Preprint
Review

This version is not peer-reviewed.

Mesenchymal Stem/Stromal Cells: A Review for Its Use After Allogeneic Hematopoietic Stem Cell Transplantation

Submitted:

24 October 2025

Posted:

28 October 2025

You are already at the latest version

Abstract
Mesenchymal stem/stromal cells (MSCs) exhibit broad differentiation capability and strong immunoregulatory potential mediated through intercellular communication and the release of diverse paracrine mediators. These features render MSCs a promising therapeutic option for managing complications associated with allogeneic hematopoietic stem cell transplantation (allo-HSCT). This review provides an updated synthesis of MSC biology, their bidirectional interaction with immune cells, and their functional contribution to the hematopoietic niche. It also evaluates current clinical evidence regarding the therapeutic roles of MSCs and MSC-derived extracellular vesicles (EVs) in acute and chronic graft-versus-host disease (aGVHD/cGVHD) as well as in poor graft function. Mechanistic insights encompass macrophage polarization toward an anti-inflammatory phenotype, inhibition of dendritic cell maturation, enhancement of regulatory T-cell expansion, and modulation of cytokine signaling pathways. Within the bone marrow milieu, MSCs contribute to stromal restoration and angiogenic repair. Data from phase II/III clinical trials and real-world cohorts in steroid-refractory aGVHD consistently confirm the safety of MSC therapy, although response rates vary—typically higher in pediatric patients and when administered early—while survival outcomes remain inconsistent. For poor graft function, limited prospective studies indicate hematopoietic recovery following third-party MSC infusions, and combination approaches such as co-administration with thrombopoietin receptor agonists are under investigation. MSC-derived EVs emulate many immunomodulatory effects of their parental cells with a potentially safer profile, though clinical validation remains in its infancy. MSC-oriented interventions hold substantial biological and therapeutic promise, offering a favorable safety margin; however, clinical translation is hindered by product variability, suboptimal engraftment and persistence, and inconsistent efficacy across studies. Future directions should emphasize standardized manufacturing and potency assays, biomarker-driven patient and timing selection, optimized conditioning and dosing strategies, and the systematic appraisal of EV-based or genetically modified MSC products through controlled trials.
Keywords: 
;  ;  ;  ;  ;  

1. Introduction

MSCs were initially defined in the late 1960s as bone marrow (BM) cells, having fibroblast-like morphology, with self-renewal capability [1]. Subsequent studies also demonstrated that MSCs were able to differentiate into several lineages, such as epithelial, neuronal, astrocytic, endothelial, and smooth muscle cells in vitro, in a coculture fashion with different cell types when subjected to characterizing conditions [2,3,4,5,6,7]. Furthermore, MSCs can differentiate into other cell lineages for the enhancement of inflammatory reactions [8]. These mirror the MSCs’ role of maintaining tissue homeostasis and improving structural integrity.
Given their MSCs’ potent immunomodulatory capacity, plasticity and self-renewal ability they have started to find space in various treatment schemes for wide array of disorders. MSCs mediate potent immunomodulatory effects via direct cell–cell contact and through release of bioactive molecules that regulate both innate and adaptive immune cells [9]. In the bone marrow, MSCs at various stages of maturation constitute the hematopoietic stem cell (HSC) niche and have an important function in HSCs maintenance and renewal [10]. These findings pave the way for using MSCs in acute graft-versus-host disease (aGVHD) and graft failures after allogeneic HSC transplants, which constitutes the major fields for MSC treatment in malignant hematology [11,12]. Survival and homing issues are the major drawbacks for the MSC treatment, thus optimizing the conditioning protocols and maintaining the intact paracrine signaling should be the first priorities mainly in the context of HSC transplantation [13].
MSC-Derived Extracellular Vesicles (MSC-EVs), offer some therapeutic potential like their parental cells. MSC-EVs contain exosomes (30–120 nm, secreted from multivesicular endocytic structures), macrovesicles (100–1000 nm, from the plasma membrane), and apoptotic bodies [9]. They control immune responses by inhibiting Th17 differentiation and induce peripheral tolerance through PD-L1 and PD-L2. There is some evidence about the use of exosomes in human disease, others are ongoing [14,15,16].
In this review, we aimed to review the relevant literature about MSC biology and its interactions with the immune system and other tissues with a special attention to HSC and bone marrow microenvironment. Also, therapeutic uses of MSCs were summarized.

2. Mesenchymal Stem Cell: Origin, Biology and Functions

The discovery of MSCs can be traced back to the mid-1970s, when fibroblast-like adherent cells were first isolated from bone marrow and shown to form colony-forming units capable of differentiating into osteogenic lineages and supporting hematopoietic growth [17]. This foundational finding established the concept of bone marrow–derived stromal cells as a distinct progenitor population. Subsequent studies demonstrated that these stromal cells retained their osteogenic potential even after extensive passaging, and by the late 1980s, they were recognized for their ability to generate bone and cartilage tissue in vivo, underscoring their multilineage differentiation capacity [17]. A major conceptual shift occurred in 1991, when the term mesenchymal stem cell was coined to describe these multipotent cells of mesodermal origin. This marked the formal recognition of MSCs as a self-renewing stem cell population distinct from fibroblasts, despite their similar morphology in vitro [18]. Many studies are using the given nomenclature in recent years.
International Society for Cellular Therapy has defined the immunophenotype of MSC [19]. Based on this definition, the cell surface markers CD73, CD90 and CD105 should be exhibited while CD14, CD34, CD45, CD11b, CD79, and histocompatibility complex molecule HLA-DR should be lacking [19]. Additionally, cell culture conditions and transforming capabilities of MSCs were addressed [19].
MSCs perform vital functions in destroyed tissue repair by immunosuppressive, antiapoptotic, anti-inflammatory, antifibrotic, pro-angiogenic, antitumorigenic, neuroprotective, antibacterial, and chemo attractive activities [20]. MSCs are expanded within the marrow microenvironment and received signals which specify their fate. These signals include cell-to-cell interactions, cell-to-matrix interactions and transcriptional program that activates and/or suppresses MSC genes [21].
In addition to their multipotency, MSCs have immunomodulatory activities that have been studied as a therapy for several immune-related diseases. They mediate potent immunomodulatory effects via direct cell–cell contact and through release of bioactive molecules that regulate both innate and adaptive immune cells [9]. Additionally, MSCs do not express human leukocyte antigen (HLA) Class II and can be administered without needing to match donors to recipients. This immunomodulatory phenotype enables MSCs to be used from one donor to various recipients without immune-mediated rejection [9,19].

