101
|
Wang S, Zhu R, Li H, Li J, Han Q, Zhao RC. Mesenchymal stem cells and immune disorders: from basic science to clinical transition. Front Med 2019; 13:138-151. [PMID: 30062557 DOI: 10.1007/s11684-018-0627-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/12/2017] [Indexed: 02/08/2023]
Abstract
As a promising candidate seed cell type in regenerative medicine, mesenchymal stem cells (MSCs) have attracted considerable attention. The unique capacity of MSCs to exert a regulatory effect on immunity in an autologous/allergenic manner makes them an attractive therapeutic cell type for immune disorders. In this review, we discussed the current knowledge of and advances in MSCs, including its basic biological properties, i.e., multilineage differentiation, secretome, and immunomodulation. Specifically, on the basis of our previous work, we proposed three new concepts of MSCs, i.e., "subtotipotent stem cell" hypothesis, MSC system, and "Yin and Yang" balance of MSC regulation, which may bring new insights into our understanding of MSCs. Furthermore, we analyzed data from the Clinical Trials database ( http://clinicaltrials.gov ) on registered clinical trials using MSCs to treat a variety of immune diseases, such as graft-versus-host disease, systemic lupus erythematosus, and multiple sclerosis. In addition, we highlighted MSC clinical trials in China and discussed the challenges and future directions in the field of MSC clinical application.
Collapse
Affiliation(s)
- Shihua Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Rongjia Zhu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Hongling Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Jing Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Qin Han
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
- Peking Union Medical College Hospital, Beijing, 100005, China.
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China.
| |
Collapse
|
102
|
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: 291] [Impact Index Per Article: 48.5] [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
|
103
|
Fisher SA, Cutler A, Doree C, Brunskill SJ, Stanworth SJ, Navarrete C, Girdlestone J, Cochrane Haematological Malignancies Group. Mesenchymal stromal cells as treatment or prophylaxis for acute or chronic graft-versus-host disease in haematopoietic stem cell transplant (HSCT) recipients with a haematological condition. Cochrane Database Syst Rev 2019; 1:CD009768. [PMID: 30697701 PMCID: PMC6353308 DOI: 10.1002/14651858.cd009768.pub2] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Recipients of allogeneic haematopoietic stem cell transplants (HSCT) can develop acute or chronic, or both forms of graft-versus-host disease (a/cGvHD), whereby immune cells of the donor attack host tissues. Steroids are the primary treatment, but patients with severe, refractory disease have limited options and a poor prognosis. Mesenchymal stromal cells (MSCs) exhibit immunosuppressive properties and are being tested in clinical trials for their safety and efficacy in treating many immune-mediated disorders. GvHD is one of the first areas in which MSCs were clinically applied, and it is important that the accumulating evidence is systematically reviewed to assess whether their use is favoured. OBJECTIVES To determine the evidence for the safety and efficacy of MSCs for treating immune-mediated inflammation post-transplantation of haematopoietic stem cells. SEARCH METHODS We searched for randomised controlled trials (RCTs) in the Cochrane Central Register of Controlled Trials (CENTRAL, the Cochrane Library 2018, Issue 12), MEDLINE (from 1946), Embase (from 1974), CINAHL (from 1937), Web of Science: Conference Proceedings Citation Index-Science (CPCI-S) (from 1990) and ongoing trial databases to 6 December 2018. No constraints were placed on language or publication status. SELECTION CRITERIA We included RCTs of participants with a haematological condition who have undergone an HSCT as treatment for their condition and were randomised to MSCs (intervention arm) or no MSCs (comparator arm), to prevent or treat GvHD. We also included RCTs which compared different doses of MSCs or MSCs of different sources (e.g. bone marrow versus cord). We included MSCs co-transplanted with haematopoietic stem cells as well as MSCs administered post-transplantation of haematopoietic stem cells. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane.We employed a random-effects model for all analyses due to expected clinical heterogeneity arising from differences in participant characteristics and interventions. MAIN RESULTS We identified 12 completed RCTs (879 participants), and 13 ongoing trials (1532 enrolled participants planned). Of 12 completed trials, 10 compared MSCs versus no MSCs and two compared different doses of MSCs. One trial was in people with thalassaemia major, the remaining trials were for haematological malignancies. Seven trials administered MSCs to prevent GvHD, whereas five trials gave MSCs to treat GvHD.In the comparison of MSCs with no MSCs, cells were administered at a dose of between 105 and 107 cells/kg in either a single dose (six trials) or in multiple doses (four trials) over a period of three days to four months. The dose-comparison trials compared 2 x 106 cells/kg with 8 x 106 cells/kg in two infusions, or 1 x 106 cells/kg with 3 x 106 cells/kg in a single infusion.The median duration of follow-up in seven trials which administered MSCs prophylactically ranged from 10 to 60 months. In three trials of MSCs as treatment for aGvHD, participants were followed up for 90 or 100 days. In two trials of MSCs as treatment for cGvHD, the mean duration of follow-up was 13.4 months (MSC group) and 23.6 months (control group) in one trial, and 56 weeks in the second trial. Five trials included adults only, six trials included adults and children, and one trial included children only. In eight trials which reported the gender distribution, the percentage of females ranged from 20% to 59% (median 35.8%).The overall quality of the included studies was low: randomisation methods were poorly reported and several of the included studies were subject to a high risk of performance bias and reporting bias. One trial which started in 2008 has not been published and the progress of this trial in unknown, leading to potential publication bias. The quality of evidence was therefore low or very low for all outcomes due to a high risk of bias as well as imprecision due to the low number of overall participants, and in some cases evidence based on a single study. We found that MSCs may make little or no difference in the risk of all-cause mortality in either prophylactic trials (HR 0.85, 95% CI 0.50 to 1.42; participants = 301; studies = 5; I2 = 34% ; low-quality evidence) or therapeutic trials (HR 1.12, 95% CI 0.80 to 1.56; participants = 244; studies = 1; very low-quality evidence), and no difference in the risk of relapse of malignant disease (prophylactic trials: RR 1.08, 95% CI 0.73 to 1.59; participants = 323; studies = 6; I2 = 0%; low-quality evidence) compared with no MSCs. MSCs were well-tolerated, no infusion-related toxicity or ectopic tissue formation was reported. No study reported health-related quality of life. In prophylactic trials, MSCs may reduce the risk of chronic GvHD (RR 0.66, 95% CI 0.49 to 0.89; participants = 283; studies = 6; I2 = 0%; low-quality evidence). This means that only 310 (95% CI 230 to 418) in every 1000 patients in the MSC arm are expected to develop chronic GvHD compared to 469 in the control arm. However, MSCs may make little or no difference to the risk of aGvHD (RR 0.86, 95% CI 0.63 to 1.17; participants = 247; studies = 6; I2 = 0%; low-quality evidence). In GvHD therapeutic trials, we are very uncertain whether MSCs improve complete response of either aGvHD (RR 1.16, 95% CI 0.79 to 1.70, participants = 260, studies = 1; very low-quality evidence) or cGvHD (RR 5.00, 95%CI 0.75 to 33.21, participants = 40, studies = 1; very low-quality evidence).In two trials which compared different doses of MSCs, we found no evidence of any differences in outcomes. AUTHORS' CONCLUSIONS MSCs are an area of intense research activity, and an increasing number of trials have been undertaken or are planned. Despite a number of reports of positive outcomes from the use of MSCs for treating acute GvHD, the evidence to date from RCTs has not supported the conclusion that they are an effective therapy. There is low-quality evidence that MSCs may reduce the risk of cGvHD. New trial evidence will be incorporated into future updates of this review, which may better establish a role for MSCs in the prevention or treatment of GvHD.
