1
|
Strategies to address mesenchymal stem/stromal cell heterogeneity in immunomodulatory profiles to improve cell-based therapies. Acta Biomater 2021; 133:114-125. [PMID: 33857693 DOI: 10.1016/j.actbio.2021.03.069] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/15/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal cells (MSCs) have gained immense attention over the past two decades due to their multipotent differentiation potential and pro-regenerative and immunomodulatory cytokine secretory profiles. Their ability to modulate the host immune system and promote tolerance has prompted several allogeneic and autologous hMSC-based clinical trials for the treatment of graft-versus-host disease and several other immune-induced disorders. However, clinical success beyond safety is still controversial and highly variable, with inconclusive therapeutic benefits and little mechanistic explanation. This clinical variability has been broadly attributed to inconsistent MSC sourcing, phenotypic characterization, variable potency, and non-standard isolation protocols, leading to functional heterogeneity among administered MSCs. Homogeneous MSC populations are proposed to yield more predictable, reliable biological responses and clinically meaningful properties relevant to cell-based therapies. Limited comparisons of heterogeneous MSCs with homogenous MSCs are reported. This review addresses this gap in the literature with a critical analysis of strategies aimed at decreasing MSC heterogeneity concerning their reported immunomodulatory profiles. STATEMENT OF SIGNIFICANCE: This review collates, summarizes, and critically analyzes published strategies that seek to improve homogeneity in immunomodulatory functioning MSC populations intended as cell therapies to treat immune-based disorders, such as graft-vs-host-disease. No such review for MSC therapies, immunomodulatory profiles and cell heterogeneity analysis is published. Since MSCs represent the most clinically studied experimental cell therapy platform globally for which there remains no US domestic marketing approval, insights into MSC challenges in therapeutic product development are imperative to providing solutions for immunomodulatory variabilities.
Collapse
|
2
|
Lee DK, Song SU. Immunomodulatory mechanisms of mesenchymal stem cells and their therapeutic applications. Cell Immunol 2017; 326:68-76. [PMID: 28919171 DOI: 10.1016/j.cellimm.2017.08.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 08/27/2017] [Accepted: 08/27/2017] [Indexed: 02/06/2023]
Abstract
In the recent years, many studies have shown that MSCs must be stimulated by pro-inflammatory cytokines or other immune mediators before they can modulate immune cells in inflamed and damaged tissues. MSCs appear to be involved in inducing several regulatory immune cells, such as Tregs, Bregs, and regulatory NK cells. This new immune milieu created by MSCs may establish a tolerogenic environment that leads to an optimal condition for the treatment of immune diseases. The mechanisms of MSC action to treat immune disorders need to be further investigated in more detail. Since there have been some contradictory outcomes of clinical trials, it is necessary to perform large-scale and randomized clinical studies, such as a phase 3 placebo-controlled double-blind study of a third party MSCs to optimize MSC administration and to prove safety and efficacy of MSC treatment. MSCs offer great therapeutic promise, especially for the treatment of difficult-to-treat immune diseases.
Collapse
Affiliation(s)
- Don K Lee
- SCM Lifesciences Co. Ltd., Incheon 22332 Republic of Korea
| | - Sun U Song
- Dept. of Integrated Biomedical Sciences, Inha University School of Medicine, Incheon 22332 Republic of Korea; SCM Lifesciences Co. Ltd., Incheon 22332 Republic of Korea.
| |
Collapse
|
3
|
Kim DS, Lee MW, Ko YJ, Park HJ, Park YJ, Kim DI, Jung HL, Sung KW, Koo HH, Yoo KH. Application of human mesenchymal stem cells cultured in different oxygen concentrations for treatment of graft-versus-host disease in mice. Biomed Res 2017; 37:311-317. [PMID: 27784875 DOI: 10.2220/biomedres.37.311] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Human mesenchymal stem cell (MSC) heterogeneity and problems associated with the ex vivo expansion of MSC are linked with the failure of MSC clinical trials. In this study, we compared the effect of MSCs cultured in different oxygen concentrations on GVHD in mice to elucidate whether hypoxia improves the immunosuppressive capacity of MSCs. Hypoxia increased the proliferative activity and the expression of several stemness and chemokine genes, such as KLF4, OCT4, C-MYC, CCL2, and CXCL10. Mice that received MSCs cultured in normoxia or hypoxia showed alleviated symptoms of GVHD and increased survival times. However, there was no significant difference in survival rates between mice that received MSCs cultured in normoxia and hypoxia. These data suggest that hypoxic culture is a useful method for maintaining and obtaining MSCs used for cell therapy and that the therapeutic potential of MSCs cultured in hypoxia warrants further investigation.