2.1. Interactions with Immune System Cells

Preclinical data suggest that MSCs control the innate and adaptive immune response with direct interaction with T cells, B cells, neutrophils, monocytes, macrophages, NK cells, and DCs, displaying broad immunomodulatory effects by cell-to-cell contact and paracrine action [22,23,24]. MSCs predominantly control macrophages by secreting soluble factors [25]. Through the production of PGE2, TGF-β, and CCL2, MSCs can induce polarization of macrophages from the pro-inflammatory M1 phenotype toward the anti-inflammatory M2 phenotype [26].
IL-17 from MSCs may activate neutrophils and enhance their phagocytic activity. MSCs obtained from bone marrow decrease the release of reactive oxygen species (ROS) from activated neutrophils and those isolated from umbilical cord tissue dampen the neutrophil-associated inflammation [27].
MSC decreases the differentiation, maturation, and antigen-presenting function of Dendritic cells (DCs) with consequent reduction in pro-inflammatory cytokines. Furthermore, MSCs block the generation of immature DCs and impair CD34+ precursor differentiation into epidermal DCs [28].
By keeping macrophages and DCs in an immature or anti-inflammatory state, MSCs reduce the activation of effector T cells and favor the generation of regulatory T (Treg) cells [29,30]. Regulatory T cells (Tregs), a sub-population of CD4+ T cells that express Forkhead box P3 (FoxP3) and CD25 (IL-2 receptor) on their surface, have strong immunosuppressive and anti-inflammatory activities [31]. MSCs might also drive the expansion and differentiation of Tregs via an HLA-G5-dependent mechanism by the PGE2 and TGF-β1 released from MSCs, and IL-10 secreted by Th2 cells [32]. MSC may also induce CD4(+) T cell towards Tregs by Notch-1 signaling. Notch1/FoxP3 pathway is also activated in CD4+ T cells co-cultured with MSCs, which promotes the percentage of CD4+CD25^highFoxP3+ Tregs [33,34]. Blockade of TGF-β as well as IL-10 abolishes the induction of Treg, emphasizing their crucial roles in immune tolerance [34].
Captivatingly, galectin-1, is expressed mostly in MSCs where it regulates cytokine secretion in GvHD and autoimmunity. Suppression of galectin-1 reverts immunosuppression of MSCs on allo-immune T cells by rescuing responses of CD4+ and CD8+ T cells [35,36]. MSCs also can express abundant Toll-like receptor (TLR) molecules—for example, TLR-3 and TLR-4—which activate nuclear factor κB (NF-κB) and subsequently enhance secretion of pro-inflammatory cytokines, including CXCL10, IL-6, and IL-8. TLR immune activation has inhibitory effect over MSC immunosuppression during RNA-virus and bacterial infections [37].

2.2. Tissue Protection and Regeneration

Others with immunomodulation, MSCs reduce the damage and restrict the repair of the tissue through paracrine actions on the endogenous recipient cells [38]. From hematology perspective, the most prominent feature of MSCs is being a potential regenerator for bone marrow stroma in poor graft function (PGF) [12].
MSCs exert a multifaceted therapeutic influence in the setting of poor graft function (PGF) following allogeneic hematopoietic stem cell transplantation. As essential components of the bone marrow microenvironment, MSCs provide structural and trophic support to hematopoietic stem and progenitor cells (HSPCs) through the secretion of stem cell factors (SCF), CXCL12, IL-6, and vascular endothelial growth factor (VEGF), all of which promote HSPC survival, homing, and proliferation [12]. In the context of PGF, where the stromal niche is often damaged or functionally exhausted, exogenously infused MSCs help restore stromal integrity and reconstitute the hematopoietic niche [12].
Beyond this supportive role, MSCs secrete angiogenic factors such as VEGF, hepatocyte growth factor (HGF), and angiopoietin-1, which facilitate endothelial repair and microvascular regeneration, thereby improving marrow perfusion and oxygenation [39]. Their potent immunomodulatory properties—marked by the suppression of alloreactive T cells and natural killer cells, expansion of regulatory T cells, and modulation of cytokine networks toward an anti-inflammatory profile—further mitigate immune-mediated marrow injury [39]. In addition, MSC-derived paracrine signals and extracellular vesicles confer anti-apoptotic and anti-fibrotic effects by enhancing progenitor cell survival and remodeling the extracellular matrix. Collectively, these biological attributes enable MSCs to simultaneously repair the marrow microenvironment, rebalance immune homeostasis, and promote durable hematopoietic recovery, providing a strong mechanistic rationale for their therapeutic application in PGF [40,41].

3. Beyond the Cells: Exosomes

An exosome is a small, membrane-bound extracellular vesicle (EV) that is released by nearly all cell types into the extracellular environment [42]. Originally believed to be degrade plasma membrane, the immunological activities of exosomes were first demonstrated in 1996 by Raposo et al. showed that MHC II molecules on CD4+ T cells can present antigens to B cell–derived exosomes [43]. This momentous discovery underscored their significance in shaping adaptive immune responses and inspired hope of exosome-based immunotherapy.
Exosomes have key functions in intercellular communication via the delivery of proteins, lipids, mRNAs, and miRNAs between cells [43]. They are associated with a variety of physiological and pathophysiological conditions including cancer [44]. MSCs exert their immunomodulatory functions partly through their secretome, a complex mixture of cytokines, growth factors, chemokines, and extracellular vesicles (MSC-EVs) that collectively regulate immune cell behavior [9]. The MSC-derived vesicles modulate immune activity by suppressing Th17 differentiation and reducing the secretion of pro-inflammatory cytokines such as TNF-α, IL-17, IL-22, and IFN-γ [16]. Additionally, MSC-EVs promote peripheral immune tolerance through the expression of checkpoint molecules PD-L1 and PD-L2 [16].

