Skip to main content

Umbilical cord mesenchymal stromal cells in serum-free defined medium display an improved safety profile

Abstract

Background

Safety evaluations in preclinical studies are needed to confirm before translating a cell-based product into clinical application. We previously developed a serum-free, xeno-free, and chemically defined media (S&XFM–CD) for the derivation of clinical-grade umbilical cord-derived MSCs (UCMSCs), and demonstrated that intraperitoneal administration of UCMSCs in S&XFM–CD (UCMSCS&XFM−CD) exhibited better therapeutic effects than UCMSCs in serum-containing media (SCM, UCMSCSCM). However, a comprehensive investigation of the safety of intraperitoneal UCMSCS&XFM−CD treatment should be performed before clinical applications.

Methods

In this study, the toxicity, immunogenicity and biodistribution of intraperitoneally transplanted UCMSCS&XFM−CD were compared with UCMSCSCM in rats via general vital signs, blood routine, blood biochemistry, subsets of T cells, serum cytokines, pathology of vital organs, antibody production and the expression of human-specific gene. The tumorigenicity and tumor-promoting effect of UCMSCS&XFM−CD were compared with UCMSCSCM in nude mice.

Results

We confirmed that intraperitoneally transplanted UCMSCS&XFM−CD or UCMSCSCM did not cause significant changes in body weight, temperature, systolic blood pressure, diastolic blood pressure, heart rate, blood routine, T lymphocyte subsets, and serum cytokines, and had no obvious histopathology change on experimental rats. UCMSCS&XFM−CD did not produce antibodies, while UCMSCSCM had very high chance of antibody production to bovine serum albumin (80%) and apolipoprotein B-100 (60%). Furthermore, intraperitoneally injected UCMSCS&XFM−CD were less likely to be blocked by the lungs and migrated more easily to the kidneys and colon tissue than UCMSCSCM. In addition, UCMSCS&XFM−CD or UCMSCSCM showed no obvious tumorigenic activity. Finally, UCMSCS&XFM−CD extended the time of tumor formation of KM12SM cells, and decreased tumor incidence than that of UCMSCSCM.

Conclusions

Taken together, our data indicate that UCMSCS&XFM−CD display an improved safety performance and are encouraged to use in future clinical trials.

Introduction

Umbilical cord (UC) tissue is easily available and is thus a useful source of mesenchymal stromal cells (MSCs) [1, 2]. UC-derived MSCs (UCMSCs) show no significant variations between donors as they are obtained from newborn infants, thus lacking age-associated differences [3] and suggesting their suitability for cell-based treatments. Positive results have been obtained on the use of USMSCs in different in vivo studies, suggesting their potential for clinical applications [4]. However, questions have been raised on their safety, immunogenicity, biodistribution, and oncological implications in cases of previous malignancy, as well as the use of allogeneic MSCs [5].

The UCMSCs used in preclinical trials are usually grown in media containing fetal bovine serum (FBS), in line with the preparation of clinical-grade MSCs [6, 7]. However, the use of FBS, an animal-derived product, has raised serious safety concerns associated with the potential transmission of unknown viruses and the induction of undesirable immunological reactions [8]. A further issue is that FBS is poorly defined, presenting difficulties for standardization, which may adversely affect the clinical application of MSCs. There is also increasing concern regarding the regulation of animal sera in cell culture materials [9, 10]. In China, the quality control guidelines for the preparation of stem cells specify that animal sera should be avoided as far as possible, and encourage the use of serum-free media instead of serum-containing media (SCM) during the preparation of stem cells for clinical applications [11, 12]. In this regard, several studies have described commercially available serum-free media that can be used for isolating and growing MSCs (listed in Table 1 in our previous article [13]). However, these disclosed or commercial defined media cannot support large-scale expansion of UCMSCs in our laboratory. We have previously developed a serum-free, xeno-free, and chemically defined medium (S&XFM–CD) for the derivation of clinical-grade UCMSCs that contains nutrients, hormones, minerals, and growth factors (see Patent No. CN. ZL201210350602.0 and [13]). A previous study confirmed the superiority of delivery by intraperitoneal, rather than intravenous or anal injection, showing higher numbers of MSCs after delivery and better recovery from experimental colitis, suggesting that this might be the best route for delivering MSCs during colitis treatment [14]. Furthermore, we demonstrated that intraperitoneal delivery of UCMSCs in S&XFM–CD medium (UCMSCS&XFM−CD) was therapeutically more effective than the administration of UCMSCs in SCM (UCMSCSCM), shown by the enhancement of macrophage polarization from the proinflammatory M1 to anti-inflammatory M2 stages in an experimental model of acute colitis [15]. Furthermore, no transformation was observed in UCMSCS&XFM−CD and both the normal karyotype and genome were preserved, allowing their successful long-term culture in vitro [13]. However, the safety of using UCMSCs in serum-free medium remains debatable due to infrequent safety analyses and a lack of comprehensive tests. In addition, the safety of intraperitoneal delivery of UCMSCS&XFM−CD requires in-depth evaluation before this strategy can be translated into clinical application. Safety is critical for clinical applications of cell-based products [2], and the safety of these products thus need to be thoroughly evaluated in both preclinical and in vitro assessments [16].