Collapse
Affiliation(s)
- Sheila A Fisher
- NHS Blood and TransplantSystematic Review InitiativeLevel 2, John Radcliffe HospitalHeadingtonOxfordOxonUKOX3 9BQ
| | - Antony Cutler
- NHS Blood and TransplantHistocompatibility & Immunogenetics Research GroupLondonUK
| | - Carolyn Doree
- NHS Blood and TransplantSystematic Review InitiativeLevel 2, John Radcliffe HospitalHeadingtonOxfordOxonUKOX3 9BQ
| | - Susan J Brunskill
- NHS Blood and TransplantSystematic Review InitiativeLevel 2, John Radcliffe HospitalHeadingtonOxfordOxonUKOX3 9BQ
| | - Simon J Stanworth
- Oxford University Hospitals NHS Foundation Trust and University of OxfordNational Institute for Health Research (NIHR) Oxford Biomedical Research CentreJohn Radcliffe Hospital, Headley WayHeadingtonOxfordUKOX3 9BQ
| | | | - John Girdlestone
- University College LondonDivision of Infection and ImmunityLondonUK
- NHS Blood and TransplantStem Cells and ImmunotherapiesHeadley WayOxfordUKOX3 9BQ
| | | |
Collapse
|
104
|
Jitschin R, Böttcher M, Saul D, Lukassen S, Bruns H, Loschinski R, Ekici AB, Reis A, Mackensen A, Mougiakakos D. Inflammation-induced glycolytic switch controls suppressivity of mesenchymal stem cells via STAT1 glycosylation. Leukemia 2019; 33:1783-1796. [PMID: 30679801 DOI: 10.1038/s41375-018-0376-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/19/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) represent key contributors to tissue homeostasis and promising therapeutics for hyperinflammatory conditions including graft-versus-host disease. Their immunomodulatory effects are controlled by microenvironmental signals. The MSCs' functional response towards inflammatory cues is known as MSC-"licensing" and includes indoleamine 2,3-dioxygenase (IDO) upregulation. MSCs use tryptophan-depleting IDO to suppress T-cells. Increasing evidence suggests that several functions are (co-)determined by the cells' metabolic commitment. MSCs are capable of both, high levels of glycolysis and of oxidative phosphorylation. Although several studies have addressed alterations of the immune regulatory phenotype elicited by inflammatory priming metabolic mechanisms controlling this process remain unknown. We demonstrate that inflammatory MSC-licensing causes metabolic shifts including enhanced glycolysis and increased fatty acid oxidation. Yet, only interfering with glycolysis impacts IDO upregulation and impedes T-cell-suppressivity. We identified the Janus kinase (JAK)/signal transducer and activator of transcription (STAT)1 pathway as a regulator of both glycolysis and IDO, and show that enhanced glucose turnover is linked to abundant STAT1 glycosylation. Inhibiting the responsible O-acetylglucosamine (O-GlcNAc) transferase abolishes STAT1 activity together with IDO upregulation. Our data suggest that STAT1-O-GlcNAcylation increases its stability towards degradation thus sustaining downstream effects. This pathway could represent a target for interventions aiming to enhance the MSCs' immunoregulatory potency.
Collapse
Affiliation(s)
- R Jitschin
- Department of Medicine 5 for Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - M Böttcher
- Department of Medicine 5 for Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - D Saul
- Department of Medicine 5 for Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - S Lukassen
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - H Bruns
- Department of Medicine 5 for Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - R Loschinski
- Department of Medicine 5 for Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - A B Ekici
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - A Reis
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - A Mackensen
- Department of Medicine 5 for Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - D Mougiakakos
- Department of Medicine 5 for Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
| |
Collapse
|
105
|
Hamilton BK. Current approaches to prevent and treat GVHD after allogeneic stem cell transplantation. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2018; 2018:228-235. [PMID: 30504315 PMCID: PMC6246030 DOI: 10.1182/asheducation-2018.1.228] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Allogeneic hematopoietic cell transplantation is a potentially curative therapy for many malignant and nonmalignant hematologic diseases. Graft-versus-host disease (GVHD) is a common complication after transplantation and remains a major cause of morbidity and mortality, limiting the success of a potentially curative transplant. This paper reviews the current and emerging strategies in GVHD prevention and treatment. New insights are leading the way to the development of novel targeted approaches to minimize the risk of disease relapse and infection. Continued collaborative efforts to conduct high-quality, multicenter clinical trials with standard end points and risk stratification are needed to determine the optimal approach to minimize GVHD and limit toxicities.
Collapse
Affiliation(s)
- Betty Ky Hamilton
- Blood and Marrow Transplant Program, Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
106
|
van Hoeven V, Munneke JM, Cornelissen AS, Omar SZ, Spruit MJ, Kleijer M, Bernink JH, Blom B, Voermans C, Hazenberg MD. Mesenchymal Stromal Cells Stimulate the Proliferation and IL-22 Production of Group 3 Innate Lymphoid Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:1165-1173. [PMID: 29980610 DOI: 10.4049/jimmunol.1700901] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 06/07/2018] [Indexed: 12/13/2022]
Abstract
Infusion of mesenchymal stromal cells (MSCs) is a promising and increasingly applied therapy for patients who suffer from a variety of inflammatory diseases, including graft-versus-host disease (GvHD), a common and life-threatening complication after allogeneic hematopoietic stem cell transplantation. The therapeutic effect of MSCs is mainly ascribed to their ability to suppress T cells and to support tissue repair. However, clinical response rates in patients with GvHD are limited to 50%, and the determinants for MSC responsiveness are unknown. We recently reported that high frequencies of activated group 3 innate lymphoid cells (ILC3s) before and after allogeneic hematopoietic stem cell transplantation were associated with a lower risk of GvHD. This may be related to IL-22 production by ILC3s, a cytokine important for intestinal epithelial cell homeostasis. In this study, we investigated whether ILC3s may contribute to the therapeutic effect of MSCs by studying the interaction between MSCs and ILC3s in vitro. ILC3s isolated from human tonsils were cocultured with human bone marrow-derived MSCs for 5 d in the presence of IL-2. Coculture with MSCs enhanced the proliferation and IL-22 production of ILC3s. Reciprocally, ILC3s promoted ICAM-1 and VCAM-1 expression on MSCs. For both directions, the activation was mainly mediated by cell-cell contact and by MSC-derived IL-7 and likely by aryl hydrocarbon receptor ligands. Thus, in addition to inhibiting the proliferation of alloreactive T cells, MSCs also promote the expansion and IL-22 production of ILC3s, which may contribute to healthy homeostasis and wound repair in the treatment of various inflammatory conditions in the intestine, including GvHD.
Collapse
Affiliation(s)
- Vera van Hoeven
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| | - J Marius Munneke
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| | - Anne S Cornelissen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, 1066 CX Amsterdam, the Netherlands
| | - Said Z Omar
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| | - Melchior J Spruit
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| | - Marion Kleijer
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, 1066 CX Amsterdam, the Netherlands
| | - Jochem H Bernink
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| | - Bianca Blom
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| | - Carlijn Voermans
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, 1066 CX Amsterdam, the Netherlands
| | - Mette D Hazenberg
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands;
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; and
| |
Collapse
|
107
|
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
|
108
|
Thielen FW, Blommestein HM, Oosten LEM, Calkoen FG, Lankester AC, Zwaginga JJ, Le Blanc K, Redondo A, Sánchez-Guijo F, Algeri M, Locatelli F, Fibbe WE, Uyl-de Groot CA. Second-line treatment for acute graft-versus-host disease with mesenchymal stromal cells: A decision model. Eur J Haematol 2018; 101:676-683. [PMID: 30084111 DOI: 10.1111/ejh.13158] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 01/09/2023]
Abstract
OBJECTIVE No standard second-line treatment exists for acute graft-versus-host disease steroid-refractory (SR-aGvHD), and long-term outcomes remain poor. Mesenchymal stromal cells (MSCs) have been evaluated as treatment, but no disease model (DM) exists that integrates and extrapolates currently available evidence. The aim of this study was to develop such a DM to describe the natural history of SR-aGvHD and to predict long-term outcomes. METHOD The DM was developed in collaboration with experts in haematology-oncology. Subsequently, a model simulation was run. Input parameters for transition and survival estimates were informed by published data of clinical trials on MSC treatment for SR-aGvHD. Parametric distributions were used to estimate long-term survival rates after MSCs. RESULTS The newly developed DM is a cohort model that consists of eight health states. For the model simulation, we obtained data on 327 patients from 14 published phase II trials. Due to limited evidence, DM structure was simplified and several assumptions had to be made. Median overall survival was 3.2 years for complete response and 0.5 years for no complete response. CONCLUSION The DM provides a comprehensive overview on the second-line treatment pathway for aGvHD and enables long-term predictions that can be used to perform a cost-effectiveness analysis comparing any treatment for SR-aGvHD.
