ArticlePDF Available

Mesenchymal Stem Cells in Clinical Trials for Immune Disorders

Georg Thieme Verlag KG
Global Medical Genetics
Authors:
Editorial
Mesenchymal Stem Cells in Clinical Trials for Immune Disorders
Zongjin Li1Zhibo Han2,3 Zhong-Chao Han3
1Department of Pathophysiology, Nankai University School of
Medicine, Tianjin, China
2Academy of Medical Engineering and Translational Medicine, Tianjin
University, Tianjin, China
3Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of
Health and Stem Cells, Health & Biotech Co., Beijing, China
Glob Med Genet 2024;11:196199.
Mesenchymal stem cells (MSCs) are a population of adult
stem cells found in various tissues, including bone marrow,
adipose tissue, and perinatal-related tissues.1MSCs have
emerged as a promising therapeutic approach due to their
unique properties, including regeneration and immunomo-
dulation. The therapeutic mechanism of MSCs is complex
and not fully understood, but they are believed to involve
several different processes. One important mechanism is the
paracrine secretion of growth factors, cytokines, and other
signaling molecules.2,3 These molecules can help to promote
tissue repair, reduce inammation, and modulate the im-
mune system by interacting with various components of
tissue microenvironments and modulating immune
responses.4The immunomodulatory effects of MSCs are
one of their most intriguing and therapeutically valuable
properties.5
A recent prospective, single-arm, phase I trial described
the efcacy and safety of human umbilical cord-derived
MSCs (UC-MSCs) in the treatment of refractory immune
thrombocytopenia (ITP), highlighting the effectiveness and
good tolerance of UC-MSCs in treating refractory ITP.6These
ndings offer insights that may advance our understanding
of the clinical application of MSCs in immune diseases. This
trial revealed that the overall response rate was 44.4% (8/18)
among all enrolled patients with mild adverse events. All
patients in the 2.0 10
6
cells/kg group achieved a platelet
count of approximately 50 10
9
/L for up to 28 weeks
(Fig. 1), which revealed a dose-dependent trend of platelet
count. Most importantly, bleeding symptoms can be
completely relieved in 60.0 to 75.0% of patients after treat-
ment, and no serious treatment-related emergent adverse
events occur.
ITP is an acquired autoimmune hemorrhagic disease
resulting from an i mbalance of immune tolerance. Refractor y
ITP refers to patients who have failed multiple therapies, and
current guidelines have limited effectiveness in alleviating
bleeding symptoms.6In this clinical trial, off-the-shelf GMP
(Good Manufacturing Practice)-grade UC-MSCs from Tianjin
Amcellgene Co., Ltd., were used.7,8 MSCs are also k nown to be
heterogeneous populations with variable functions. Many
studies have comparatively analyzed the differential proper-
ties and biological functions of MSCs derived from perinatal
and adult tissues, including their molecular proles, differ-
entiation potentials, proliferation/clonogenic formation ca-
pacities, immunomodulatory functions, and hematopoietic
support abilit ies. MSCs derived from perinatal tissues such as
the umbilical cord and placenta have attracted attention
because of their noninvasive isolation methods and minimal
ethical issues. Moreover, compared with ad ult tissue-derived
MSCs, perinatal tissue-derived MSCs are young cells without
increased possibilities of mutation.1,5 The banking of UC-
MSCs represents a noninvasive, simple, and safe means for
Address for correspondence
Zongjin Li, MD, PhD, Nankai
University School of Medicine,
Tianjin, China
(e-mail: zongjinli@nankai.edu.cn).
Zhibo Han, MD, Academy of Medical
Engineering and Translational
Medicine, Tianjin University,
Tianjin, China (e-mail: hanzhibo@
health-biotech.com).
Zhong-Chao Han, MD, PhD, Beijing
Engineering Laboratory of Perinatal
Stem Cells,Beijing Instituteof Health
and StemCells, Health & BiotechCo.,
Beijing, China
(e-mail: hanzhongchao@
health-biotech.com).
DOI https://doi.org/
10.1055/s-0044-1788044.
ISSN 2699-9404.
© 2024. The Author(s).
This is an open access article published by Thieme under the terms of the
Creative Commons Attribution License, permitting unrestricted use,
distribution, and reproduction so long as the original work is properly cited.
(https://creativecommons.org/licenses/by/4.0/).
