical Center of Federal Medical‐Biologic AgencyMoscowRussia
| | - Victor Lesnyak
- Federal Research and Clinical Center of Federal Medical‐Biologic AgencyMoscowRussia
| | - Vladimir Kalsin
- Federal Research and Clinical Center of Federal Medical‐Biologic AgencyMoscowRussia
| | - Olesya Danilevskaya
- Federal Research and Clinical Center of Federal Medical‐Biologic AgencyMoscowRussia
- Pulmonology Scientific Research Institute under Federal Medical‐Biologic AgencyMoscowRussia
| | - Alexey Nikitin
- Federal Research and Clinical Center of Federal Medical‐Biologic AgencyMoscowRussia
- Pulmonology Scientific Research Institute under Federal Medical‐Biologic AgencyMoscowRussia
| | - Anna Sotnikova
- Federal Research and Clinical Center of Federal Medical‐Biologic AgencyMoscowRussia
- Pulmonology Scientific Research Institute under Federal Medical‐Biologic AgencyMoscowRussia
| | - Svetlana Kotova
- Institute for Regenerative MedicineI. M.Sechenov First Moscow State Medical UniversityMoscowRussia
- Semenov Institute of Chemical PhysicsMoscow
| | - Vladimir Baklaushev
- Federal Research and Clinical Center of Federal Medical‐Biologic AgencyMoscowRussia
- Pulmonology Scientific Research Institute under Federal Medical‐Biologic AgencyMoscowRussia
| |
Collapse
|
55
|
Hejretová L, Čedíková M, Dolejšová M, Vlas T, Jindra P, Lysák D, Holubová M. Comparison of the immunomodulatory effect of single MSC batches versus pooled MSC products. Cell Tissue Bank 2019; 21:119-129. [PMID: 31863261 DOI: 10.1007/s10561-019-09805-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022]
Abstract
Severe corticosteroid-refractory graft-versus-host-disease (GVHD) is a major non-relapse cause of mortality and morbidity after an allogeneic hematopoietic stem cell transplantation (allo-HSCT). One of the most promising treatment options is using advanced therapy medicinal products based on mesenchymal stem cells (MSCs) immunomodulation ability. The protocols of MSC application differ in many parameters including a source of MSC, a dose, a number of doses or way of preparation of the medicinal product. The process is limited by the need for laborious and expensive manufacturing processes fraught with batch-to-batch variability. In our study, we compared the immunomodulatory effects of different MSC batches versus pooled MSC, specifically the influence on lymphocyte proliferation, the metabolic activity, and the expression of activation markers on T cells. Our goal was to determine whether the effect depends on donor-to-donor heterogeneity and if pooling of MSCs could increase their immunomodulatory ability. All tested batches showed an immunomodulatory effect, with no significant differences between the groups. Our study suggests that immunosuppressive potential is comparable in single batches and pooled products, and the use of products got from individual donors is suitable to treat corticosteroid-refractory GVHD.
Collapse
Affiliation(s)
- L Hejretová
- Department of Haematology and Oncology, University Hospital, Pilsen, Alej Svobody 80, 304 60, Pilsen, Czech Republic.,Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - M Čedíková
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - M Dolejšová
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - T Vlas
- Institute of Immunology and Allergology, University Hospital and Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - P Jindra
- Department of Haematology and Oncology, University Hospital, Pilsen, Alej Svobody 80, 304 60, Pilsen, Czech Republic
| | - D Lysák
- Department of Haematology and Oncology, University Hospital, Pilsen, Alej Svobody 80, 304 60, Pilsen, Czech Republic
| | - M Holubová
- Department of Haematology and Oncology, University Hospital, Pilsen, Alej Svobody 80, 304 60, Pilsen, Czech Republic. .,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| |
Collapse
|
56
|
Bonig H, Kuçi Z, Kuçi S, Bakhtiar S, Basu O, Bug G, Dennis M, Greil J, Barta A, Kállay KM, Lang P, Lucchini G, Pol R, Schulz A, Sykora KW, Teichert von Luettichau I, Herter-Sprie G, Ashab Uddin M, Jenkin P, Alsultan A, Buechner J, Stein J, Kelemen A, Jarisch A, Soerensen J, Salzmann-Manrique E, Hutter M, Schäfer R, Seifried E, Paneesha S, Novitzky-Basso I, Gefen A, Nevo N, Beutel G, Schlegel PG, Klingebiel T, Bader P. Children and Adults with Refractory Acute Graft-versus-Host Disease Respond to Treatment with the Mesenchymal Stromal Cell Preparation "MSC-FFM"-Outcome Report of 92 Patients. Cells 2019; 8:cells8121577. [PMID: 31817480 PMCID: PMC6952775 DOI: 10.3390/cells8121577] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/24/2019] [Accepted: 11/30/2019] [Indexed: 01/22/2023] Open
Abstract
(1) Background: Refractory acute graft-versus-host disease (R-aGvHD) remains a leading cause of death after allogeneic stem cell transplantation. Survival rates of 15% after four years are currently achieved; deaths are only in part due to aGvHD itself, but mostly due to adverse effects of R-aGvHD treatment with immunosuppressive agents as these predispose patients to opportunistic infections and loss of graft-versus-leukemia surveillance resulting in relapse. Mesenchymal stromal cells (MSC) from different tissues and those generated by various protocols have been proposed as a remedy for R-aGvHD but the enthusiasm raised by initial reports has not been ubiquitously reproduced. (2) Methods: We previously reported on a unique MSC product, which was generated from pooled bone marrow mononuclear cells of multiple third-party donors. The products showed dose-to-dose equipotency and greater immunosuppressive capacity than individually expanded MSCs from the same donors. This product, MSC-FFM, has entered clinical routine in Germany where it is licensed with a national hospital exemption authorization. We previously reported satisfying initial clinical outcomes, which we are now updating. The data were collected in our post-approval pharmacovigilance program, i.e., this is not a clinical study and the data is high-level and non-monitored. (3) Results: Follow-up for 92 recipients of MSC-FFM was reported, 88 with GvHD ≥°III, one-third only steroid-refractory and two-thirds therapy resistant (refractory to steroids plus ≥2 additional lines of treatment). A median of three doses of MSC-FFM was administered without apparent toxicity. Overall response rates were 82% and 81% at the first and last evaluation, respectively. At six months, the estimated overall survival was 64%, while the cumulative incidence of death from underlying disease was 3%. (4) Conclusions: MSC-FFM promises to be a safe and efficient treatment for severe R-aGvHD.
Collapse
Affiliation(s)
- Halvard Bonig
- Goethe University Medical Center, Institute of Transfusion Medicine and Immunohematology, and German Red Cross Blood Center Frankfurt, Frankfurt am Main, Frankfurt 60528, Germany; (R.S.); (E.S.)
