- Note
- Open access
- Published:
Optimization of transplantation methods using isolated mesenchymal stem/stromal cells: clinical trials of inflammatory bowel diseases as an example
Inflammation and Regeneration volume 44, Article number: 37 (2024)
Abstract
Mesenchymal stem/stromal cells (MSCs) are distributed in various tissues and are used in clinical applications as a source of transplanted cells because of their easy harvestability. Although MSCs express numerous cell-surface antigens, single-cell analyses have revealed a highly heterogeneous cell population depending on the original tissue and donor conditions, including age and interindividual differences. This heterogeneity leads to differences in their functions, such as multipotency and immunomodulatory effects, making it challenging to effectively treat targeted diseases. The therapeutic efficacy of MSCs is controversial and depends on the implantation site. Thus, there is no established recipe for the transplantation of MSCs (including the type of disease, type of origin, method of cell culture, form of transplanted cells, and site of delivery). Our recent preclinical study identified appropriate MSCs and their suitable transplantation routes in a mouse model of inflammatory bowel disease (IBD). Three-dimensional (3D) cultures of MSCs have been demonstrated to enhance their properties and sustain engraftment at the lesion site. In this note, we explore the methods of MSC transplantation for treating IBDs, especially Crohn’s disease, from clinical trials published over the past decade. Given the functional changes in MSCs in 3D culture, we also investigate the clinical trials using 3D constructs of MSCs and explore suitable diseases that might benefit from this approach. Furthermore, we discuss the advantages of the prospective isolation of MSCs in terms of interindividual variability. This note highlights the need to define the method of MSC transplantation, including interindividual variability, the culture period, and the transplantation route.
Background
The number of patients with inflammatory bowel diseases (IBDs), including ulcerative colitis (UC) and Crohn’s disease (CD), was approximately 5 million worldwide in 2019 [1]. The number is increasing in industrialized countries, including Asia, and is estimated to reach 10 million by 2050 [2, 3]. Although IBDs are caused by chronic inflammation, UC involves inflammation of the rectum to the small intestine, whereas CD involves inflammation of the whole gastrointestinal tract from the gut to the mouth [4]. Inflammation is promoted by genetic and environmental factors such as the gut microbiome [5, 6]. Considering its ability to suppress inflammation, 5-aminosalicylic acid (5-ASA) is prescribed to patients with IBD as a first-line treatment [7]. However, the anti-inflammatory effect of 5-ASA is insufficient for durable remission and mucosal healing [8, 9]. Mucosal healing, denoting the regeneration of the damaged intestinal mucosa, is essential for long-term clinical remission in patients with IBDs [10]. Recently, novel therapeutics, such as biologics and small molecules, have been developed to support mucosal healing. Anti-tumor necrosis factor (TNF)-α therapies (e.g., infliximab and adalimumab) are typical biologics and are often used in patients unresponsive to 5-ASA and corticosteroid treatments [11, 12]. However, a certain population of patients with CD are either unresponsive or lose their response to biologics [13, 14]. Although 70% of patients with CD display small bowel lesions [15], the poor therapeutic efficacy of biologics in treating small bowel lesions might explain the lack of improvement in outcomes, including hospitalization [10, 16, 17]. Another potential reason could be the low treatment efficacy of refractory perianal fistulas [18]. There is no definitive evidence that new biologics and small molecules in long-term use have totally closed intestinal or anal fistula lesions in CD that cause a poor outcome and operation. Therefore, developing novel therapies for long-term clinical remission of patients with CD would be important.
Recently, stem cell transplantation therapy for IBDs has attracted attention because of its immunomodulatory function and promotion of mucosal healing [19, 20]. Mesenchymal stem/stromal cells (MSCs) are good cell source candidates owing to their immunosuppressive effects and ability to migrate to inflammatory sites [20,21,22,23]. In the USA, EU, and Japan, darvadstrocel, a dispersion of expanded allogeneic, human adipose tissue–derived MSCs (AD-MSCs), is approved for the treatment of patients with an inadequate response to at least one conventional therapy or biologics with complex perianal fistulas with non-active or mildly active luminal CD [24]. However, a recent announcement underpinned that the primary endpoint of combined remission at 24 weeks could not be achieved in the phase III ADMIRE-CD II study [25]. In addition, a suitable transplantation method for MSCs, including the delivery route (i.e., intravenous, intraperitoneal, or anal injection) and cell form, has not yet been established for the treatment of IBDs [21]. Therefore, further developing stem cell–based therapies and investigating transplantation methods suitable for difficult-to-treat conditions, such as complex perianal fistulas, are necessary.
MSCs are a cell population that adheres to a plastic dish under culture conditions and can be obtained from diverse tissues, such as the bone marrow, adipose tissue, placenta, dental pulp, and skin [26,27,28,29,30]. Although bone marrow–derived MSCs (BM-MSCs) have been the most studied and are used in many clinical applications [31], they are present in only 0.001–0.01% of the bone marrow and exhibit invasive issues at the harvesting stage [32, 33]. Adipose tissues contain the MSC fraction with 1–10% stromal cells and are attracting attention as a cell source for clinical applications owing to their accessibility [34, 35]. Previously, we compared the percentage of the MSC fraction among subcutaneous, amnion, chorion, villus, umbilical cord, and visceral fat and demonstrated that it is the highest in subcutaneous fat [28]. MSCs express specific cell-surface antigens (CD44, CD73, CD90, and CD105) and lack hematopoietic cell antigens (CD14, CD19, CD34, and CD45), endothelial marker (CD31), costimulatory molecules (CD40, CD80, and CD86), and MHC molecules (HLA class II) [26]. The positive markers depend on the species and tissue of origin [27, 36,37,38,39,40,41]. Considering the differences in cell-surface markers and gene expression profiles [42], the function of MSCs in migration at the inflammation site and their immunomodulatory features may vary [43, 44]. Additionally, it has been noted that the characterization of MSCs is altered during the clinical-scale expansion because of cellular senescence [45,46,47].
In this paper, we review clinical trials of MSCs in patients with IBDs, especially CD, from 2012 to 2023 in terms of the cell culture period for transplantation, transplantation methods, and their mode of action. Furthermore, we describe our novel findings demonstrating the advantages of isolating MSCs using fluorescence-activated cell sorting (FACS) in mouse and human studies.
Methods
Search strategy and study selection
Published papers were sorted using the following bibliographic databases: PubMed (from January 2012 to December 2023) and Google Scholar (from January 2012 to December 2023). The search terms in Table 1 include variations of “inflammatory bowel disease,” “Crohn’s disease,” “ulcerative colitis,” and “mesenchymal stem/stromal cells.” The search terms related to Table 2 included variations of “three-dimensional,” “organoid,” “spheroid,” “scaffold,” “spheroid-free,” “tissue engineering,” and “mesenchymal stem/stromal cells.”