Table 1 Vital sign assessment

Potential adverse events related to MSC use, including the risk of biodistributed, immunological, and oncological safety concerns, have received increasing attention in the last decade [17]. In addition, differences in cell culture condition may affect MSC characteristics, which may influence their safety, biological actions, and therapeutic effectiveness [18]. Biosafety analyses related to biodistribution, tumor-promoting effects, and the immunological consequences of UCMSCS&XFM−CD and UCMSCSCM administration require further comparison and evaluation. Hence, in this study, we compared the differences in safety-related biological characteristics between UCMSCS&XFM−CD and UCMSCSCM such as toxicity, immunogenicity, biodistribution, tumorigenicity, and tumor-promoting effects, and comprehensively evaluated the biosafety of UCMSCS&XFM−CD.

Materials and methods

UCMSCs isolation

Human umbilical cord tissue was obtained at the First Affiliated Hospital of Baotou Medical College from a full-term healthy infant delivered after a cesarean section, whose mother had signed an informed consent form and the procedure was approved by the Medical Ethics Committee of Baotou Medical College (No: 2021-033). Isolation and culture of UCMSCs were performed as previously described [19]. Briefly, the UC was rinsed with sterile phosphate-buffered saline (PBS) to remove any remaining blood, and the vessels were separated from the cord segments. After thorough washing with PBS, Wharton’s jelly was initially chopped and then sliced into pieces (0.5‒1 mm) which, following removal of the cord segments, were incubated for five days in either S&XFM–CD or SCM (IMDM with 10% FBS) at 37 °C and 5% CO2. Approximately half of the culture medium was replaced every two to three days. UCMSCs were passaged when 80‒90% confluent to densities of 3000 cells/cm2.

Flow cytometry

UCMSCS&XFM−CD and UCMSCSCM from the fifth passage were harvested, resuspended as single-cell suspensions, and incubated (1 h, 4 °C, in the dark) with commercial antibodies against CD14, CD19, CD29, CD34, CD44, CD45, CD73, CD90, CD105, and HLA-DR. The antibody details are provided in Additional file 1: Table S1 (all reagents were from BD). A FACSCalibur flow cytometer (Becton Dickinson, Franklin Lakes, NJ, USA) was used with data analysis with CellQuest Pro 3.7 software (Becton Dickinson).

In the animal experiments, blood samples were taken from the orbital venous plexus of the rats. After isolation, mononuclear cells were incubated as above with commercial antibodies against CD3, CD4, and CD8 (all reagents were from BD, Additional file 1: Table S1). The proportions of CD3+, CD4+, and CD8+ T cells, and the CD4+/CD8+ T cell ratios were analyzed on the flow cytometer as above.

Lineage differentiation and staining

UCMSCS&XFM−CD and UCMSCSCM at passage 5 were differentiated into adipocytes, osteocytes, and chondrocytes. Histochemistry was performed as described previously [20] (All reagents were from Cyagen, Guangzhou, China).

Animal experiments

All animals were provided and raised by the Experimental Animal Center of the First Affiliated Hospital of Baotou Medical College, with 12-h light–dark cycle and free access to food and water. All animal operations were approved by the Ethics Committee of Baotou Medical College (No: 2021-033) and followed the guidelines for the care and use of experimental animals of the National Institutes of Health. Then the animals were euthanized by intraperitoneal injection of pentobarbital sodium (180 mg/kg), and the follow-up experiments were carried out.

Toxicity, immunogenicity, and biodistribution assays

15 six-week-old male Sprague–Dawley (SD) rats (weighing 200 ± 20 g) were randomly divided into three groups, with 5 rats in each group. Of these, the UCMSCS&XFM−CD and UCMSCSCM groups received respective doses of UCMSCS&XFM−CD and UCMSCSCM (2 × 106 cells/kg body weight) suspended in 0.5 mL PBS via intraperitoneal injection once at the beginning of the study, and the PBS group received 0.5 mL of PBS.

Antibody detection experiments

Rats received intraperitoneal injections of UCMSCS&XFM−CD or UCMSCSCM (2 × 106 cells/kg body weight) in 0.5 ml of PBS (n = 5/group). Further injections were given weekly for the subsequent three weeks. Blood samples for antibodies against bovine serum albumin (BSA) and apolipoprotein B-100 (apoB-100) were collected before the injections and one week following the last injection.

Tumorigenicity assay

20 six-week-old male nude mice (weighing 20 ± 2 g) were randomly assigned to three groups, with 5 mice in each group. The groups received single subcutaneous inoculations of UCMSCS&XFM−CD, UCMSCSCM, and human colon cancer KM12SM cells (2 × 106 cells/mouse), respectively, into the lateral trunk region. The mice were checked twice a week thereafter for tumor growth (for 3 months).

Tumor-promoting assay

Nude mice as above were randomly divided into three groups. Single subcutaneous inoculations of KM12SM cells (2 × 106 cells/mouse) were given into the lateral trunk region alone or in conjunction with UCMSCS&XFM−CD or UCMSCSCM at a 1:1 ratio. After inoculation, the mice were observed twice a week for tumor growth. After sacrifice, the tumors were weighed and the volumes were calculated as ½ × minor axis2 × major axis.

General vital sign detection

The vital signs of the rats, including anal temperature, weight, blood pressure, and heart rate were observed and recorded. The injection sites were checked, anal temperatures were measured with thermometer (Ji Nuo Tai Technology, Beijing, China), and the blood pressure and heart rates with a Softron 2006A meter (Softron Biotechnology, Beijing, China).

Blood routine and biochemical index detection

Blood samples were collected from orbital venous plexus, and performed to detect and compare blood routine and biochemical indexes by using the kits (Jiancheng, Nanjing, China).