- Correspondence: or ; Tel.: +49696782177
| | - Zyrafete Kuçi
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Frankfurt am Main, Frankfurt 60590, Germany; (Z.K.); (S.K.); (S.B.); (A.J.); (J.S.); (E.S.-M.); (M.H.); (T.K.); (P.B.)
| | - Selim Kuçi
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Frankfurt am Main, Frankfurt 60590, Germany; (Z.K.); (S.K.); (S.B.); (A.J.); (J.S.); (E.S.-M.); (M.H.); (T.K.); (P.B.)
| | - Shahrzad Bakhtiar
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Frankfurt am Main, Frankfurt 60590, Germany; (Z.K.); (S.K.); (S.B.); (A.J.); (J.S.); (E.S.-M.); (M.H.); (T.K.); (P.B.)
| | - Oliver Basu
- University Children’s Hospital Essen, Essen 45122, Germany;
| | - Gesine Bug
- Department of Medicine 2, Hematology and Oncology, University Hospital, Goethe University Frankfurt, Frankfurt am Main 60590, Germany;
| | - Mike Dennis
- Christie Hospital, Department of Haematology, Manchester M20 4BX, UK;
| | - Johann Greil
- University Children’s Hospital Heidelberg, Heidelberg 69120, Germany;
| | - Aniko Barta
- Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Department for Haematology and SCT, Budapest H1097, Hungary;
| | - Krisztián M. Kállay
- Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Pediatric Hematology and Stem Cell Transplantation Department, Budapest H1097, Hungary;
| | - Peter Lang
- University Children’s Hospital Tübingen, Tübingen 72076, Germany;
| | - Giovanna Lucchini
- Great Ormond Street Hospital, Department of Hematology/Oncology, London WC1N 3JH, UK;
| | - Raj Pol
- Department of Haematology, University of Sheffield, Sheffield S10 2TN, UK;
| | - Ansgar Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm 89070, Germany;
| | - Karl-Walter Sykora
- Children’s Hospital, Medizinische Hochschule Hannover, Hannover 30625, Germany;
| | - Irene Teichert von Luettichau
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Kinderklinik München Schwabing, Klinikum Rechts der Isar, Technische Universität München, München 80804, Germany;
| | - Grit Herter-Sprie
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, University of Cologne, Cologne 50937, Germany;
| | - Mohammad Ashab Uddin
- Department for Stem Cells & Immunotherapies, NHSBT, Birmingham B15 2SG, UK; (M.A.U.); (P.J.)
| | - Phil Jenkin
- Department for Stem Cells & Immunotherapies, NHSBT, Birmingham B15 2SG, UK; (M.A.U.); (P.J.)
| | - Abdulrahman Alsultan
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children’s Hospital, Riyadh 14611, Saudi Arabia;
| | - Jochen Buechner
- Oslo University Hospital, Department of Pediatric Hematology and Oncology, Oslo 0424, Norway;
| | - Jerry Stein
- Schneider Children’s Medical Center of Israel, Department for Hemato-Oncology, Petach Tikva 4920235, Israel;
| | - Agnes Kelemen
- B-A-Z County Hospital, Pediatric Haematology and Stem Cell Transplantation Unit, Miskolc 3526, Hungary;
| | - Andrea Jarisch
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Frankfurt am Main, Frankfurt 60590, Germany; (Z.K.); (S.K.); (S.B.); (A.J.); (J.S.); (E.S.-M.); (M.H.); (T.K.); (P.B.)
| | - Jan Soerensen
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Frankfurt am Main, Frankfurt 60590, Germany; (Z.K.); (S.K.); (S.B.); (A.J.); (J.S.); (E.S.-M.); (M.H.); (T.K.); (P.B.)
| | - Emilia Salzmann-Manrique
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Frankfurt am Main, Frankfurt 60590, Germany; (Z.K.); (S.K.); (S.B.); (A.J.); (J.S.); (E.S.-M.); (M.H.); (T.K.); (P.B.)
| | - Martin Hutter
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Frankfurt am Main, Frankfurt 60590, Germany; (Z.K.); (S.K.); (S.B.); (A.J.); (J.S.); (E.S.-M.); (M.H.); (T.K.); (P.B.)
| | - Richard Schäfer
- Goethe University Medical Center, Institute of Transfusion Medicine and Immunohematology, and German Red Cross Blood Center Frankfurt, Frankfurt am Main, Frankfurt 60528, Germany; (R.S.); (E.S.)
| | - Erhard Seifried
- Goethe University Medical Center, Institute of Transfusion Medicine and Immunohematology, and German Red Cross Blood Center Frankfurt, Frankfurt am Main, Frankfurt 60528, Germany; (R.S.); (E.S.)
| | - Shankara Paneesha
- Department of Haematology & Stem Cell Transplantation, Birmingham Heartlands Hospital, Birmingham B9 5SS, UK;
| | | | - Aharon Gefen
- Rambam Medical Center, Ruth Rappaport Children’s Hospital, Pediatric Hematology Oncology Division, The Reiner-Shudi Pediatric Bone Marrow Transplantation Unit, Haifa 3109601, Israel; (A.G.); (N.N.)
| | - Neta Nevo
- Rambam Medical Center, Ruth Rappaport Children’s Hospital, Pediatric Hematology Oncology Division, The Reiner-Shudi Pediatric Bone Marrow Transplantation Unit, Haifa 3109601, Israel; (A.G.); (N.N.)
| | - Gernot Beutel
- Hannover Medical School (MHH), Hannover, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover 30625, Germany;
| | | | - Thomas Klingebiel
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Frankfurt am Main, Frankfurt 60590, Germany; (Z.K.); (S.K.); (S.B.); (A.J.); (J.S.); (E.S.-M.); (M.H.); (T.K.); (P.B.)
| | - Peter Bader
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Frankfurt am Main, Frankfurt 60590, Germany; (Z.K.); (S.K.); (S.B.); (A.J.); (J.S.); (E.S.-M.); (M.H.); (T.K.); (P.B.)
| |
Collapse
|
57
|
Godoy JAP, Paiva RMA, Souza AM, Kondo AT, Kutner JM, Okamoto OK. Clinical Translation of Mesenchymal Stromal Cell Therapy for Graft Versus Host Disease. Front Cell Dev Biol 2019; 7:255. [PMID: 31824942 PMCID: PMC6881464 DOI: 10.3389/fcell.2019.00255] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/15/2019] [Indexed: 12/15/2022] Open
Abstract
Graft versus host disease (GVHD) is a common condition in patients subjected to allogeneic hematopoietic stem cell transplantation (HSCT). The immune cells derived from the grafted stem cells attack recipient's tissues, including those from the skin, liver, eyes, mouth, lungs, gastrointestinal tract, neuromuscular system, and genitourinary tract, may lead to severe morbidity and mortality. Acute GVHD can occur within few weeks after the allogeneic cells have engrafted in the recipient while chronic GVHD may occur any time after transplant, typically within months. Although treatable by systemic corticosteroid administration, effective responses are not achieved for a significant proportion of patients, a condition associated with poor prognosis. The use of multipotent mesenchymal stromal cells (MSCs) as an alternative to treat steroid-refractory GVHD had improved last decade, but the results are still controversial. Some studies have shown improvement in the life quality of patients after MSCs treatment, while others have found no significant benefits. In addition to variations in trial design, discrepancies in protocols for MSCs isolation, characterization, and ex vivo manipulation, account for inconsistent clinical results. In this review, we discuss the immunomodulatory properties supporting the therapeutic use of MSCs in GVHD and contextualize the main clinical findings of recent trials using these cells. Critical parameters for the clinical translation of MSCs, including consistent production of MSCs according to Good Manufacturing Practices (GMPs) and informative potency assays for product quality control (QC), are addressed.