Collapse
Affiliation(s)
- Dae Seong Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Miyamura K. Insurance approval of mesenchymal stem cell for acute GVHD in Japan: need of follow up for some remaining concerns. Int J Hematol 2016; 103:155-64. [PMID: 26759322 DOI: 10.1007/s12185-015-1930-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 12/15/2015] [Indexed: 10/22/2022]
Abstract
Acute graft-versus-host disease (aGVHD) is a major obstacle following allogeneic hematopoietic stem cell transplantation. Steroid is the standard treatment for aGVHD grade II-IV; however, nearly half of patients do not respond to the therapy. Many drugs have been proposed, but no standard therapy has been determined. This is because of the resistance to these drugs and of infections due to prolonged immunosuppressive states. Over the past decade a new approach using mesenchymal stem cells (MSCs) has been emerging in Japan and western countries. MSCs have unique characteristics such as specific immunosuppressive properties, no immunogenicity on their own and supportive activity for hematopoiesis. Most of the published trials have reported a favorable effect in acute GVHD, but a phase III trial failed to reach the primary endpoint, although, subgroup analyses found significant effects on gut and liver GVHD in the patients with MSCs infusion. In Japan several institutes are trying to develop MSC for clinical use in post HSCT patients. However, several limitations make it difficult to use MSC in clinical practice. Recently we conducted a phase II/III study using MSC (JR-031) for patients with steroid-refractory grade III or IV aGVHD. From the feasible clinical results, JR-031 was approved by PMDA as the first product which meets the Act to Revise the Pharmaceutical Affairs Act and the Act to Ensure the Safety of Regenerative Medicine. The cost of one series of the treatment is more than ten million yen. Now we encounter new issues such as cost, indication, safety and efficacy. The mechanism of MSC is still unclear and potential concerns about ectopic tissue formation and MSC related malignancy in vivo remain. In conclusion, MSC infusions are well tolerated and show benefit in some patients without adverse safety effects; however, long-term follow-up is needed to be more certain of this.
Collapse
Affiliation(s)
- Koichi Miyamura
- Department of Hematology, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan.
| |
Collapse
|
5
|
Yi HG, Yahng SA, Kim I, Lee JH, Min CK, Kim JH, Kim CS, Song SU. Allogeneic clonal mesenchymal stem cell therapy for refractory graft-versus-host disease to standard treatment: a phase I study. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 20:63-7. [PMID: 26807024 PMCID: PMC4722192 DOI: 10.4196/kjpp.2016.20.1.63] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 08/27/2015] [Accepted: 09/11/2015] [Indexed: 01/17/2023]
Abstract
Severe graft-versus-host disease (GVHD) is an often lethal complication of allogeneic hematopoietic stem cell transplantation (HSCT). The safety of clinical-grade mesenchymal stem cells (MSCs) has been validated, but mixed results have been obtained due to heterogeneity of the MSCs. In this phase I study, the safety of bone marrow-derived homogeneous clonal MSCs (cMSCs) isolated by a new subfractionation culturing method was evaluated. cMSCs were produced in a GMP facility and intravenously administered to patients who had refractory GVHD to standard treatment resulting after allogeneic HSCT for hematologic malignancies. After administration of a single dose (1×106 cells/kg), 11 patients were evaluated for cMSC treatment safety and efficacy. During the trial, nine patients had 85 total adverse events and the rate of serious adverse events was 27.3% (3/11 patients). The only one adverse drug reaction related to cMSC administration was grade 2 myalgia in one patient. Treatment response was observed in four patients: one with acute GVHD (partial response) and three with chronic GVHD. The other chronic patients maintained stable disease during the observation period. This study demonstrates single cMSC infusion to have an acceptable safety profile and promising efficacy, suggesting that we can proceed with the next stage of the clinical trial.