Georg Thieme Verlag KG, digerstraße 14, 70469 Stuttgart,
Germany
Editorial
THIEME
196
Article published online: 2024-06-27
harvesting MSCs.9In this phase I trial, systemic infusion
of UC-MSCs was applied, and the circulation dynamics of
UC-MSCs were detected by the SRY gene in peripheral blood.
However, UC-MSCs were not detectable in the blood 8 hours
after injection. The therapeutic mechanisms of UC-MSCs
were investigated, and the data revealed a temporary de-
crease in T-cell percentages during the infusion of UC-MSCs,
followed by a gradual increase in the proportion of CD8 þ
CD28suppressive T-cell subsets.
MSCs interact with various components of the immune
system and modulate immune responses in several ways.
First, MSCs can directly interact with T cells through cell
surface molecules such as PD-L1, which binds to the PD-1
receptor on T cells, leading to reduced T-cell proliferation and
activation.10 Secondly, MSCs secrete various soluble factors,
such as prostaglandin E2 (PGE2), indoleamine 2,3-dioxyge-
nase (IDO), transforming growth factor-beta (TGF-β), and
hepatocyte growth factor, all of which contribute to the
suppression of T-cell proliferation and activity.11 In addition,
MSCs inhibit B-cell maturation through the release of soluble
factors such as PGE2 and by producing IDO, decreasing the
differentiation of B cells into plasma cells and reducing
antibody production.12 However, opposite results were ob-
served in which MSCs promoted the proliferation and differ-
entiation of B cells.2Furthermore, MSCs reduce the cytotoxic
activity of natural k iller (NK) cells and their ability to produce
proinammatory cytokines. MSCs inuence macrophages to
adopt an anti-inammatory M2 phenotype rather than a
proinammatory M1 phenotype mediated by MSC-derived
IL-10, TGF-β, and PGE2.13 The immunomodulatory capacity
of MSCs is a critical aspect of their therapeutic potential,
enabling them to create a more favorable environment for
tissue repair and regeneration by modulating immune
responses in a controlled manner (Fig. 2). The immuno-
modulatory properties of MSCs have signicant implications
for treating various inammatory and autoimmune condi-
tions, such as graft-versus-host disease, rheumatoid arthri-
tis, multiple sclerosis, systemic lupus erythematosus,
Crohns disease, and ulcerative colitis.12
For MSC therapy, intravenous infusions of MSCs are a
prominent priority for repeated injection, noninvasive pro-
cedures, and easy operation. The intravenously trans-
planted MSCs disappeared in a short time, and an
increase in the number of MSCs might not prolong cell
retention. MSCs might interact within the blood circulation
after intravenous administration to function in immuno-
modulation (Fig. 3). MSCs can modulate the immune
response by interacting with adaptive immune cells and
NK cells and exerting paracrine effects. However, most
MSCs return to and reach the lung after intravenous ad-
ministration.14 Therefore, how intravenously infused MSCs
bridge the spatial gap between the lung and other organs to
achieve immunomodulatory effects remains a challenge.
Recently, the process by which exogenous cells in an organ
affect distant recipient cells in another organ has been recog-
nized as transorgan communication.15 A recent study demon-
strated that intravenous infusion of MSCs could secrete
extracellular vesicles (EVs) and contribute to the therapeutic
effects of MSCs.14 This study revealed that intravenously
transplanted MSCs could serve as an endocrine reservoir to
secrete EVs into the blood continuously and gradually to
promote transorgan communication.
In summary, this phase I trial showed that UC-MSCs can
increase platelet counts and improve bleeding symptoms in
Fig. 1 Platelet counts over time in all patients. Platelet counts of all enrolled patients from baselin eto 24 weeks after the completion of UC-MSC
administration. Reproduced with permission from Chen et al 2024.6UC-MSC, umbilical cord-derived mesenchymal stem cell.
Global Medical Genetics Vol. 11 No. 3/2024 © 2024. The Author(s).
Editorial 197
Fig. 3 Interactions of MSCs with tissue microenvironments after intravenous administration. Reproduced with permission from Leibacher and
Henschler 2016.16 MSC, mesenchymal stem cell.
Fig. 2 MSCs act as immunomodulators, inuencing the behavior of other immunecells to create a more balancedimmune response. This proper ty makes
them a potentialtreatment for a wide range ofdiseases characterizedby an imbalanced immune response, such as autoimmune disorders, graft-versus-host
disease, and inammatory conditions. Reproduced with permission from Huang et al 2022.12 MSC, mesenchymal stem cell.