Collapse
Affiliation(s)
- Juliana A. P. Godoy
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Raquel M. A. Paiva
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Aline M. Souza
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Andrea T. Kondo
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Jose M. Kutner
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Oswaldo K. Okamoto
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
58
|
Central metabolism of functionally heterogeneous mesenchymal stromal cells. Sci Rep 2019; 9:15420. [PMID: 31659213 PMCID: PMC6817850 DOI: 10.1038/s41598-019-51937-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 10/08/2019] [Indexed: 12/16/2022] Open
Abstract
Metabolism and mitochondrial biology have gained a prominent role as determinants of stem cell fate and function. In the context of regenerative medicine, innovative parameters predictive of therapeutic efficacy could be drawn from the association of metabolic or mitochondrial parameters to different degrees of stemness and differentiation potentials. Herein, this possibility was addressed in human mesenchymal stromal/stem cells (hMSC) previously shown to differ in lifespan and telomere length. First, these hMSC were shown to possess significantly distinct proliferation rate, senescence status and differentiation capacity. More potential hMSC were associated to higher mitochondrial (mt) DNA copy number and lower mtDNA methylation. In addition, they showed higher expression levels of oxidative phosphorylation subunits. Consistently, they exhibited higher coupled oxygen consumption rate and lower transcription of glycolysis-related genes, glucose consumption and lactate production. All these data pointed at oxidative phosphorylation-based central metabolism as a feature of higher stemness-associated hMSC phenotypes. Consistently, reduction of mitochondrial activity by complex I and III inhibitors in higher stemness-associated hMSC triggered senescence. Finally, functionally higher stemness-associated hMSC showed metabolic plasticity when challenged by glucose or glutamine shortage, which mimic bioenergetics switches that hMSC must undergo after transplantation or during self-renewal and differentiation. Altogether, these results hint at metabolic and mitochondrial parameters that could be implemented to identify stem cells endowed with superior growth and differentiation potential.
Collapse
|
59
|
Bozkurt C, Karaöz E, Adaklı Aksoy B, Aydoğdu S, Fışgın T. The Use of Allogeneic Mesenchymal Stem Cells in Childhood Steroid-Resistant Acute Graft-Versus-Host Disease: A Retrospective Study of a Single-Center Experience. Turk J Haematol 2019; 36:186-192. [PMID: 31208159 PMCID: PMC6682770 DOI: 10.4274/tjh.galenos.2019.2019.0090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Objective: Steroid-resistant acute graft-versus-host disease (srAGVHD) is the most important cause of morbidity and mortality after allogeneic stem cell transplantation. There are several treatment methods available, including mesenchymal stem cell (MSC) application. The aim of this study was to evaluate the results of MSC therapy performed in children with srAGVHD. Materials and Methods: MSC therapy was used in our center between November 2014 and December 2017 for 22 patients who developed srAGVHD. The patients were retrospectively evaluated in terms of treatment response and survival. Results: After application of MSCs, complete response was obtained in 45.5% of the subjects, partial response was obtained in 13.6%, and no response was obtained in 40.9%. We found that 45.5% of the patients were alive and 54.5% had died and our treatment results were similar to those in the literature. Response to MSC treatment was found to be the only prognostic marker affecting mortality. Conclusion: MSC application is a treatment method that can be used safely together with other treatment methods in srAGVHD, a condition that has a high mortality rate. There are almost no acute side effects. There are also no serious long-term side effects in the literature. Prospective randomized studies are required to obtain high-quality data.
Collapse
Affiliation(s)
- Ceyhun Bozkurt
- İstinye University Faculty of Medicine, Department of Pediatrics, İstanbul, Turkey,Altınbaş University Faculty of Medicine, Bahçelievler Medical Park Hospital Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Erdal Karaöz
- İstinye University Faculty of Medicine, Department of Histology-Embryology, İstanbul, Turkey,İstinye University Faculty of Medicine, Stem Cell and Tissue Engineering Research and Application Center, İstanbul, Turkey,Liv Hospital, Regenerative Medicine, Stem Cell Production Center, İstanbul, Turkey
| | - Başak Adaklı Aksoy
- İstinye University Faculty of Medicine, Department of Pediatrics, İstanbul, Turkey,Altınbaş University Faculty of Medicine, Bahçelievler Medical Park Hospital Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Selime Aydoğdu
- Altınbaş University Faculty of Medicine, Bahçelievler Medical Park Hospital Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Tunç Fışgın
- Altınbaş University Faculty of Medicine, Bahçelievler Medical Park Hospital Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| |
Collapse
|
60
|
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: 42] [Impact Index Per Article: 7.0] [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
|
61
|
Bremm M, Pfeffermann LM, Cappel C, Katzki V, Erben S, Betz S, Quaiser A, Merker M, Bonig H, Schmidt M, Klingebiel T, Bader P, Huenecke S, Rettinger E. Improving Clinical Manufacturing of IL-15 Activated Cytokine-Induced Killer (CIK) Cells. Front Immunol 2019; 10:1218. [PMID: 31214182 PMCID: PMC6554420 DOI: 10.3389/fimmu.2019.01218] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/13/2019] [Indexed: 12/18/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are an immunotherapeutic approach to combat relapse following allogeneic hematopoietic stem cell transplantation (HSCT) in acute leukemia or myelodysplastic syndrome (MDS) patients. Prompt and sequential administration of escalating cell doses improves the efficacy of CIK cell therapy without exacerbating graft vs. host disease (GVHD). This study addresses manufacturing-related issues and aimed to develop a time-, personal- and cost-saving good manufacturing process (GMP)-compliant protocol for the generation of ready-for-use therapeutic CIK cell doses starting from one unstimulated donor-derived peripheral blood (PB) or leukocytapheresis (LP) products. Culture medium with or without the addition of either AB serum, fresh frozen plasma (FFP) or platelet lysate (PL) was used for culture. Fresh and cryopreserved CIK cells were compared regarding expansion rate, viability, phenotype, and ability to inhibit leukemia growth. Cell numbers increased by a median factor of 10-fold in the presence of FFP, PL, or AB serum, whereas cultivation in FFP/PL-free or AB serum-free medium failed to promote adequate CIK cell proliferation (p < 0.01) needed to provide clinical doses of 1 × 106 T cells/kG, 5 × 106 T cells/kG, 1 × 107 T cells/kG, and 1 × 108 T cells/kG recipient body weight. CIK cells consisting of T cells, T- natural killer (T-NK) cells and a minor fraction of NK cells were not significantly modified by different medium supplements. Moreover, neither cytotoxic potential against leukemic THP-1 cells nor cell activation shown by CD25 expression were significantly influenced. Moreover, overnight and long-term cryopreservation had no significant effect on the composition of CIK cells, their phenotype or cytotoxic potential. A viability of almost 93% (range: 89–96) and 89.3% (range: 84–94) was obtained after freeze-thawing procedure and long-term storage, respectively, whereas viability was 96% (range: 90-97) in fresh CIK cells. Altogether, GMP-complaint CIK cell generation from an unstimulated donor-derived PB or LP products was feasible. Introducing FFP, which is easily accessible, into CIK cell cultures was time- and cost-saving without loss of viability and potency in a 10-12 day batch culture. The feasibility of cryopreservation enabled storage and delivery of sequential highly effective ready-for-use CIK cell doses and therefore reduced the number of manufacturing cycles.