Collapse
Affiliation(s)
- Hyeon Gyu Yi
- Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon 22332, Korea
| | - Seung-Ah Yahng
- Department of Internal Medicine, Incheon St. Mary's Hospital, The Catholic University of Korea, Incheon 21431, Korea
| | - Inho Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University School of Medicine, Seoul 03080, Korea
| | - Je-Hwan Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Chang-Ki Min
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul 06591, Korea
| | - Jun Hyung Kim
- Translational Research Center, Inha University School of Medicine and SCM Lifescience Co., Ltd., Incheon 22332, Korea
| | - Chul Soo Kim
- Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon 22332, Korea
| | - Sun U Song
- Translational Research Center, Inha University School of Medicine and SCM Lifescience Co., Ltd., Incheon 22332, Korea
| |
Collapse
|
6
|
Fierabracci A, Del Fattore A, Luciano R, Muraca M, Teti A, Muraca M. Recent advances in mesenchymal stem cell immunomodulation: the role of microvesicles. Cell Transplant 2013; 24:133-149. [PMID: 24268069 DOI: 10.3727/096368913x675728] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells are the most widely used cell phenotype for therapeutic applications, the main reasons being their well-established abilities to promote regeneration of injured tissues and to modulate immune responses. Efficacy was reported in the treatment of several animal models of inflammatory and autoimmune diseases and, in clinical settings, for the management of disorders such as GVHD, systemic lupus erythematosus, multiple sclerosis, and inflammatory bowel disease. The effects of mesenchymal stem cells are believed to be largely mediated by paracrine signals, and several secreted molecules have been identified as contributors to the net biological effect. Recently, it has been recognized that bioactive molecules can be shuttled from cell to cell packed in microvesicles, tiny portions of cytoplasm surrounded by a membrane. Coding and noncoding RNAs are also carried in such microvesicles, transferring relevant biological activity to target cells. Several reports indicate that the regenerative effect of mesenchymal stem cells can be reproduced by microvesicles isolated from their culture medium. More recent evidence suggests that the immunomodulatory effects of mesenchymal stem cells are also at least partially mediated by secreted microvesicles. These findings allow better understanding of the mechanisms involved in cell-to-cell interaction and may have interesting implications for the development of novel therapeutic tools in place of the parent cells.
Collapse
|
7
|
Unrelated allogeneic bone marrow-derived mesenchymal stem cells for steroid-refractory acute graft-versus-host disease: a phase I/II study. Int J Hematol 2013; 98:206-13. [PMID: 23860964 DOI: 10.1007/s12185-013-1399-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/05/2013] [Accepted: 07/09/2013] [Indexed: 02/07/2023]
Abstract
We conducted a multicenter phase I/II study using mesenchymal stem cells (MSCs) manufactured from the bone marrow of healthy unrelated volunteers to treat steroid-refractory acute graft-versus-host disease (aGVHD). Fourteen patients with hematological malignancies who suffered from grade II (9 patients) or III aGVHD (5) were treated. Affected organs were gut (10 patients), skin (9 patients), and liver (3 patients). Seven patients had two involved organs. The median age was 52. No other second-line agents were given. MSCs were given at a dose of 2 × 10(6) cells/kg for each infusion twice a week for 4 weeks. If needed, patients were continuously given MSCs weekly for an additional 4 weeks. By week 4, 13 of 14 patients (92.9 %) had responded to MSC therapy with a complete response (CR; n = 8) or partial response (PR; n = 5). At 24 weeks, 11 patients (10 with CR and 1 with PR) were alive. At 96 weeks, 8 patients were alive in CR. A total of 6 patients died, attributable to the following: underlying disease relapse (2 patients), breast cancer relapse (1), veno-occlusive disease (1), ischemic cholangiopathy (1), and pneumonia (1). No clear adverse effects associated with MSC infusion were observed. Third party-derived bone marrow MSCs may be safe and effective for patients with steroid-refractory aGVHD.
Collapse
|
8
|
Wong RSY, Cheong SK. Role of mesenchymal stem cells in leukaemia: Dr. Jekyll or Mr. Hyde? Clin Exp Med 2013; 14:235-48. [DOI: 10.1007/s10238-013-0247-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 06/08/2013] [Indexed: 01/19/2023]
|
9
|
Tomchuck SL, Norton EB, Garry RF, Bunnell BA, Morris CA, Freytag LC, Clements JD. Mesenchymal stem cells as a novel vaccine platform. Front Cell Infect Microbiol 2012; 2:140. [PMID: 23162801 PMCID: PMC3499769 DOI: 10.3389/fcimb.2012.00140] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Accepted: 10/22/2012] [Indexed: 01/14/2023] Open
Abstract
Vaccines are the most efficient and cost-effective means of preventing infectious disease. However, traditional vaccine approaches have thus far failed to provide protection against human immunodeficiency virus (HIV), tuberculosis, malaria, and many other diseases. New approaches to vaccine development are needed to address some of these intractable problems. In this report, we review the literature identifying stimulatory effects of mesenchymal stem cells (MSC) on immune responses and explore the potential for MSC as a novel, universal vaccination platform. MSC are unique bone marrow-derived multipotent progenitor cells that are presently being exploited as gene therapy vectors for a variety of conditions, including cancer and autoimmune diseases. Although MSC are predominantly known for anti-inflammatory properties during allogeneic MSC transplant, there is evidence that MSC can actually promote adaptive immunity under certain settings. MSC have also demonstrated some success in anti-cancer therapeutic vaccines and anti-microbial prophylactic vaccines, as we report, for the first time, the ability of modified MSC to express and secrete a viral antigen that stimulates antigen-specific antibody production in vivo. We hypothesize that the unique properties of modified MSC may enable MSC to serve as an unconventional but innovative, vaccine platform. Such a platform would be capable of expressing hundreds of proteins, thereby generating a broad array of epitopes with correct post-translational processing, mimicking natural infection. By stimulating immunity to a combination of epitopes, it may be possible to develop prophylactic and even therapeutic vaccines to tackle major health problems including those of non-microbial and microbial origin, including cancer, or an infectious disease like HIV, where traditional vaccination approaches have failed.