Collapse
Affiliation(s)
- Frederick W Thielen
- Erasmus School of Health Policy and Management, Erasmus University, Rotterdam, The Netherlands
| | - Hedwig M Blommestein
- Erasmus School of Health Policy and Management, Erasmus University, Rotterdam, The Netherlands
- Comprehensive Cancer Organisation, Utrecht, The Netherlands
| | - Liesbeth E M Oosten
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Friso G Calkoen
- Department of Paediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Arjan C Lankester
- Department of Paediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap J Zwaginga
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
- Center for Clinical Transfusion Research, Sanquin Research, Leiden, The Netherlands
| | - Katarina Le Blanc
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Alba Redondo
- IBSAL-Hospital Universitario de Salamanca, Salamanca, Spain
| | | | | | | | - Wim E Fibbe
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Carin A Uyl-de Groot
- Erasmus School of Health Policy and Management, Erasmus University, Rotterdam, The Netherlands
| |
Collapse
|
109
|
Torres Crigna A, Daniele C, Gamez C, Medina Balbuena S, Pastene DO, Nardozi D, Brenna C, Yard B, Gretz N, Bieback K. Stem/Stromal Cells for Treatment of Kidney Injuries With Focus on Preclinical Models. Front Med (Lausanne) 2018; 5:179. [PMID: 29963554 PMCID: PMC6013716 DOI: 10.3389/fmed.2018.00179] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/24/2018] [Indexed: 12/18/2022] Open
Abstract
Within the last years, the use of stem cells (embryonic, induced pluripotent stem cells, or hematopoietic stem cells), Progenitor cells (e.g., endothelial progenitor cells), and most intensely mesenchymal stromal cells (MSC) has emerged as a promising cell-based therapy for several diseases including nephropathy. For patients with end-stage renal disease (ESRD), dialysis or finally organ transplantation are the only therapeutic modalities available. Since ESRD is associated with a high healthcare expenditure, MSC therapy represents an innovative approach. In a variety of preclinical and clinical studies, MSC have shown to exert renoprotective properties, mediated mainly by paracrine effects, immunomodulation, regulation of inflammation, secretion of several trophic factors, and possibly differentiation to renal precursors. However, studies are highly diverse; thus, knowledge is still limited regarding the exact mode of action, source of MSC in comparison to other stem cell types, administration route and dose, tracking of cells and documentation of therapeutic efficacy by new imaging techniques and tissue visualization. The aim of this review is to provide a summary of published studies of stem cell therapy in acute and chronic kidney injury, diabetic nephropathy, polycystic kidney disease, and kidney transplantation. Preclinical studies with allogeneic or xenogeneic cell therapy were first addressed, followed by a summary of clinical trials carried out with autologous or allogeneic hMSC. Studies were analyzed with respect to source of cell type, mechanism of action etc.
Collapse
Affiliation(s)
- Adriana Torres Crigna
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Cristina Daniele
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Carolina Gamez
- Department for Experimental Orthopaedics and Trauma Surgery, Medical Faculty Mannheim, Orthopaedic and Trauma Surgery Centre (OUZ), Heidelberg University, Mannheim, Germany
| | - Sara Medina Balbuena
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Diego O. Pastene
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Daniela Nardozi
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Cinzia Brenna
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Benito Yard
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Norbert Gretz
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Karen Bieback
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| |
Collapse
|
110
|
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
|
111
|
Ringden O, Baygan A, Remberger M, Gustafsson B, Winiarski J, Khoein B, Moll G, Klingspor L, Westgren M, Sadeghi B. Placenta-Derived Decidua Stromal Cells for Treatment of Severe Acute Graft-Versus-Host Disease. Stem Cells Transl Med 2018. [PMID: 29533533 PMCID: PMC5866941 DOI: 10.1002/sctm.17-0167] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Severe acute graft‐versus‐host disease (GVHD) is a life‐threatening complication after allogeneic hematopoietic stem cell transplantation (HSCT). The placenta protects the fetus from the mother's immune system. We evaluated placenta‐derived decidua stromal cells (DSCs), which differ from bone marrow mesenchymal stromal cells (BM‐MSCs), as a treatment for severe acute GVHD. DSCs were obtained from term placentas. The DSCs were given to 38 patients with severe acute GVHD; 25 were steroid refractory (SR). DSCs were thawed and infused in buffer supplemented with either 10% AB plasma (group 1, n = 17), or 5% albumin (group 2, n = 21). The viability of cells was higher when thawed in albumin rather than AB plasma (p < .001). Group 1 received a higher cell dose (p < .001), cells of lower passage number (p < .001), and fewer infusions (p = .002) than group 2. The GVHD response (no/partial/complete) was 7/5/5 in group 1 and 0/10/11 in group 2 (p = .01). One‐year survival in the two groups was 47% (95% confidence interval [CI] 23–68) and 76% (95% CI 51–89), respectively (p = .016). For the SR patients, 1‐year survival was 73% (95% CI 37–90) in SR group 2 (n = 11), which was better than 31% (95% CI 11–54) in SR group 1 (n = 13; p = .02), 20% (95% CI 5–42) in BM‐MSC treated (n = 15; p = .0015), and 3% (95% CI 0–14) in historic controls (n = 32; p < .001). DSCs are a promising new treatment for severe acute GVHD. Prospective randomized trials are needed for evaluation of efficacy. (Clinical trial NCT‐02172937.) stemcellstranslationalmedicine2018;7:325–332
Collapse
Affiliation(s)
- Olle Ringden
- Translational Cell Therapy Research (TCR), Department of Laboratory Medicine
| | - Arjang Baygan
- Translational Cell Therapy Research (TCR), Department of Laboratory Medicine
| | - Mats Remberger
- Center for Allogeneic Stem Cell Transplantation, Department of Oncology and Pathology
| | - Britt Gustafsson
- Division of Pediatrics, Department of Clinical Intervention and Technology, CLINTEC
| | - Jacek Winiarski
- Division of Pediatrics, Department of Clinical Intervention and Technology, CLINTEC
| | - Bita Khoein
- Translational Cell Therapy Research (TCR), Department of Laboratory Medicine
| | - Guido Moll
- Berlin-Brandenburg Center and School for Regenerative Therapies (BCRT/BSRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Lena Klingspor
- Division of Clinical Microbiology, Department of Laboratory Medicine, CLINTEC, Karolinska Institutet and Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Magnus Westgren
- Division of Obstetrics and Gynaecology, Department of Clinical Intervention and Technology, CLINTEC, Karolinska Institutet and Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Behnam Sadeghi
- Translational Cell Therapy Research (TCR), Department of Laboratory Medicine
| |
Collapse
|
112
|
Keto J, Kaartinen T, Salmenniemi U, Castrén J, Partanen J, Hänninen A, Korhonen M, Lähteenmäki K, Itälä-Remes M, Nystedt J. Immunomonitoring of MSC-Treated GvHD Patients Reveals Only Moderate Potential for Response Prediction but Indicates Treatment Safety. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018. [PMID: 29516024 PMCID: PMC5834657 DOI: 10.1016/j.omtm.2018.02.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Mesenchymal stromal cells (MSCs) are used as salvage therapy to treat steroid-refractory acute graft-versus-host disease (aGvHD). We studied the immunological response to MSC treatment in 16 aGvHD patients by assessing lymphocyte profiles and three proposed aGvHD serum markers during the MSC treatment. Surprisingly, there were no obvious differences in the lymphocyte profiles between the responders and non-responders. The numbers of T, B, and NK cells were below the normal reference interval in all patients. CD4+ T helper (Th) cell levels remained particularly low throughout the follow-up period. The relative proportion of Th1 cells decreased, while regulatory T cells remained unaltered, and only very few Th2 and Th17 cells could be detected. Serum concentrations of regenerating islet-derived protein 3-alpha, cytokeratin-18 fragments (CK18F), and elafin were significantly elevated in patient samples compared with healthy controls, but only CK18F showed any potential in the prediction of patients’ response to MSCs. No obvious markers for MSC therapy response were revealed in this study, but the results suggest that allogeneic MSCs do not provoke overt T cell-mediated immune responses at least in immunosuppressed aGvHD patients. The results advocate for the safety of MSC therapy and bring new insights in MSC immunomodulation mechanisms.