Collapse
Affiliation(s)
- Melanie Bremm
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | | | - Claudia Cappel
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Verena Katzki
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Stephanie Erben
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Sibille Betz
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Andrea Quaiser
- Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Michael Merker
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Halvard Bonig
- Division for Translational Development of Cellular Therapeutics, Institute for Transfusion Medicine and Immunohematology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Michael Schmidt
- Division for Translational Development of Cellular Therapeutics, Institute for Transfusion Medicine and Immunohematology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Thomas Klingebiel
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Peter Bader
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Sabine Huenecke
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Eva Rettinger
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| |
Collapse
|
62
|
Perspectives for Clinical Translation of Adipose Stromal/Stem Cells. Stem Cells Int 2019; 2019:5858247. [PMID: 31191677 PMCID: PMC6525805 DOI: 10.1155/2019/5858247] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/26/2019] [Accepted: 03/07/2019] [Indexed: 12/15/2022] Open
Abstract
Adipose stromal/stem cells (ASCs) are an ideal cell type for regenerative medicine applications, as they can easily be harvested from adipose tissue in large quantities. ASCs have excellent proliferation, differentiation, and immunoregulatory capacities that have been demonstrated in numerous studies. Great interest and investment have been placed in efforts to exploit the allogeneic use and immunomodulatory and anti-inflammatory effects of ASCs. However, bridging the gap between in vitro and in vivo studies and moving into clinical practice remain a challenge. For the clinical translation of ASCs, several issues must be considered, including how to characterise such a heterogenic cell population and how to ensure their safety and efficacy. This review explores the different phases of in vitro and preclinical ASC characterisation and describes the development of appropriate potency assays. In addition, good manufacturing practice requirements are discussed, and cell-based medicinal products holding marketing authorisation in the European Union are reviewed. Moreover, the current status of clinical trials applying ASCs and the patent landscape in the field of ASC research are presented. Overall, this review highlights the applicability of ASCs for clinical cell therapies and discusses their potential.
Collapse
|
63
|
Bieback K, Kuçi S, Schäfer R. Production and quality testing of multipotent mesenchymal stromal cell therapeutics for clinical use. Transfusion 2019; 59:2164-2173. [DOI: 10.1111/trf.15252] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/10/2019] [Accepted: 02/10/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty MannheimGerman Red Cross Blood Donor Service Baden‐Württemberg‐Hessen gGmbH, Heidelberg University Mannheim Germany
- FlowCore Mannheim, Medical Faculty MannheimHeidelberg University Germany
| | - Selim Kuçi
- Department for Children and Adolescents, Division for Stem Cell Transplantation and ImmunologyUniversity Hospital Frankfurt Frankfurt am Main Germany
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohaematology, German Red Cross Blood Donor Service Baden‐Württemberg‐Hessen gGmbHGoethe University Hospital Frankfurt am Main Germany
| |
Collapse
|
64
|
Elgaz S, Kuçi Z, Kuçi S, Bönig H, Bader P. Clinical Use of Mesenchymal Stromal Cells in the Treatment of Acute Graft-versus-Host Disease. Transfus Med Hemother 2019; 46:27-34. [PMID: 31244579 DOI: 10.1159/000496809] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 01/10/2019] [Indexed: 12/12/2022] Open
Abstract
Acute graft-versus-host disease (aGvHD) continues to impact morbidity and mortality after allogeneic stem cell transplantation (allo-SCT). First-line therapy for aGvHD still remains the use of high-dose corticosteroids. Unfortunately, 40-60% of patients with aGvHD exhibit steroid resistance, which is associated with a very poor prognosis. As no effective second-line therapy existed, in recent decades various treatment options were considered for the treatment of therapy-refractory GvHD. Based on their in vitro immunomodulatory properties, the use of mesenchymal stromal cells (MSCs) in the treatment of aGvHD has been introduced. However, most of the clinical data are generated from uncontrolled trials and case series, showing clinical responses to MSCs. Clinical results are more consistent in children despite the use of MSC preparations of various provenance and manufacturing protocols. While these data support the therapeutic principle, the great variability of outcomes strongly suggests that not all MSC preparations are equal and that the specific manufacturing protocols influence therapeutic success in vivo.
Collapse
Affiliation(s)
- Sümeyye Elgaz
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Zyrafete Kuçi
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Selim Kuçi
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Halvard Bönig
- German Red Cross Blood Center Frankfurt and Institute of Transfusion Medicine and Immunohematology, Goethe University Medical Center, Frankfurt am Main, Germany
| | - Peter Bader
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
65
|
Zuo R, Liu M, Wang Y, Li J, Wang W, Wu J, Sun C, Li B, Wang Z, Lan W, Zhang C, Shi C, Zhou Y. BM-MSC-derived exosomes alleviate radiation-induced bone loss by restoring the function of recipient BM-MSCs and activating Wnt/β-catenin signaling. Stem Cell Res Ther 2019; 10:30. [PMID: 30646958 PMCID: PMC6334443 DOI: 10.1186/s13287-018-1121-9] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 12/12/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023] Open
Abstract
Background Radiotherapy to cancer patients is inevitably accompanied by normal tissue injury, and the bone is one of the most commonly damaged tissues. Damage to bone marrow mesenchymal stem cells (BM-MSCs) induced by radiation is thought to be a major cause of radiation-induced bone loss. Exosomes exhibit great therapeutic potential in the treatment of osteoporosis, but whether exosomes are involved in radiation-induced bone loss has not been thoroughly elucidated to date. The main purpose of this study is to investigate the role of exosomes derived from BM-MSCs in restoring recipient BM-MSC function and alleviating radiation-induced bone loss. Methods BM-MSC-derived exosomes were intravenously injected to rats immediately after irradiation. After 28 days, the left tibiae were harvested for micro-CT and histomorphometric analysis. The effects of exosomes on antioxidant capacity, DNA damage repair, proliferation, and cell senescence of recipient BM-MSCs were determined. Osteogenic and adipogenic differentiation assays were used to detect the effects of exosomes on the differentiation potential of recipient BM-MSCs, and related genes were measured by qRT-PCR and Western blot analysis. β-Catenin expression was detected at histological and cytological levels. Results BM-MSC-derived exosomes can attenuate radiation-induced bone loss in a rat model that is similar to mesenchymal stem cell transplantation. Exosome-treated BM-MSCs exhibit reduced oxidative stress, accelerated DNA damage repair, and reduced proliferation inhibition and cell senescence-associate protein expression compared with BM-MSCs that exclusively received irradiation. Following irradiation, exosomes promote β-catenin expression in BM-MSCs and restore the balance between adipogenic and osteogenic differentiation. Conclusions Our findings indicate that BM-MSC-derived exosomes take effects by restoring the function of recipient BM-MSCs. Therefore, exosomes may represent a promising cell-free therapeutic approach for the treatment of radiation-induced bone loss. Electronic supplementary material The online version of this article (10.1186/s13287-018-1121-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rui Zuo
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Minghan Liu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Yanqiu Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Jie Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Wenkai Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Junlong Wu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Chao Sun
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Bin Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Ziwen Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University(Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Weiren Lan
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Chao Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University(Third Military Medical University), Chongqing, 400038, People's Republic of China.