Global Medical Genetics Vol. 11 No. 3/2024 © 2024. The Author(s).
Editorial198
refractory ITP patients. MSCs have immense potential for
treating various diseases because of their immunomodulatory
properties. Research to address the distribution and metabo-
lism of MSCs in organ tissues is vital to translate thispotential
into effective and widely available clinical applications. As
our understanding of MSCs and their interactions with the
immune system deepens, we can look forward to a future
where MSC therapy becomes a mainstay in treating a broad
spectrum of immune-related conditions, including refrac-
tory ITP.
AuthorsContributions
Z.L. wrote the paper, and Z.H. and Z.C.H. revised and
approved the nal manuscript.
Funding
This work was supported by grants from the Tianjin Natural
Science Foundation (21JCZDJC00070, 22JCZXJC00170) and
the Nankai University Eye Institute (NKYKD202203).
Conict of Interest
None declared.
References
1Li Z, Han ZC. Introduction of perinatal tissue-derived stem cells..
In:Han ZC, Takahashi TA, Li Z, eds. Perinatal Stem Cells: Biology,
Manufacturing and Translational Medicine. Singapore; Springer
Singapore;2019:17
2Ji YR, Yang ZX, Han Z-B, et al. Mesenchymal stem cells support
proliferation and terminal differentiation of B cells. Cell Physiol
Biochem 2012;30(06):15261537
3Zhao Q, Han Z, Wang J, Han Z. Development and investigational
new drug application of mesenchymal stem/stromal cells prod-
ucts in China. Stem Cells Transl Med 2021;10(Suppl 2):S18S30
4Hezam K, Fu E, Zhang J, Li Z. Therapeutic trends of priming
mesenchymal stem cells: a bibliometric analysis. Biochem Bio-
phys Rep 2024;38:101708
5Shi Y, Wang Y, Li Q, et al. Immunoregulatory mechanisms of
mesenchymal stem and stromal cells in inammatory diseases.
Nat Rev Nephrol 2018;14(08):493507
6Chen Y, Xu Y, Chi Y, et al. Efcacy and safety of human umbilical
cord-derived mesenchymal stem cells in the treatment of refrac-
tory immune throm bocytopenia: a prospective, single arm , phase
I trial. Signal Transduct Target Ther 2024;9(01):102
7Chen H. Stem cell translational medicine: the Tianjin model
revisited. Stem Cells Transl Med 2021;10(Suppl 2):S4S9
8Fang L, Meikuang L, Ye G, et al. Successful treatment of a 19-
month-old boy with hepatitis associated aplastic anemia by
infusion of umbilical cord-derived mesenchymal stromal cells:
a case report. Cell Transplant 2021;30:963689720977144
9Han ZC, Takahashi TA, Han Z, Li Z . Perinatal Stem Cells. Singapore::
Springer; 2019
10 Liang W, Chen X, Zhang S, et al. Mesenchymal stem cells as a
double-edged sword in tumor growth: focusing on MSC-derived
cytokines. Cell Mol Biol Lett 2021;26(01):3
11 Du WJ, Chi Y, Yang ZX, et al. Heterogeneity of proangiogenic featuresin
mesenchymal stem cells derived from bone marrow, adipose tissue,
umbilical cord, and placenta. Stem Cell Res Ther 2016;7(01):163
12 Huang Y, Wu Q, Tam PKH. Immunomodulatory mechanisms of
mesenchymal stem cells and their potential clinical applications.
Int J Mol Sci 2022;23(17):10023
13 Cao X, Duan L, Hou H, et al. IGF-1C hydrogel improves the thera-
peutic effectsof MSCs on colitis in mice throughPGE
2
-mediatedM2
macrophage polarization. Theranostics 2020;10(17):76977709
14 Huang A, Liu Y, Qi X, et al. Intravenously transplanted mesenchy-
mal stromal cells: a new endocr ine reservoir for cardioprotection.
Stem Cell Res Ther 2022;13(01):253
15 Wysoczynski M, Khan A, Bolli R. New paradigms in cell therapy:
repeated dosing, intravenous delivery, immunomodulatory
actions, and new cell types. Circ Res 2018;123(02):138158
16 Leibacher J, Henschler R. Biodistribution, migration and homing
of systemically applied mesenchymal stem/stromal cells. Stem
Cell Res Ther 2016;7:7
Global Medical Genetics Vol. 11 No. 3/2024 © 2024. The Author(s).