Enzyme-linked immunosorbent assays

Blood was collected and assayed for antibodies against BSA and apoB-100 by ELISA (Absin, Shanghai, China) as previously described [8, 21]. Interferon (IFN)-γ, tumor necrosis factor (TNF)-α, interleukin (IL)-4, and IL-6 levels in serum were also measured by ELISA (Neobioscience, Shenzhen, China).

Histological analysis

Fixed tissues (lung, liver, kidney, heart, testis, and colon) were paraffin-embedded, sectioned (5 µm), and stained with hematoxylin and eosin. The stained sections were observed by a blinded histopathologist under light microscopy to check for pathological changes.

qRT-PCR

Standard curves for the assessment of the biodistribution of UCMSCS&XFM−CD and UCMSCSCM were generated by administering serial dilutions of UCMSCs to mouse tissues. UCMSCs (2 × 10, 2 × 102, 2 × 103, 2 × 104, or 2 × 105) were added to the whole mouse tissues prior to homogenization. Total RNA was extracted from the hearts, brains, lungs, livers, kidneys, spleens, quadriceps, and iliac marrow of the rats using TRIzol reagent (Invitrogen, USA) and reverse-transcribed to cDNA using the QuantiTect Reverse Transcription Kit (Qiagen, Germany). The cDNA was amplified by qRT-PCR using Platinum SYBR Green PCR Mix (Invitrogen, USA) and a 7700 Sequence Detector (Applied Biosystems, USA) with human and mouse GAPDH primers (Human GAPDH primers: Forward primer, TGC TTT TAA CTC TGG TAA AGT GGA TA; Reverse primer, GTG GAA TCA TAT TGG AAC ATG TAA AC. Mouse GAPDH primers: forward primer, CAG CGA CAC CCA CTC CTC CAC CTT; reverse primer, CAT GAG GTC CAC CAC CCT GTT GCT).

Statistical analysis

The data were presented as mean ± standard deviation (SD). Differences between two groups were analyzed by t-tests and those between multiple groups by one-way analysis of variance (ANOVA) followed by Tukey’s multiple comparisons tests. P < 0.05 was considered statistically significant. Data were analyzed using SPSS 17.0 (SPSS, IL, USA).

Results

Characterization of UCMSCS&XFM−CD and UCMSCSCM

While both UCMSCS&XFM−CD and UCMSCSCM exhibited the essential fibroblast-like structure of MSCs, UCMSCS&XFM−CD appeared slender and bright (Fig. 1A). As shown by flow cytometry, both cell types showed high levels (> 95%) of CD29, CD44, CD73, CD90, and CD105 but poor expression (< 2%) of the CD14, CD19, CD34, CD45, and HLA-DR surface antigens (Fig. 1B). The abilities of UCMSCS&XFM−CD and UCMSCSCM to differentiate into osteocytes, chondrocytes, and adipocytes were confirmed by Alizarin Red, Alcian Blue, and Oil Red-O staining, respectively (Fig. 1C).

Fig. 1
figure 1

The characterization analysis of UCMSCS&XFM−CD and UCMSCSCM. A Morphology characteristics of UCMSCS&XFM−CD and UCMSCSCM. Scale bar = 100 μm. B Flow cytometry revealed that UCMSCS&XFM−CD and UCMSCSCM showed high expression of CD29, CD44, CD73, CD90 and CD105, but lacked expression of CD14, CD19, CD34, CD45 and HLA-DR surface antigens. C Representative images of osteogenic, chondrogenic, and adipogenic differentiation of UCMSCS&XFM−CD and UCMSCSCM after specific inductions and staining. Scale bars = 100 μm

Toxicity evaluation of UCMSCS&XFM−CD and UCMSCSCM

To evaluate potential toxic effects, rats received intraperitoneal injections of UCMSCSCM, UCMSCS&XFM−CD, or PBS followed by assessments on days 14 and 35. The rats in all three groups survived. The vital signs, including body weight, temperature, heart rate, and blood pressure (both systolic and diastolic) of rats in the UCMSCSCM and UCMSCS&XFM−CD groups were not significantly different from those in the PBS group at days 14 and 35 after cell transplantation (Table 1). Compared with the PBS group, alkaline phosphatase levels were significantly increased in the UCMSCSCM group on the 14th day (P < 0.05), but no changes were observed in the UCMSCS&XFM−CD group (Table 2). Albumin, urea nitrogen, and glucose in both the UCMSCSCM and UCMSCSCM groups were significantly increased (all P < 0.05), while there were no significant changes in other blood biochemical indices (Table 2). On day 35, none of the blood biochemical indices in the UCMSCS&XFM−CD and UCMSCSCM groups differed significantly from those in the PBS group (Table 2). Microscopic diagnosis showed no obvious histopathological changes associated with MSCs in the six tissues analyzed (lung, liver, kidney, heart, testis, and colon) in the UCMSCS&XFM−CD and UCMSCSCM groups (Fig. 2).

Table 2 Biochemical indicators test
Fig. 2
figure 2

Toxicity evaluation of UCMSCS&XFM−CD and UCMSCSCM. H&E-stained lung, liver, kidney, heart, testicle and colon in PBS, UCMSCS&XFM−CD or UCMSCSCM group for 28 days. Images were captured at magnification of 10×

Immunogenicity of UCMSCS&XFM−CD and UCMSCSCM

No significant changes were observed in the routine blood measurements between the UCMSCSCM and UCMSCS&XFM−CD groups compared with the PBS group on days 14 and 35 (Table 3). The CD3+, CD4+, and CD8+ T lymphocyte proportions and CD4+/CD8+ ratios in the UCMSCS&XFM−CD group were not significantly different from those in the UCMSCSCM group (Table 4). One rat was found to be positive for serum IFN-γ in the UCMSCSCM group but no IFN-γ positivity was observed in the PBS and UCMSCS&XFM−CD groups. All rats were negative for IL-4 and IL-10, and the serum TNF-α level did not change in the UCMSCSCM group relative to the PBS group (Table 5). All rats were evaluated for the production of antibodies binding to BSA and apoB-100. As expected, no antibodies were detected in the UCMSCS&XFM−CD group on days 21, 28, and 35. While in the UCMSCSCM group, one, three and four rats had developed antibody against BSA on days 21, 28, and 35, and one, two and three rats for antibody against apoB-100, respectively (Table 6).