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China.
| |
Collapse
|
66
|
Leyendecker A, Pinheiro CCG, Amano MT, Bueno DF. The Use of Human Mesenchymal Stem Cells as Therapeutic Agents for the in vivo Treatment of Immune-Related Diseases: A Systematic Review. Front Immunol 2018; 9:2056. [PMID: 30254638 PMCID: PMC6141714 DOI: 10.3389/fimmu.2018.02056] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022] Open
Abstract
Background: One of the greatest challenges for medicine is to find a safe and effective treatment for immune-related diseases. However, due to the low efficacy of the treatment available and the occurrence of serious adverse effects, many groups are currently searching for alternatives to the traditional therapy. In this regard, the use of human mesenchymal stem cells (hMSCs) represents a great promise for the treatment of a variety of immune-related diseases due to their potent immunomodulatory properties. The main objective of this study is, therefore, to present and summarize, through a systematic review of the literature, in vivo studies in which the efficacy of the administration of hMSCs for the treatment of immune-related diseases was evaluated. Methods: The article search was conducted in PubMed/MEDLINE, Scopus and Web of Science databases. Original research articles assessing the therapeutic potential of hMSCs administration for the in vivo treatment immune-related diseases, published from 1984 to December 2017, were selected and evaluated. Results: A total of 132 manuscripts formed the basis of this systematic review. Most of the studies analyzed reported positive results after hMSCs administration. Clinical effects commonly observed include an increase in the survival rates and a reduction in the severity and incidence of the immune-related diseases studied. In addition, hMSCs administration resulted in an inhibition in the proliferation and activation of CD19+ B cells, CD4+ Th1 and Th17 cells, CD8+ T cells, NK cells, macrophages, monocytes, and neutrophils. The clonal expansion of both Bregs and Tregs cells, however, was stimulated. Administration of hMSCs also resulted in a reduction in the levels of pro-inflammatory cytokines such as IFN-γ, TNF-α, IL-1, IL-2, IL-12, and IL-17 and in an increase in the levels of immunoregulatory cytokines such as IL-4, IL-10, and IL-13. Conclusions: The results obtained in this study open new avenues for the treatment of immune-related diseases through the administration of hMSCs and emphasize the importance of the conduction of further studies in this area.
Collapse
|
67
|
Seng A, Dunavin N. Mesenchymal stromal cell infusions for acute graft-versus-host disease: Rationale, data, and unanswered questions. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/acg2.14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Amara Seng
- Department of Microbiology; Molecular Genetics and Immunology; University of Kansas Medical Center; Kansas City Kansas
| | - Neil Dunavin
- Division of Hematological Malignancies and Cellular Therapeutics; Department of Internal Medicine; University of Kansas Medical Center; Kansas City Kansas
| |
Collapse
|
68
|
Česen Mazič M, Girandon L, Kneževič M, Avčin SL, Jazbec J. Treatment of Severe Steroid-Refractory Acute-Graft-vs.-Host Disease With Mesenchymal Stem Cells-Single Center Experience. Front Bioeng Biotechnol 2018; 6:93. [PMID: 30087891 PMCID: PMC6066564 DOI: 10.3389/fbioe.2018.00093] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/21/2018] [Indexed: 12/13/2022] Open
Abstract
The most effective treatment of steroid refractory acute graft vs. host disease (aGvHD) is not yet established and mesenchymal stem cells (MSC) appear to be a promising therapy for the condition. We report single center case series of three patients, who underwent allogeneic hematopoietic cell transplantation and later developed steroid refractory graft-vs.-host disease, treated with MSC infusions. Two patients achieved complete remission and one patient partial remission of skin and/or gastrointestinal aGvHD. We demonstrated application of MSC for treatment of severe steroid refractory aGvHD is feasible in clinical practice. Detailed description of patient's features and MSC production protocol is crucial for future comparison on efficacy and safety of cell-based therapies. However, for any substantial conclusions regarding efficacy of MSC higher patient numbers will be required.
Collapse
Affiliation(s)
- Maja Česen Mazič
- Department for Pediatric Hematology and Oncology, University Children Hospital Ljubljana, Ljubljana, Slovenia
| | | | | | - Simona L Avčin
- Department for Pediatric Hematology and Oncology, University Children Hospital Ljubljana, Ljubljana, Slovenia
| | - Janez Jazbec
- Department for Pediatric Hematology and Oncology, University Children Hospital Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
69
|
Trento C, Bernardo ME, Nagler A, Kuçi S, Bornhäuser M, Köhl U, Strunk D, Galleu A, Sanchez-Guijo F, Gaipa G, Introna M, Bukauskas A, Le Blanc K, Apperley J, Roelofs H, Van Campenhout A, Beguin Y, Kuball J, Lazzari L, Avanzini MA, Fibbe W, Chabannon C, Bonini C, Dazzi F. Manufacturing Mesenchymal Stromal Cells for the Treatment of Graft-versus-Host Disease: A Survey among Centers Affiliated with the European Society for Blood and Marrow Transplantation. Biol Blood Marrow Transplant 2018; 24:2365-2370. [PMID: 30031938 PMCID: PMC6299357 DOI: 10.1016/j.bbmt.2018.07.015] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/09/2018] [Indexed: 12/11/2022]
Abstract
The immunosuppressive properties of mesenchymal stromal cells (MSC) have been successfully tested to control clinical severe graft-versus host disease and improve survival. However, clinical studies have not yet provided conclusive evidence of their efficacy largely because of lack of patients' stratification criteria. The heterogeneity of MSC preparations is also a major contributing factor, as manufacturing of therapeutic MSC is performed according to different protocols among different centers. Understanding the variability of the manufacturing protocol would allow a better comparison of the results obtained in the clinical setting among different centers. In order to acquire information on MSC manufacturing we sent a questionnaire to the European Society for Blood and Marrow Transplantation centers registered as producing MSC. Data from 17 centers were obtained and analyzed by means of a 2-phase questionnaire specifically focused on product manufacturing. Gathered information included MSC tissue sources, MSC donor matching, medium additives for ex vivo expansion, and data on MSC product specification for clinical release. The majority of centers manufactured MSC from bone marrow (88%), whilst only 2 centers produced MSC from umbilical cord blood or cord tissue. One of the major changes in the manufacturing process has been the replacement of fetal bovine serum with human platelet lysate as medium supplement. 59% of centers used only third-party MSC, whilst only 1 center manufactured exclusively autologous MSC. The large majority of these facilities (71%) administered MSC exclusively from frozen batches. Aside from variations in the culture method, we found large heterogeneity also regarding product specification, particularly in the markers used for phenotypical characterization and their threshold of expression, use of potency assays to test MSC functionality, and karyotyping. The initial data collected from this survey highlight the variability in MSC manufacturing as clinical products and the need for harmonization. Until more informative potency assays become available, a more homogeneous approach to cell production may at least reduce variability in clinical trials and improve interpretation of results.
Collapse
Affiliation(s)
- Cristina Trento
- School of Cancer & Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Maria Ester Bernardo
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele Telethon Institute for Gene Therapy (TIGET), San Raffaele Scientific Institute, Milan, Italy
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Selim Kuçi
- Division for Stem Cell Transplantation and Immunology, University Hospital for Children and Adolescents, Frankfurt am Main, Germany
| | - Martin Bornhäuser
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Germany
| | - Ulrike Köhl
- Institute of Clinical Immunology, University Leipzig, Leipzig, Germany; Fraunhofer Institute of Cellular Therapy and Immunology and Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - Dirk Strunk
- Experimental & Clinical Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria
| | - Antonio Galleu
- School of Cancer & Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Fermin Sanchez-Guijo
- Hematology Department, IBSAL-Hospital Universitario de Salamanca, University of Salamanca, Salamanca, Spain
| | - Giuseppe Gaipa
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST Monza, Monza, Italy
| | - Martino Introna
- USS Centro di Terapia Cellulare "G.Lanzani", ASST Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | - Adomas Bukauskas
- Center of Hematology, Oncology and Transfusion Medicine, Vilnius University Hospital Santariskiu Klinikos, Vilnius, Lithuania
| | - Katarina Le Blanc
- Department of Haematology, Karolinska University Hospital, Sweden, Stockholm
| | - Jane Apperley
- Department of Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Helene Roelofs
- Leiden University Medical Centre, Leiden, The Netherlands
| | - Ann Van Campenhout
- Transplantation Laboratory Hematology, U.Z. Gasthuisberg, Leuven, Belgium
| | - Yves Beguin
- Laboratory of Cell and Gene Therapy, Clinical Haematology, CHU of Liège, Liège, Belgium
| | - Jürgen Kuball
- Department of Hematology and Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lorenza Lazzari
- Cell Factory GMP, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Maria Antonietta Avanzini
- Laboratorio Immunologia e dei Trapianti, Cell Factory, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Willem Fibbe
- Leiden University Medical Centre, Leiden, The Netherlands
| | - Christian Chabannon
- Institut Paoli Calmettes & Inserm CBT-1409, Centre d'Investigations Cliniques en Biothérapies, Marseille, France
| | - Chiara Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, University Vita-Salute San Raffaele and Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Dazzi
- School of Cancer & Pharmaceutical Sciences, King's College London, London, United Kingdom.