Editorial 199
... MSCs possess immunosuppressive properties that enable them to modulate immune responses. 22 They can inhibit the proliferation of T cells 23,24 and the activation of natural killer (NK) cells, making them attractive candidates for applications in treating autoimmune diseases and enhancing graft survival in transplantation settings. ...
... MSCs not only possess self-renewal ability and multidirectional differentiation potential, but also play important roles in biological processes such as immune regulation and tissue repair. 22,25,26 In clinical applications, MSCs have been explored for the treatment of various diseases, such as acute and chronic inflammation, 27 autoimmune diseases, 28,29 bone and soft tissue injuries, 30,31 due to their low immunogenicity and good safety. In addition, with the rapid development of regenerative medicine, MSCs have a broader application prospect in cardiovascular and cerebrovascular diseases, diabetes, and nervous system diseases. ...
Article
Full-text available
Mesenchymal stem cells (MSCs), as a stem cell type with multiple differentiation potentials and immune regulatory abilities, have shown broad prospects in the treatment of ischemic stroke in recent years. The main characteristics of MSCs include their self-renewal ability, differentiation potential for different types of cells, and the ability to secrete various bioactive factors such as cytokines, chemokines, and growth factors, which play a key role in tissue repair and regeneration. In the treatment of ischemic stroke, MSCs exert therapeutic effects through various mechanisms, including promoting vascular regeneration of damaged brain tissue, reducing inflammatory responses, and protecting neurons from damage caused by apoptosis. Research have shown that MSCs can promote the repair of ischemic areas by releasing neurotrophic factors and angiogenic factors, while inhibiting immune responses triggered by ischemia, thereby improving neurological function. With the in-depth study of its biological mechanism, MSCs have gradually shown good safety and effectiveness in clinical applications. Therefore, fully exploring and utilizing the potential of MSCs in the treatment of ischemic stroke may provide new ideas and solutions for future neural repair and regenerative medicine.
... MSCs, a promising cell therapy strategy in tissue repair and regenerative medicine, exert their therapeutic effects, including immune regulation, antifibrosis effects and the promotion of tissue regeneration via direct differentiation and paracrine functions [21][22][23]. Recent studies have indicated that the therapeutic potential of MSCs relies mainly on EVs secreted by MSCs [24,25]. ...
Article
Full-text available
Background Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease (ESRD) globally, presenting a significant therapeutic challenge. Extracellular vesicles (EVs) from mesenchymal stem cells (MSCs) have emerged as promising therapeutic agents. This study explored the therapeutic effects and mechanisms of EVs derived from human placental mesenchymal stem cells (hP-MSCs) on DKD. Methods EVs were isolated from cultured hP-MSCs and administered to streptozotocin (STZ)-induced diabetic mice and high glucose–treated glomerular mesangial cells. The therapeutic impact of EVs was assessed through histological analysis and biochemical assays. miR-99b-5p expression in EVs and its role in modulating the mechanistic target of rapamycin (mTOR)/autophagy pathway were examined via western blotting and RT‒qPCR. Results Treatment with hP-MSC-derived EVs significantly alleviated renal fibrosis and improved renal function in DKD models. These EVs were enriched with miR-99b-5p, which targeted and inhibited mTOR signaling, thereby increasing autophagic activity and reducing cellular proliferation and extracellular matrix accumulation in renal tissues. Conclusions hP-MSC-derived EVs can mitigate renal injury in DKD by modulating the miR-99b-5p/mTOR/autophagy pathway. These findings suggest a potential cell-free therapeutic strategy for managing DKD.
Article
Interleukin-4 (IL-4), which is traditionally associated with inflammation, has emerged as a key player in tissue regeneration. Produced primarily by T-helper 2 (Th2) and other immune cells, IL-4 activates endogenous lymphocytes and promotes M2 macrophage polarization, both of which are crucial for tissue repair. Moreover, IL-4 stimulates the proliferation and differentiation of various cell types, contributing to efficient tissue regeneration, and shows promise for promoting tissue regeneration after injury. This review explores the multifaceted roles of IL-4 in tissue repair, summarizing its mechanisms and potential for clinical application. This review delves into the multifaceted functions of IL-4, including its immunomodulatory effects, its involvement in tissue regeneration, and its potential therapeutic applications. We discuss the mechanisms underlying IL-4-induced M2 macrophage polarization, a crucial process for tissue repair. Additionally, we explore innovative strategies for delivering IL-4, including gene therapy, protein-based therapies, and cell-based therapies. By leveraging the regenerative properties of IL-4, we can potentially develop novel therapies for various diseases, including chronic inflammatory disorders, autoimmune diseases, and organ injuries. While early research has shown promise for the application of IL-4 in regenerative medicine, further studies are needed to fully elucidate its therapeutic potential and optimize its use.