4. MSCs for the Treatment of GVHD

The attack of the host tissues by donor T cells is known as GVHD and is a well-documented response following allogeneic HSCT. It is typically divided into acute (aGVHD) or chronic (cGVHD), depending on whether it occurs before or after post-transplant day 100, as historically defined [45]. Yet this distinction is arbitrary along the time axis, since aGVHD can continue over 100 days, and there is often an overlap between the acute and chronic form [45].
Graft-versus-host disease (GVHD) is initiated by tissue damage incurred by the conditioning, which liberates pro-inflammatory cytokines that serve prime donor T cells. These donor T cells respond to host major and minor histocompatibility antigens, migrate to target organs, and recruit effector cells, including cytotoxic T cells and NK cells. Effector cells induce tissue damage through either direct cytotoxicity and/or release of cytokines to augment inflammation. The skin, the GI tract, and the liver are the most frequently involved organs [46]. GVHD causes death in 8–16% of adult HSCT recipients [47], but this is likely an underestimation since GVHD increases the risk for secondary causes of death, i.e., infection, organ failure, and bleeding.
Chronic GVHD (cGVHD) is associated with persistent inflammation; elimination of tolerance; dysregulation of T cells and B cells, and defective regulatory subsets. Activated (i.e., matrix-producing) myofibroblasts, activated by cytokines such as platelet-derived growth factor α (PDGFα) and transforming growth factor β (TGFβ), provoke fibrosis that can lead to involvement of multiple tissues. Cytokines such as inflammatory chemokines, CXCL9, CXCL10, and B-cell activating factor (BAFF) have shown to be increased in cGVHD [48]. Elevated CXCL9 at day +100 has been linked with progressive severe cGVHD, and elevated CXCL10 was described in patients with early rather than chronic manifestations [49]. Different metabolic profiles may be also useful to differentiate active inflammation from irreversible tissue destruction [50,51].
Corticosteroids continue to be the mainstay of primary treatment for aGVHD and cGVHD, although outcomes are mixed and poor in steroid-refractory patients [52]. Several other agents like Janus Kinase (JAK) inhibitor ruxolitinib and Rho-associated coiled-coil containing protein kinase 2 (ROCK2) inhibitor belumosidil were demonstrated as effective options and was approved by the Food and Drug Administration (FDA) for acute and chronic forms of the disease [53,54]. Nevertheless, 50–60% of patients require additional “steroid-sparing” modalities at any point of the course [52].
Based on early in vitro evidence of the potent immunomodulatory properties of MSCs, their adoptive transfer was proposed for the treatment of steroid-refractory acute graft-versus-host disease (SR-aGVHD) [11]. The first successful case in 2004 showed resolution of severe gastrointestinal and hepatic GVHD after infusion of haploidentical maternal MSCs [11], followed by a European phase II trial in which 55 patients achieved an overall response (OR) rate of 71% with significantly improved survival among complete responders [55]. Subsequent meta-analyses confirmed the safety of systemic MSC therapy with minimal toxicity and no increased risk of relapse or malignancy, though occasional late infectious events were reported [56,57,58,59,60]. In later phase II–III studies, including a U.S. multicenter trial of Remestemcel-L (Prochymal), MSCs were well tolerated but failed to meet primary endpoints; subgroup analyses suggested higher response rates in liver GVHD and among pediatric patients [61,62]. Consistent findings were reported in European, Asian, and Chinese studies showing ORs of 48–71% and improved survival among responders [34,63,64,65,66,67,68]. Real-world data from Japan with Temcell demonstrated organ-specific ORs (36–64%) and highlighted favorable outcomes in younger patients and early intervention [69]. Collectively, these findings indicate that MSCs provide a safe adjunctive option for SR-aGVHD. A summary of relevant studies displayed in Table 1.

5. MSC for the Treatment of Poor Graft Function

Poor graft function (PGF) is a rare yet serious complication following allogeneic hematopoietic cell transplantation (alloHCT), distinct from graft failure or GVHD. It is characterized by persistent cytopenia requiring frequent transfusions or growth factor support, despite full donor chimerism and the absence of relapse or other causes [81]. PGF leads to significant morbidity, including recurrent infections, bleeding, iron overload, and repeated hospitalizations [81,82]. Current management strategies—such as thrombopoietin receptor agonists (TPO-RAs), donor stem cell boosts, or second transplants—are often limited by inconsistent efficacy, treatment-related toxicity, and the risk of GVHD [83,84,85]. In this context, MSCs have emerged as a promising alternative. The bone marrow microenvironment is frequently damaged in transplant recipients due to underlying malignancy, conditioning therapy, or immune injury [85]. Since MSCs play a key role in maintaining hematopoietic homeostasis through secretion of cytokines and growth factors and by modulating immune responses, their use may restore stromal support and promote hematopoietic recovery in PGF [86].
Clinical studies have explored various strategies combining MSCs with other agents to improve hematopoietic recovery in PGF. In one study, Zhu et al. treated 16 patients with severe post-transplant thrombocytopenia using weekly infusions of umbilical cord–derived MSCs (1×10⁶ cells/kg for 4–6 doses) plus avatrombopag, a second-generation thrombopoietin receptor agonist (TPO-RA), achieving platelet counts >50×10⁹/L in 13 patients within a median of 32 days [87]. Similarly, a multicenter prospective trial using single or repeated intravenous infusions of third-party bone marrow–derived MSCs (1–2×10⁶ cells/kg) in 30 PGF patients produced complete hematologic responses in one-third of cases [12].
As of June 2021, 43 interventional or expanded-access MSC trials for GVHD had been registered on clinicaltrials.gov, including 19 completed and 10 with published outcomes. A broader review of 55 clinical studies encompassing 2,696 patients confirmed the excellent safety profile of MSC therapy, with only transient fever reported as a consistent adverse event and no observed increase in infusion-related toxicity, infection, thrombosis, death, or secondary malignancy [88].

6. Conclusions and Future Perspectives

Because of the multipotent nature and the immunomodulatory capacity of MSCs, they are highly promising for applications in regenerative medicine and immune-related diseases. These effects are mainly mediated by direct contact with immune cells along with paracrine action of chemokines, cytokines, growth factors, other inflammatory signals. Moreover, MSCs derived exosomes are an exciting therapeutic candidate, bearing the therapeutic attributes of their mother cells and with improved safety profile which minimizes concerns on tumorigenesis and genetic instability.
However, the clinical use of MSCs as a therapy is still at issue even though remarkable advances have been achieved. As of now, robust and conclusive evidence with respect to all patient cohorts has not been provided. Importantly, MSCs are heterogeneous, and their function mathematically modeled which depends greatly on anti-inflammatory as well as inflammatory microenvironments has been leading to the difference in immunomodulatory results. Acute and chronic inflammation-induced alterations of MSC function need to be further studied.
Problems remaining are the survival time and homing capacity of infused MSCs, which are determined by the local microenvironment around lesions. However, optimizing the preconditioning protocols for improving the therapeutic efficacy and paracrine stability of MSCs and maximizing their therapeutic effects is still a priority, especially in the clinical context of transplantation.

Author Contributions

Conceptualization: A.D., U.H., H.E., T.U., M.S.D., F.A.; Methodology: A.D., U.H., H.E., T.U., M.S.D., F.A.; Writing—original draft preparation, A.D., U.H., H.E., T.U., M.S.D., F.A; writing—review and editing, A.D., U.H., H.E., T.U., M.S.D., F.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare that there is no conflict of interest.