Table 3 Blood routine test
Table 4 The sub-population of T-cells
Table 5 Cytokine levels
Table 6 Incidence of antibody to BSA and apoB-100

Biodistribution of UCMSCS&XFM−CD and UCMSCSCM

Previously, we found that intraperitoneal injection of both UCMSCS&XFM−CD and UCMSCSCM resulted in minimal recruitment and persistence of the cells in the colon after 72 h, shown by fluorescence imaging [15]. For further tracking and quantification of UCMSCs, UCMSCS&XFM−CD, and UCMSCSCM (2 × 106 cells) after intraperitoneal injection, the lungs, kidneys, livers, testes, hearts, and colons of the mice were collected at 24 and 72 h after transplantation and were quantified by qRT-PCR measurements and the construction of standard curves, as described previously [22]. Both UCMSCS&XFM−CD and UCMSCSCM were observed in the hearts, lungs, livers, testes, kidneys, and colons, and a large number of injected cells were blocked in the lungs at 24 h after intraperitoneal injection in both groups (Fig. 3A). Then, the number of UCMSCS&XFM−CD blocked in the lungs quickly decreased, and significantly fewer UCMSCS&XFM−CD than UCMSCSCM were in the lungs at 72 h after injection (P < 0.05, Fig. 3B, C). However, more UCMSCS&XFM−CD than UCMSCSCM retained in the kidneys and colon (both P < 0.05–0.01, Fig. 3B, C).

Fig. 3
figure 3

Migration and tumorigenic potential of UCMSCS&XFM−CD and UCMSCSCM. Percentages of UCMSCSCM or UCMSCS&XFM−CD in lung, kidney, liver, testicle, heart, and colon at 24 (A) and 72 h (B) after intraperitoneally injection. C The fold decrease in the number of UCMSCS&XFM−CD and UCMSCSCM in each organ from 24 to 72 h. D Representative images of tumorigenic activity of UCMSCS&XFM−CD and UCMSCSCM

Tumorigenic potential of UCMSCS&XFM−CD and UCMSCSCM

Mice inoculated with the KM12SM cell line formed progressive nodules at the injection site, which were found after sacrifice to cover a macroscopic tumor. The mice treated with UCMSCSCM, UCMSCS&XFM−CD, or PBS showed no clinical signs during the experimental period (Fig. 3D).

Tumor-promoting effect of UCMSCS&XFM−CD and UCMSCSCM

We next investigated the role of UCMSCS&XFM−CD and UCMSCSCM on tumor growth in nude mice. Subcutaneous injections of 2 × 106 KM12SM cells were given into the right flanks of the mice, together with equal numbers of UCMSCS&XFM−CD or UCMSCSCM. The control group was injected with PBS only. Co-injection of KM12SM cells with UCMSCSCM reduced the time taken for tumor formation to 13 days compared with 15 days in the group co-injected with UCMSCS&XFM−CD and 21 days in the PBS group (Fig. 4A). UCMSCSCM also increased the incidence of tumor development in the mice to 90% compared with 80% in mice co-injected with UCMSCS&XFM−CD and 50% in the PBS group (Fig. 4A). However, neither the weights nor volumes of the tumors differed significantly between the UCMSCS&XFM−CD and UCMSCSCM groups (Fig. 4B–D).

Fig. 4
figure 4

Tumor-promoting effect of UCMSCS&XFM−CD and UCMSCSCM. A Kaplan–Meier curve. The weights B and volumes C of the excised tumors between the UCMSCS&XFM−CD and UCMSCSCM groups. D Representative images of the excised tumors between the UCMSCS&XFM−CD and UCMSCSCM groups

Discussion

The evaluation of the safety of UCMSCs, including assessment of toxicity, abnormal immunological reactions, biodistribution, and tumorigenicity, is a major focus in the development of “cell-based therapies” [23]. UCMSCs cultured in FBS or platelet lysates have been increasingly shown to be safe without deterioration [24,25,26]. However, the usage of FBS or platelet lysates makes standardization of cell manufacturing process difficult because of the poorly defined nature. The use of S&XFM–CD, as an ideal cell culture medium for cell therapy, has been proposed to replace SCM during the preparation of stem cells [10]. In this study, we demonstrated that intraperitoneally transplanted UCMSCS&XFM−CD did not cause significant changes in general vital signs, histopathology, routine blood indices, T lymphocyte subsets, and serum cytokine levels, and showed no obvious tumorigenic activity in experimental rats. Furthermore, UCMSCS&XFM−CD did not result in the production of anti-BSA and apoB-100 antibodies, in contrast to UCMSCSCM which has a very high chance of producing antibodies to BSA (80%) and apoB-100 (60%). In addition, UCMSCS&XFM−CD extended the time taken for KM12SM cell tumor formation and decreased the tumor incidence compared with UCMSCSCM. Finally, intraperitoneally injected UCMSCS&XFM−CD were less likely to be blocked by the lungs and migrated more easily to the kidneys and colon than UCMSCSCM and had almost disappeared 72 h after transplantation. Thus, these findings indicate that UCMSCS&XFM−CD displayed an improved safety performance.