Collapse
Affiliation(s)
- Joni Keto
- Finnish Red Cross Blood Service, Kivihaantie 7, FI-00310 Helsinki, Finland
| | - Tanja Kaartinen
- Finnish Red Cross Blood Service, Kivihaantie 7, FI-00310 Helsinki, Finland
| | - Urpu Salmenniemi
- Division of Medicine, Department of Hematology and Stem Cell Transplantation Unit, Turku University Hospital, Hämeentie 11, FI-20521 Turku, Finland
| | - Johanna Castrén
- Finnish Red Cross Blood Service, Kivihaantie 7, FI-00310 Helsinki, Finland
| | - Jukka Partanen
- Finnish Red Cross Blood Service, Kivihaantie 7, FI-00310 Helsinki, Finland
| | - Arno Hänninen
- Department of Medical Microbiology and Immunology, University of Turku, Kiinamyllynkatu 13, FI-20520 Turku, Finland
| | - Matti Korhonen
- Finnish Red Cross Blood Service, Kivihaantie 7, FI-00310 Helsinki, Finland
| | | | - Maija Itälä-Remes
- Division of Medicine, Department of Hematology and Stem Cell Transplantation Unit, Turku University Hospital, Hämeentie 11, FI-20521 Turku, Finland
| | - Johanna Nystedt
- Finnish Red Cross Blood Service, Kivihaantie 7, FI-00310 Helsinki, Finland
| |
Collapse
|
113
|
Ringdén O, Labopin M, Sadeghi B, Mailhol A, Beelen D, Fløisand Y, Ghavamzadeh A, Finke J, Ehninger G, Volin L, Socié G, Kröger N, Stuhler G, Ganser A, Schmid C, Giebel S, Mohty M, Nagler A. What is the outcome in patients with acute leukaemia who survive severe acute graft-versus-host disease? J Intern Med 2018; 283:166-177. [PMID: 29027756 DOI: 10.1111/joim.12695] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Acute graft-versus-host disease (aGVHD) is a major complication of allogeneic haematopoietic stem cell transplantation (HSCT). With new promising therapies, survival may improve for severe aGVHD. OBJECTIVES We wanted to analyze the long-term outcome in patients who survive severe aGVHD. METHODS This study was a landmark analysis of 23 567 patients with acute Leukaemia who survived for more than 6 months after HSCT, 2002-2014. Patients alive after severe aGVHD (n = 1738) were compared to controls. RESULTS Patients with severe aGVHD had higher non-relapse mortality (NRM) and higher rate of extensive chronic GVHD (cGVHD) than the controls (P < 10-5 ). The probability of relapse was significantly lower in the severe aGVHD group, but Leukaemia-free survival (LFS) and overall survival were significantly lower than for the controls (P < 10-5 ). Five-year LFS in patients with severe aGVHD was 49%, as opposed to 61% in controls with no or mild GVHD and 59% in patients with moderate GVHD. CONCLUSIONS HSCT patients who survive severe aGVHD have higher risk of developing extensive cGVHD, a higher NRM, a lower relapse probability, and lower LFS than other HSCT patients. This study is a platform for outcome analysis in patients treated with novel therapies for acute GVHD.
Collapse
Affiliation(s)
- O Ringdén
- Division of Therapeutic Immunology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - M Labopin
- Hôpital Saint Antoine, Paris, France
| | - B Sadeghi
- Division of Therapeutic Immunology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - A Mailhol
- Hôpital Saint Antoine, Paris, France
| | - D Beelen
- University of Essen, Duisburg, Germany
| | - Y Fløisand
- Department of Hematology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - A Ghavamzadeh
- Shariati Hospital, Hematology-Oncology and BMT Research, Tehran, Iran
| | - J Finke
- Department of Medicine-Hematology, Oncology, University of Freiburg, Freiburg, Germany
| | - G Ehninger
- Medizinische Klinik und Poliklinik 1, Universitätsklinikum Dresden, Dresden, Germany
| | - L Volin
- Comprehensive Cancer Center, Stem Cell Transplantation Unit, Helsinki University Hospital, Helsinki, Finland
| | - G Socié
- Department of Hematology - BMT, Hopital St. Louis, Paris, France
| | - N Kröger
- Department of Stem cell Transplantation, University Hospital Eppendorf, Hamburg, Germany
| | - G Stuhler
- Deutsche Klinik für Diagnostik, KMT Zentrum, Wiesbaden, Germany
| | - A Ganser
- Medical University Hannover, Hannover, Germany
| | - C Schmid
- University of Munich, Munich, Germany
| | - S Giebel
- Department of Bone Marrow Transplantation and Oncohematology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - M Mohty
- Hôpital Saint Antoine, Paris, France
| | - A Nagler
- Chaim Sheba Medical Center, Tel-Hashomer, Israel
| |
Collapse
|
114
|
Xu T, Zhang Y, Chang P, Gong S, Shao L, Dong L. Mesenchymal stem cell-based therapy for radiation-induced lung injury. Stem Cell Res Ther 2018; 9:18. [PMID: 29386045 PMCID: PMC5793340 DOI: 10.1186/s13287-018-0776-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Since radiotherapy is widely used in managing thoracic tumors, physicians have begun to realize that radiation-induced lung injury (RILI) seriously limits the effects of radiotherapy. Unfortunately, there are still no effective methods for controlling RILI. Over the last few decades numerous studies have reported the beneficial effects of mesenchymal stem cells (MSCs) on tissue repair and regeneration. MSCs can not only differentiate into lung alveolar epithelial cells and secrete anti-inflammatory factors, but they also deliver some vehicles for gene therapy in repairing the injured lung, which provides new ideas for managing RILI. Thus, many scientists have attempted to manage RILI using MSC-based therapy. However, as a novel therapy MSCs still face various limitations. Herein, we shed light on the current understanding of MSC-based therapy for RILI, including the feasibility, molecular mechanisms, animal studies, and clinical research of MSC-based therapy for RILI. We also present an overview of RILI and MSCs.
Collapse
Affiliation(s)
- Tiankai Xu
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Yuyu Zhang
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Pengyu Chang
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Shouliang Gong
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130021, China.,Key Laboratory of Radiobiology, Ministry of health, School of Public Health, Jilin University, Changchun, 130021, China
| | - Lihong Shao
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Lihua Dong
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
115
|
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: 81] [Impact Index Per Article: 11.6] [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
|
116
|
Hill L, Alousi A, Kebriaei P, Mehta R, Rezvani K, Shpall E. New and emerging therapies for acute and chronic graft versus host disease. Ther Adv Hematol 2018; 9:21-46. [PMID: 29317998 PMCID: PMC5753923 DOI: 10.1177/2040620717741860] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/10/2017] [Indexed: 12/14/2022] Open
Abstract
Graft versus host disease (GVHD) remains a major cause of morbidity and mortality following allogeneic hematopoietic stem-cell transplantation (HSCT). Despite the use of prophylactic GVHD regimens, a significant proportion of transplant recipients will develop acute or chronic GVHD following HSCT. Corticosteroids are standard first-line therapy, but are only effective in roughly half of all cases with ~50% of patients going on to develop steroid-refractory disease, which increases the risk of nonrelapse mortality. While progress has been made with improvements in survival outcomes over time, corticosteroids are associated with significant toxicities, and many currently available salvage therapies are associated with increased immunosuppression, infectious complications, and potential loss of the graft versus leukemia (GVL) effect. Thus, there is an unmet need for development of newer treatment strategies for both acute and chronic GVHD to improve long-term post-transplant outcomes and quality of life for HSCT recipients. Here, we provide a concise review of major emerging therapies currently being studied in the treatment of acute and chronic GVHD.