Clinical trials were extracted from the papers searched by one author. Another author validated the list and selected reports that matched the purpose of this review. Eighteen papers of 29 cases were selected in Table 1. Complex perianal fistulas not associated with IBDs were excluded. The cases using adipose-derived stromal vascular fraction were excluded. Nineteen papers of 20 cases were selected in Table 2. MSC preparations consisting of cell sheets were excluded.
mRNA sequencing (mRNA-seq) and data reanalysis
Principal component and gene expression analyses of MSC markers were performed based on an mRNA-seq dataset from our previous study [85]. The Euclidian distance was calculated based on the total genes (81,602 transcripts) after log2 transformation in the mRNA-seq dataset. Data analyses and visualizations were conducted using the RStudio environment and a Tag-Count Comparison Graphical User Interface (TCC-GUI).
Statistical analysis
All statistical analyses were performed using the statistical programming language R version 4.3.3 (2024–02-29). Statistical significance was determined using the Wilcoxon rank-sum test.
Main
Current status of clinical applications of MSCs in IBD treatment from published papers
Eighteen clinical trials for treating CD have been published in the past decade (Table 1) [48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65]. Of the 18 studies, 10 used BM-MSCs, six used AD-MSCs, and two used umbilical cord–derived MSCs (UC-MSCs) as transplanted cells. These trials included four cases using MSC preparations (i.e., remestemcel-L, darvadstrocel, and TH-SC01) [55, 60,61,62]. Five of the 18 studies were trials on the intravenous injection of MSCs in patients with luminal CD [48,49,50,51,52]. One study used autologous BM-MSCs for intravenous injection, whereas others used allogeneic BM- or UC-MSCs. No such trials have been conducted using AD-MSCs. For the treatment of fistulizing CD, infusion into the lesion site was performed because of accessibility, with 13 of the 18 reports involving intralesional injections [54,55,56,57,58,59,60,61,62]. Of the studies using intralesional injections, nine targeted perianal fistulas, two targeted peripouch or rectovaginal fistulas, and two targeted luminal CD, including strictures. One of these reports showed the serial intravenous and intralesional injections of allogeneic BM-MSCs in patients with perianal fistulizing CD [53]. Three studies reported the efficacy and safety of combination therapy with autologous AD-MSCs and bioabsorbable matrices as a scaffold in intralesional injections [63,64,65]. All the studies used allogeneic MSCs in intralesional injections without scaffolds. Although some trials have reported a few adverse effects, it is important to note that all studies unequivocally demonstrated the safety of MSC transplantation. Almost all studies have shown benefits regarding the efficacy of MSC therapy; however, in a few cases (i.e., patients with luminal CD), they have not shown a significant advantage. For example, Lightner et al. performed intralesional transplantation of allogeneic BM-MSCs (1.5–3.0 × 108) into four patients with luminal CD [55]. They reported that the indices of clinical remission and response, with a simple endoscopic score for CD (SES-CD), dropped from 17 to 5, the Crohn’s disease activity index score (CDAI) dropped from 228 to 200, and C-reactive protein (CRP) remained at 0.20–0.30. In contrast, in the control group, the SES-CD increased from 15.5 to 25.0, the CDAI increased from 146 to 158, and the CRP dropped from 3.65 to 1.50 at 3 months. No patients in the treatment group showed clinical remission or response. One patient received anti-TNF-α monoclonal antibody treatment (i.e., Cimzia) before MSC therapy. Previous studies have shown that patients with pre-treated anti-TNF-α treatment have a lower response to subsequent therapies than naive patients [86, 87]. Lightner et al. indicated that achieving efficacy when attempting new therapeutics for treatment-refractory patients is challenging. They also performed intralesional transplantation of allogeneic BM-MSCs (7.5 × 107) to treat peripouch fistulas in patients with ileal pouch-anal anastomosis [56]. Four of 13 patients (31%) in the treated group and one of six (20%) in the control had complete clinical and radiographic healing at 6 months. Vieujean et al. showed a 40% complete stricture resolution at 48 weeks in patients with luminal CD [57]. On the other hand, Molendijk et al. demonstrated the efficacy of intravenous injection of allogeneic BM-MSCs in treating refractory perianal fistulas in patients with CD [54]. Low-dose administration (3 × 107) resulted in greater fistula healing than high-dose administration (9 × 107). The dose in Lightner’s trial was higher than that in Molendijk’s trial; this may also have caused these differences in efficacy. Thus, further analysis is needed to determine the relationship between efficacy and dose dependency on the CD condition, especially luminal CD.
The culture period of MSCs for transplantation is yet to be clearly defined. Of the 18 reports, only three clearly described the total culture period [48, 51, 57]. Others were either partially or not described. In the papers described, the shortest culture period was 2 weeks, and the maximum was 4 or 6 weeks in five or fewer passages. Previous studies have shown that MSCs gradually lose specific capacities such as proliferation, differentiation, and secretion during expansion [45,46,47]. Moreover, there are differences in transcriptional characteristics between cultured and freshly isolated MSCs [42]. Even within the same MSC preparation, functions may differ depending on the culture period. Thus, culture days and the number of passages before transplantation should be defined for optimal efficacy.
The key players in the pathogenesis of IBDs are T helper (Th) cells and regulatory T cells (Tregs) [88, 89]. The imbalance in these cells activates other immune cells (i.e., macrophages and B cells) exacerbating inflammation in the gut mucosa [5]. Almost all studies have proposed an immunomodulatory function as a mode of action for MSC therapy. In a preclinical study, MSCs inhibited the function of Th cells and increased the population of Tregs by modulating the secretion of numerous factors, such as pro-inflammatory (e.g., interferon-γ, TNF-α, and interleukin [IL]-17) and anti-inflammatory (IL-10 and transforming growth factor-β) cytokines [90, 91]. Despite analyses of lymphocytes in peripheral blood and biopsy samples, the mode of action of MSCs in humans still needs to be fully understood.