Although the levels of albumin, urea nitrogen, and glucose sugar were significantly elevated in the UCMSCS&XFM−CD group at day 14, the levels of these indicators are still within the reference range [27, 28]. According to the general guidelines for clinicopathological data analysis, clinicopathological parameters cannot be evaluated in isolation, and damage to organs is only likely when the activities or concentrations of these indicators are higher than twice the normal upper limit [28, 29]. Moreover, no significant differences were shown between the UCMSCS&XFM−CD and UCMSCSCM group, suggesting that the changes in blood biochemical indices caused by the intraperitoneal administration of UCMSCs were independent of the cell culture conditions. In addition, the levels of these three indicators did not differ significantly between the UCMSCS&XFM−CD and PBS groups on day 35. This indicates that changes in blood biochemical indices resulting from transplantation of UCMSCs into the abdominal cavity occur over a period of time, and that the indicators will gradually return to normal over time.

Intraperitoneally administered UCMSCS&XFM−CD did not induce immune changes, including alterations in the levels of routine blood indices, inflammatory cytokines, and T-cell sub-populations, which is consistent with the low expression of HLA-DR in UCMSCS&XFM−CD [13]. This suggested low immunogenicity of the allogeneic UCMSCs in S&XFM–CD, as they did not cause alloimmune reactions. Alloantibodies can also result from exposure to FBS [8, 30] and MSCs are often grown in medium containing FBS. Uptake of FBS by MSCs has been observed, together with the presence of FBS components on the cell surfaces [31]. It has also been reported that anti-BSA antibodies interfered with the efficacy of treatment [32] and an animal study observed marked humoral responses after administration of MSCs cultured in 20% FBS [31]. UCMSCSCM thus have a very high chance of inducing antibody production, while no antibodies are produced in UCMSCS&XFM−CD due to the absence of animal-derived ingredients. This is easy to understand as serum-free media have no animal-derived components. This feature of UCMSCS&XFM−CD is thus a great advantage in clinical applications, especially for repeat infusions.

The in vivo distribution of MSCs is closely related to the transplantation route [33]. The distribution of MSCs after intravenous infusion has been reported in many studies [34, 35]. A previous study confirmed that delivery by intraperitoneal injection resulted in greater MSC distribution and better recovery from experimental colitis than either intravenous or anal injections [14], and intraperitoneally administered UCMSCS&XFM−CD showed greater therapeutic efficacy than UCMSCSCM. Interestingly, we found that intraperitoneal infusion resulted in a similar cell distribution to intravenous infusion, such as lung preference within 24 h, but changing to a gradual predominance of kidney and colon distribution at 48 h [36]. We found that intraperitoneally injected UCMSCS&XFM−CD are less likely to be blocked by the lungs, which might reduce the risk of vascular embolism. The lung is the organ in which the injected cells are blocked most heavily compared with other organs and pulmonary complications are more likely to be life-threatening. This is related to the size of the cells, consistent with the findings of previous studies [24]. However, it is inconsistent with low expression of SDF-1 in UCMSCS&XFM−CD [13, 37], suggesting that cell size plays a greater role than SDF-1 expression. Of course, other unknown factors cannot be ruled out. In addition, UCMSCS&XFM−CD migrated more easily to the colon than UCMSCSCM, which may be related to the intraperitoneal injection route. This may be more beneficial for colitis. Although more UCMSCS&XFM−CD migrated to the kidneys in our study, in view of the differences about cell homing in healthy and colitis rats, more experiments are still needed to further confirm the distribution of cells in colitis rats. In this study, we used normal rats with normal immune function, while most of the patients in the clinic would have abnormal immune function, especially immune-related diseases. Thus, the life span and migration rate of UCMSCs may differ between the normal and injured colon due to the inflammatory microenvironment, and the secretion of inflammatory factors may attract UCMSCs homing to and staying in the injury area, which is supported by our previous findings [15].

Although there is still controversy surrounding the ability of MSCs to induce tumors, some studies have indicated that MSCs can indeed induce both tumor development and metastasis [38, 39]. The developers of cell therapy products are required to conduct tumor-promoting tests and consult with regulators. Our results show that UCMSCS&XFM−CD have a lower tumor-promoting effect than UCMSCSCM. We speculate that this may be due to the reduced release of SDF-1 by UCMSCS&XFM−CD [13, 40]. However, our previous results showed that UCMSCS&XFM−CD expressed high levels of PDGF and IGF-1 [13], and it has been found that these cytokines enhance MSC-mediated tumor promotion [41, 42]. This finding conflicts with our results and may be the consequence of different cell sources [43], different culture conditions, or different tumors [44, 45]. In short, UCMSCS&XFM−CD have low tumor-promoting ability, which would likely increase the use of cells. However, further research on the exact mechanism underlying the reduced tumor-promoting effect is needed.

Conclusions

In conclusion, the present study provided evidence that UCMSCS&XFM−CD display an improved safety performance. In combination with our previous reports that UCMSCS&XFM−CD are more effective in promoting recovery from experimental colitis [13, 15], we encourage the use of UCMSCS&XFM−CD in future clinical trials.

Availability of data and materials

The data presented in this study are available on request from the corresponding author.