| |
Collapse
|
70
|
Moutuou MM, Pagé G, Zaid I, Lesage S, Guimond M. Restoring T Cell Homeostasis After Allogeneic Stem Cell Transplantation; Principal Limitations and Future Challenges. Front Immunol 2018; 9:1237. [PMID: 29967605 PMCID: PMC6015883 DOI: 10.3389/fimmu.2018.01237] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/17/2018] [Indexed: 11/29/2022] Open
Abstract
For several leukemia patients, allogeneic stem cell transplantation (allogeneic-SCT) is the unique therapeutic modality that could potentially cure their disease. Despite significant progress made in clinical management of allogeneic-SCT, acute graft-versus-host disease (aGVHD) and infectious complications remain the second and third cause of death after disease recurrence. Clinical options to restore immunocompetence after allogeneic-SCT are very limited as studies have raised awareness about the safety with regards to graft-versus-host disease (GVHD). Preclinical works are now focusing on strategies to improve thymic functions and to restore the peripheral niche that have been damaged by alloreactive T cells. In this mini review, we will provide a brief overview about the adverse effects of GVHD on the thymus and the peripheral niche and the resulting negative outcome on peripheral T cell homeostasis. Finally, we will discuss the potential relevance of coordinating our studies on thymic rejuvenation and improvement of the peripheral lymphoid niche to achieve optimal T cell regeneration in GVHD patients.
Collapse
Affiliation(s)
- Moutuaata M Moutuou
- Division d'Hématologie-Oncologie, Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université of Montréal, Montréal, QC, Canada
| | - Gabriel Pagé
- Département de Microbiologie, Infectiologie et Immunologie, Université of Montréal, Montréal, QC, Canada
| | - Intesar Zaid
- Division d'Hématologie-Oncologie, Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université of Montréal, Montréal, QC, Canada
| | - Sylvie Lesage
- Division d'Hématologie-Oncologie, Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université of Montréal, Montréal, QC, Canada
| | - Martin Guimond
- Division d'Hématologie-Oncologie, Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université of Montréal, Montréal, QC, Canada
| |
Collapse
|
71
|
Servais S, Baron F, Lechanteur C, Seidel L, Selleslag D, Maertens J, Baudoux E, Zachee P, Van Gelder M, Noens L, Kerre T, Lewalle P, Schroyens W, Ory A, Beguin Y. Infusion of bone marrow derived multipotent mesenchymal stromal cells for the treatment of steroid-refractory acute graft-versus-host disease: a multicenter prospective study. Oncotarget 2018; 9:20590-20604. [PMID: 29755674 PMCID: PMC5945536 DOI: 10.18632/oncotarget.25020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/17/2018] [Indexed: 12/12/2022] Open
Abstract
The prognosis of steroid-refractory acute graft-versus-host disease (aGVHD) remains poor and better treatments are urgently needed. Multipotent mesenchymal stromal cell (MSC)-based therapy emerged as a promising approach but response rates were highly variable across studies. We conducted a multicenter prospective study assessing the efficacy of 1-2 infusion(s) of cryopreserved, third-party donor bone marrow-derived MSCs for treating grade II-IV steroid-refractory or -dependent aGVHD in a series of 33 patients. MSCs were produced centrally and distributed to 8 hospitals throughout Belgium to be infused in 2 consecutive cohorts of patients receiving 1-2 or 3-4 × 106 MSCs/kg per dose, respectively. All patients received MSCs as the first rescue therapy after corticosteroids, with the exception for one patient who received prior treatment with mycophenolate mofetil (that was still ongoing by the time of MSC therapy). In these conditions, MSC therapy resulted in at least a partial response in 13 patients (40.6%) at day 30 and in 15 patients (46%) within 90 days after first MSC infusion. The corresponding complete response rates were 21.6% (7 patients) and 30% (10 patients), respectively. Only 5 patients achieved a sustained complete response, lasting for at least 1 month. The 1-year overall survival was 18.2% (95% CI: 8.82-37.5%). Higher response and survival rates were observed among patients receiving 3-4 × 106 MSCs/kg for first infusion, as compared with patients receiving 1-2 × 106 MSCs/ kg. Response and survival with MSC therapy for SR/SD-aGVHD remains to be optimized.
Collapse
Affiliation(s)
- Sophie Servais
- Department of Hematology, CHU of Liège, 4000 Liège, Belgium
- Laboratory of Cell and Gene Therapy, CHU of Liège, 4000 Liège, Belgium
| | - Frédéric Baron
- Department of Hematology, CHU of Liège, 4000 Liège, Belgium
- Laboratory of Cell and Gene Therapy, CHU of Liège, 4000 Liège, Belgium
| | - Chantal Lechanteur
- Department of Hematology, CHU of Liège, 4000 Liège, Belgium
- Laboratory of Cell and Gene Therapy, CHU of Liège, 4000 Liège, Belgium
| | - Laurence Seidel
- Department of biostatistics, SIMÉ, CHU of Liège, 4000 Liège, Belgium
| | | | - Johan Maertens
- Department of Hematology, AZ Gasthuisberg, 3000 Leuven, Belgium
| | - Etienne Baudoux
- Department of Hematology, CHU of Liège, 4000 Liège, Belgium
- Laboratory of Cell and Gene Therapy, CHU of Liège, 4000 Liège, Belgium
| | - Pierre Zachee
- Department of Hematology, ZNA Stuivenberg, 2060 Antwerp, Belgium
| | - Michel Van Gelder
- Department of Internal Medicine, Hematology Division, Maastricht University Medical Center, 6202 AZ Maastricht, The Nertherlands
| | - Lucien Noens
- Department of Hematology, UZ Gent, 9000 Ghent, Belgium
| | - Tessa Kerre
- Department of Hematology, UZ Gent, 9000 Ghent, Belgium
| | - Philippe Lewalle
- Department of Hematology, Institut Jules-Bordet, 1000 Brussels, Belgium
| | - Wilfried Schroyens
- Department of Hematology, Antwerp University Hospital, 2650 Edegem and University of Antwerp, 2610 Antwerp, Belgium
| | - Aurélie Ory
- Clinical Research Associate of the Belgian Hematology Society, CHU Sart-Tilman, 4000 Liège, Belgium
| | - Yves Beguin
- Department of Hematology, CHU of Liège, 4000 Liège, Belgium
- Laboratory of Cell and Gene Therapy, CHU of Liège, 4000 Liège, Belgium
| |
Collapse
|
72
|
Bader P, Kuçi Z, Bakhtiar S, Basu O, Bug G, Dennis M, Greil J, Barta A, Kállay KM, Lang P, Lucchini G, Pol R, Schulz A, Sykora KW, von Luettichau I, Herter-Sprie G, Uddin MA, Jenkin P, Alsultan A, Buechner J, Stein J, Kelemen A, Jarisch A, Soerensen J, Salzmann-Manrique E, Hutter M, Schäfer R, Seifried E, Klingebiel T, Bonig H, Kuçi S. Effective treatment of steroid and therapy-refractory acute graft-versus-host disease with a novel mesenchymal stromal cell product (MSC-FFM). Bone Marrow Transplant 2018; 53:852-862. [PMID: 29379171 PMCID: PMC6039391 DOI: 10.1038/s41409-018-0102-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 12/27/2022]
Abstract
The inability to generate mesenchymal stromal cells (MSCs) of consistent potency likely is responsible for inconsistent clinical outcomes of patients with aGvHD receiving MSC products. We developed a novel MSC manufacturing protocol characterized by high in vitro potency and near-identity of individual doses, referred to as “MSC-Frankfurt am Main (MSC-FFM)”. Herein, we report outcomes of the 69 patients who have received MSC-FFM. These were 51 children and 18 adults with refractory aGvHD grade II (4%), III (36%) or IV (59%). Patients were refractory either to frontline therapy (steroids) (29%) or to steroids and 1–5 additional lines of immunosuppressants (71%) were given infusions in four weekly intervals. The day 28 overall response rate was 83%; at the last follow-up, 61% and 25% of patients were in complete or partial remission. The median follow-up was 8.1 months. Six-month estimate for cumulative incidence of non-relapse mortality was 27% (range, 16–38); leukemia relapse mortality was 2% (range, 0–5). This was associated with a superior six-month overall survival (OS) probability rate of 71% (range, 61–83), compared to the outcome of patients not treated with MSC-FFM. This novel product was effective in children and adults, suggesting that MSC-FFM represents a promising therapy for steroid refractory aGvHD.