Clinical trials using the three-dimensional construct of MSCs
Previous studies have shown that spheroids derived from three-dimensional (3D) MSC cultures exhibit enhanced functions such as immunomodulatory effects, angiogenesis, and multipotency by altering the expression of genes such as TNF-α-stimulated gene 6 (TSG-6), fibroblast growth factor-2 (FGF-2), angiogenin, and OCT4 [92,93,94]. Preclinical studies demonstrated the advantage of 3D culture–derived MSC spheroid transplantation in several animal models [95,96,97,98,99]. In the model of skin ulcers and bone defects, as surgically accessible tissues, the intralesional injection of MSC spheroids led to markedly superior and faster regeneration than that seen with 2D-cultured MSCs [95,96,97]. These results suggest that these phenotypes are attributed to enhanced gene expression related to the extracellular matrix (ECM), angiogenesis, and migration (e.g., fibronectin, FGF-1, CXC chemokine receptor 4, and integrin α2). In myocardial infarction models, functional recovery was higher in the case of the intramyocardial injection of MSC spheroids than that of 2D culture–derived MSCs [98]. Xu et al. demonstrated that the intralesional injection of AD-MSC spheroids was more beneficial to functional recovery than that of 2D culture–derived MSCs in a kidney injury model [99]. In clinical trials of IBD treatment, the scaffold has been used to sustain transplanted MSCs at the lesion site for an extended time. However, no clinical trials have been conducted on treating IBDs using 3D-structured MSCs without scaffolds. Here, we describe the published clinical trials using 3D structures with MSCs for other diseases (Table 2) [66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84].
Seventeen of the 19 studies used bioabsorbable scaffolds, whereas two did not. In scaffold-free trials, 3D structures were generated by expanding cultures of autologous MSCs, followed by 3D cultures in osteogenic differentiation media or media containing ascorbate [66, 67]. 3D cultures have been performed to improve the effectiveness of bone reconstitution by facilitating osteogenesis and angiogenesis or improving adhesiveness to the cartilaginous matrix, which has been used to treat degenerative disc disease or knee chondral lesions. In contrast, in the trials with the MSC plus scaffold construct, 13 papers were trials for osteochondral and tendon regeneration, two for spinal cord and spine regeneration, one for wound healing of ulcers, and one for ischemic heart disease. Fourteen of the 19 papers used 3D constructs for bone and cartilage regeneration, indicating that surgically accessible tissues were targeted [67,68,69,70,71,72,73,74,75,76,77,78,79,80]. MSCs originated from various tissues, including the bone marrow (eight reports), umbilical cord (three reports), adipose tissue (two reports), synovium (two reports), alveolar bone (two reports), gingiva (one report), and Wharton’s jelly (one report). Lamas et al. reported treating patients with full-thickness rotator cuff tears using autologous BM-MSCs with type I collagen membrane [76]. They used a scaffold to augment the ability of MSCs because MSCs alone are insufficient to improve the healing of repaired tendons. Morrison et al. also reported that MSCs alone cannot restore bone structure and that a supportive matrix is required [80]. Depending on the tissue of origin, MSCs have different differentiation abilities [41]. Zhuang et al. used BM-MSCs for treating bone defects because the MSCs can prompt attachment to the surface and inner space of porous scaffolds, such as a β-tricalcium phosphate, efficiently constructing a bioactive composite [72]. Although synovium-derived MSCs have more prominent chondrogenic and less osteogenic differentiation abilities than those derived from the bone marrow or periosteum, Akgun et al. used synovium-derived MSCs with type I/III collagen membranes to treat chondral defects of the knee [70]. Apatzidou et al. reported improvements in osteoarthritis using umbilical cord blood-derived MSCs, which promoted the differentiation of endogenous chondroprogenitors through paracrine effects [78]. In the case of periodontal intrabony defects, autologous gingiva-derived MSCs alter the components of the extracellular matrix in gingival fibroblasts by migrating into periodontal defects [77]. Thus, several trials employ MSCs to promote the multipotency of MSCs or the differentiation of endogenous cells in the lesion site rather than immunomodulatory functions; therefore, the MSCs used tend to differ depending on the type of scaffold used or the target disease.
Regarding the culture period, although eight studies did not describe the MSC expansion time, the period ranged from 14 to 50 days in the papers described. Generating 3D constructs without scaffolds requires a longer culture period (2–3 weeks or 40 days) [66, 67]. In contrast, when using scaffolds, the incubation period was mostly 2 days, with the shortest period being 4 h and the longest being 20–30 days. In particular, 3D cultures with osteogenic differentiation require extended culture periods. Notably, eight of the 19 reports performed in vitro 3D culture, whereas the others reported short incubation times ranging from 10 to 60 min in syringes or lacked details. Because the 3D structure of MSCs by self-aggregation enhances their immunomodulatory effects, a scaffold-free 3D MSC construct for treating inflammatory diseases, including IBDs, may be useful in clinical trials. However, evidence on the efficacy of MSC spheroids in clinical trials is currently insufficient; in the future, it is considered necessary to advance clinical research, including IBDs.
Advantages of prospective isolated MSCs using specific cell-surface markers
MSCs exhibit cellular heterogeneity with various cell-surface antigens [100]. Even when similar cell-surface markers are expressed on MSCs, their functions, including differentiation potential and immunomodulatory effects, differ depending on the tissue of origin. For example, previous studies have shown that UC-MSCs have higher proliferation, colony-forming, and immunomodulation abilities than AD-MSCs [101, 102]. Furthermore, it is noted that adhering MSCs on plastic dishes are heterogeneous cell populations with donor-to-donor variability in clinical trials [58, 103]. This interindividual variability is a significant problem when autologous MSCs are used. In preclinical studies, high-quality BM-MSCs have been isolated using several specific markers by FACS (i.e., for mice, a combination of CD140a and Sca-1; for humans, a combination of CD90, CD271, and CD146) [27, 37]. These prospectively isolated MSCs have a high potential for colony formation. In addition, we previously demonstrated that AD-MSCs isolated using CD73 molecules (termed CD73+ cells) have a higher colony-forming ability than conventional heterogeneous adherent MSCs (termed cMSCs) from human and rodent experiments [28]. Intranasal injection of CD73+ cells reduces macrophage infiltration and suppresses fibrosis at the lesion site in mice with bleomycin-induced pulmonary fibrosis [28]. Moreover, in a mouse model of dextran sulfate sodium-induced colitis, intravenous injection of CD73+ cells significantly attenuated tissue destruction compared to that observed with cMSCs [85]. We also demonstrated that compared with cMSCs, human adipose tissue–derived CD73+ cells downregulated the expression of genes related to “immune response,” “inflammatory response,” and “neutrophil chemotaxis” (e.g., IL-1β, IL-6, IL-19, C–C motif chemokine ligand 2 [CCL2], CCL3, and CCL4), as revealed via a transcriptome analysis (Fig. 1) [85]. In contrast, the terms for the upregulated genes were “GTP biosynthetic process,” “UTP biosynthetic process,” “negative regulation of platelet-derived growth factor receptor signaling pathway,” and “negative regulation of intrinsic apoptotic signaling pathway.” These features indicate that CD73 is an ectonucleotidase that metabolizes extracellular adenosine triphosphate [104].