Collapse
Affiliation(s)
- LaQuisa Hill
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Amin Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer, Houston, TX, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer, Houston, TX, USA
| | - Rohtesh Mehta
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer, Houston, TX, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer, Houston, TX, USA
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 0423, Houston, TX 77030-4000, USA
| |
Collapse
|
117
|
Fujii S, Miura Y, Fujishiro A, Shindo T, Shimazu Y, Hirai H, Tahara H, Takaori-Kondo A, Ichinohe T, Maekawa T. Graft-Versus-Host Disease Amelioration by Human Bone Marrow Mesenchymal Stromal/Stem Cell-Derived Extracellular Vesicles Is Associated with Peripheral Preservation of Naive T Cell Populations. Stem Cells 2017; 36:434-445. [PMID: 29239062 DOI: 10.1002/stem.2759] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/30/2017] [Accepted: 12/03/2017] [Indexed: 12/13/2022]
Abstract
A substantial proportion of patients with acute graft-versus-host disease (aGVHD) respond to cell therapy with culture-expanded human bone marrow mesenchymal stromal/stem cells (BM-MSCs). However, the mechanisms by which these cells can ameliorate aGVHD-associated complications remain to be clarified. We show here that BM-MSC-derived extracellular vesicles (EVs) recapitulated the therapeutic effects of BM-MSCs against aGVHD. Systemic infusion of human BM-MSC-derived EVs prolonged the survival of mice with aGVHD and reduced the pathologic damage in multiple GVHD-targeted organs. In EV-treated GVHD mice, CD4+ and CD8+ T cells were suppressed. Importantly, the ratio of CD62L-CD44+ to CD62L + CD44- T cells was decreased, suggesting that BM-MSC-derived EVs suppressed the functional differentiation of T cells from a naive to an effector phenotype. BM-MSC-derived EVs also preserved CD4 + CD25 + Foxp3+ regulatory T cell populations. In a culture of CD3/CD28-stimulated human peripheral blood mononuclear cells with BM-MSC-derived EVs, CD3+ T cell activation was suppressed. However, these cells were not suppressed in cultures with EVs derived from normal human dermal fibroblasts (NHDFs). NHDF-derived EVs did not ameliorate the clinical or pathological characteristics of aGVHD in mice, suggesting an immunoregulatory function unique to BM-MSC-derived EVs. Microarray analysis of microRNAs in BM-MSC-derived EVs versus NHDF-derived EVs showed upregulation of miR-125a-3p and downregulation of cell proliferative processes, as identified by Gene Ontology enrichment analysis. Collectively, our findings provide the first evidence that amelioration of aGVHD by therapeutic infusion of BM-MSC-derived EVs is associated with the preservation of circulating naive T cells, possibly due to the unique microRNA profiles of BM-MSC-derived EVs. Stem Cells 2018;36:434-445.
Collapse
Affiliation(s)
- Sumie Fujii
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan.,Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuo Miura
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan.,Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Aya Fujishiro
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan.,Division of Gastroenterology and Hematology, Department of Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Takero Shindo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yutaka Shimazu
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideyo Hirai
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
| | - Hidetoshi Tahara
- Department of Cellular and Molecular Biology, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Taira Maekawa
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
118
|
Cirillo M, Tan P, Sturm M, Cole C. Cellular Immunotherapy for Hematologic Malignancies: Beyond Bone Marrow Transplantation. Biol Blood Marrow Transplant 2017; 24:433-442. [PMID: 29102721 DOI: 10.1016/j.bbmt.2017.10.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/25/2017] [Indexed: 02/06/2023]
Abstract
Immunotherapy has changed treatment practices for many hematologic malignancies. Even in the current era of targeted therapy, chemotherapy remains the backbone of treatment for many hematologic malignancies, especially in acute leukemias, where relapse remains the major cause of mortality. Application of novel immunotherapies in hematology attempts to harness the killing power of the immune system against leukemia and lymphoma. Cellular immunotherapy is evolving rapidly for high-risk hematologic disorders. Recent advances include chimeric antigen-receptor T cells, mesenchymal stromal/stem cells, dendritic cell tumor vaccines, cytokine-induced killer cells, and virus-specific T cells. The advantages of nontransplantation cellular immunotherapy include suitability for patients for whom transplantation has failed or is contraindicated, and a potentially less-toxic treatment alternative to transplantation for relapsed/refractory patients. This review examines those emerging cellular immunotherapies that are changing treatment paradigms for patients with hematologic malignancies.
Collapse
Affiliation(s)
- Melita Cirillo
- Department of Haematology Cell and Tissue Therapies, Royal Perth Hospital, Perth, Western Australia, Australia.
| | - Peter Tan
- Department of Haematology Cell and Tissue Therapies, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Marian Sturm
- Department of Haematology Cell and Tissue Therapies, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Catherine Cole
- Department of Haematology Cell and Tissue Therapies, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
119
|
Stem cells in cardiovascular diseases: turning bad days into good ones. Drug Discov Today 2017; 22:1730-1739. [DOI: 10.1016/j.drudis.2017.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 06/28/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022]
|
120
|
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
|
121
|
Gustafsson B, Frisk P, Szakos A, Sadeghi B, Ringdén O, Frost BM. Successful treatment with placenta-derived decidual stromal cells in a pediatric patient with life-threatening acute gastrointestinal graft-versus-host disease. Pediatr Transplant 2017; 21. [PMID: 28612364 DOI: 10.1111/petr.12990] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2017] [Indexed: 12/14/2022]
Abstract
Severe aGvHD is a life-threatening complication after allogeneic HSCT. The GI tract is considered to play a key role in aGvHD, where the disease process can start and is one of the major target organs. Here, we present a case of a one-year-old child with a life-threatening GI-aGvHD stage IV, post-HSCT, resistant to steroids and MMF for 4 weeks. He was successfully treated with placenta-derived DSC.
Collapse
Affiliation(s)
- Britt Gustafsson
- Department of Clinical Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Women's and Children's Health, Pediatric Oncology, Children's University Hospital, Uppsala, Sweden
| | - Per Frisk
- Department of Women's and Children's Health, Pediatric Oncology, Children's University Hospital, Uppsala, Sweden.,Department of Women's and Children's Health, Uppsala University Hospital, Uppsala, Sweden
| | - Attilla Szakos
- Department of Pathology and Cytologi, Karolinska University Hospital, Stockholm, Sweden
| | - Behnam Sadeghi
- Department of Laboratory Medicine, Division of Therapeutic Immunology, Karolinska Institutet, Stockholm, Sweden
| | - Olle Ringdén
- Department of Laboratory Medicine, Division of Therapeutic Immunology, Karolinska Institutet, Stockholm, Sweden
| | - Britt-Marie Frost
- Department of Women's and Children's Health, Pediatric Oncology, Children's University Hospital, Uppsala, Sweden.,Department of Women's and Children's Health, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
122
|
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: 3.8] [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
|
123
|
Mattei A, Magalon J, Bertrand B, Philandrianos C, Veran J, Giovanni A. Cell therapy and vocal fold scarring. Eur Ann Otorhinolaryngol Head Neck Dis 2017; 134:339-345. [PMID: 28689790 DOI: 10.1016/j.anorl.2017.06.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Vocal fold microstructure is complex and can be affected by laryngeal microsurgery, inducing scarring that prevents mechanical uncoupling of epithelium and muscle, leading to vibration disorder and disabling dysphonia. Treatment options presently are few, and often without efficacy for vibration, having only an impact on volume to reduce glottal closure defect. The present review of the literature had two aims: (i) to report the current state of the literature on cell therapy in vocal fold scarring; and (ii) to analyze the therapeutic interest of the adipose-derived stromal vascular fraction in the existing therapeutic armamentarium. A PubMed® search conducted in September 2016 retrieved English or French-language original articles on the use of stem cells to treat vocal fold scarring. Twenty-seven articles published between 2003 and 2016 met the study selection criteria. Mesenchymal stem cells were most widely used, mainly derived from bone marrow or adipose tissue. Four studies were performed in vitro on fibroblasts, and 18 in vivo on animals. End-points comprised: (i) scar analysis (macro- and micro-scopic morphology, viscoelastic properties, extracellular matrix, fibroblasts); and (ii) assessment of stem cell survival and differentiation. The studies testified to the benefit of mesenchymal stem cells, and especially those of adipose derivation. The stromal vascular fraction exhibits properties that might improve results by facilitating production logistics.