Collapse
Affiliation(s)
- Peter Bader
- Department for Children and Adolescents, Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Frankfurt am Main, Germany.
| | - Zyrafete Kuçi
- Department for Children and Adolescents, Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Shahrzad Bakhtiar
- Department for Children and Adolescents, Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Oliver Basu
- University Children's Hospital, Essen, Germany
| | - Gesine Bug
- Department of Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Michael Dennis
- Department of Haematology, Christie Hospital, Manchester, United Kingdom
| | - Johann Greil
- University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Aniko Barta
- Department for Haematology and SCT, St. István and St. László Hospital, Budapest, Hungary
| | - Krisztián M Kállay
- Department for Haematology and SCT, St. István and St. László Hospital, Budapest, Hungary
| | - Peter Lang
- University Children's Hospital Tübingen, Tübingen, Germany
| | - Giovanna Lucchini
- Department of Hematology/Oncology, Great Ormond Street Hospital, London, United Kingdom
| | - Raj Pol
- Department of Haematology, University of Sheffield, Sheffield, United Kingdom
| | | | | | - Irene von Luettichau
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Kinderklinik München Schwabing, Klinikum Rechts der Isar, Technische Universität München, München, Germany
| | - Grit Herter-Sprie
- Department I for Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Mohammad Ashab Uddin
- Department for Stem Cells & Immunotherapies, NHSBT, Birmingham, Great Britain, UK
| | - Phil Jenkin
- Department for Stem Cells & Immunotherapies, NHSBT, Birmingham, Great Britain, UK
| | - Abdulrahman Alsultan
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children's Hospital, Riyadh, Saudi Arabia
| | - Jochen Buechner
- Oslo University Hospital Rikshospitalet, Department of Pediatric Medicine, Section for Pediatric Hematology/Oncology, Oslo, Norway
| | - Jerry Stein
- Department for Hemato-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Agnes Kelemen
- B-A-Z County Hospital, Pediatric Haematology and Stem Cell Transplantation Unit, Miskolc, Hungary
| | - Andrea Jarisch
- Department for Children and Adolescents, Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Jan Soerensen
- Department for Children and Adolescents, Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Emilia Salzmann-Manrique
- Department for Children and Adolescents, Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Martin Hutter
- Department for Children and Adolescents, Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Richard Schäfer
- German Red Cross Blood Center Frankfurt and Institute of Transfusion Medicine and Immunohematology, Goethe University Medical Center, Frankfurt am Main, Germany
| | - Erhard Seifried
- German Red Cross Blood Center Frankfurt and Institute of Transfusion Medicine and Immunohematology, Goethe University Medical Center, Frankfurt am Main, Germany
| | - Thomas Klingebiel
- Department for Children and Adolescents, Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Halvard Bonig
- German Red Cross Blood Center Frankfurt and Institute of Transfusion Medicine and Immunohematology, Goethe University Medical Center, Frankfurt am Main, Germany
| | - Selim Kuçi
- Department for Children and Adolescents, Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
73
|
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: 130] [Impact Index Per Article: 16.3] [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
|
74
|
Shanbhag S, Stavropoulos A, Suliman S, Hervig T, Mustafa K. Efficacy of Humanized Mesenchymal Stem Cell Cultures for Bone Tissue Engineering: A Systematic Review with a Focus on Platelet Derivatives. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:552-569. [PMID: 28610481 DOI: 10.1089/ten.teb.2017.0093] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fetal bovine serum (FBS) is the most commonly used supplement for ex vivo expansion of human mesenchymal stem cells (hMSCs) for bone tissue engineering applications. However, from a clinical standpoint, it is important to substitute animal-derived products according to current good manufacturing practice (cGMP) guidelines. Humanized alternatives to FBS include three categories of products: human serum (HS), human platelet derivatives (HPDs)-including platelet lysate (PL) or platelet releasate (PR), produced by freeze/thawing or chemical activation of platelet concentrates, respectively, and chemically defined media (serum-free) (CDM). In this systematic literature review, the in vitro and in vivo osteogenic potential of hMSCs expanded in humanized (HS-, HPD-, or CDM-supplemented) media versus hMSCs expanded in FBS-supplemented media, was compared. In addition, PL and PR were compared in terms of their growth factor (GF)/cytokine-content and cell-culture efficacy. When using either 10-20% autologous or pooled HS, 3-10% pooled HPDs or CDM supplemented with GFs, in comparison with 10-20% FBS, a majority of studies reported similar or superior in vitro proliferation and osteogenic differentiation, and in vivo bone formation in ectopic or orthotopic rodent models. Moreover, a trend for higher GF content was observed in PL versus PR, although evidence for cell culture efficacy is limited. In summary, humanized supplements seem at least equally effective as FBS for hMSC expansion and osteogenic differentiation. Although pooled HPDs appear to be the most favorable supplement for large-scale hMSC expansion, further efforts are needed to standardize the preparation and composition of these products in compliance with cGMP standards.