Collapse
Affiliation(s)
- Suzanne L Tomchuck
- Department of Microbiology and Immunology, Tulane University School of Medicine New Orleans, LA, USA
| | | | | | | | | | | | | |
Collapse
|
10
|
Double allogenic mesenchymal stem cells transplantations could not enhance therapeutic effect compared with single transplantation in systemic lupus erythematosus. Clin Dev Immunol 2012; 2012:273291. [PMID: 22829849 PMCID: PMC3399403 DOI: 10.1155/2012/273291] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 04/30/2012] [Indexed: 12/21/2022]
Abstract
The clinical trial of allogenic mesenchymal stem cells (MSCs) transplantation for refractory SLE patients has shown significant safety and efficacy profiles. However, the optimum frequency of the MSCs transplantation (MSCT) is unknown. This study was undertaken to observe whether double transplantations of MSCs is superior to single transplantation. Fifty-eight refractory SLE patients were enrolled in this study, in which 30 were randomly given single MSCT, and the other 28 were given double MSCT. Patients were followed up for rates of survival, disease remission, and relapse, as well as transplantation-related adverse events. SLE disease activity index (SLEDAI) and serologic features were evaluated. Our results showed that no remarkable differences between single and double allogenic MSCT were found in terms of disease remission and relapse, amelioration of disease activity, and serum indexes in an SLE clinical trial with more than one year followup. This study demonstrated that single MSCs transplantation at the dose of one million MSCs per kilogram of body weight was sufficient to induce disease remission for refractory SLE patients.
Collapse
|
11
|
Toll-like receptor 3 and suppressor of cytokine signaling proteins regulate CXCR4 and CXCR7 expression in bone marrow-derived human multipotent stromal cells. PLoS One 2012; 7:e39592. [PMID: 22745793 PMCID: PMC3382127 DOI: 10.1371/journal.pone.0039592] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 05/25/2012] [Indexed: 01/07/2023] Open
Abstract
Background The use of bone marrow-derived human multipotent stromal cells (hMSC) in cell-based therapies has dramatically increased in recent years, as researchers have exploited the ability of these cells to migrate to sites of tissue injury, inflammation, and tumors. Our group established that hMSC respond to “danger” signals – by-products of damaged, infected or inflamed tissues – via activation of Toll-like receptors (TLRs). However, little is known regarding downstream signaling mediated by TLRs in hMSC. Methodology/Principal Findings We demonstrate that TLR3 stimulation activates a Janus kinase (JAK) 2/signal transducer and activator of transcription (STAT) 1 pathway, and increases expression of suppressor of cytokine signaling (SOCS) 1 and SOCS3 in hMSC. Our studies suggest that each of these SOCS plays a distinct role in negatively regulating TLR3 and JAK/STAT signaling. TLR3-mediated interferon regulatory factor 1 (IRF1) expression was inhibited by SOCS3 overexpression in hMSC while SOCS1 overexpression reduced STAT1 activation. Furthermore, our study is the first to demonstrate that when TLR3 is activated in hMSC, expression of CXCR4 and CXCR7 is downregulated. SOCS3 overexpression inhibited internalization of both CXCR4 and CXCR7 following TLR3 stimulation. In contrast, SOCS1 overexpression only inhibited CXCR7 internalization. Conclusion/Significance These results demonstrate that SOCS1 and SOCS3 each play a functionally distinct role in modulating TLR3, JAK/STAT, and CXCR4/CXCR7 signaling in hMSC and shed further light on the way hMSC respond to danger signals.