Our recent study also revealed upregulation of the expression of ECM-related genes (e.g., fibronectin 1, S100A13, SLC3A2, integrin β3, periostin, and FGF-1) in CD73+ cells compared with that in cMSCs (Fig. 1) [85]. Furthermore, the 3D culture of CD73+ cells enhanced their characteristics, such as upregulation of the expression of ECM remodeling and Wnt signaling genes (e.g., WNT11) and downregulation of the expression of inflammatory genes (Fig. 1). Upregulation of the expression of immunomodulatory genes (e.g., matrix Gla protein and TSG-6) was also observed in 3D culture-derived CD73+ spheroids (Fig. 1). Considering these properties, CD73+ spheroids may be suitable for transanal transplantation to treat IBDs. Indeed, our preclinical study showed that CD73+ spheroids increased the engraftment rate into the lesion site compared to CD73+ cells, preventing mucosal atrophy in a mouse model of colitis. The proportion of endogenous CD140a+ fibroblasts was altered after CD73+ spheroid transplantation in the lesion site. Supernatants of CD73+ spheroids, including their secretory factors, affect the gene expression profiles of fibroblasts, such as ECM remodeling and integrin downstream genes in vitro. These findings suggest that transanal transplantation of CD73+ spheroids exerts a potential therapeutic effect against IBDs via the paracrine effects of secreted factors caused by sustained engraftment.
Although the efficacy of CD73+ cells and CD73+ spheroids in treating inflammatory diseases in these animal models is promising, whether the CD73+ cell population is homogeneous remains unknown. Therefore, we reanalyzed the gene expression profiles based on transcription analysis of CD73+ cells and cMSCs. No significant differences in the expression of MSC markers, including CD73, CD44, CD90, CD105, CD146, and CD271, were observed between the CD73+ cell populations and cMSCs (Fig. 2a). Principal component analysis showed that one donor (donor #1) was distant from the others (Fig. 2b). We compared the distance based on their gene expression between CD73+ cells and cMSCs. Notably, the distance between inter-CD73+ cells was significantly smaller than that between cMSCs (P = 0.026; Fig. 2c). These results indicate that the CD73+ cell population has less donor-to-donor variability than heterogeneously adherent MSCs, which makes them clinically applicable.
Conclusions
In clinical trials of MSCs, the optimal transplantation method for target diseases must be defined; thus, the original tissue, isolation, form, and delivery route of MSCs must be adapted according to the disease. 3D culture–derived MSC constructs may be suitable for treating surgically accessible bone and cartilage defects or IBDs, especially perianal fistulizing CD (Fig. 1). In addition, it is desirable to use MSCs isolated with specific cell-surface markers to avoid interindividual deviation. However, several challenges persist, such as the development of clinical antibodies for isolation and residual antibodies in transplanted MSCs; furthermore, their efficacy and safety need to be considered. Nevertheless, the properties of MSCs render them promising in regenerative medicine.
Availability of data and materials
The mRNA-seq data included those described in previous studies and were reanalyzed. The mRNA-seq data supporting the findings of the present study were deposited in the Gene Expression Omnibus SuperSeries dataset GSE211637 and GSE265844.
Abbreviations
- AD-MSCs:
-
Adipose tissue-derived MSCs
- 5-ASA:
-
5-Aminosalicylic acid
- BM-MSCs:
-
Bone marrow-derived MSCs
- CCL:
-
C–C motif chemokine ligand
- CD:
-
Crohn’s disease
- CDAI:
-
Crohn’s disease activity index score
- CRP:
-
C-reactive protein
- ECM:
-
Extracellular matrix
- FACS:
-
Fluorescence-activated cell sorting
- FGF:
-
Fibroblast growth factor
- IBDs:
-
Inflammatory bowel diseases
- IL:
-
Interleukin
- MSCs:
-
Mesenchymal stem/stromal cells
- TCC-GUI:
-
Tag-Count Comparison Graphical User Interface
- Th:
-
T helper
- 3D:
-
Three-dimensional
- TNF-α:
-
Tumor necrosis factor-alpha
- TSG-6:
-
TNF-α-stimulated gene 6
- UC:
-
Ulcerative colitis
- UC-MSCs:
-
Umbilical cord-derived MSCs
References
Wang R, Li Z, Liu S, Zhang D. Global, regional and national burden of inflammatory bowel disease in 204 countries and territories from 1990 to 2019: a systematic analysis based on the Global Burden of Disease Study 2019. BMJ Open. 2023;13(3): e065186.
Kaplan GG, Windsor JW. The four epidemiological stages in the global evolution of inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 2021;18(1):56–66.
Sebastian S, Siegmund B, Teferra F, McGovern DPB, Queiroz NSF, van der Woude CJ, Sharma V. Promoting equity in inflammatory bowel disease: a global approach to care. Lancet Gastroenterol Hepatol. 2024;9(3):192–4.
Sairenji T, Collins KL, Evans DV. An update on inflammatory bowel disease. Prim Care. 2017;44(4):673–92.
Ramos GP, Papadakis KA. Mechanisms of disease: inflammatory bowel diseases. Mayo Clin Proc. 2019;94(1):155–65.
Sugihara K, Kamada N. Metabolic network of the gut microbiota in inflammatory bowel disease. Inflamm Regen. 2024;44(1):11.
Mishra R, Dhawan P, Srivastava AS, Singh AB. Inflammatory bowel disease: therapeutic limitations and prospective of the stem cell therapy. World J Stem Cells. 2020;12(10):1050–66.
Abraham BP, Ahmed T, Ali T. Inflammatory bowel disease: pathophysiology and current therapeutic approaches. Handb Exp Pharmacol. 2017;239:115–46.
Wright EK, Ding NS, Niewiadomski O. Management of inflammatory bowel disease. Med J Aust. 2018;209(7):318–23.
Okamoto R, Mizutani T, Shimizu H. Development and application of regenerative medicine in inflammatory bowel disease. Digestion. 2023;104(1):24–9.
Carter MJ, Lobo AJ, Travis SP. Guidelines for the management of inflammatory bowel disease in adults. Gut. 2004;53 Suppl 5(Suppl 5):V1-16.
Kornbluth A, Sachar DB. Ulcerative colitis practice guidelines in adults (update): American College of Gastroenterology Practice Parameters Committee. Am J Gastroenterol. 2004;99(7):1371–85.
Li Y, Chen J, Bolinger AA, Chen H, Liu Z, Cong Y, Brasier AR, Pinchuk IV, Tian B, Zhou J. Target-based small molecule drug discovery towards novel therapeutics for inflammatory bowel diseases. Inflamm Bowel Dis. 2021;27(Suppl 2):S38–62.