Collapse
Affiliation(s)
- Siddharth Shanbhag
- 1 Department of Clinical Dentistry, Centre for Clinical Dental Research, University of Bergen , Bergen, Norway
| | - Andreas Stavropoulos
- 2 Department of Periodontology, Faculty of Odontology, Malmö University , Malmö, Sweden
| | - Salwa Suliman
- 1 Department of Clinical Dentistry, Centre for Clinical Dental Research, University of Bergen , Bergen, Norway
| | - Tor Hervig
- 3 Department of Immunology and Transfusion Medicine, Haukeland University Hospital , Bergen, Norway
| | - Kamal Mustafa
- 1 Department of Clinical Dentistry, Centre for Clinical Dental Research, University of Bergen , Bergen, Norway
| |
Collapse
|
75
|
Jurado M, De La Mata C, Ruiz-García A, López-Fernández E, Espinosa O, Remigia MJ, Moratalla L, Goterris R, García-Martín P, Ruiz-Cabello F, Garzón S, Pascual MJ, Espigado I, Solano C. Adipose tissue-derived mesenchymal stromal cells as part of therapy for chronic graft-versus-host disease: A phase I/II study. Cytotherapy 2017; 19:927-936. [PMID: 28662983 DOI: 10.1016/j.jcyt.2017.05.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 04/21/2017] [Accepted: 05/08/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS Despite the efficacy of allogeneic hematopoietic stem cell transplantation (allo-HSCT), the procedure is still associated with high toxicity in patients with refractory graft-versus-host disease (GvHD). Mesenchymal stromal cells (MSCs) are a new mode of therapy in the context of allo-HSCT. The objective of this study was to evaluate the safety and feasibility of the use of adipose tissue-derived MSCs (AT-MSCs) in patients with chronic GvHD. METHODS Fourteen patients with moderate (n = 7) or severe (n = 7) chronic GvHD received 1 × 106/kg (group A, n = 9) or 3 × 106/kg (group B, n = 5) AT-MSCs with cyclosporine and prednisone as first-line therapy. RESULTS Ten of the 14 patients were able to continue under the protocol: 80% were in complete remission, and 100% were off of steroids at week 56. The remaining 4 patients either worsened from chronic GvHD (n = 3) or abandoned the study (n = 1). At the end of the study, 11 of 14 patients are alive (overall survival 71.4%, median survival of 45.3 weeks). No suspected unexpected serious adverse reactions occurred during the trial. Neither relapse of underlying disease nor mortality due to infection was observed in this cohort. Biological studies showed increased CD19, CD4 and tumor necrosis factor-α with a temporary decrease in natural killer cells. DISCUSSION AT-MSCs, in combination with immunosuppressive therapy, may be considered feasible and safe and likely would have an impact on the course of chronic GvHD. More studies are warranted to understand the potential benefits of AT-MSCs in these patients.
Collapse
Affiliation(s)
- Manuel Jurado
- Department of Hematology, Complejo Hospitalario Universitario, Granada, Spain; Genyo Pfizer, Universidad de Granada, Junta de Andalucía, Centre for Genomics and Oncological Research (GENYO), Granada, Spain.
| | - Claudia De La Mata
- Department of Hematology, Complejo Hospitalario Universitario, Granada, Spain
| | - Antonio Ruiz-García
- Cellular manufacturing Unit, Instituto de Investigación Biosanitaria (IBS), Complejo Hospitalario Universitario, Granada, Spain
| | | | - Olga Espinosa
- Cellular manufacturing Unit, Instituto de Investigación Biosanitaria (IBS), Complejo Hospitalario Universitario, Granada, Spain
| | | | - Lucía Moratalla
- Department of Hematology, Complejo Hospitalario Universitario, Granada, Spain
| | - Rosa Goterris
- Department of Hematology, Hospital Clínico, Valencia, Spain
| | | | | | | | | | | | - Carlos Solano
- Department of Hematology, Hospital Clínico, Valencia, Spain; School of Medicine, University of Valencia, Spain
| |
Collapse
|
76
|
Shearer JJ, Figueiredo Neto M, Umbaugh CS, Figueiredo ML. In Vivo Exposure to Inorganic Arsenic Alters Differentiation-Specific Gene Expression of Adipose-Derived Mesenchymal Stem/Stromal Cells in C57BL/6J Mouse Model. Toxicol Sci 2017; 157:172-182. [PMID: 28206643 PMCID: PMC5837658 DOI: 10.1093/toxsci/kfx026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The number of mesenchymal stem cell (MSC) therapeutic modalities has grown in recent years. Adipose-derived mesenchymal stem/stromal cells (ASCs) can be isolated and expanded relatively easily as compared with their bone-marrow counterparts, making them a particularly promising source of MSCs. And although the biological mechanisms surrounding ASCs are actively being investigated, little is known about the effects that in vivo environmental exposures might have on their ability to properly differentiate. Therefore, we hypothesized that ASCs isolated from mice exposed to inorganic arsenic (iAs) would have an altered response towards adipogenic, osteogenic, and/or chondrogenic differentiation. To test this hypothesis, C57BL/6J male mice were provided drinking water containing 0, 300, or 1000 ppb iAs. ASCs were then isolated and differentiated, which was assessed by immunocytochemistry and real-time quantitative PCR (RT-qPCR). Our results showed that total urinary arsenic equilibrated within 1 week of exposure to iAs and was maintained throughout the study. ASCs isolated from each exposure group maintained differentiation capabilities for each lineage. The magnitude of differentiation-specific gene expression, however, appeared to be concentration dependent. For osteogenesis and chondrogenesis, differentiation-specific gene expression decreased, whereas adipogenesis showed a biphasic response with an initial decrease followed by an increase in adipogenic-related gene expression following iAs exposure. These results suggest that the level in which differentiation-specific genes are induced within these stromal cells might be sensitive to environmental contaminants. These findings highlight the need to take into account potential environmental exposures prior to selecting stromal cell donors, so ASCs can achieve optimal efficiency in regenerative therapy applications.
Collapse
Affiliation(s)
- Joseph J. Shearer
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana 47907
| | - Manoel Figueiredo Neto
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana 47907
| | - C. Samuel Umbaugh
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana 47907
| | - Marxa L. Figueiredo
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana 47907
| |
Collapse
|
77
|
Carty F, Mahon BP, English K. The influence of macrophages on mesenchymal stromal cell therapy: passive or aggressive agents? Clin Exp Immunol 2017; 188:1-11. [PMID: 28108980 DOI: 10.1111/cei.12929] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 01/16/2017] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stromal cells (MSC) have emerged as promising cell therapies for multiple conditions based on demonstrations of their potent immunomodulatory and regenerative capacities in models of inflammatory disease. Understanding the effects of MSC on T cells has dominated the majority of work carried out in this field to date; recently, however, a number of studies have shown that the therapeutic effect of MSC requires the presence of macrophages. It is timely to review the mechanisms and manner by which MSC modulate macrophage populations in order to design more effective MSC therapies and clinical studies. A complex cross-talk exists through which MSC and macrophages communicate, a communication that is not controlled exclusively by MSC. Here, we examine the evidence that suggests that MSC not only respond to inflammatory macrophages and adjust their secretome accordingly, but also that macrophages respond to encounters with MSC, creating a feedback loop which contributes to the immune regulation observed following MSC therapy. Future studies examining the effects of MSC on macrophages should consider the antagonistic role that macrophages play in this exchange.
Collapse
Affiliation(s)
- F Carty
- Institute of Immunology, Department of Biology, Maynooth University, Maynooth, County Kildare, Ireland
| | - B P Mahon
- Institute of Immunology, Department of Biology, Maynooth University, Maynooth, County Kildare, Ireland
| | - K English
- Institute of Immunology, Department of Biology, Maynooth University, Maynooth, County Kildare, Ireland
| |
Collapse
|