Article
Full-text available
Patients with refractory immune thrombocytopenia (ITP) frequently encounter substantial bleeding risks and demonstrate limited responsiveness to existing therapies. Umbilical cord-derived mesenchymal stem cells (UC-MSCs) present a promising alternative, capitalizing on their low immunogenicity and potent immunomodulatory effects for treating diverse autoimmune disorders. This prospective phase I trial enrolled eighteen eligible patients to explore the safety and efficacy of UC-MSCs in treating refractory ITP. The research design included administering UC-MSCs at escalating doses of 0.5 × 106 cells/kg, 1.0 × 106 cells/kg, and 2.0 × 106 cells/kg weekly for four consecutive weeks across three cohorts during the dose-escalation phase, followed by a dose of 2.0 × 106 cells/kg weekly for the dose-expansion phase. Adverse events, platelet counts, and changes in peripheral blood immunity were monitored and recorded throughout the administration and follow-up period. Ultimately, 12 (with an addition of three patients in the 2.0 × 106 cells/kg group due to dose-limiting toxicity) and six patients were enrolled in the dose-escalation and dose-expansion phase, respectively. Thirteen patients (13/18, 72.2%) experienced one or more treatment emergent adverse events. Serious adverse events occurred in four patients (4/18, 22.2%), including gastrointestinal hemorrhage (2/4), profuse menstruation (1/4), and acute myocardial infarction (1/4). The response rates were 41.7% in the dose-escalation phase (5/12, two received 1.0 × 106 cells/kg per week, and three received 2.0 × 106 cells/kg per week) and 50.0% (3/6) in the dose-expansion phase. The overall response rate was 44.4% (8/18) among all enrolled patients. To sum up, UC-MSCs are effective and well tolerated in treating refractory ITP (ClinicalTrials.gov ID: NCT04014166).
Article
Full-text available
Mesenchymal stem cells (MSCs) have gained substantial attention in regenerative medicine due to their multi-lineage differentiation potential and immunomodulatory capabilities. MSCs have demonstrated therapeutic promise in numerous preclinical and clinical studies across a variety of diseases, including neurodegenerative disorders, cardiovascular diseases, and autoimmune conditions. Recently, priming MSCs has emerged as a novel strategy to enhance their therapeutic efficacy by preconditioning them for optimal survival and function in challenging in vivo environments. This study presented a comprehensive bibliometric analysis of research activity in the field of priming mesenchymal stem cells (MSCs) from 2003 to 2023. Utilizing a dataset of 585 documents, we explored research trends, leading authors and countries, productive journals, and frequently used keywords. We also explored priming strategies to augment the therapeutic efficacy of MSCs. Our findings show increasing research productivity with a peak in 2019, identified the United States as the leading contributor, and highlighted WANG JA as the most prolific author. The most published journal was Stem Cell Research & Therapy. Keyword analysis revealed core research areas emerging hotspots, while coword and cited sources visualizations elucidated the conceptual framework and key information sources. Further studies are crucial to advance the translation of primed MSCs from bench to bedside, potentially revolutionizing the landscape of regenerative medicine.
Article
Full-text available
Mesenchymal stem cells (MSCs) are multipotent stem cells with the capacity of self-renewal, homing, and low immunogenicity. These distinct biological characteristics have already shown immense potential in regenerative medicine. MSCs also possess immunomodulatory properties that can maintain immune homeostasis when the immune response is over-activated or under-activated. The secretome of MSCs consists of cytokines, chemokines, signaling molecules, and growth factors, which effectively contribute to the regulation of immune and inflammatory responses. The immunomodulatory effects of MSCs can also be achieved through direct cell contact with microenvironmental factors and immune cells. Furthermore, preconditioned and engineered MSCs can specifically improve the immunomodulation effects in diverse clinical applications. These multifunctional properties of MSCs enable them to be used as a prospective therapeutic strategy to treat immune disorders, including autoimmune diseases and incurable inflammatory diseases. Here we review the recent exploration of immunomodulatory mechanisms of MSCs and briefly discuss the promotion of the genetically engineered MSCs. Additionally, we review the potential clinical applications of MSC-mediated immunomodulation in four types of immune diseases, including systemic lupus erythematosus, Crohn’s disease, graft-versus-host disease, and COVID-19.