Roda G, Jharap B, Neeraj N, Colombel J-F. Loss of response to anti-TNFs: definition, epidemiology, and management. Clin Transl Gastroenterol. 2016;7(1): e135.
Peyrin-Biroulet L, Loftus EV Jr, Colombel JF, Sandborn WJ. The natural history of adult Crohn’s disease in population-based cohorts. Am J Gastroenterol. 2010;105(2):289–97.
Lichtenstein GR, Rutgeerts P. Importance of mucosal healing in ulcerative colitis. Inflamm Bowel Dis. 2009;16(2):338–46.
Takenaka K, Fujii T, Suzuki K, Shimizu H, Motobayashi M, Hibiya S, Saito E, Nagahori M, Watanabe M, Ohtsuka K. Small bowel healing detected by endoscopy in patients with Crohn’s disease after treatment with antibodies against tumor necrosis factor. Clin Gastroenterol Hepatol. 2020;18(7):1545–52.
Yzet C, Brazier F, Sabbagh C, Fumery M. Managing complex perianal disease after anti-TNF failure: where to go next? Curr Res Pharmacol Drug Discov. 2022;3:100081.
Zhang H-M, Yuan S, Meng H, Hou X-T, Li J, Xue J-C, Li Y, Wang Q, Nan J-X, Jin X-J, et al. Stem cell-based therapies for inflammatory bowel disease. Int J Mol Sci. 2022;23(15):8494.
Tian CM, Zhang Y, Yang MF, Xu HM, Zhu MZ, Yao J, Wang LS, Liang YJ, Li DF. Stem cell therapy in inflammatory bowel disease: a review of achievements and challenges. J Inflamm Res. 2023;16:2089–119.
Wang M, Liang C, Hu H, Zhou L, Xu B, Wang X, Han Y, Nie Y, Jia S, Liang J, et al. Intraperitoneal injection (IP), intravenous injection (IV) or anal injection (AI)? Best way for mesenchymal stem cells transplantation for colitis. Sci Rep. 2016;6:30696.
Caplan AI, Correa D. The MSC: an injury drugstore. Cell Stem Cell. 2011;9(1):11–5.
Bianco P, Cao X, Frenette PS, Mao JJ, Robey PG, Simmons PJ, Wang CY. The meaning, the sense and the significance: translating the science of mesenchymal stem cells into medicine. Nat Med. 2013;19(1):35–42.
Meng ZW, Baumgart DC. Darvadstrocel for the treatment of perianal fistulas in Crohn’s disease. Expert Rev Gastroenterol Hepatol. 2020;14(6):405–10.
Ilic D, Liovic M. Industry updates from the field of stem cell research and regenerative medicine in October 2023. Regen Med. 2024;19(2):69–82.
Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop D, Horwitz E. Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement. Cytotherapy. 2006;8(4):315–7.
Mabuchi Y, Morikawa S, Harada S, Niibe K, Suzuki S, Renault-Mihara F, Houlihan DD, Akazawa C, Okano H, Matsuzaki Y. LNGFR(+)THY-1(+)VCAM-1(hi+) cells reveal functionally distinct subpopulations in mesenchymal stem cells. Stem Cell Reports. 2013;1(2):152–65.
Suto EG, Mabuchi Y, Toyota S, Taguchi M, Naraoka Y, Itakura N, Matsuoka Y, Fujii Y, Miyasaka N, Akazawa C. Advantage of fat-derived CD73 positive cells from multiple human tissues, prospective isolated mesenchymal stromal cells. Sci Rep. 2020;10(1):15073.
Yasui T, Mabuchi Y, Toriumi H, Ebine T, Niibe K, Houlihan DD, Morikawa S, Onizawa K, Kawana H, Akazawa C, et al. Purified human dental pulp stem cells promote osteogenic regeneration. J Dent Res. 2016;95(2):206–14.
Festa E, Fretz J, Berry R, Schmidt B, Rodeheffer M, Horowitz M, Horsley V. Adipocyte lineage cells contribute to the skin stem cell niche to drive hair cycling. Cell. 2011;146(5):761–71.
Tsuchiya A, Kojima Y, Ikarashi S, Seino S, Watanabe Y, Kawata Y, Terai S. Clinical trials using mesenchymal stem cells in liver diseases and inflammatory bowel diseases. Inflamm Regen. 2017;37:16.
Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143–7.
Hernigou P, Poignard A, Beaujean F, Rouard H. Percutaneous autologous bone-marrow grafting for nonunions: influence of the number and concentration of progenitor cells. JBJS. 2005;87(7):1430–7.
Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, Benhaim P, Lorenz HP, Hedrick MH. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7(2):211–28.
De Ugarte DA, Morizono K, Elbarbary A, Alfonso Z, Zuk PA, Zhu M, Dragoo JL, Ashjian P, Thomas B, Benhaim P. Comparison of multi-lineage cells from human adipose tissue and bone marrow. Cells Tissues Organs. 2003;174(3):101–9.
Sacchetti B, Funari A, Michienzi S, Di Cesare S, Piersanti S, Saggio I, Tagliafico E, Ferrari S, Robey PG, Riminucci M. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell. 2007;131(2):324–36.
Morikawa S, Mabuchi Y, Kubota Y, Nagai Y, Niibe K, Hiratsu E, Suzuki S, Miyauchi-Hara C, Nagoshi N, Sunabori T, et al. Prospective identification, isolation, and systemic transplantation of multipotent mesenchymal stem cells in murine bone marrow. J Exp Med. 2009;206(11):2483–96.
Mendez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA, Scadden DT. Ma’ayan A, Enikolopov GN, Frenette PS: Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466(7308):829–34.
Tormin A, Li O, Brune JC, Walsh S, Schütz B, Ehinger M, Ditzel N, Kassem M, Scheding S. CD146 expression on primary nonhematopoietic bone marrow stem cells is correlated with in situ localization. Blood, J Am Soc Hematol. 2011;117(19):5067–77.
Zhou BO, Yue R, Murphy MM, Peyer JG, Morrison SJ. Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. Cell Stem Cell. 2014;15(2):154–68.
Ogata Y, Mabuchi Y, Yoshida M, Suto EG, Suzuki N, Muneta T, Sekiya I, Akazawa C. Purified human synovium mesenchymal stem cells as a good resource for cartilage regeneration. PLoS One. 2015;10(6):e0129096.
Zhang P, Dong J, Fan X, Yong J, Yang M, Liu Y, Zhang X, Lv L, Wen L, Qiao J, et al. Characterization of mesenchymal stem cells in human fetal bone marrow by single-cell transcriptomic and functional analysis. Signal Transduct Target Ther. 2023;8(1):126.