Collapse
Affiliation(s)
- A Mattei
- Aix Marseille université, 13000 Marseille, France; Service d'oto-rhino-laryngologie et chirurgie cervicofaciale, La Conception, Assistance publique-Hôpitaux de Marseille, 147, boulevard Baille, 13005 Marseille, France.
| | - J Magalon
- VRCM Inserm UMR 1076, faculté de pharmacie de Marseille, Aix Marseille université, 27, boulevard Jean-Moulin, 13385 Marseille cedex 5, France; Inserm CBT-1409, laboratoire de culture et thérapie cellulaire, La Conception, Assistance publique-Hôpitaux de Marseille, 13005 Marseille, France
| | - B Bertrand
- Service de chirurgie plastique et réparatrice, La Conception, Assistance publique-Hôpitaux de Marseille, 13005 Marseille, France
| | - C Philandrianos
- Service de chirurgie plastique et réparatrice, La Conception, Assistance publique-Hôpitaux de Marseille, 13005 Marseille, France
| | - J Veran
- Inserm CBT-1409, laboratoire de culture et thérapie cellulaire, La Conception, Assistance publique-Hôpitaux de Marseille, 13005 Marseille, France
| | - A Giovanni
- Service d'oto-rhino-laryngologie et chirurgie cervicofaciale, La Conception, Assistance publique-Hôpitaux de Marseille, 147, boulevard Baille, 13005 Marseille, France; CNRS, laboratoire parole et langage, Aix Marseille université, 5, avenue Pasteur, 13100 Aix-en-Provence, France
| |
Collapse
|
124
|
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
|
125
|
Zocchi E, Hontecillas R, Leber A, Einerhand A, Carbo A, Bruzzone S, Tubau-Juni N, Philipson N, Zoccoli-Rodriguez V, Sturla L, Bassaganya-Riera J. Abscisic Acid: A Novel Nutraceutical for Glycemic Control. Front Nutr 2017; 4:24. [PMID: 28660193 PMCID: PMC5468461 DOI: 10.3389/fnut.2017.00024] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 05/19/2017] [Indexed: 01/03/2023] Open
Abstract
Abscisic acid is naturally present in fruits and vegetables, and it plays an important role in managing glucose homeostasis in humans. According to the latest U.S. dietary survey, about 92% of the population might have a deficient intake of ABA due to their deficient intake of fruits and vegetables. This review summarizes the in vitro, preclinical, mechanistic, and human translational findings obtained over the past 15 years in the study of the role of ABA in glycemic control. In 2007, dietary ABA was first reported to ameliorate glucose tolerance and obesity-related inflammation in mice. The most recent findings regarding the topic of ABA and its proposed receptor lanthionine synthetase C-like 2 in glycemic control and their interplay with insulin and glucagon-like peptide-1 suggest a major role for ABA in the physiological response to a glucose load in humans. Moreover, emerging evidence suggests that the ABA response might be dysfunctional in diabetic subjects. Follow on intervention studies in healthy individuals show that low-dose dietary ABA administration exerts a beneficial effect on the glycemia and insulinemia profiles after oral glucose load. These recent findings showing benefits in humans, together with extensive efficacy data in mouse models of diabetes and inflammatory disease, suggest the need for reference ABA values and its possible exploitation of the glycemia-lowering effects of ABA for preventative purposes. Larger clinical studies on healthy, prediabetic, and diabetic subjects are needed to determine whether addressing the widespread dietary ABA deficiency improves glucose control in humans.
Collapse
Affiliation(s)
- Elena Zocchi
- Department of Experimental Medicine, Section of Biochemistry and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Raquel Hontecillas
- BioTherapeutics Inc., Blacksburg, VA, United States.,Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, Blacksburg, VA, United States
| | - Andrew Leber
- BioTherapeutics Inc., Blacksburg, VA, United States
| | | | - Adria Carbo
- BioTherapeutics Inc., Blacksburg, VA, United States
| | - Santina Bruzzone
- Department of Experimental Medicine, Section of Biochemistry and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Nuria Tubau-Juni
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, Blacksburg, VA, United States
| | | | | | - Laura Sturla
- Department of Experimental Medicine, Section of Biochemistry and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Josep Bassaganya-Riera
- BioTherapeutics Inc., Blacksburg, VA, United States.,Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
126
|
Intravenous mesenchymal stromal cell therapy for inflammatory bowel disease: Lessons from the acute graft versus host disease experience. Cytotherapy 2017; 19:655-667. [PMID: 28433516 DOI: 10.1016/j.jcyt.2017.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 02/01/2017] [Accepted: 03/06/2017] [Indexed: 12/18/2022]
Abstract
Bone marrow-derived mesenchymal stromal cells (BMSCs) are primitive, supportive cells of the bone marrow with tri-lineage potential to differentiate into bone, cartilage, fat and muscle. These cells possess both in vitro and in vivo immunomodulatory and wound-healing properties. Several studies have demonstrated efficacy of intravenously administered BMSCs in treating acute graft-versus-host disease (GvHD). Use of intravenous (IV) BMSCs in inflammatory bowel diseases (IBD) in humans has been limited to small studies in adults, but results have been promising. There remain many unanswered questions regarding safety, tolerability, effectiveness and optimal use of BMSCs to treat IBD, particularly in immunocompromised patients. This article reviews the evidence for using BMSCs to treat acute GvHD and how this experience may inform the potential use of BMSCs as a treatment for IBD.
Collapse
|
127
|
Sugino N, Ichinohe T, Takaori-Kondo A, Maekawa T, Miura Y. Pharmacological targeting of bone marrow mesenchymal stromal/stem cells for the treatment of hematological disorders. Inflamm Regen 2017; 37:7. [PMID: 29259706 PMCID: PMC5725802 DOI: 10.1186/s41232-017-0038-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/20/2017] [Indexed: 01/16/2023] Open
Abstract
The therapeutic effects of mesenchymal stromal/stem cells (MSCs) are mainly based on three characteristics: immunomodulation, tissue regeneration, and hematopoietic support. Cell therapy using culture-expanded MSCs is effective in some intractable bone and hemato-immune disorders; however, its efficacy is limited. In this article, we review the previous efforts to improve the clinical outcomes of cell therapy using MSCs for such disorders. We describe pharmacological targeting of endogenous bone marrow-derived MSCs as a crucial quality-based intervention to establish more effective MSC-based therapies.
Collapse
Affiliation(s)
- Noriko Sugino
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan.,Department of Hematology/Oncology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507 Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 734-8553 Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology/Oncology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507 Japan
| | - Taira Maekawa
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Yasuo Miura
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan
| |
Collapse
|
128
|
Dotoli GM, De Santis GC, Orellana MD, de Lima Prata K, Caruso SR, Fernandes TR, Rensi Colturato VA, Kondo AT, Hamerschlak N, Simões BP, Covas DT. Mesenchymal stromal cell infusion to treat steroid-refractory acute GvHD III/IV after hematopoietic stem cell transplantation. Bone Marrow Transplant 2017; 52:859-862. [DOI: 10.1038/bmt.2017.35] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 01/12/2017] [Accepted: 01/27/2017] [Indexed: 02/06/2023]
|
129
|
Bouchlaka MN, Moffitt AB, Kim J, Kink JA, Bloom DD, Love C, Dave S, Hematti P, Capitini CM. Human Mesenchymal Stem Cell-Educated Macrophages Are a Distinct High IL-6-Producing Subset that Confer Protection in Graft-versus-Host-Disease and Radiation Injury Models. Biol Blood Marrow Transplant 2017; 23:897-905. [PMID: 28257800 DOI: 10.1016/j.bbmt.2017.02.018] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 02/27/2017] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) have immunosuppressive and tissue repair properties, but clinical trials using MSCs to prevent or treat graft-versus-host disease (GVHD) have shown mixed results. Macrophages (MØs) are important regulators of immunity and can promote tissue regeneration and remodeling. We have previously shown that MSCs can educate MØs toward a unique anti-inflammatory immunophenotype (MSC-educated MØs [MEMs]); however, their implications for in vivo models of inflammation have not been studied yet. We now show that in comparison with MØs, MEMs have increased expression of the inhibitory molecules PD-L1, PD-L2, in addition to markers of alternatively activated MØs: CD206 and CD163. RNA-Seq analysis of MEMs, as compared with MØs, show a distinct gene expression profile that positively correlates with multiple pathways important in tissue repair. MEMs also show increased expression of IL-6, transforming growth factor-β, arginase-1, CD73, and decreased expression of IL-12 and tumor necrosis factor-α. We show that IL-6 secretion is controlled in part by the cyclo-oxygenase-2, arginase, and JAK1/STAT1 pathway. When tested in vivo, we show that human MEMs significantly enhance survival from lethal GVHD and improve survival of mice from radiation injury. We show these effects could be mediated in part through suppression of human T cell proliferation and may have attenuated host tissue injury in part by enhancing murine fibroblast proliferation. MEMs are a unique MØ subset with therapeutic potential for the management of GVHD and/or protection from radiation-induced injury.