Collapse
|
12
|
Strioga M, Viswanathan S, Darinskas A, Slaby O, Michalek J. Same or not the same? Comparison of adipose tissue-derived versus bone marrow-derived mesenchymal stem and stromal cells. Stem Cells Dev 2012; 21:2724-52. [PMID: 22468918 DOI: 10.1089/scd.2011.0722] [Citation(s) in RCA: 598] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) comprise a heterogeneous population of cells with multilineage differentiation potential, the ability to modulate oxidative stress, and secrete various cytokines and growth factors that can have immunomodulatory, angiogenic, anti-inflammatory and anti-apoptotic effects. Recent data indicate that these paracrine factors may play a key role in MSC-mediated effects in modulating various acute and chronic pathological conditions. MSCs are found in virtually all organs of the body. Bone marrow-derived MSCs (BM-MSCs) were discovered first, and the bone marrow was considered the main source of MSCs for clinical application. Subsequently, MSCs have been isolated from various other sources with the adipose tissue, serving as one of the alternatives to bone marrow. Adipose tissue-derived MSCs (ASCs) can be more easily isolated; this approach is safer, and also, considerably larger amounts of ASCs can be obtained compared with the bone marrow. ASCs and BM-MSCs share many biological characteristics; however, there are some differences in their immunophenotype, differentiation potential, transcriptome, proteome, and immunomodulatory activity. Some of these differences may represent specific features of BM-MSCs and ASCs, while others are suggestive of the inherent heterogeneity of both BM-MSC and ASC populations. Still other differences may simply be related to different isolation and culture protocols. Most importantly, despite the minor differences between these MSC populations, ASCs seem to be as effective as BM-MSCs in clinical application, and, in some cases, may be better suited than BM-MSCs. In this review, we will examine in detail the ontology, biology, preclinical, and clinical application of BM-MSCs versus ASCs.
Collapse
Affiliation(s)
- Marius Strioga
- Department of Immunology, Center of Oncosurgery, Institute of Oncology, Vilnius University, Vilnius, Lithuania.
| | | | | | | | | |
Collapse
|
13
|
Yi T, Song SU. Immunomodulatory properties of mesenchymal stem cells and their therapeutic applications. Arch Pharm Res 2012; 35:213-21. [PMID: 22370776 DOI: 10.1007/s12272-012-0202-z] [Citation(s) in RCA: 210] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Revised: 10/31/2011] [Accepted: 11/02/2011] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells that can be isolated from most adult tissues, including bone marrow, adipose, liver, amniotic fluid, lung, skeletal muscle and kidney. The term MSC is currently being used to represent both mesenchymal stem cells and multipotent mesenchymal stromal cells. Numerous reports on systemic administration of MSCs leading to functional improvements based on the paradigm of engraftment and differentiation have been published. However, it is not only difficult to demonstrate extensive engraftment of cells, but also no convincing clinical results have been generated from phase 3 trials as of yet and prolonged responses to therapy have been noted after identification of MSCs had discontinued. It is now clear that there is another mechanism by which MSCs exert their reparative benefits. Recently, MSCs have been shown to possess immunomodulatory properties. These include suppression of T cell proliferation, influencing dendritic cell maturation and function, suppression of B cell proliferation and terminal differentiation, and immune modulation of other immune cells such as NK cells and macrophages. In terms of the clinical applications of MSCs, they are being tested in four main areas: tissue regeneration for cartilage, bone, muscle, tendon and neuronal cells; as cell vehicles for gene therapy; enhancement of hematopoietic stem cell engraftment; and treatment of immune diseases such as graft-versus-host disease, rheumatoid arthritis, experimental autoimmune encephalomyelitis, sepsis, acute pancreatitis and multiple sclerosis. In this review, the mechanisms of immunomodulatory effects of MSCs and examples of animal and clinical uses of their immunomodulatory effects are described.
Collapse
Affiliation(s)
- Tacghee Yi
- Clinical Research Center, Inha Research Institute, Inha University School of Medicine, Incheon 400-712, Korea
| | | |
Collapse
|
14
|
Kikuchi-Taura A, Taguchi A, Kanda T, Inoue T, Kasahara Y, Hirose H, Sato I, Matsuyama T, Nakagomi T, Yamahara K, Stern D, Ogawa H, Soma T. Human umbilical cord provides a significant source of unexpanded mesenchymal stromal cells. Cytotherapy 2012; 14:441-50. [PMID: 22339605 DOI: 10.3109/14653249.2012.658911] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AIMS Human mesenchymal stromal cells (MSC) have considerable potential for cell-based therapies, including applications for regenerative medicine and immune suppression in graft-versus-host disease (GvHD). However, harvesting cells from the human body can cause iatrogenic disorders and in vitro expansion of MSC carries a risk of tumorigenesis and/or expansion of unexpected cell populations. METHODS Given these problems, we have focused on umbilical cord, a tissue obtained with few ethical problems that contains significant numbers of MSC. We have developed a modified method to isolate MSC from umbilical cord, and investigated their properties using flow cytometry, mRNA analysis and an in vivo GvHD model. RESULTS Our study demonstrates that, using umbilical cord, large numbers of MSC can be safely obtained using a simple procedure without in vitro expansion, and these non-expanded MSC have the potential to suppress GvHD. CONCLUSIONS Our results suggest that the combined banking of umbilical cord-derived MSC and identical cord blood-derived hematopoietic stem cell banking, where strict inspection of the infectious disease status of donors is performed, as well as further benefits of HLA-matched mesenchymal cells, could become one of the main sources of cells for cell-based therapy against various disorders.