References

  1. Salem HK, Thiemermann C. Mesenchymal stromal cells: current understanding and clinical status. Stem Cells. 2010; 28(3): 585-96. [CrossRef]
  2. Spees JL, Olson SD, Ylostalo J, Lynch PJ, Smith J, Perry A, Peister A, Wang MY, Prockop DJ. Differentiation, cell fusion, and nuclear fusion during ex vivo repair of epithelium by human adult stem cells from bone marrow stroma. Proc Natl Acad Sci U S A. 2003; 100(5): 2397-402. [CrossRef]
  3. Phinney DG, Isakova I. Plasticity and therapeutic potential of mesenchymal stem cells in the nervous system. Curr Pharm Des. 2005; 11(10): 1255-65. [CrossRef]
  4. Tropel P, Platet N, Platel JC, Noël D, Albrieux M, Benabid AL, Berger F. Functional neuronal differentiation of bone marrow-derived mesenchymal stem cells. Stem Cells. 2006; 24(12): 2868-76. Erratum in: Stem Cells. 2007; 25(2): 543. [CrossRef]
  5. Sivamani RK, Schwartz MP, Anseth KS, Isseroff RR. Keratinocyte proximity and contact can play a significant role in determining mesenchymal stem cell fate in human tissue. FASEB J. 2011; 25(1): 122-31. [CrossRef]
  6. Reyhani S, Abbaspanah B, Mousavi SH. Umbilical cord-derived mesenchymal stem cells in neurodegenerative disorders: from literature to clinical practice. Regen Med. 2020; 15(4): 1561-1578. [CrossRef]
  7. Yang G, Fan X, Mazhar M, Yang S, Xu H, Dechsupa N, Wang L. Mesenchymal Stem Cell Application and Its Therapeutic Mechanisms in Intracerebral Hemorrhage. Front Cell Neurosci. 2022; 16: 898497. [CrossRef]
  8. Leuning DG, Beijer NRM, du Fossé NA, Vermeulen S, Lievers E, van Kooten C, Rabelink TJ, Boer J. The cytokine secretion profile of mesenchymal stromal cells is determined by surface structure of the microenvironment. Sci Rep. 2018; 8(1): 7716. [CrossRef]
  9. Li N, Hua J. Interactions between mesenchymal stem cells and the immune system. Cell Mol Life Sci. 2017; 74(13): 2345-2360. [CrossRef]
  10. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008; 8(9): 726-36. [CrossRef]
  11. Le Blanc K, Rasmusson I, Sundberg B, Götherström C, Hassan M, Uzunel M, Ringdén O. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet. 2004; 363(9419): 1439-41. [CrossRef]
  12. Liu X, Wu M, Peng Y, Chen X, Sun J, Huang F, Fan Z, Zhou H, Wu X, Yu G, Zhang X, Li Y, Xiao Y, Song C, Xiang AP, Liu Q. Improvement in poor graft function after allogeneic hematopoietic stem cell transplantation upon administration of mesenchymal stem cells from third-party donors: a pilot prospective study. Cell Transplant. 2014; 23(9): 1087-98. [CrossRef]
  13. 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. [CrossRef]
  14. Ghannam S, Pène J, Moquet-Torcy G, Jorgensen C, Yssel H. Mesenchymal stem cells inhibit human Th17 cell differentiation and function and induce a T regulatory cell phenotype. J Immunol. 2010; 185(1): 302-12. Erratum in: J Immunol. 2013; 191(11): 5777. [CrossRef]
  15. Davies LC, Heldring N, Kadri N, Le Blanc K. Mesenchymal Stromal Cell Secretion of Programmed Death-1 Ligands Regulates T Cell Mediated Immunosuppression. Stem Cells. 2017; 35(3): 766-776. [CrossRef]
  16. Tang Y, Zhou Y, Li HJ. Advances in mesenchymal stem cell exosomes: a review. Stem Cell Res Ther. 2021; 12(1): 71. [CrossRef]
  17. Han X, Liao R, Li X, Zhang C, Huo S, Qin L, Xiong Y, He T, Xiao G, Zhang T. Mesenchymal stem cells in treating human diseases: molecular mechanisms and clinical studies. Signal Transduct Target Ther. 2025; 10(1): 262. [CrossRef]
  18. Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991; 9(5): 641-50. [CrossRef]
  19. Mahla RS. Stem Cells Applications in Regenerative Medicine and Disease Therapeutics. Int J Cell Biol. 2016; 2016: 6940283. [CrossRef]
  20. Galipeau J, Sensébé L. Mesenchymal Stromal Cells: Clinical Challenges and Therapeutic Opportunities. Cell Stem Cell. 2018; 22(6): 824-833. [CrossRef]
  21. Sobacchi C, Palagano E, Villa A, Menale C. Soluble Factors on Stage to Direct Mesenchymal Stem Cells Fate. Front Bioeng Biotechnol. 2017; 5: 32. [CrossRef]
  22. Zhou Y, Yamamoto Y, Xiao Z, Ochiya T. The Immunomodulatory Functions of Mesenchymal Stromal/Stem Cells Mediated via Paracrine Activity. J Clin Med. 2019; 8(7): 1025. [CrossRef]
  23. Chen J, Zheng CX, Jin Y, Hu CH. Mesenchymal stromal cell-mediated immune regulation: A promising remedy in the therapy of type 2 diabetes mellitus. Stem Cells. 2021; 39(7): 838-852. [CrossRef]
  24. Reis M, Mavin E, Nicholson L, Green K, Dickinson AM, Wang XN. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Attenuate Dendritic Cell Maturation and Function. Front Immunol. 2018; 9: 2538. [CrossRef]
  25. Lu D, Xu Y, Liu Q, Zhang Q. Mesenchymal Stem Cell-Macrophage Crosstalk and Maintenance of Inflammatory Microenvironment Homeostasis. Front Cell Dev Biol. 2021; 9: 681171. [CrossRef]
  26. Vasandan AB, Jahnavi S, Shashank C, Prasad P, Kumar A, Prasanna SJ. Human Mesenchymal stem cells program macrophage plasticity by altering their metabolic status via a PGE2-dependent mechanism. Sci Rep. 2016; 6: 38308. [CrossRef]
  27. Salami F, Tavassoli A, Mehrzad J, Parham A. Immunomodulatory effects of mesenchymal stem cells on leukocytes with emphasis on neutrophils. Immunobiology. 2018; 223(12): 786-791. [CrossRef]
  28. Ramasamy R, Fazekasova H, Lam EW, Soeiro I, Lombardi G, Dazzi F. Mesenchymal stem cells inhibit dendritic cell differentiation and function by preventing entry into the cell cycle. Transplantation. 2007; 83(1): 71-6. Erratum in: Transplantation. 2007; 83(11): 1524. [CrossRef]