Ho AD, Wagner W, Franke W. Heterogeneity of mesenchymal stromal cell preparations. Cytotherapy. 2008;10(4):320–30.
Costa LA, Eiro N, Fraile M, Gonzalez LO, Saá J, Garcia-Portabella P, Vega B, Schneider J, Vizoso FJ. Functional heterogeneity of mesenchymal stem cells from natural niches to culture conditions: implications for further clinical uses. Cell Mol Life Sci. 2021;78:447–67.
Tanabe S, Sato Y, Suzuki T, Suzuki K, Nagao T, Yamaguchi T. Gene expression profiling of human mesenchymal stem cells for identification of novel markers in early- and late-stage cell culture. J Biochem. 2008;144(3):399–408.
Churchman SM, Ponchel F, Boxall SA, Cuthbert R, Kouroupis D, Roshdy T, Giannoudis PV, Emery P, McGonagle D, Jones EA. Transcriptional profile of native CD271+ multipotential stromal cells: evidence for multiple fates, with prominent osteogenic and Wnt pathway signaling activity. Arthritis Rheum. 2012;64(8):2632–43.
Kilpinen L, Tigistu-Sahle F, Oja S, Greco D, Parmar A, Saavalainen P, Nikkila J, Korhonen M, Lehenkari P, Kakela R, et al. Aging bone marrow mesenchymal stromal cells have altered membrane glycerophospholipid composition and functionality. J Lipid Res. 2013;54(3):622–35.
Dhere T, Copland I, Garcia M, Chiang KY, Chinnadurai R, Prasad M, Galipeau J, Kugathasan S. The safety of autologous and metabolically fit bone marrow mesenchymal stromal cells in medically refractory Crohn’s disease - a phase 1 trial with three doses. Aliment Pharmacol Ther. 2016;44(5):471–81.
Knyazev O, Parfenov A, Shcherbakov P, Ruchkina I, Konoplyannikov A. Cell therapy of refractory Crohn’s disease. Bull Exp Biol Med. 2013;156:139–45.
Forbes GM, Sturm MJ, Leong RW, Sparrow MP, Segarajasingam D, Cummins AG, Phillips M, Herrmann RP. A phase 2 study of allogeneic mesenchymal stromal cells for luminal Crohn’s disease refractory to biologic therapy. Clin Gastroenterol Hepatol. 2014;12(1):64–71.
Gregoire C, Briquet A, Pirenne C, Lechanteur C, Louis E, Beguin Y. Allogeneic mesenchymal stromal cells for refractory luminal Crohn’s disease: a phase I-II study. Dig Liver Dis. 2018;50(11):1251–5.
Zhang J, Lv S, Liu X, Song B, Shi L. Umbilical cord mesenchymal stem cell treatment for Crohn’s disease: a randomized controlled clinical trial. Gut Liver. 2018;12(1):73–8.
Knyazev O, Kagramanova A, Lischinskaya A, Korneeva I, Zvyaglova M, Babayan A, Konoplyannikov A, Parfenov A. Stem cell therapy for perianal Crohn’s disease. Proc Latv Acad Sci Sect B. 2020;74(2):68–74.
Molendijk I, Bonsing BA, Roelofs H, Peeters KC, Wasser MN, Dijkstra G, van der Woude CJ, Duijvestein M, Veenendaal RA, Zwaginga JJ, et al. Allogeneic bone marrow-derived mesenchymal stromal cells promote healing of refractory perianal fistulas in patients with Crohn’s disease. Gastroenterology. 2015;149(4):918-927 e916.
Lightner AL, Ream J, Nachand D, Fulmer C, Regueiro M, Steele SR. Remestemcel-L allogeneic bone marrow-derived mesenchymal stem cell product to treat medically refractory Crohn’s colitis: preliminary phase IB/IIA study. Br J Surg. 2022;109(8):653–5.
Lightner AL, Reese J, Ream J, Nachand D, Jia X, Pineiro AO, Dadgar N, Steele S, Hull T. A phase IB/IIA study of allogeneic, bone marrow-derived, mesenchymal stem cells for the treatment of refractory ileal-anal anastomosis and peripouch fistulas in the setting of Crohn’s disease of the pouch. J Crohns Colitis. 2023;17(4):480–8.
Vieujean S, Loly JP, Boutaffala L, Meunier P, Reenaers C, Briquet A, Lechanteur C, Baudoux E, Beguin Y, Louis E. Mesenchymal stem cell injection in Crohn’s disease strictures: a phase I-II clinical study. J Crohns Colitis. 2022;16(3):506–10.
Lightner AL, Reese J, Ream J, Nachand D, Jia X, Dadgar N, Steele SR, Hull T. A phase IB/IIA study of ex vivo expanded allogeneic bone marrow-derived mesenchymal stem cells for the treatment of perianal fistulizing Crohn’s disease. Dis Colon Rectum. 2023;66(10):1359–72.
de la Portilla F, Alba F, Garcia-Olmo D, Herrerias JM, Gonzalez FX, Galindo A. Expanded allogeneic adipose-derived stem cells (eASCs) for the treatment of complex perianal fistula in Crohn’s disease: results from a multicenter phase I/IIa clinical trial. Int J Colorectal Dis. 2013;28(3):313–23.
Panes J, Garcia-Olmo D, Van Assche G, Colombel JF, Reinisch W, Baumgart DC, Dignass A, Nachury M, Ferrante M, Kazemi-Shirazi L, et al. Expanded allogeneic adipose-derived mesenchymal stem cells (Cx601) for complex perianal fistulas in Crohn’s disease: a phase 3 randomised, double-blind controlled trial. Lancet. 2016;388(10051):1281–90.
Furukawa S, Mizushima T, Nakaya R, Shibata M, Yamaguchi T, Watanabe K, Futami K. Darvadstrocel for complex perianal fistulas in Japanese adults with Crohn’s disease: a phase 3 study. J Crohns Colitis. 2023;17(3):369–78.
Wei J, Zhang Y, Chen C, Feng X, Yang Z, Feng J, Jiang Q, Fu J, Xuan J, Gao H, et al. Efficacy and safety of allogeneic umbilical cord-derived mesenchymal stem cells for the treatment of complex perianal fistula in Crohn’s disease: a pilot study. Stem Cell Res Ther. 2023;14(1):311.
Dietz AB, Dozois EJ, Fletcher JG, Butler GW, Radel D, Lightner AL, Dave M, Friton J, Nair A, Camilleri ET, et al. Autologous mesenchymal stem cells, applied in a bioabsorbable matrix, for treatment of perianal fistulas in patients with Crohn’s disease. Gastroenterology. 2017;153(1):59-62 e52.