Collapse
Affiliation(s)
- Myriam N Bouchlaka
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Andrea B Moffitt
- Department of Medicine, Duke University, Durham, North Carolina; Duke Center of Genomic and Computational Biology, Durham, North Carolina
| | - Jaehyup Kim
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - John A Kink
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Debra D Bloom
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Cassandra Love
- Department of Medicine, Duke University, Durham, North Carolina; Duke Center of Genomic and Computational Biology, Durham, North Carolina
| | - Sandeep Dave
- Department of Medicine, Duke University, Durham, North Carolina; Duke Center of Genomic and Computational Biology, Durham, North Carolina
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Christian M Capitini
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; University of Wisconsin Carbone Cancer Center, Madison, Wisconsin.
| |
Collapse
|
130
|
Funaki M, Janmey PA. Technologies to Engineer Cell Substrate Mechanics in Hydrogels. BIOLOGY AND ENGINEERING OF STEM CELL NICHES 2017:363-373. [DOI: 10.1016/b978-0-12-802734-9.00023-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
131
|
Grégoire C, Lechanteur C, Briquet A, Baudoux É, Baron F, Louis E, Beguin Y. Review article: mesenchymal stromal cell therapy for inflammatory bowel diseases. Aliment Pharmacol Ther 2017; 45:205-221. [PMID: 27878827 DOI: 10.1111/apt.13864] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/21/2016] [Accepted: 10/25/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Inflammatory bowel diseases (IBD) are chronic relapsing diseases in which pro-inflammatory immune cells and cytokines induce intestinal tissue damage and disability. Mesenchymal stromal cells (MSCs) exert powerful immunomodulatory effects and stimulate tissue repair. AIM To review the current data on mesenchymal stromal cell therapy in IBD. METHOD We searched PubMed and 'ClinicalTrials.gov' databases using the terms 'mesenchymal stromal cells', 'mesenchymal stem cell transplantation', 'inflammatory bowel diseases', 'Crohn disease' and 'colitis, ulcerative'. Additional publications were identified from individual article reference lists. RESULTS MSCs include inhibition of Th1/Th17 lymphocytes and recruitment of regulatory T lymphocytes, induction of antigen-presenting cells into a regulatory-like profile, and stimulation of epithelial cell differentiation and proliferation. More than 200 patients with refractory fistulas have been treated with local injections of MSCs, resulting in complete response in more than half, and in overall response in approximately two thirds of patients. In refractory luminal Crohn's disease, 49 cases of systemic MSC infusions have been reported, while trials with autologous MSCs resulted in mitigated responses, studies using allogeneic MSCs were promising, with around 60% of patients experiencing a response and around 40% achieving clinical remission. CONCLUSIONS Mesenchymal stromal cells might represent a promising therapy for IBD, especially for Crohn's disease. There remain many unsolved questions concerning the optimal origin and source of mesenchymal stromal cells, dosage and modalities of administration. Moreover, mesenchymal stromal cells still need to prove their effectiveness compared with conventional treatments in randomised controlled trials.
Collapse
Affiliation(s)
- C Grégoire
- Unit of Haematology, Department of Haematology, CHU of Liège, GIGA-I3, University of Liège, Liège, Belgium
| | - C Lechanteur
- Laboratory of Cell and Gene Therapy (LTCG), CHU of Liège, Liège, Belgium
| | - A Briquet
- Laboratory of Cell and Gene Therapy (LTCG), CHU of Liège, Liège, Belgium
| | - É Baudoux
- Laboratory of Cell and Gene Therapy (LTCG), CHU of Liège, Liège, Belgium
| | - F Baron
- Unit of Haematology, Department of Haematology, CHU of Liège, GIGA-I3, University of Liège, Liège, Belgium
| | - E Louis
- Department of Gastroenterology, CHU of Liège, University of Liège, Liège, Belgium
| | - Y Beguin
- Unit of Haematology, Department of Haematology, CHU of Liège, GIGA-I3, University of Liège, Liège, Belgium.,Laboratory of Cell and Gene Therapy (LTCG), CHU of Liège, Liège, Belgium
| |
Collapse
|
132
|
|
133
|
Salmenniemi U, Itälä-Remes M, Nystedt J, Putkonen M, Niittyvuopio R, Vettenranta K, Korhonen M. Good responses but high TRM in adult patients after MSC therapy for GvHD. Bone Marrow Transplant 2016; 52:606-608. [PMID: 27941780 DOI: 10.1038/bmt.2016.317] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- U Salmenniemi
- Department of Clinical Hematology and Stem Cell Transplantation, Turku University Hospital and University of Turku, Turku, Finland
| | - M Itälä-Remes
- Department of Clinical Hematology and Stem Cell Transplantation, Turku University Hospital and University of Turku, Turku, Finland
| | - J Nystedt
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - M Putkonen
- Department of Clinical Hematology and Stem Cell Transplantation, Turku University Hospital and University of Turku, Turku, Finland
| | - R Niittyvuopio
- Comprehensive Cancer Center, Stem Cell Transplantation Unit, Helsinki University Hospital, Helsinki, Finland
| | - K Vettenranta
- Helsinki University Children's Hospital and University of Helsinki, Helsinki, Finland
| | - M Korhonen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| |
Collapse
|
134
|
Girdlestone J. Mesenchymal stromal cells with enhanced therapeutic properties. Immunotherapy 2016; 8:1405-1416. [DOI: 10.2217/imt-2016-0098] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
135
|
Nitkin CR, Bonfield TL. Concise Review: Mesenchymal Stem Cell Therapy for Pediatric Disease: Perspectives on Success and Potential Improvements. Stem Cells Transl Med 2016; 6:539-565. [PMID: 28191766 PMCID: PMC5442806 DOI: 10.5966/sctm.2015-0427] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 08/04/2016] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) represent a potentially revolutionary therapy for a wide variety of pediatric diseases, but the optimal cell-based therapeutics for such diversity have not yet been specified. The published clinical trials for pediatric pulmonary, cardiac, orthopedic, endocrine, neurologic, and hematologic diseases provide evidence that MSCs are indeed efficacious, but the significant heterogeneity in therapeutic approaches between studies raises new questions. The purpose of this review is to stimulate new preclinical and clinical trials to investigate these factors. First, we discuss recent clinical trials for pediatric diseases studying MSCs obtained from bone marrow, umbilical cord and umbilical cord blood, placenta, amniotic fluid, and adipose tissue. We then identify factors, some unique to pediatrics, which must be examined to optimize therapeutic efficacy, including route of administration, dose, timing of administration, the role of ex vivo differentiation, cell culture techniques, donor factors, host factors, and the immunologic implications of allogeneic therapy. Finally, we discuss some of the practicalities of bringing cell-based therapy into the clinic, including regulatory and manufacturing considerations. The aim of this review is to inform future studies seeking to maximize therapeutic efficacy for each disease and for each patient. Stem Cells Translational Medicine 2017;6:539-565.