Abbreviations

UC:

Umbilical cord

MSCs:

Mesenchymal stromal cells

UCMSCs:

UC-derived MSCs

FBS:

Fetal bovine serum

SCM:

Serum-containing media

S&XFM–CD:

Serum-free, xeno-free, and chemically defined medium

PBS:

Phosphate-buffered saline

SD:

Sprague–Dawley

BSA:

Bovine serum albumin

apoB-100:

Apolipoprotein B-100

IFN:

Interferon

TNF:

Tumor necrosis factor

IL:

Interleukin

References

  1. Najar M, Melki R, Khalife F, Lagneaux L, Bouhtit F, Moussa Agha D, Fahmi H, Lewalle P, Fayyad-Kazan M, Merim M. Therapeutic mesenchymal stem/stromal cells: value, challenges and optimization. Front Cell Dev Biol. 2021;9:716853.

    Article  PubMed  Google Scholar 

  2. Viswanathan S, Shi Y, Galipeau J, Krampera M, Leblanc K, Martin I, Nolta J, Phinney DG, Sensebe L. Mesenchymal stem versus stromal cells: international society for cell & gene therapy (isct(r)) mesenchymal stromal cell committee position statement on nomenclature. Cytotherapy. 2019;21:1019–24.

    Article  CAS  PubMed  Google Scholar 

  3. Raileanu VN, Whiteley J, Chow T, Kollara A, Mohamed A, Keating A, Rogers IM. Banking mesenchymal stromal cells from umbilical cord tissue: large sample size analysis reveals consistency between donors. Stem Cells Transl Med. 2019;8:1041–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kabat M, Bobkov I, Kumar S, Grumet M. Trends in mesenchymal stem cell clinical trials 2004–2018: is efficacy optimal in a narrow dose range? Stem Cells Transl Med. 2019;9:17.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Merimi M, Fahmi H, De Kock J, Beguin C, Burny A, Moll G, Poggi A, Najar M. Mesenchymal stem/stromal cells as a therapeutic tool in cell-based therapy and regenerative medicine: an introduction expertise to the topical collection. Cells. 2022;11:3158.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Phinney DG, Galipeau J. C. Msc Committee Of The International Society Of, and T Gene, Manufacturing mesenchymal stromal cells for clinical applications: a survey of Good Manufacturing Practices at U.S. academic centers. Cytotherapy. 2019;21:782–92.

    Article  PubMed  Google Scholar 

  7. Solomon J, Csontos L, Clarke D, Bonyhadi M, Zylberberg C, McNiece I, Kurtzberg J, Bell R, Deans R. Current perspectives on the use of ancillary materials for the manufacture of cellular therapies. Cytotherapy. 2016;18:1–12.

    Article  CAS  PubMed  Google Scholar 

  8. Owens SD, Kol A, Walker NJ, Borjesson DL. Allogeneic mesenchymal stem cell treatment induces specific alloantibodies in horses. Stem Cells Int. 2016;2016:5830103.

    Article  PubMed  PubMed Central  Google Scholar 

  9. van der Valk J, Bieback K, Buta C, Cochrane B, Dirks WG, Fu J, Hickman JJ, Hohensee C, Kolar R, Liebsch M, Pistollato F, Schulz M, Thieme D, Weber T, Wiest J, Winkler S, Gstraunthaler G. Fetal bovine serum (FBS): past - present - future. Altex. 2018;35:99–118.

    Article  PubMed  Google Scholar 

  10. Nguyen LT, Tran NT, Than UTT, Nguyen MQ, Tran AM, Do PTX, Chu TT, Nguyen TD, Bui AV, Ngo TA, Hoang VT, Hoang NTM. Optimization of human umbilical cord blood-derived mesenchymal stem cell isolation and culture methods in serum- and xeno-free conditions. Stem Cell Res Ther. 2022;13:15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cao J, Hao J, Wang L, Tan Y, Tian Y, Li S, Ma A, Fu B, Dai J, Zhai P, Xiang P, Zhang Y, Cheng T, Peng Y, Zhou Q, Zhao T. Developing standards to support the clinical translation of stem cells. Stem Cells Transl Med. 2021;10(Suppl 2):S85–95.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Zhao Q, Han Z, Wang J, Han Z. Development and investigational new drug application of mesenchymal stem/stromal cells products in China. Stem Cells Transl Med. 2021;10(Suppl 2):S18–30.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Wu X, Ma Z, Wu D. Derivation of clinical-grade mesenchymal stromal cells from umbilical cord under chemically defined culture condition - platform for future clinical application. Cytotherapy. 2020;22:377.

    Article  CAS  PubMed  Google Scholar 

  14. Wang M, Liang C, Hu H, Zhou L, Xu B, Wang X, Han Y, Nie Y, Jia S, Liang J, Wu K. Intraperitoneal injection (IP), Intravenous injection (IV) or anal injection (AI)? Best way for mesenchymal stem cells transplantation for colitis. Sci Rep. 2016;6:30696.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Wu X, Wu D, Mu Y, Zhao Y, Ma Z. Serum-free medium enhances the therapeutic effects of umbilical cord mesenchymal stromal cells on a murine model for acute colitis. Front Bioeng Biotechnol. 2020;8:586.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Lindblad RW, Ibenana L, Wagner JE, McKenna DH Jr, Hei DJ, Hematti P, Couture LA, Silberstein LE, Armant M, Rooney CM, Gee AP, Welniak LA, Heath Mondoro T, Wood DA, Styers D. Cell therapy product administration and safety: data capture and analysis from the Production assistance for cellular therapies (PACT) program. Transfusion. 2015;55:674–9.