Lightner AL, Dozois EJ, Dietz AB, Fletcher JG, Friton J, Butler G, Faubion WA. Matrix-delivered autologous mesenchymal stem cell therapy for refractory rectovaginal Crohn’s fistulas. Inflamm Bowel Dis. 2020;26(5):670–7.
Dozois EJ, Lightner AL, Dietz AB, Fletcher JG, Lee YS, Friton JJ, Faubion WA. Durable response in patients with refractory fistulizing perianal Crohn’s disease using autologous mesenchymal stem cells on a dissolvable matrix: results from the phase I stem cell on matrix plug trial. Dis Colon Rectum. 2023;66(2):243–52.
Fomekong E, Dufrane D, Berg BV, Andre W, Aouassar N, Veriter S, Raftopoulos C. Application of a three-dimensional graft of autologous osteodifferentiated adipose stem cells in patients undergoing minimally invasive transforaminal lumbar interbody fusion: clinical proof of concept. Acta Neurochir (Wien). 2017;159(3):527–36.
Shimomura K, Yasui Y, Koizumi K, Chijimatsu R, Hart DA, Yonetani Y, Ando W, Nishii T, Kanamoto T, Horibe S, et al. First-in-human pilot study of implantation of a scaffold-free tissue-engineered construct generated from autologous synovial mesenchymal stem cells for repair of knee chondral lesions. Am J Sports Med. 2018;46(10):2384–93.
Giannotti S, Trombi L, Bottai V, Ghilardi M, D’Alessandro D, Danti S, Dell’Osso G, Guido G, Petrini M. Use of autologous human mesenchymal stromal cell/fibrin clot constructs in upper limb non-unions: long-term assessment. PLoS One. 2013;8(8):e73893.
Kim YS, Choi YJ, Suh DS, Heo DB, Kim YI, Ryu JS, Koh YG. Mesenchymal stem cell implantation in osteoarthritic knees: is fibrin glue effective as a scaffold? Am J Sports Med. 2015;43(1):176–85.
Akgun I, Unlu MC, Erdal OA, Ogut T, Erturk M, Ovali E, Kantarci F, Caliskan G, Akgun Y. Matrix-induced autologous mesenchymal stem cell implantation versus matrix-induced autologous chondrocyte implantation in the treatment of chondral defects of the knee: a 2-year randomized study. Arch Orthop Trauma Surg. 2015;135(2):251–63.
Baba S, Yamada Y, Komuro A, Yotsui Y, Umeda M, Shimuzutani K, Nakamura S. Phase I/II trial of autologous bone marrow stem cell transplantation with a three-dimensional woven-fabric scaffold for periodontitis. Stem Cells Int. 2016;2016:6205910.
Zhuang Y, Gan Y, Shi D, Zhao J, Tang T, Dai K. A novel cytotherapy device for rapid screening, enriching and combining mesenchymal stem cells into a biomaterial for promoting bone regeneration. Sci Rep. 2017;7(1):15463.
Sponer P, Kucera T, Brtkova J, Urban K, Koci Z, Mericka P, Bezrouk A, Konradova S, Filipova A, Filip S. Comparative study on the application of mesenchymal stromal cells combined with tricalcium phosphate scaffold into femoral bone defects. Cell Transpl. 2018;27(10):1459–68.
Redondo LM, Garcia V, Peral B, Verrier A, Becerra J, Sanchez A, Garcia-Sancho J. Repair of maxillary cystic bone defects with mesenchymal stem cells seeded on a cross-linked serum scaffold. J Craniomaxillofac Surg. 2018;46(2):222–9.
Gjerde C, Mustafa K, Hellem S, Rojewski M, Gjengedal H, Yassin MA, Feng X, Skaale S, Berge T, Rosen A, et al. Cell therapy induced regeneration of severely atrophied mandibular bone in a clinical trial. Stem Cell Res Ther. 2018;9(1):213.
Lamas JR, Garcia-Fernandez C, Tornero-Esteban P, Lopiz Y, Rodriguez-Rodriguez L, Ortega L, Fernandez-Gutierrez B, Marco F. Adverse effects of xenogenic scaffolding in the context of a randomized double-blind placebo-controlled study for repairing full-thickness rotator cuff tears. Trials. 2019;20(1):387.
Abdal-Wahab M, Abdel Ghaffar KA, Ezzatt OM, Hassan AAA, El Ansary MMS, Gamal AY. Regenerative potential of cultured gingival fibroblasts in treatment of periodontal intrabony defects (randomized clinical and biochemical trial). J Periodontal Res. 2020;55(3):441–52.
Apatzidou DA, Bakopoulou AA, Kouzi-Koliakou K, Karagiannis V, Konstantinidis A. A tissue-engineered biocomplex for periodontal reconstruction. A proof-of-principle randomized clinical study. J Clin Periodontol. 2021;48(8):1111–25.
Park YB, Ha CW, Lee CH, Yoon YC, Park YG. Cartilage regeneration in osteoarthritic patients by a composite of allogeneic umbilical cord blood-derived mesenchymal stem cells and hyaluronate hydrogel: results from a clinical trial for safety androof-of-concept with 7 years of extended follow-up. Stem Cells Transl Med. 2017;6(2):613–21.
Morrison DA, Kop AM, Nilasaroya A, Sturm M, Shaw K, Honeybul S. Cranial reconstruction using allogeneic mesenchymal stromal cells: a phase 1 first-in-human trial. J Tissue Eng Regen Med. 2018;12(2):341–8.
Xiao Z, Tang F, Zhao Y, Han G, Yin N, Li X, Chen B, Han S, Jiang X, Yun C, et al. Significant improvement of acute complete spinal cord injury patients diagnosed by a combined criteria implanted with NeuroRegen scaffolds and mesenchymal stem cells. Cell Transpl. 2018;27(6):907–15.
de Garcia Frutos A, Gonzalez-Tartiere P, Coll Bonet R, Ubierna Garces MT, Del Arco Churruca A, Rivas Garcia A, Matamalas Adrover A, Salo Bru G, Velazquez JJ, Vila-Canet G, et al. Randomized clinical trial: expanded autologous bone marrow mesenchymal cells combined with allogeneic bone tissue, compared with autologous iliac crest graft in lumbar fusion surgery. Spine J. 2020;20(12):1899–910.
Hashemi SS, Mohammadi AA, Kabiri H, Hashempoor MR, Mahmoodi M, Amini M, Mehrabani D. The healing effect of Wharton’s jelly stem cells seeded on biological scaffold in chronic skin ulcers: a randomized clinical trial. J Cosmet Dermatol. 2019;18(6):1961–7.