Collapse
Affiliation(s)
- Akie Kikuchi-Taura
- Department of Regenerative Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Wang Y, Han ZB, Ma J, Zuo C, Geng J, Gong W, Sun Y, Li H, Wang B, Zhang L, He Y, Han ZC. A toxicity study of multiple-administration human umbilical cord mesenchymal stem cells in cynomolgus monkeys. Stem Cells Dev 2011; 21:1401-8. [PMID: 21958114 DOI: 10.1089/scd.2011.0441] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Therapies based on stem cells have shown very attractive potential in many clinical studies. However, the data about the safety of stem cells application remains insufficient. The present study was designed to evaluate the overall toxicology of human umbilical cord mesenchymal stem cells (hUC-MSCs) in cynomolgus monkeys with repeated administrations. hUC-MSCs were administered by intravenous injection once every 2 weeks for 6 weeks. The dose levels employed in this study were 2×10(6), 1×10(7) cells/kg body weight. Toxicity was evaluated by clinical observations (body weight, body temperature, and ophthalmology exams), pathology (blood cell counts, clinical biochemistry, urinalysis, and bone marrow smears), immunologic consequences (lymphoproliferation assay, the secretion of interferon-γ and interleukin-4, the percentage of CD3, CD4, CD8 T cells, and the ratio of CD4 and CD8 T cells) and anatomic pathology. Pharmacodynamics in blood and distribution of hUC-MSCs in the tissues of cynomolgus monkeys were measured by real-time polymerase chain reaction. All animals survived until scheduled euthanasia. No stem cells transplantation-related toxicity was found in this study. hUC-MSCs could be found in the blood of cynomolgus monkeys beyond 8 h. The findings of this 6-week toxicity study showed that the transplantation of hUC-MSCs did not affect the general health of cynomolgus monkeys.
Collapse
Affiliation(s)
- Youwei Wang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Hematology and Blood Diseases Hospital, Tianjin, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Zhang H, Chen Z, Bie P. Bone marrow-derived mesenchymal stem cells as immunosuppressants in liver transplantation: a review of current data. Transfus Med Rev 2011; 26:129-41. [PMID: 22015073 DOI: 10.1016/j.tmrv.2011.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
This article provides an overview of the current knowledge relating to the potential use of bone marrow-derived mesenchymal stem cells (BM-MSCs) acting as immunosuppressants after liver transplantation. Clinical use of BM-MSCs in liver transplantation remains experimental, as there is uncertainty as to their mechanism of action, conflicting studies in animal models, and the possibility of their cellular fusion with hepatocytes leading to potentially genetically unstable hepatocytes. These obstacles, to their underuse, have been decreasing, and BM-MSCs have elicited great interest for possible use in solid organ transplantation. Bone marrow-derived-MSCs, when transplanted systemically, might positively influence grafted organ outcome through cell-cell contact or the secretion of soluble factors that are immunomodulatory. Thus, the use of BM-MSCs to modulate organ rejection may directly or indirectly influence the survival properties of transplanted livers.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Hepatobiliary Surgery, SouthWestern Hospital, Chongqing, China.
| | | | | |
Collapse
|
17
|
Patel SA, Rameshwar P. Stem Cell Transplantation for Hematological Malignancies: Prospects for Personalized Medicine and Co-therapy with Mesenchymal Stem Cells. ACTA ACUST UNITED AC 2011; 9:229-239. [PMID: 21892378 DOI: 10.2174/187569211796957548] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bone marrow transplantation is a form of cell therapy that has been in practice for decades for the treatment of hematological disorders and solid tumors. Immunosuppressive therapy has been a mainstay for treatment, but the severity of the adverse effects has made it an undesirable choice. Mesenchymal stem cells (MSCs), which reside in the vascular regions of the bone marrow, have been shown to serve as cellular support for the hematopoietic stem cell (HSC) niche. Furthermore, the immune suppressive properties of MSCs have been explored in the treatment of inflammatory and autoimmune disorders. Thus, co-therapy with MSCs has been shown to facilitate engraftment of hematopoietic cells by suppressive graft versus host disease (GvHD). Although the mechanism by which MSCs suppress GvHD is unclear, the experimental evidence suggests that this partly occurs by modulation of immune response such as the induction of regulatory T cells. This paper discusses the role of MSCs as co-therapy for the future of stem cell transplantation, with the overarching theme of personalized medicine for cell-based health interventions.