Article
Full-text available
Background Intravenous administration of mesenchymal stromal cells (MSCs) has an acknowledged competence of cardiac repair, despite a lack of systematic description of the underlying biological mechanisms. The lung, but not the heart, is the main trapped site for intravenously transplanted MSCs, which leaves a spatial gap between intravenously transplanted MSCs and the injured myocardium. How lung-trapped MSCs after intravenous transplantation rejuvenate the injured myocardium remains unknown. Methods MSCs were isolated from human placenta tissue, and DF-MSCs or Gluc-MSCs were generated by transduced with firefly luciferase (Fluc)/enhanced green fluorescence protein (eGFP) or Gaussia luciferase (Gluc) lactadherin fusion protein. The therapeutic efficiency of intravenously transplanted MSCs was investigated in a murine model of doxorubicin (Dox)-induced cardiotoxicity. Trans-organ communication from the lung to the heart with the delivery of blood was investigated by testing the release of MSC-derived extracellular vesicles (MSC-EVs), and the potential miRNA inner MSC-EVs were screened out and verified. The potential therapeutic miRNA inner MSC-EVs were then upregulated or downregulated to assess the further therapeutic efficiency Results Dox-induced cardiotoxicity, characterized by cardiac atrophy, left ventricular dysfunction, and injured myocardium, was alleviated by consecutive doses of MSCs. These cardioprotective effects might be attributed to suppressing GRP78 triggering endoplasmic reticulum (ER) stress-induced apoptosis in cardiomyocytes. Our results confirmed that miR-181a-5p from MSCs-derived EVs (MSC-EVs) inhibited GRP78. Intravenous DF-MSCs were trapped in lung vasculature, secreted a certain number of EVs into serum, which could be confirmed by the detection of eGFP+ EVs. GLuc activity was increased in serum EVs from mice administrated with GLuc-MSCs. MiR-181a-5p, inhibiting GRP78 with high efficacy, was highly expressed in serum EVs and myocardium after injecting consecutive doses of MSCs into mice treated with Dox. Finally, upregulation or downregulation of miR-181a-5p levels in MSC-EVs enhanced or weakened therapeutic effects on Dox-induced cardiotoxicity through modulating ER stress-induced apoptosis. Conclusions This study identifies intravenously transplanted MSCs, as an endocrine reservoir, to secrete cardioprotective EVs into blood continuously and gradually to confer the trans-organ communication that relieves Dox-induced cardiotoxicity.
Article
Full-text available
Stem cells hold great promise for cell therapy to treat a wide spectrum of intractable diseases. Despite enthusiasm for stem cell therapy, the clinical and translational research of stem cells overall has been a slow and cumbersome process. This article uses the “technological system” as a framework to analyze the Tianjin model of stem cell translational medicine. It shows how heterogeneous elements interact with one another and relate to scientific, technological, social, economic, and political variables in order to fulfill the system goal of producing cell therapy in China. Then the strengths and weaknesses of the Tianjin model are compared with translational programs in other countries and the implications for the cell therapy industry are discussed.
Article
Full-text available
Mesenchymal stem/stromal cells (MSCs) have broad application prospects for regenerative medicine due to their self‐renewal, high plasticity, ability for differentiation, and immune response and modulation. Interest in turning MSCs into clinical applications has never been higher than at present. Many biotech companies have invested great effort from development of clinical grade MSC product to investigational new drug (IND) enabling studies. Therefore, the growing demand for publication of MSC regulation in China necessitates various discussions in accessible professional journals. The National Medical Products Administration has implemented regulations on the clinical application of MSCs therapy. The regulations for MSCs products as drug have been updated in recent years in China. This review will look over the whole procedure in allogeneic MSC development, including regulations, guidance, processes, quality management, pre‐IND meeting, and IND application for obtaining an approval to start clinical trials in China. The review focused on process and regulatory challenges in the development of MSCs products, with the goal of providing strategies to meet regulatory demands. This article describes a path for scientists, biotech companies, and clinical trial investigators toward the successful development of MSC‐based therapeutic product.