He X, Wang Q, Zhao Y, Zhang H, Wang B, Pan J, Li J, Yu H, Wang L, Dai J, et al. Effect of intramyocardial grafting collagen scaffold with mesenchymal stromal cells in patients with chronic ischemic heart disease: a randomized clinical trial. JAMA Netw Open. 2020;3(9):e2016236.
Hisamatsu D, Itakura N, Mabuchi Y, Ozaki R, Suto EG, Naraoka Y, Ikeda A, Ito L, Akazawa C. CD73-positive cell spheroid transplantation attenuates colonic atrophy. Pharmaceutics. 2023;15(3):845.
Sands BE, Feagan BG, Rutgeerts P, Colombel J-F, Sandborn WJ, Sy R, D’Haens G, Ben-Horin S, Xu J, Rosario M. Effects of vedolizumab induction therapy for patients with Crohn’s disease in whom tumor necrosis factor antagonist treatment failed. Gastroenterology. 2014;147(3):618-627. e613.
Sands BE, Sandborn WJ, Van Assche G, Lukas M, Xu J, James A, Abhyankar B, Lasch K. Vedolizumab as induction and maintenance therapy for Crohn’s disease in patients naïve to or who have failed tumor necrosis factor antagonist therapy. Inflamm Bowel Dis. 2017;23(1):97–106.
Neurath MF. Targeting immune cell circuits and trafficking in inflammatory bowel disease. Nat Immunol. 2019;20(8):970–9.
Iwata K, Mikami Y, Kato M, Yahagi N, Kanai T. Pathogenesis and management of gastrointestinal inflammation and fibrosis: from inflammatory bowel diseases to endoscopic surgery. Inflamm Regen. 2021;41(1):21.
Singer NG, Caplan AI. Mesenchymal stem cells: mechanisms of inflammation. Annu Rev Pathol. 2011;6:457–78.
Ghannam S, Pène J, Torcy-Moquet 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.
Bartosh TJ, Ylostalo JH, Mohammadipoor A, Bazhanov N, Coble K, Claypool K, Lee RH, Choi H, Prockop DJ. Aggregation of human mesenchymal stromal cells (MSCs) into 3D spheroids enhances their antiinflammatory properties. Proc Natl Acad Sci U S A. 2010;107(31):13724–9.
Niibe K, Ohori-Morita Y, Zhang M, Mabuchi Y, Matsuzaki Y, Egusa H. A shaking-culture method for generating bone marrow derived mesenchymal stromal/stem cell-spheroids with enhanced multipotency in vitro. Front Bioeng Biotechnol. 2020;8:590332.
Bhang SH, Lee S, Shin JY, Lee TJ, Kim BS. Transplantation of cord blood mesenchymal stem cells as spheroids enhances vascularization. Tissue Eng Part A. 2012;18(19–20):2138–47.
Hsu SH, Hsieh PS. Self-assembled adult adipose-derived stem cell spheroids combined with biomaterials promote wound healing in a rat skin repair model. Wound Repair Regen. 2015;23(1):57–64.
Amos PJ, Kapur SK, Stapor PC, Shang H, Bekiranov S, Khurgel M, Rodeheaver GT, Peirce SM, Katz AJ. Human adipose-derived stromal cells accelerate diabetic wound healing: impact of cell formulation and delivery. Tissue Eng Part A. 2010;16(5):1595–606.
Yanagihara K, Uchida S, Ohba S, Kataoka K, Itaka K. Treatment of bone defects by transplantation of genetically modified mesenchymal stem cell spheroids. Mol Ther Methods Clin Dev. 2018;9:358–66.
Liu BH, Yeh HY, Lin YC, Wang MH, Chen DC, Lee BH, Hsu SH. Spheroid formation and enhanced cardiomyogenic potential of adipose-derived stem cells grown on chitosan. Biores Open Access. 2013;2(1):28–39.
Xu Y, Shi T, Xu A, Zhang L. 3D spheroid culture enhances survival and therapeutic capacities of MSCs injected into ischemic kidney. J Cell Mol Med. 2016;20(7):1203–13.
Mabuchi Y, Okawara C, Mendez-Ferrer S, Akazawa C. Cellular heterogeneity of mesenchymal stem/stromal cells in the bone marrow. Front Cell Dev Biol. 2021;9:689366.
Kern S, Eichler H, Stoeve J, Klüter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem cells. 2006;24(5):1294–301.
Lee M, Jeong SY, Ha J, Kim M, Jin HJ, Kwon S-J, Chang JW, Choi SJ, Oh W, Yang YS. Low immunogenicity of allogeneic human umbilical cord blood-derived mesenchymal stem cells in vitro and in vivo. Biochem Biophys Res Commun. 2014;446(4):983–9.
Raynaud CM, Rafii A. The necessity of a systematic approach for the use of MSCs in the clinical setting. Stem Cells Int. 2013;2013:892340.
Kepp O, Bezu L, Yamazaki T, Di Virgilio F, Smyth MJ, Kroemer G, Galluzzi L. ATP and cancer immunosurveillance. EMBO J. 2021;40(13):e108130.
Acknowledgements
We thank Dr. Rica Tanaka (Juntendo University School of Medicine) for providing clinical samples. We also acknowledge all the laboratory members involved in this project.
Funding
This work was financially supported by Otsuka Holdings Co., Ltd., Japan, and the Japan Society for the Promotion of Science (JSPS)/Ministry of Education, Culture, Sports, Science, and Technology (MEXT) KAKENHI Grant-in-Aid for Scientific Research (B) (grant number 21H03328). This work was partially supported by the JSPS/MEXT KAKENHI Grant-in-Aid for Early-Career Scientists (grant number 21K15888).
Author information
Authors and Affiliations
Contributions
DH designed and conceptualized the study. DH and TY prepared the data. DH performed data analysis. DH and AI prepared the tables. DH wrote the manuscript. YM, MW, and CA reviewed the manuscript. All the authors have read and approved the final draft of the manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
The study was conducted following the Declaration of Helsinki, and the protocol was approved by the Institutional Review Board (IRB) of Juntendo University Hospital (IRB #M16–0102; date of approval: February 21, 2020). Informed consent was obtained from all subjects involved in the study.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interest.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Hisamatsu, D., Ikeba, A., Yamato, T. et al. Optimization of transplantation methods using isolated mesenchymal stem/stromal cells: clinical trials of inflammatory bowel diseases as an example. Inflamm Regener 44, 37 (2024). https://doi.org/10.1186/s41232-024-00350-5
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s41232-024-00350-5