Collapse
Affiliation(s)
- Shyam A Patel
- Department of Medicine, Division of Hematology/Oncology, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, NJ, USA
| | | |
Collapse
|
18
|
Wernicke CM, Grunewald TG, Hendrik J, Kuci S, Kuci Z, Koehl U, Mueller I, Doering M, Peters C, Lawitschka A, Kolb HJ, Bader P, Burdach S, von Luettichau I. Mesenchymal stromal cells for treatment of steroid-refractory GvHD: a review of the literature and two pediatric cases. Int Arch Med 2011; 4:27. [PMID: 21843360 PMCID: PMC3169455 DOI: 10.1186/1755-7682-4-27] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Accepted: 08/15/2011] [Indexed: 12/11/2022] Open
Abstract
Severe acute graft versus host disease (GvHD) is a life-threatening complication after allogeneic hematopoietic stem cell transplantation. Human mesenchymal stromal cells (MSCs) play an important role in endogenous tissue repair and possess strong immune-modulatory properties making them a promising tool for the treatment of steroid-refractory GvHD. To date, a few reports exist on the use of MSCs in treatment of GvHD in children indicating that children tend to respond better than adults, albeit with heterogeneous results. We here present a review of the literature and the clinical course of two instructive pediatric patients with acute steroid-refractory GvHD after haploidentical stem cell transplantation, which exemplify the beneficial effects of third-party transplanted MSCs in treatment of acute steroid-refractory GvHD. Moreover, we provide a meta-analysis of clinical studies addressing the outcome of patients with steroid-refractory GvHD and treatment with MSCs in adults and in children (n = 183; 122 adults, 61 children). Our meta-analysis demonstrates that the overall response-rate is high (73.8%) and confirms, for the first time, that children indeed respond better to treatment of GvHD with MSCs than adults (complete response 57.4% vs. 45.1%, respectively). These data emphasize the significance of this therapeutic approach especially in children and indicate that future prospective studies are needed to assess the reasons for the observed differential response-rates in pediatric and adult patients.
Collapse
Affiliation(s)
- Caroline M Wernicke
- Children's Cancer Research and Roman Herzog Comprehensive Cancer Center, Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Kölner Platz 1, 80804 Munich, Germany
| | - Thomas Gp Grunewald
- Children's Cancer Research and Roman Herzog Comprehensive Cancer Center, Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Kölner Platz 1, 80804 Munich, Germany.,Medical Life Science and Technology Center, TUM Graduate School, Technische Universität München, Boltzmannstrasse 17, 85748 Garching, Germany
| | - Juenger Hendrik
- Children's Cancer Research and Roman Herzog Comprehensive Cancer Center, Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Kölner Platz 1, 80804 Munich, Germany
| | - Selim Kuci
- Division for Stem Cell Transplantation, Department of Hematology, Oncology and Hemostasis, Hospital for Children and Adolescents, University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Zyrafete Kuci
- Division for Stem Cell Transplantation, Department of Hematology, Oncology and Hemostasis, Hospital for Children and Adolescents, University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Ulrike Koehl
- Division for Stem Cell Transplantation, Department of Hematology, Oncology and Hemostasis, Hospital for Children and Adolescents, University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Ingo Mueller
- University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Michaela Doering
- University Children's Hospital, Hoppe-Seyler-Strasse 1, 72076 Tuebingen, Germany
| | - Christina Peters
- St. Anna Children's Hospital, Kinderspitalgasse 6, 1090 Vienna, Austria
| | - Anita Lawitschka
- St. Anna Children's Hospital, Kinderspitalgasse 6, 1090 Vienna, Austria
| | - Hans-Jochem Kolb
- Division for Stem Cell Transplantation, Department of Medicine III, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, 81675 Munich, Germany
| | - Peter Bader
- Division for Stem Cell Transplantation, Department of Hematology, Oncology and Hemostasis, Hospital for Children and Adolescents, University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Stefan Burdach
- Children's Cancer Research and Roman Herzog Comprehensive Cancer Center, Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Kölner Platz 1, 80804 Munich, Germany
| | - Irene von Luettichau
- Children's Cancer Research and Roman Herzog Comprehensive Cancer Center, Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Kölner Platz 1, 80804 Munich, Germany
| |
Collapse
|
19
|
Landgraf K, Brunauer R, Lepperdinger G, Grubeck-Loebenstein B. The suppressive effect of mesenchymal stromal cells on T cell proliferation is conserved in old age. Transpl Immunol 2011; 25:167-72. [PMID: 21726642 DOI: 10.1016/j.trim.2011.06.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 06/17/2011] [Accepted: 06/18/2011] [Indexed: 12/29/2022]
Abstract
Mesenchymal stromal cells (MSC) have become a useful tool in curing graft versus host disease (GVHD) after transplantation. No information is presently available whether the immunosuppressive properties of this cell type are maintained in old age. It was therefore the aim of our study to analyze the immunoregulatory effect of MSC on peripheral blood mononuclear cells (PBMC) in old age. We studied the proliferation, activation and cytokine production of PBMC following co-culture with MSC from young (<30 years) and old (>61 years) donors. Our results demonstrate that MSC from elderly donors exhibit the same suppressive effects on T cell proliferation as their young counterparts. In both age groups T cell activation was not influenced by co-culture with MSC from young and elderly donors. With the exception of IL-6, cytokine production by unstimulated or stimulated PBMC was also not affected by MSC from either age group. IL-6 production was increased during co-culture of PBMC and MSC and was higher when MSC from elderly donors were used. After PHA stimulation, however, this age-specific difference was balanced and appeared even. As high IL-6 production is a prerequisite for an effective suppression of T cell proliferation, MSC can be considered a powerful tool for immunoregulatory therapies in old age.