Article
Full-text available
Here we presented a case of a 19-month-old boy who developed severe aplastic anemia postacute hepatitis. He was treated successfully with the umbilical cord-derived mesenchymal stromal cells (UC-MSCs) infusion and cyclosporine A (CsA). The boy achieved both hematopoietic recovery and normal lymphocyte proportion. So far, his condition still remains stable. To our knowledge, there is a rare previous report on the utility of MSCs infusion for the treatment of hepatitis-associated aplastic anemia (HAAA). Considering the efficacy, safety, and strong operability, particularly for pediatric patient, the infusion of UC-MSCs combined with CsA could be an effective alternative for the treatment of HAAA.
Article
Full-text available
Mesenchymal stem cells (MSCs) show homing capacity towards tumor sites. Numerous reports indicate that they are involved in multiple tumor-promoting processes through several mechanisms, including immunosuppression; stimulation of angiogenesis; transition to cancer-associated fibroblasts; inhibition of cancer cell apoptosis; induction of epithelial–mesenchymal transition (EMT); and increase metastasis and chemoresistance. However, other studies have shown that MSCs suppress tumor growth by suppressing angiogenesis, incrementing inflammatory infiltration, apoptosis and cell cycle arrest, and inhibiting the AKT and Wnt signaling pathways. In this review, we discuss the supportive and suppressive impacts of MSCs on tumor progression and metastasis. We also discuss MSC-based therapeutic strategies for cancer based on their potential for homing to tumor sites.
Article
Full-text available
Background: Mesenchymal stem cell (MSC)-based therapies hold great promise for the treatment of inflammatory bowel disease (IBD). In order to optimize and maximize the therapeutic benefits of MSCs, we investigated whether cotransplantation of a chitosan (CS)-based injectable hydrogel with immobilized IGF-1 C domain peptide (CS-IGF-1C) and human placenta-derived MSCs (hP-MSCs) could ameliorate colitis in mice. ¬ Methods: IGF-1C hydrogel was generated by immobilizing IGF-1C to CS hydrogel. Colitis was induced by 2,4,6-trinitrobenzene sulfonic acid (TNBS) in mice. We initially applied hP-MSCs and CS-IGF-1C hydrogel for the treatment of colitis by in situ injection, and molecular imaging methods were used for real-time imaging of reactive oxygen species (ROS) and tracking of transplanted hP-MSCs by bioluminescence imaging (BLI). Furthermore, the effects of CS-IGF-1C hydrogel on prostaglandin E2 (PGE2) secretion of hP-MSCs and polarization of M2 macrophages were investigated as well. Results: The CS-IGF-1C hydrogel significantly increased hP-MSC proliferation and promoted the production of PGE2 from hP-MSCs in vitro. Moreover, in vivo studies indicated that the CS-IGF-1C hydrogel promoted hP-MSC survival as visualized by BLI and markedly alleviated mouse colitis, which was possibly mediated by hP-MSC production of PGE2 and interleukin-10 (IL-10) production by polarized M2 macrophages. Conclusions: The CS-IGF-1C hydrogel improved the engraftment of transplanted hP-MSCs, ameliorated inflammatory responses, and further promoted the functional and structural recovery of colitis through PGE2-mediated M2 macrophage polarization. Molecular imaging approaches and therapeutic strategies for hydrogel application provide a versatile platform for exploring the promising therapeutic potential of MSCs in the treatment of IBD.
Book
This book provides a comprehensive introduction to various types of perinatal stem cells. Given their unique regenerative abilities, stem cells offer a promising avenue in the treatment of degenerative diseases or injury. Currently, the limitations of postnatal cell sources and expanding efficiency may limit autologous stem cell therapies. Although embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) can be cultured indefinitely ex vivo, and can differentiate into three germ layers, ethical issues, the teratoma formation of ESCs and oncogenic risk of iPSCs are major obstacles to their clinical application. More recently, perinatal stem cells have been isolated from the umbilical cord, Wharton’s Jelly, placenta, amniotic membrane and amniotic fluid, which are normally discarded as medical waste. This book, after describing perinatal stem cells in detail, introduces readers to the various types of perinatal stem cells, addressing their characterization, banking, quality control and stability. Importantly, it also reviews the clinical applications of perinatal stem cells to therapy of diseases. Accordingly, it offers a valuable resource for clinicians, researchers and graduate students alike.