  29. Gao F, Chiu SM, Motan DA, Zhang Z, Chen L, Ji HL, Tse HF, Fu QL, Lian Q. Mesenchymal stem cells and immunomodulation: current status and future prospects. Cell Death Dis. 2016; 7(1): e2062. [CrossRef]
  30. Jiang W, Xu J. Immune modulation by mesenchymal stem cells. Cell Prolif. 2020; 53(1): e12712. [CrossRef]
  31. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008; 133(5): 775-87. [CrossRef]
  32. Di Ianni M, Del Papa B, De Ioanni M, Moretti L, Bonifacio E, Cecchini D, Sportoletti P, Falzetti F, Tabilio A. Mesenchymal cells recruit and regulate T regulatory cells. Exp Hematol. 2008; 36(3): 309-18. [CrossRef]
  33. Del Papa B, Sportoletti P, Cecchini D, Rosati E, Balucani C, Baldoni S, Fettucciari K, Marconi P, Martelli MF, Falzetti F, Di Ianni M. Notch1 modulates mesenchymal stem cells mediated regulatory T-cell induction. Eur J Immunol. 2013; 43(1): 182-7. [CrossRef]
  34. Hong JW, Lim JH, Chung CJ, Kang TJ, Kim TY, Kim YS, Roh TS, Lew DH. Immune Tolerance of Human Dental Pulp-Derived Mesenchymal Stem Cells Mediated by CD4⁺CD25⁺FoxP3⁺ Regulatory T-Cells and Induced by TGF-β1 and IL-10. Yonsei Med J. 2017; 58(5): 1031-1039. [CrossRef]
  35. Gieseke F, Böhringer J, Bussolari R, Dominici M, Handgretinger R, Müller I. Human multipotent mesenchymal stromal cells use galectin-1 to inhibit immune effector cells. Blood. 2010; 116(19): 3770-9. [CrossRef]
  36. Laranjeira P, Pedrosa M, Pedreiro S, Gomes J, Martinho A, Antunes B, Ribeiro T, Santos F, Trindade H, Paiva A. Effect of human bone marrow mesenchymal stromal cells on cytokine production by peripheral blood naive, memory, and effector T cells. Stem Cell Res Ther. 2015; 6(1): 3. [CrossRef]
  37. Liotta F, Angeli R, Cosmi L, Filì L, Manuelli C, Frosali F, Mazzinghi B, Maggi L, Pasini A, Lisi V, Santarlasci V, Consoloni L, Angelotti ML, Romagnani P, Parronchi P, Krampera M, Maggi E, Romagnani S, Annunziato F. Toll-like receptors 3 and 4 are expressed by human bone marrow-derived mesenchymal stem cells and can inhibit their T-cell modulatory activity by impairing Notch signaling. Stem Cells. 2008; 26(1): 279-89. [CrossRef]
  38. Spees JL, Lee RH, Gregory CA. Mechanisms of mesenchymal stem/stromal cell function. Stem Cell Res Ther. 2016; 7(1): 125. [CrossRef]
  39. Wu Y, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells. 2007; 25(10): 2648-59. [CrossRef]
  40. Lai RC, Arslan F, Lee MM, Sze NS, Choo A, Chen TS, Salto-Tellez M, Timmers L, Lee CN, El Oakley RM, Pasterkamp G, de Kleijn DP, Lim SK. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010; 4(3): 214-22. [CrossRef]
  41. Rani S, Ryan AE, Griffin MD, Ritter T. Mesenchymal Stem Cell-derived Extracellular Vesicles: Toward Cell-free Therapeutic Applications. Mol Ther. 2015; 23(5): 812-823. [CrossRef]
  42. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013; 200(4): 373-83. [CrossRef]
  43. Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV, Melief CJ, Geuze HJ. B lymphocytes secrete antigen-presenting vesicles. J Exp Med. 1996; 183(3): 1161-72. [CrossRef]
  44. Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, Curry WT Jr, Carter BS, Krichevsky AM, Breakefield XO. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008; 10(12): 1470-6. [CrossRef]
  45. Garnett C, Apperley JF, Pavlů J. Treatment and management of graft-versus-host disease: improving response and survival. Ther Adv Hematol. 2013; 4(6): 366-78. [CrossRef]
  46. Socié G, Ritz J. Current issues in chronic graft-versus-host disease. Blood. 2014; 124(3): 374-84. [CrossRef]
  47. Phelan R, Arora M, Chen M. Current Use and Outcome of Hematopoietic Stem Cell Transplantation: CIBMTR US Summary Slides. (2020). Available at: https://www.cibmtr.org/ReferenceCenter/SlidesReports/SummarySlides/ Pages/index.aspx.
  48. Wolff D, Greinix H, Lee SJ, Gooley T, Paczesny S, Pavletic S, Hakim F, Malard F, Jagasia M, Lawitschka A, Hansen JA, Pulanic D, Holler E, Dickinson A, Weissinger E, Edinger M, Sarantopoulos S, Schultz KR. Biomarkers in chronic graft-versus-host disease: quo vadis? Bone Marrow Transplant. 2018; 53(7): 832-837. [CrossRef]
  49. Goklemez S, Im AP, Cao L, Pirsl F, Steinberg SM, Curtis LM, Mitchell SA, Cowen EW, Baruffaldi J, Rose J, Mays J, Ostojic A, Holtzman NG, Hakim FT, Pavletic SZ. Clinical characteristics and cytokine biomarkers in patients with chronic graft-vs-host disease persisting seven or more years after diagnosis. Am J Hematol. 2020; 95(4): 387-394. [CrossRef]
  50. Subburaj D, Ng B, Kariminia A, Abdossamadi S, Lauener M, Nemecek ER, Rozmus J, Kharbanda S, Kitko CL, Lewis VA, Schechter-Finklestein T, Jacobsohn DA, Harris AC, Pulsipher MA, Bittencourt H, Choi SW, Caywood EH, Kasow KA, Bhatia M, Oshrine BR, Coulter D, Chewning JH, Joyce M, Pawlowska AB, Megason GC, Lawitschka A, Ostroumov E, Klein Geltink R, Cuvelier GDE, Schultz KR. Metabolomic identification of α-ketoglutaric acid elevation in pediatric chronic graft-versus-host disease. Blood. 2022; 139(2): 287-299. [CrossRef]
  51. Cuvelier GDE, Ng B, Abdossamadi S, Nemecek ER, Melton A, Kitko CL, Lewis VA, Schechter T, Jacobsohn DA, Harris AC, Pulsipher MA, Bittencourt H, Choi SW, Caywood EH, Kasow KA, Bhatia M, Oshrine BR, Chaudhury S, Coulter D, Chewning JH, Joyce M, Savaşan S, Pawlowska AB, Megason GC, Mitchell D, Cheerva AC, Lawitschka A, Ostroumov E, Schultz KR. A diagnostic classifier for pediatric chronic graft-versus-host disease: results of the ABLE/PBMTC 1202 study. Blood Adv. 2023; 7(14): 3612-3623. [CrossRef]