Collapse
Affiliation(s)
- Katja Landgraf
- Institute for Biomedical Aging Research, Austrian Academy of Sciences, Rennweg 10, 6020 Innsbruck, Austria.
| | | | | | | |
Collapse
|
20
|
Sinkovics JG. Antileukemia and antitumor effects of the graft-versus-host disease: a new immunovirological approach. Acta Microbiol Immunol Hung 2010; 57:253-347. [PMID: 21183421 DOI: 10.1556/amicr.57.2010.4.2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In leukemic mice, the native host's explicit and well-defined immune reactions to the leukemia virus (a strong exogenous antigen) and to leukemia cells (pretending in their native hosts to be protected "self" elements) are extinguished and replaced in GvHD (graft-versus-host disease) by those of the immunocompetent donor cells. In many cases, the GvHD-inducer donors display genetically encoded resistance to the leukemia virus. In human patients only antileukemia and anti-tumor cell immune reactions are mobilized; thus, patients are deprived of immune reactions to a strong exogenous antigen (the elusive human leukemia-sarcoma retroviruses). The innate and adaptive immune systems of mice have to sustain the immunosuppressive effects of leukemia-inducing retroviruses. Human patients due to the lack of leukemiainducing retroviral pathogens (if they exist, they have not as yet been discovered), escape such immunological downgrading. After studying leukemogenic retroviruses in murine and feline (and other mammalian) hosts, it is very difficult to dismiss retroviral etiology for human leukemias and sarcomas. Since no characterized and thus recognized leukemogenic-sarcomagenic retroviral agents are being isolated from the vast majority of human leukemias-sarcomas, the treatment for these conditions in mice and in human patients vastly differ. It is immunological and biological modalities (alpha interferons; vaccines; adoptive lymphocyte therapy) that dominate the treatment of murine leukemias, whereas combination chemotherapy remains the main remission-inducing agent in human leukemias-lymphomas and sarcomas (as humanized monoclonal antibodies and immunotoxins move in). Yet, in this apparently different backgrounds in Mus and Homo, GvHD, as a treatment modality, appears to work well in both hosts, by replacing the hosts' anti-leukemia and anti-tumor immune faculties with those of the donor. The clinical application of GvHD in the treatment of human leukemias-lymphomas and malignant solid tumors remains a force worthy of pursuit, refinement and strengthening. Graft engineering and modifications of the inner immunological environment of the recipient host by the activation or administration of tumor memory T cells, selected Treg cells and natural killer (NKT) cell classes and cytokines, and the improved pharmacotherapy of GvHD without reducing its antitumor efficacy, will raise the value of GvHD to the higher ranks of the effective antitumor immunotherapeutical measures. Clinical interventions of HCT/HSCT (hematopoietic cell/stem cell transplants) are now applicable to an extended spectrum of malignant diseases in human patients, being available to elderly patients, who receive non-myeloablative conditioning, are re-enforced by post-transplant donor lymphocyte (NK cell and immune T cell) infusions and post-transplant vaccinations, and the donor cells may derive from engineered grafts, or from cord blood with reduced GvHD, but increased GvL/GvT-inducing capabilities (graft-versus leukemia/tumor). Post-transplant T cell transfusions are possible only if selected leukemia antigen-specific T cell clones are available. In verbatim quotation: "Ultimately, advances in separation of GvT from GvHD will further enhance the potential of allogeneic HCT as a curative treatment for hematological malignancies" (Rezvani, A.R. and Storb, R.F., Journal of Autoimmunity 30:172-179, 2008 (see in the text)). It may be added: for cure, a combination of the GvL/T effects with new targeted therapeutic modalities, as elaborated on in this article, will be necessary.
Collapse
Affiliation(s)
- Joseph G Sinkovics
- The University of South Florida College of Medicine, St. Joseph Hospital's Cancer Institute, Affiliated with the H. L. Moffitt Comprehensive Cancer Center, Tampa, FL 33607-6307, USA.
| |
Collapse
|