    Article  PubMed  Google Scholar 

  17. Toyserkani NM, Jorgensen MG, Tabatabaeifar S, Jensen CH, Sheikh SP, Sorensen JA. Concise review: a safety assessment of adipose-derived cell therapy in clinical trials: a systematic review of reported adverse events. Stem Cells Transl Med. 2017;6:1786–94.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Egger D, Lavrentieva A, Kugelmeier P, Kasper C. Physiologic isolation and expansion of human mesenchymal stem/stromal cells for manufacturing of cell-based therapy products. Eng Life Sci. 2022;22:361–72.

    Article  CAS  PubMed  Google Scholar 

  19. Wu X, Kang H, Liu X, Gao J, Zhao K, Ma Z. Serum and xeno-free, chemically defined, no-plate-coating-based culture system for mesenchymal stromal cells from the umbilical cord. Cell Prolif. 2016;49:579–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wu X, Mu Y, Yao J, Lin F, Wu D, Ma Z. Adipose-derived stem cells from patients with ulcerative colitis exhibit impaired immunosuppressive function. Front Cell Dev Biol. 2022;10:822772.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Sakamoto N, Tsuji K, Muul LM, Lawler AM, Petricoin EF, Candotti F, Metcalf JA, Tavel JA, Lane HC, Urba WJ, Fox BA, Varki A, Lunney JK, Rosenberg AS. Bovine apolipoprotein B-100 is a dominant immunogen in therapeutic cell populations cultured in fetal calf serum in mice and humans. Blood. 2007;110:501–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Song WJ, Li Q, Ryu MO, Ahn JO, Ha Bhang D, Chan Jung Y, Youn HY. TSG-6 secreted by human adipose tissue-derived mesenchymal stem cells ameliorates DSS-induced colitis by inducing M2 macrophage polarization in mice. Sci Rep. 2017;7:5187.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Zhuang WZ, Lin YH, Su LJ, Wu MS, Jeng HY, Chang HC, Huang YH, Ling TY. Mesenchymal stem/stromal cell-based therapy: mechanism, systemic safety and biodistribution for precision clinical applications. J Biomed Sci. 2021;28:28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Liang Q, Li Q, Ren B, Yin ZQ. Intravenous infusion of small umbilical cord mesenchymal stem cells could enhance safety and delay retinal degeneration in RCS rats. BMC Ophthalmol. 2022;22:67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Shi L, Zhang Y, Dong X, Pan Y, Ying H, Chen J, Yang W, Zhang Y, Fei H, Liu X, Wei C, Lin H, Zhou H, Zhao C, Yang A, Zhou F, Zhang S. Toxicity from a single injection of human umbilical cord mesenchymal stem cells into rat ovaries. Reprod Toxicol. 2022;110:9–18.

    Article  CAS  PubMed  Google Scholar 

  26. Li X, Huang Q, Zhang X, Xie C, Liu M, Yuan Y, Feng J, Xing H, Ru L, Yuan Z, Xu Z, Yang Y, Long Y, Xing C, Song J, Hu X, Xu Q. Reproductive and developmental toxicity assessment of human umbilical cord mesenchymal stem cells in rats. Front Cell Dev Biol. 2022;10:883996.

    Article  PubMed  PubMed Central  Google Scholar 

  27. R.L. Hall, Clinical pathology of laboratory animals, 2015.

  28. Dirven H, Vist GE, Bandhakavi S, Mehta J, Fitch SE, Pound P, Ram R, Kincaid B, Leenaars CHC, Chen M, Wright RA, Tsaioun K. Performance of preclinical models in predicting drug-induced liver injury in humans: a systematic review. Sci Rep. 2021;11:6403.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Tayebi B, Babaahmadi M, Pakzad M, Hajinasrollah M, Mostafaei F, Jahangiri S, Kamali A, Baharvand H, Baghaban Eslaminejad M, Hassani SN, Hajizadeh-Saffar E. Standard toxicity study of clinical-grade allogeneic human bone marrow-derived clonal mesenchymal stromal cells. Stem Cell Res Therapy. 2022;13:213.

    Article  CAS  Google Scholar 

  30. Sundin M, Ringden O, Sundberg B, Nava S, Gotherstrom C, Le Blanc K. No alloantibodies against mesenchymal stromal cells, but presence of anti-fetal calf serum antibodies, after transplantation in allogeneic hematopoietic stem cell recipients. Haematologica. 2007;92:1208–15.

    Article  CAS  PubMed  Google Scholar 

  31. Spees JL, et al. Internalized antigens must be removed to prepare hypoimmunogenic mesenchymal stem cells for cell and gene therapy. Mol Therapy J Am Soc Gene Therapy. 2004;9(5):747.

    Article  CAS  Google Scholar 

  32. Horwitz EM, Gordon PL, Koo WK, Marx JC, Neel MD, McNall RY, Muul L, Hofmann T. Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: Implications for cell therapy of bone. Proc Natl Acad Sci USA. 2002;99:8932–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kamiyama Y, Naritomi Y, Moriya Y, Yamamoto S, Kitahashi T, Maekawa T, Yahata M, Hanada T, Uchiyama A, Noumaru A, Koga Y, Higuchi T, Ito M, Komatsu H, Miyoshi S, Kimura S, Umeda N, Fujita E, Tanaka N, Sugita T, Takayama S, Kurogi A, Yasuda S, Sato Y. Biodistribution studies for cell therapy products: current status and issues. Regen Ther. 2021;18:202–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Srinivasan RC, Kannisto K, Strom SC, Gramignoli R. Evaluation of different routes of administration and biodistribution of human amnion epithelial cells in mice. Cytotherapy. 2019;21:113–24.