  52. Flowers ME, Martin PJ. How we treat chronic graft-versus-host disease. Blood. 2015; 125(4): 606-15. [CrossRef]
  53. Food and Drug Administration. FDA Approves Ruxolitinib for Acute Graft Versus-Host Disease (2019). Available at: https://www.fda.gov/drugs/ resources-information-approved-drugs/fda-approves-ruxolitinib-acute-graft versus-host-disease (Accessed September 29, 2021).
  54. Food and Drug Administration. FDA Approves Belumosudil for Chronic Graft-Versus-Host Disease (2021). Available at: https://www.fda.gov/drugs/ drug-approvals-and-databases/fda-approves-belumosudil-chronic-graft versus-host-disease (Accessed September 29, 2021).
  55. Le Blanc K, Frassoni F, Ball L, Locatelli F, Roelofs H, Lewis I, Lanino E, Sundberg B, Bernardo ME, Remberger M, Dini G, Egeler RM, Bacigalupo A, Fibbe W, Ringdén O; Developmental Committee of the European Group for Blood and Marrow Transplantation. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet. 2008; 371(9624): 1579-86. [CrossRef]
  56. 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(7): 852-862. [CrossRef]
  57. Lalu MM, McIntyre L, Pugliese C, Fergusson D, Winston BW, Marshall JC, Granton J, Stewart DJ; Canadian Critical Care Trials Group. Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systematic review and meta-analysis of clinical trials. PLoS One. 2012; 7(10): e47559. [CrossRef]
  58. Müller I, Kordowich S, Holzwarth C, Isensee G, Lang P, Neunhoeffer F, Dominici M, Greil J, Handgretinger R. Application of multipotent mesenchymal stromal cells in pediatric patients following allogeneic stem cell transplantation. Blood Cells Mol Dis. 2008; 40(1): 25-32. [CrossRef]
  59. von Bahr L, Batsis I, Moll G, Hägg M, Szakos A, Sundberg B, Uzunel M, Ringden O, Le Blanc K. Analysis of tissues following mesenchymal stromal cell therapy in humans indicates limited long-term engraftment and no ectopic tissue formation. Stem Cells. 2012; 30(7): 1575-8. [CrossRef]
  60. Stoma I, Karpov I, Krivenko S, Iskrov I, Milanovich N, Koritko A, Uss A. Mesenchymal stem cells transplantation in hematological patients with acute graft-versus-host disease: characteristics and risk factors for infectious complications. Ann Hematol. 2018; 97(5): 885-891. [CrossRef]
  61. Kebriaei P, Hayes J, Daly A, Uberti J, Marks DI, Soiffer R, Waller EK, Burke E, Skerrett D, Shpall E, Martin PJ. A Phase 3 Randomized Study of Remestemcel-L versus Placebo Added to Second-Line Therapy in Patients with Steroid-Refractory Acute Graft-versus-Host Disease. Biol Blood Marrow Transplant. 2020; 26(5): 835-844. [CrossRef]
  62. Ringdén O, Uzunel M, Rasmusson I, Remberger M, Sundberg B, Lönnies H, Marschall HU, Dlugosz A, Szakos A, Hassan Z, Omazic B, Aschan J, Barkholt L, Le Blanc K. Mesenchymal stem cells for treatment of therapy-resistant graft-versus-host disease. Transplantation. 2006; 81(10): 1390-7. [CrossRef]
  63. Fernández-Maqueda C, Gonzalo-Daganzo R, Regidor C, Martín-Donaire T, Sánchez R, Bueno JL, Bautista G, De Liglesia A, Gutiérrez Y, García-Berciano M, Forés R, Royuela A, Fernández MN, Duarte RF, Cabrera-Marín JR. Mesenchymal stromal cells for steroid-refractory acute GvHD. Bone Marrow Transplant. 2017; 52(11): 1577-1579. [CrossRef]
  64. Muroi K, Miyamura K, Okada M, Yamashita T, Murata M, Ishikawa T, Uike N, Hidaka M, Kobayashi R, Imamura M, Tanaka J, Ohashi K, Taniguchi S, Ikeda T, Eto T, Mori M, Yamaoka M, Ozawa K. Bone marrow-derived mesenchymal stem cells (JR-031) for steroid-refractory grade III or IV acute graft-versus-host disease: a phase II/III study. Int J Hematol. 2016; 103(2): 243-50. [CrossRef]
  65. Zhao K, Lou R, Huang F, Peng Y, Jiang Z, Huang K, Wu X, Zhang Y, Fan Z, Zhou H, Liu C, Xiao Y, Sun J, Li Y, Xiang P, Liu Q. Immunomodulation effects of mesenchymal stromal cells on acute graft-versus-host disease after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2015; 21(1): 97-104. [CrossRef]
  66. 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. 2017; 52(4): 606-608. [CrossRef]
  67. Galleu A, Milojkovic D, Deplano S, Szydlo R, Loaiza S, Wynn R, Marks DI, Richardson D, Orchard K, Kanfer E, Tholouli E, Saif M, Sivaprakasam P, Lawson S, Bloor A, Pagliuca A, Potter V, Mehra V, Snowden JA, Vora A, Kishore B, Hunter H, Apperley JF, Dazzi F. Mesenchymal stromal cells for acute graft-versus-host disease: response at 1 week predicts probability of survival. Br J Haematol. 2019; 185(1): 89-92. [CrossRef]
  68. Sánchez-Guijo F, Caballero-Velázquez T, López-Villar O, Redondo A, Parody R, Martínez C, Olavarría E, Andreu E, Prósper F, Díez-Campelo M, Regidor C, Villaron E, López-Corral L, Caballero D, Cañizo MC, Pérez-Simon JA. Sequential third-party mesenchymal stromal cell therapy for refractory acute graft-versus-host disease. Biol Blood Marrow Transplant. 2014; 20(10): 1580-5. [CrossRef]
  69. Murata M, Terakura S, Wake A, Miyao K, Ikegame K, Uchida N, Kataoka K, Miyamoto T, Onizuka M, Eto T, Doki N, Ota S, Sato M, Hashii Y, Ichinohe T, Fukuda T, Atsuta Y, Okamoto S, Teshima T. Off-the-shelf bone marrow-derived mesenchymal stem cell treatment for acute graft-versus-host disease: real-world evidence. Bone Marrow Transplant. 2021; 56(10): 2355-2366. [CrossRef]