Collapse
Affiliation(s)
- Christopher R. Nitkin
- Division of Neonatology, Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
| | - Tracey L. Bonfield
- Division of Pulmonology, Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
136
|
Rizk M, Monaghan M, Shorr R, Kekre N, Bredeson CN, Allan DS. Heterogeneity in Studies of Mesenchymal Stromal Cells to Treat or Prevent Graft-versus-Host Disease: A Scoping Review of the Evidence. Biol Blood Marrow Transplant 2016; 22:1416-1423. [PMID: 27130504 DOI: 10.1016/j.bbmt.2016.04.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/11/2016] [Indexed: 12/29/2022]
Abstract
Effective treatments are lacking for the treatment of steroid-refractory graft-versus-host disease (GVHD), a major cause of morbidity and mortality after allogeneic hematopoietic cell transplantation. Mesenchymal stromal cells (MSCs) have demonstrated promise but there is uncertainty regarding their clinical effectiveness. A systematic scoping review of the literature was performed to characterize the heterogeneity of published studies and identify opportunities for standardization. Thirty studies were identified, including 19 studies (507 patients) addressing the treatment of acute or chronic GVHD and 11 prevention studies (277 patients). Significant heterogeneity was observed in the age and diagnoses of study subjects, intensity and specifics of the conditioning regimens, degree of HLA matching, and source of hematopoietic cells. MSCs were derived from bone marrow (83% of studies), cord blood (13%), or adipose tissue (3%) and were cryopreserved from third-party allogeneic donors in the majority of studies (91% of prevention studies and 63% of treatment studies). Culture conditions and media supplements were highly variable and characterization of MSCs did not conform to all International Society for Cellular Therapy criteria in any study. MSCs were harvested from cell culture at passage 1 to 7 and the dosage of MSCs ranged from 0.3 to 10 × 10(6)/kg, using varying schedules of administration. Treatment response criteria were not standardized and effectiveness in controlled treatment studies (5 studies) was unconvincing. Details of actively recruiting trials suggest heterogeneity still persists with only 53% of registered trials describing the use of standard GVHD response criteria and few detailing methods of MSC manufacturing. Future studies will need to make substantial coordinated efforts to reduce study heterogeneity and clarify the role of MSCs in GVHD.
Collapse
Affiliation(s)
- Mina Rizk
- Blood and Marrow Transplantation, Department of Medicine, The Ottawa Hospital and University of Ottawa and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Madeline Monaghan
- Blood and Marrow Transplantation, Department of Medicine, The Ottawa Hospital and University of Ottawa and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Risa Shorr
- Blood and Marrow Transplantation, Department of Medicine, The Ottawa Hospital and University of Ottawa and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Natasha Kekre
- Blood and Marrow Transplantation, Department of Medicine, The Ottawa Hospital and University of Ottawa and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Christopher N Bredeson
- Blood and Marrow Transplantation, Department of Medicine, The Ottawa Hospital and University of Ottawa and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - David S Allan
- Blood and Marrow Transplantation, Department of Medicine, The Ottawa Hospital and University of Ottawa and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
137
|
Locatelli F, Algeri M, Trevisan V, Bertaina A. Remestemcel-L for the treatment of graft versus host disease. Expert Rev Clin Immunol 2016; 13:43-56. [PMID: 27399600 DOI: 10.1080/1744666x.2016.1208086] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Remestemcel-L, a third-party, off-the-shelf preparation of bone-marrow derived mesenchymal stromal cells (MSCs), has been developed for experimental use in acute graft-versus-host disease (aGvHD) and other immune-mediated conditions. Several preclinical and clinical studies have indeed suggested the potential of human mesenchymal stromal cells (MSCs) as an effective treatment for steroid-refractory aGvHD. However, an unambiguous demonstration of efficacy is still lacking. Areas covered: This review critically examines the biologic rationale supporting MSCs use in aGvHD and analyzes the results of published clinical trials in this setting, with a particular focus on the potential benefits and drawbacks of Remestemcel-L. For this purpose, a systematic literature search was performed in PubMed using the following keywords: 'mesenchymal stromal cells', 'mesenchymal progenitor cells', 'multipotent stromal cells', 'mesenchymal cells', 'MSC', 'Remestemcel-L', 'Prochymal', and 'graft-versus-host disease' or 'GvHD'. Expert commentary: Remestemcel-L represents a promising alternative to second-line immunosuppressive agents for the treatment of steroid-refractory aGvHD. Despite the safety and the favorable risk/benefit profile of this cell product, which has been demonstrated in several phase I-II studies, large and prospective randomized trials are required to confirm its efficacy in aGvHD and to define the optimal schedule of administration in terms of infusion timing, cell dose and pharmacological synergism.
Collapse
Affiliation(s)
- F Locatelli
- a Department of Pediatric Hematology-Oncology , IRCCS, Bambino Gesù Children's Hospital , Rome , Italy.,b Department of Pediatrics , University of Pavia , Pavia , Italy
| | - M Algeri
- a Department of Pediatric Hematology-Oncology , IRCCS, Bambino Gesù Children's Hospital , Rome , Italy
| | - V Trevisan
- a Department of Pediatric Hematology-Oncology , IRCCS, Bambino Gesù Children's Hospital , Rome , Italy
| | - A Bertaina
- a Department of Pediatric Hematology-Oncology , IRCCS, Bambino Gesù Children's Hospital , Rome , Italy
| |
Collapse
|
138
|
Abstract
INTRODUCTION Graft-versus-host disease (GVHD) leads to significant morbidity and mortality after allogeneic stem cell transplantation. While corticosteroids alone are adequate in some cases, they are often insufficient, leading to poor quality of life associated with the symptoms of disease, or mortality from infection and GVHD. Moreover, corticosteroids have significant side effects and often do not lead to durable responses. New therapies are needed to improve the development and progression of acute and chronic GVHD. AREAS COVERED We discuss the spectrum of emerging drugs for GVHD prevention and therapy. Cellular therapies will be briefly discussed. The available pre-clinical and clinical data regarding monoclonal antibodies, interleukin-2, alpha-1 antitrypsin, histone deacetylase inhibitors, tyrosine kinase inhibitors, and proteasome inhibitors will be reviewed. EXPERT OPINION Although therapies emerging for GVHD remain promising, most of these drugs are still in early phase clinical trials and require randomized comparisons before formal conclusions can be drawn. It is likely that in the near future some of these agents will show improvements in response when compared with corticosteroids alone. Although it is difficult to predict which of these agents will be most promising, alpha-1 antitrypsin, ruxolitinib and interleukin-2 have demonstrated encouraging results.
Collapse
Affiliation(s)
- Natasha Kekre
- a Division of Hematology , Ottawa Health Research Institute, The Ottawa Hospital and University of Ottawa , Ottawa , ON , Canada
| | - Joseph H Antin
- b Blood and Marrow Transplantation Program, Division of Hematologic Malignancies , Dana-Farber Cancer Institute, Harvard Medical School , Boston , MA , USA
| |
Collapse
|
139
|
Renteria AS, Levine JE, Ferrara JLM. Therapeutic targets and emerging treatment options in gastrointestinal acute graft-versus-host disease. Expert Opin Orphan Drugs 2016; 4:469-484. [PMID: 30057862 DOI: 10.1517/21678707.2016.1166949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction Graft-versus-host disease (GVHD) continues to be the major lethal complication of allogeneic hematopoietic stem cell transplantation (HCT) but the standard of care, high dose steroids, has not changed in 40 years. Approximately 50% of GVHD patients will develop steroid refractory disease, typically involving the gastrointestinal (GI) tract, which has a very poor prognosis. Newly developed GVHD biomarker-based risk scores provide the first opportunity to treat patients at the onset of symptoms according to risk of steroid failure. Furthermore, improvements in our understanding of the pathobiology of GVHD, its different signaling pathways, involved cytokines, and the role of post-translational and epigenetic modifications, has identified new therapeutic targets for clinical trials. Areas covered This manuscript summarizes the pathophysiology, diagnosis, staging, current and new targeted therapies for GVHD, with an emphasis on GI GVHD. A literature search on PubMed was undertaken and the most relevant references included. Expert Opinion The standard treatment for GVHD, high dose steroids, offers less than optimal outcomes as well as significant toxicities. Better treatments, especially for GI GVHD, are needed to reduce non-relapse mortality after allogeneic HCT. The identification of high risk patients through a biomarker-defined scoring system offers a personalized approach to a disease that still requires significant research attention.
Collapse
Affiliation(s)
- Anne S Renteria
- Blood and Marrow Transplantation Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John E Levine
- Blood and Marrow Transplantation Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James L M Ferrara
- Hematologic Malignancies Translational Research Center, Blood and Marrow Transplantation Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|