    Article  CAS  PubMed  Google Scholar 

  35. Brooks A, Futrega K, Liang X, Hu X, Liu X, Crawford DHG, Doran MR, Roberts MS, Wang H. Concise review: quantitative detection and modeling the in vivo kinetics of therapeutic mesenchymal stem/stromal cells. Stem Cells Transl Med. 2018;7:78–86.

    Article  PubMed  Google Scholar 

  36. Aguilera Y, Mellado-Damas N, Olmedo-Moreno L, Lopez V, Panadero-Moron C, Benito M, Guerrero-Cazares H, Marquez-Vega C, Martin-Montalvo A, Capilla-Gonzalez V. Preclinical safety evaluation of intranasally delivered human mesenchymal stem cells in juvenile mice. Cancers. 2021;13:1169.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Luo Q, Zhang B, Kuang D, Song G. Role of stromal-derived factor-1 in mesenchymal stem cell paracrine-mediated tissue repair. Curr Stem Cell Res Ther. 2016;11:585–92.

    Article  CAS  PubMed  Google Scholar 

  38. Lan T, Luo M, Wei X. Mesenchymal stem/stromal cells in cancer therapy. J Hematol Oncol. 2021;14:195.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Antoon R, Wang XH, Saleh AH, Warrington J, Hedley DW, Keating A. Pancreatic cancer growth promoted by bone marrow mesenchymal stromal cell-derived IL-6 is reversed predominantly by IL-6 blockade. Cytotherapy. 2022;24:699–710.

    Article  CAS  PubMed  Google Scholar 

  40. Pan C, Fang Q, Liu P, Ma D, Cao S, Zhang L, Chen Q, Hu T, Wang J. Mesenchymal stem cells with cancer-associated fibroblast-like phenotype stimulate SDF-1/CXCR4 axis to enhance the growth and invasion of b-cell acute lymphoblastic leukemia cells through cell-to-cell communication. Front Cell Dev Biol. 2021;9:708513.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Li X, Fan Q, Peng X, Yang S, Wei S, Liu J, Yang L, Li H. Mesenchymal/stromal stem cells: necessary factors in tumour progression. Cell Death Discov. 2022;8:333.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Raghavan S, Snyder CS, Wang A, McLean K, Zamarin D, Buckanovich RJ, Mehta G. Carcinoma-associated mesenchymal stem cells promote chemoresistance in ovarian cancer stem cells via PDGF signaling. Cancers. 2020;12:2063.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kidd S, Spaeth E, Watson K, Burks J, Lu H, Klopp A, Andreeff M, Marini FC. Origins of the tumor microenvironment: quantitative assessment of adipose-derived and bone marrow-derived stroma. PLoS ONE. 2012;7:e30563.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lee HY, Hong IS. Double-edged sword of mesenchymal stem cells: Cancer-promoting versus therapeutic potential. Cancer Sci. 2017;108:1939–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Xuan X, Tian C, Zhao M, Sun Y, Huang C. Mesenchymal stem cells in cancer progression and anticancer therapeutic resistance. Cancer Cell Int. 2021;21:595.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (No. 81960252), Natural Science Foundation of Beijing (No. 7222267) and Natural Science Foundation of Inner Mongolia (Nos. 2021LHMS08019 and 2021LHMS08023). The funding bodies played no role in the design of the study and collection, analysis and interpretation of data and in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

XW and YY: conception and design, financial support, collection and/or assembly of data, data analysis and interpretation, manuscript writing, and final approval of manuscript; YM and ZM: provision of study material, collection and/or assembly of data, data analysis, interpretation, and final approval of manuscript; DW: conception and design, manuscript writing, data analysis, interpretation, and final approval of manuscript.

Corresponding author

Correspondence to Daocheng Wu.

Ethics declarations

Ethics approval and consent to participate

Human umbilical cord tissue for UCMSC isolation and culture was obtained at the First Affiliated Hospital of Baotou Medical College from a full-term healthy infant delivered after a cesarean section, whose mother had signed an informed consent form and the procedure was approved by the Medical Ethics Committee of Baotou Medical College (approved project: Mechanism of mesenchymal stem cells regulating macrophage polarization through microRNA-21 in the treatment of ulcerative colitis, approval number: 2021-033, Date of approval: April 19, 2021). All animals were provided and raised by the Experimental Animal Center of the First Affiliated Hospital of Baotou Medical College. All animal operations were approved by the Ethics Committee of Baotou Medical College (approved project: Mechanism of mesenchymal stem cells regulating macrophage polarization through microRNA-21 in the treatment of ulcerative colitis, approval number: 2021-033, Date of approval: April 19, 2021) and followed the guidelines for the care and use of experimental animals of the National Institutes of Health. Our manuscript is reported following the ARRIVE guidelines.

Consent for publication

Not applicable.

Competing interests

XW is a cofounder and CEO of Beijing Stem Cell (ProterCell) Biotechnology Co., Ltd. and Inner Mongolia Stem Cell (ProterCell) Biotechnology Co., Ltd. YY is a co-researcher for Beijing Stem Cell (ProterCell) Biotechnology Co., Ltd. and Research Center for Hua-Da Precision Medicine of Inner Mongolia Autonomous Region. All other authors declare that they have no competing interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1

. Table S1: Flow cytometry antibodies from BD bioscience.

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/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, X., Ma, Z., Yang, Y. et al. Umbilical cord mesenchymal stromal cells in serum-free defined medium display an improved safety profile. Stem Cell Res Ther 14, 360 (2023). https://doi.org/10.1186/s13287-023-03604-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-023-03604-0

Keywords