  70. Resnick IB, Barkats C, Shapira MY, Stepensky P, Bloom AI, Shimoni A, Mankuta D, Varda-Bloom N, Rheingold L, Yeshurun M, Bielorai B, Toren A, Zuckerman T, Nagler A, Or R. Treatment of severe steroid resistant acute GVHD with mesenchymal stromal cells (MSC). Am J Blood Res. 2013; 3(3): 225-38.
  71. Introna M, Lucchini G, Dander E, Galimberti S, Rovelli A, Balduzzi A, Longoni D, Pavan F, Masciocchi F, Algarotti A, Micò C, Grassi A, Deola S, Cavattoni I, Gaipa G, Belotti D, Perseghin P, Parma M, Pogliani E, Golay J, Pedrini O, Capelli C, Cortelazzo S, D’Amico G, Biondi A, Rambaldi A, Biagi E. Treatment of graft versus host disease with mesenchymal stromal cells: a phase I study on 40 adult and pediatric patients. Biol Blood Marrow Transplant. 2014; 20(3): 375-81. [CrossRef]
  72. Te Boome LC, Mansilla C, van der Wagen LE, Lindemans CA, Petersen EJ, Spierings E, Thus KA, Westinga K, Plantinga M, Bierings M, Broers AE, Cuijpers ML, van Imhoff GW, Janssen JJ, Huisman C, Zeerleder S, Huls G, Boelens JJ, Wulffraat NM, Slaper-Cortenbach IC, Kuball J. Biomarker profiling of steroid-resistant acute GVHD in patients after infusion of mesenchymal stromal cells. Leukemia. 2015; 29(9): 1839-46. [CrossRef]
  73. von Dalowski F, Kramer M, Wermke M, Wehner R, Röllig C, Alakel N, Stölzel F, Parmentier S, Sockel K, Krech M, Schmitz M, Platzbecker U, Schetelig J, Bornhäuser M, von Bonin M. Mesenchymal Stromal Cells for Treatment of Acute Steroid-Refractory Graft Versus Host Disease: Clinical Responses and Long-Term Outcome. Stem Cells. 2016; 34(2): 357-66. [CrossRef]
  74. 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(6): 859-862. [CrossRef]
  75. 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(29): 20590-20604. [CrossRef]
  76. Galleu A, Milojkovic D, Deplano S, Szydlo R, Loaiza S, Wynn R, Marks DI, Richardson D, Orchard K, Kanfer E, Tholouli E, Saif M, Sivaprakasam P, Lawson S, Bloor A, Pagliuca A, Potter V, Mehra V, Snowden JA, Vora A, Kishore B, Hunter H, Apperley JF, Dazzi F. Mesenchymal stromal cells for acute graft-versus-host disease: response at 1 week predicts probability of survival. Br J Haematol. 2019; 185(1): 89-92. [CrossRef]
  77. Hinden L, Avner M, Stepensky P, Or R, Almogi-Hazan O. Lymphocyte counts may predict a good response to mesenchymal stromal cells therapy in graft versus host disease patients. PLoS One. 2019; 14(6): e0217572. [CrossRef]
  78. Kurtzberg J, Abdel-Azim H, Carpenter P, Chaudhury S, Horn B, Mahadeo K, Nemecek E, Neudorf S, Prasad V, Prockop S, Quigg T, Satwani P, Cheng A, Burke E, Hayes J, Skerrett D; MSB-GVHD001/002 Study Group. A Phase 3, Single-Arm, Prospective Study of Remestemcel-L, Ex Vivo Culture-Expanded Adult Human Mesenchymal Stromal Cells for the Treatment of Pediatric Patients Who Failed to Respond to Steroid Treatment for Acute Graft-versus-Host Disease. Biol Blood Marrow Transplant. 2020; 26(5): 845-854. [CrossRef]
  79. Zhao K, Lin R, Fan Z, Chen X, Wang Y, Huang F, Xu N, Zhang X, Zhang X, Xuan L, Wang S, Lin D, Deng L, Nie D, Weng J, Li Y, Zhang X, Li Y, Xiang AP, Liu Q. Mesenchymal stromal cells plus basiliximab, calcineurin inhibitor as treatment of steroid-resistant acute graft-versus-host disease: a multicenter, randomized, phase 3, open-label trial. J Hematol Oncol. 2022; 15(1): 22. [CrossRef]
  80. Ulu BU, Hindilerden IY, Yigenoglu TN, Tiryaki TO, Erkurt MA, Korkmaz G, Namdaroglu S, Aksoy E, Korkmaz S, Seyhan M, Yilmaz S, Besisik SK, Dal MS, Ulas T, Altuntas F. Are mesenchymal stem cells still effective in acute GvHD management? Transfus Apher Sci. 2025; 64(1): 104051. [CrossRef]
  81. Prabahran A, Koldej R, Chee L, Ritchie D. Clinical features, pathophysiology, and therapy of poor graft function post-allogeneic stem cell transplantation. Blood Adv. 2022; 6(6): 1947-1959. [CrossRef]
  82. Prabahran A, Koldej R, Chee L, Wong E, Ritchie D. Evaluation of risk factors for and subsequent mortality from poor graft function (PGF) post allogeneic stem cell transplantation. Leuk Lymphoma. 2021; 62(6): 1482-1489. [CrossRef]
  83. Mahat U, Rotz SJ, Hanna R. Use of Thrombopoietin Receptor Agonists in Prolonged Thrombocytopenia after Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant. 2020; 26(3): e65-e73. [CrossRef]
  84. Shahzad M, Siddiqui RS, Anwar I, Chaudhary SG, Ali T, Naseem M, Ahmed TF, Ahmed Z, Khurana S, Ahmed N, Balusu R, Singh AK, Hematti P, Callander NS, Abhyankar SH, McGuirk JP, Mushtaq MU. Outcomes with CD34-Selected Stem Cell Boost for Poor Graft Function after Allogeneic Hematopoietic Stem Cell Transplantation: A Systematic Review and Meta-Analysis. Transplant Cell Ther. 2021; 27(10): 877.e1-877.e8. [CrossRef]
  85. Man Y, Lu Z, Yao X, Gong Y, Yang T, Wang Y. Recent Advancements in Poor Graft Function Following Hematopoietic Stem Cell Transplantation. Front Immunol. 2022; 13: 911174. [CrossRef]
  86. Li T, Luo C, Zhang J, Wei L, Sun W, Xie Q, Liu Y, Zhao Y, Xu S, Wang L. Efficacy and safety of mesenchymal stem cells co-infusion in allogeneic hematopoietic stem cell transplantation: a systematic review and meta-analysis. Stem Cell Res Ther. 2021; 12(1): 246. [CrossRef]
  87. Zhu L, Liu J, Kong P, Gao S, Wang L, Liu H, Zhang C, Gao L, Feng Y, Chen T, Gao L, Zhang X. Analysis of the Efficacy and Safety of Avatrombopag Combined with MSCs for the Treatment of Thrombocytopenia After Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol. 2022; 13: 910893. [CrossRef]
  88. Thompson M, Mei SHJ, Wolfe D, Champagne J, Fergusson D, Stewart DJ, Sullivan KJ, Doxtator E, Lalu M, English SW, Granton J, Hutton B, Marshall J, Maybee A, Walley KR, Santos CD, Winston B, McIntyre L. Cell therapy with intravascular administration of mesenchymal stromal cells continues to appear safe: An updated systematic review and meta-analysis. EClinicalMedicine. 2020; 19: 100249. [CrossRef]
  89. Huang R, Chen T, Wang S, Wang J, Su Y, Liu J, Zhang Y, Ma X, Wen Q, Kong P, Zhang C, Gao L, Zhong JF, Gao L, Zhang X. Mesenchymal Stem Cells for Prophylaxis of Chronic Graft-vs-Host Disease After Haploidentical Hematopoietic Stem Cell Transplant: An Open-Label Randomized Clinical Trial. JAMA Oncol. 2024; 10(2): 220-226. [CrossRef]
  90. Yao H, Huang R, Fu H, Lin R, Zhang Y, Feng Y, Wang Y, Chen T, Wang X, Zhu L, Liu J, Liu Y, Zhao L, Wang L, Kong P, Wen Q, Zhang C, Gao L, Gao L, Liu Q, Zhang X, Huang X, Zhang X. Sequential Infusion of Mesenchymal Stem Cell for Graft-Versus-Host Disease Prevention in Haploidentical Hematopoietic Stem Cell Transplantation: An Open-Label, Multicenter, Randomized Controlled Clinical Trial. J Clin Oncol. 2025; 43(17): 1997-2006. [CrossRef]
Table 1. MSC treatment studies with 25 participants or more.
Table 1. MSC treatment studies with 25 participants or more.
Reference, year Cohort Acute GVHD grade Dose of MSCs, cells/kg Number of doses Response rate Day +28 Survival
Le Blanc et al. 2008 [55] n=55 (25 children, 30 adults) II: n=5, III: n=25, IV: n=25 1.4×106 1 (n=27), 2 (n=5), 3–5 (n=6) CR=54.5%, PR=16%, OR=70.8% 2-year OS 35%
Resnick et al. 2013 [70] n=50 (25 children, 25 adults) II–III: n=8, IV: n=42 1.05×106 (avg first dose) 1–6 CR=34%, PR=16%, OR=50% 3.6-year DFS 56%
Sánchez-Guijo et al. 2014 [68] n=25 adults II: n=7, III: n=15, IV: n=3 1.1×106 2 (n=4), 3 (n=7), 4 (n=14) CR=44%, PR=27%, OR=71% 1-year OS 40%
Introna et al. 2014 [71] n=40 (15 children, 25 adults) II–IV: n=20 GVHD overlap: n=20 1.5×106 3 (children 2–7), 2 (adults 2–11) CR=57.1%, PR=40%, OR=60% 2-year OS 50% children, 38.6% adults
Zhao et al. 2015 [65] n=28 (age 14–54) II: n=4, III: n=8, IV: n=16 1×106 4 (2–8) MSC vs. ctrl CR: 75% vs. 42%, OR: 60% vs. 26% M vs. C
1-year OS: 46% vs. 26%, (1.1–5.4m)
Te Boome et al. 2015 [72] n=48 (7 children, 41 adults) II: n=3, III: n=33, IV: n=12 - - CR=25% 1-year OS 44%
von Dalowski et al. 2016 [73] n=58 adults II–III: n=12, IV: n=46 0.99×106 ≤2 (n=40), >3 (n=18) CR=9%, PR=38%, OR=47% 100-day OS 34.5%, 2-year OS 16.6%
Fernández-Maqueda et al. 2017 [63] n=33 adults II: n=17, III: n=7, IV: n=9 1.06×106 4 (1–16) CR=33%, PR=48%, NR=15% 1-year OS 79% in CR patients vs. 25% in PR/NR
Salmenniemi et al. 2017 [66] n=30 (8 children, 22 adults) II: n=2, III: n=14, IV: n=14, cGVHD: n=4 2×106 Up to 6 doses CR=23%, VGPR=13%, PR=17%, OR=53% 6-month OS 54%, 2-year OS 29%
Dotoli et al. 2017 [74] n=46 (16 children, 30 adults) II: n=10, III: n=20, IV: n=16 Cumulative dose 6.81×106 3 (1–7) CR=6.5%, PR=43.5%, OR=50% 100-day OS 34.4%, 2-year OS 17.4%
Servais et al. 2018 [75] n=33 (4 children, 29 adults) II: n=5, III: n=15, IV: n=13 1–2×106, 3–4×106 1 (n=25), 2 (n=8) CR=21.9%, OR=40% 1-year OS 18%
Bader et al. 2018 [56] n=69 (51 children, 18 adults) II: n=10, III: n=18, IV: n=41 1–2×106 1–4 CR=31.9%, PR=50.7%, OR=82.6% 6-month OS 71.6%
Galleu et al. 2019 [76] n=60 (4 months–68 years) II–III: n=5, IV–V: n=55 2.6×106 1 (n=34), 2 (n=16), 3 (n=6), 6 (n=1) NRe OS 104 days (0–215)
Hinden L, et al. 2019 [77] n=26 (both <18 and >18) II: 3 responders, 2 non-responders, III–IV: 10 responders, 11 non-responders 0.59–1.8x 106 1 (n=26) NRe OS 40 days 11 (84.6% responders) and 4 (30.8% non-responders)
Kebriaei P, et al. 2020 [61] n=163 with MSCs (M) and 81 controls (C) II: 37 M, 21 C, III: 82 M, 44 C, IV: 44 M, 14 C 2×106 8 given over 4 weeks; PR maintained for an additional 4 weeks DCR=38.5%, MCR=32.1, OR=53.8% M vs. 54.3% C 180 days M: 34% C: 42%
Kurtzberg J, et al. 2020 [78] n=54 II: n=23, III: n=26, IV: n=5 2×106 8 given over 4 weeks CR=25.9%, PR=40.7%, OR=66.6% 180 days, 68.5%
Ke Zhao et al. 2022 [79] n=101 with MSCs (M) and n=102 controls without MSCs (C) II:36M,37C III:41M,44C IV:22M,18C 1×106 Once weekly for 4 consecutive weeks CR=56.6%,
PR=26.3%
OR=82.9%
Ruihao Huang et al.2024
Preemptive therapy after haplo HSCT [89]
n=74 with MSCs (M) and n=74 controls standard prophylaxis (C) III and IV n=2M,
III and IV n=10C
1 × 106 4(every 2 weeks) 2-year cumulative incidence of severe cGVHD in the M group n:4 and C group:13
Ulu et al.2025 [80] n=36 with MSCs III and IV n=24 1.72x106 2(every 2 weeks) PR+CR=20% 6-month OS 33.3%
Han Yao et al.2025
Preemptive therapy after haplo HSCT* [90]
n = 96 with MSCs (M) and 96 controls (C) III and IV M group n=2
III and IV C group n:21
1 × 106 8 (eight doses within 3 months) 3-year GFRS rate in the M group n:59 and C group:30
Abbreviations: aGvHD, acute graft-versus-host disease; cGvHD, chronic graft-versus-host disease; GRFS,GVHD-free and relapse-free survival rate; MSCs, mesenchymal stem/stromal cells; OS, overall survival; DFS, disease-free survival; CR, complete response; DCR, durable complete response; MCR, major complete response VGPR, very good partial response; PR, partial response; OR, overall response; NRe, not reported; M, MSC treatment arm; C, control arm.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated