Skip to main content

Potentiation of cord blood cell therapy with erythropoietin for children with CP: a 2 × 2 factorial randomized placebo-controlled trial

Abstract

Background

Concomitant administration of allogeneic umbilical cord blood (UCB) infusion and erythropoietin (EPO) showed therapeutic efficacy in children with cerebral palsy (CP). However, no clinical studies have investigated the effects of UCB and EPO combination therapy using a 2 × 2 four-arm factorial blinded design with four arms. This randomized placebo-controlled trial aimed to identify the synergistic and individual efficacies of UCB cell and EPO for the treatment of CP.

Methods

Children diagnosed with CP were randomly segregated into four groups: (A) UCB+EPO, (B) UCB+placebo EPO, (C) placebo UCB+EPO, and (D) placebo UCB+placebo EPO. Based on the UCB unit selection criteria of matching for ≥ 4/6 of human leukocyte antigen (HLA)-A, -B, and DRB1 and total nucleated cell (TNC) number of ≥ 3 × 107/kg, allogeneic UCB was intravenously infused and 500 IU/kg human recombinant EPO was administered six times. Functional measurements, brain imaging studies, and electroencephalography were performed from baseline until 12 months post-treatment. Furthermore, adverse events were closely monitored.

Results

Eighty-eight of 92 children enrolled (3.05 ± 1.22 years) completed the study. Change in gross motor performance measure (GMPM) was greater in group A than in group D at 1 month (2.30 vs. 0.71, P = 0.025) and 12 months (6.85 vs. 2.34, P = 0.018) post-treatment. GMPM change ratios were calculated to adjust motor function at the baseline. Group A showed a larger improvement in the GMPM change ratio at 1 month and 12 months post-treatment than group D. At 12 months post-treatment, the GMPM change ratios were in the order of groups A, B, C, and D. These results indicate synergistic effect of UCB and EPO combination better than each single therapy. In diffusion tensor imaging, the change ratio of fractional anisotropy at spinothalamic radiation was higher in group A than group D in subgroup of age ≥ 3 years. Additionally, higher TNC and more HLA-matched UCB units led to better gross motor outcomes in group A. Adverse events remained unchanged upon UCB or EPO administration.

Conclusions

These results indicate that the efficacy of allogeneic UCB cell could be potentiated by EPO for neurological recovery in children with CP without harmful effects.

Trial registration

ClinicalTrials.gov, NCT01991145, registered 25 November 2013.

Background

Cerebral palsy (CP), the leading cause of motor impairment in early childhood, causes life-long disabilities [1, 2]. Clinical improvements following conventional rehabilitation or surgical therapies are limited [1]. Children with CP also present motor improvement to an extent until certain age [3]. Thereafter, it is difficult to acquire higher gross motor function and further functional decline may be observed in severely disabled children [4]. Lasting neuroinflammation and apoptosis occur in brains of CP patients, which cannot be corrected with conventional therapeutic approaches [5]. These disruptions influence the endogenous repair and regeneration after primary insult to the immature brain, known as a tertiary pathomechanism [6]. Cell and growth factor therapies are suggested to have therapeutic effects against this pathogenesis [6, 7].

Cell therapy in CP has been investigated for more than 10 years [1, 8]. The cell types used in clinical trials were umbilical cord blood (UCB) cells, olfactory ensheathing cells, neural stem cells, and neural progenitor cells [9]. Among these various cell types, the UCB containing stem cells are reportedly safe even for newborns [9,10,11]. Since its first use in 1988, UCB has been administered in over 100 indications including neurological disorders without reports of harmful effects [12,13,14]. UCB has been suggested to exert neuroprotective, anti-inflammatory, and anti-apoptotic effects [15]. Although autologous UCB may be ideal with positive results in previous clinical trials, most children with CP do not possess their own UCBs [16, 17]. UCB has substantial advantages over other cell sources because UCB has been banked worldwide and allogeneic UCB can be an alternative option with advantage of immune-tolerant characteristics [18].

So far, cell therapy has shown its efficacy mostly in preclinical stem cell researches. The main reasons that clinical applications of cell therapies for CP remain in the experimental stage are safety concerns and insufficient efficacy issues. Growth factors such as erythropoietin (EPO) and the granulocyte colony-stimulating factors have been introduced to potentiate the efficacy of cell therapy [19, 20]. EPO was reported to exert neuroprotective and neural repair effects, particularly in a neonatal hypoxic/ischemic brain injury CP model [21]. In a rat model of stroke, combination therapy with UCB cell and EPO exerted synergistic effects on neurological recovery, characterized by neurogenesis and angiogenesis, compared to UCB or EPO monotherapy [22]. Since both UCB and EPO could stimulate the same Akt signaling pathway, the effect of UCB might be reinforced by EPO [23, 24]. Furthermore, the clinical use of EPO showed neuroprotective effects among preterm infants [25, 26].

In our previous clinical trial, children with CP-administered intravenous allogeneic UCB infusion with EPO showed better outcomes than those administered EPO alone and control groups [27]. A subsequent trial assessing the therapeutic efficacy of UCB monotherapy suggested a therapeutic potential of UCB with its immunomodulatory characteristics including systemic pentraxin 3 (PTX3) upregulation [28]. However, the synergistic effect of UCB and EPO has not been assessed by direct group comparisons. This 2 × 2 factorial-designed double-blind placebo-controlled randomized trial was performed to identify the individual and/or synergistic efficacies of UCB and EPO combination therapy in children with CP for 1 year, with a longer period than that of our previous trials. In addition to the assessment of the functional changes, we assessed changes in the brain tissue through brain imaging and electroencephalography (EEG). Molecules potentially associated with neurological recovery were assayed and specific conditions of UCB and its recipients, serving as potential indicators of treatment effectiveness were also analyzed herein.

Methods

Participants

The inclusion criteria were children diagnosed with CP between 10 months and 6 years of age who had (i) allogeneic UCB units with criteria of ≥ 3 × 107/kg total nucleated cell (TNC) number and matched for ≥ 4/6 of the human leukocyte antigen (HLA)-A, B, and DRB1 at high resolution and (ii) a hemoglobin level ≤ 13.6 g/dL. Parents or representatives provided written informed consent to participate in the study. The exclusion criteria were aspiration pneumonia, genetic diseases, hypersensitivity to the study medications, coagulopathy, intractable epilepsy, hypertension, hepatic or renal impairments, malignancies, and absolute neutrophil count ≤ 500/dL. The protocol was approved by the institutional review board (No. 2013-04-41) and the Korean Ministry of Food and Drug Safety (No. 12515) (Clinicaltrials.gov NCT01991145) [29].

Study design and masking

The procedure was conducted as a double-blind placebo-controlled randomized trial. Participants were assigned into four groups using a block randomization code generated with SAS version 9.2 (SAS Institute Inc., Cary, NC, USA): (A) UCB+EPO, (B) UCB+placebo EPO (P-EPO), (C) placebo UCB (P-UCB)+EPO, and (D) P-UCB+P-EPO. Randomization was stratified by 2 factors: age (< 3 vs ≥ 3 years) and severity in the gross motor function classification system (GMFCS) level (GMFCS I–III, vs GMFCS IV–V) to ensure an even distribution into the allocation arms. The sample size was planned to recruit 30 patients per each group, total number of 120, based on central limit theorem [29]. To maintain blindness of the study of all participants, researchers, and outcome assessors to the treatment, an elaborate cooperation protocol was used (Fig. 1) [29]. Placebo materials of UCB, EPO, and cyclosporine were used. P-UCB was made from the subject’s own peripheral blood by UCB managers on the day of UCB therapy with the same appearance of UCB. Laboratory results such as the levels of hemoglobin affected by EPO and cyclosporine in the placebo groups which may affect the blindness of investigators were given artificial values by a designated investigator in the Department of Laboratory Medicine. The sham results were replaced by true values after completion of the study.

Fig. 1
figure 1

Screening, randomization, and follow-up. a The timeline of the study, b the cooperation of investigators to maintain double-blindness, and c the study flow. CP, cerebral palsy; DTI, diffusion tensor image; EEG, electroencephalogram; EPO, erythropoietin; FA, fractional anisotropy; GMFCS, gross motor function classification system; HLA, human leukocyte antigen; MRI, magnetic resonance imaging; PET, positron emission tomography; UCB, umbilical cord blood

All data were recorded on government-sponsored online case reporting system using the internet-based Clinical Research and Trial management system, Korea (C140005), and managed independently.

Procedures

Allogeneic UCB units were selected from the affiliated CHA cord blood bank after approval of Korean Organ Sharing Center. ABO blood types were matched, and two units of UCB were allowed to maintain the cell dose. Before administration, each unit was washed to eliminate dimethyl sulfoxide [30]. A single intravenous infusion of UCB or its placebo was performed. Groups A and B were administered with oral cyclosporine (ChongKunDang Pharm, Corp., Korea) at a dose of 7 mg/kg bid per day starting from 3 days before UCB administration; the same prescription was continued for 16 days (D− 3 to D+ 12 days). Groups C and D were administered placebos of UCB prepared from autologous peripheral blood and cyclosporine vehicle.

All participants in group A and C were administered EPO (Espogen®, LG Chem, Ltd., Korea) intravenously at a dose of 500 IU/kg at 2 h before UCB or placebo infusion. Subsequently, from D+ 3, each subject was injected five additional times with EPO at the same dose subcutaneously at 3-day intervals. Groups B and D were administered the EPO vehicle as a placebo. The vehicle placebo cyclosporine and EPO were provided by their own pharmaceutical companies.

All participants continued their conventional rehabilitation and were monitored for adverse events (AEs) (Fig. 1).

Outcomes

Functional outcomes

Primary outcomes were the total scores of the gross motor performance measure (GMPM) [31], gross motor functional measure (GMFM) [32], and raw scores of mental and motor scales of the Bayley Scales for Infant Development-II (BSID-II) [33] which were assessed at baseline and 1, 3, 6, and 12 months after treatment (Additional file 1). The reliabilities of the primary outcomes among assessors were established by the clinical study team [34,35,36].

Subgroup analyses were conducted to estimate favorable indications for treatment according to the following clinical conditions: gestational age (GA) on birth divided as term (GA ≥ 37 weeks) vs preterm (GA < 37 weeks); severity in the motor function impairment divided as mild (GMFCS levels I–III) vs severe (GMFCS levels IV–V) impairment; and age at the time of the procedure divided as younger (< 3 years) vs older (≥ 3 years) ages.

Secondary outcome measures were other functional measures including GMFCS [37], Pediatric Evaluation of Disability Inventory [38], Functional Independence Measure for Children [39], summed scores on muscular strength by Medical Research Council scale [40], Beery-Buktenica developmental test of visual-motor integration [41], selective control assessment of lower extremity [42], modified Ashworth scale [43], modified Tardieu scale [44], and Quality of Upper Extremity Skills Test [45] (Additional file 2). All functional outcomes were assessed as planned in the trial protocol by trained assessors who were not aware of group assignment.

Survey of parent perception of the intervention

The subjective satisfaction towards the intervention was surveyed among the caregivers of the patients at completion of the study before the group allocation was open (Additional file 2).

Imaging studies and electroencephalogram (EEG)

Brain magnetic resonance imaging (MRI) and 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-PET/CT) images were acquired at baseline and at 12 months after intervention. Diffusion tensor imaging (DTI) data from brain MRI were obtained to determine the effects of treatment on the white matter integration. Fractional anisotropy (FA) values were calculated by a voxel-based approach using the Tract-Based Spatial Statistics tool in an automated process [46, 47]. There are a total of 17 different white-mater tracts—single corpus callosum and bilateral fibers of eight tracts such as the anterior thalamic radiation (ATR), the cingulum in the cingulate cortex area, the cingulum in the hippocampal area, the corticospinal tract, the inferior fronto-occipital fasciculus, the superior and the inferior longitudinal fasciculus, and the uncinate fasciculus (Additional file 3)—from JHU white matter tractography atlases [48]. 18F-PET/CT images were acquired to assess differences in the regional brain glucose metabolism between groups and between the pre-treatment and the post-treatment imaging data (Additional file 4). Furthermore, sleeping asleep EEG was performed at baseline and 12 months after treatment. The average delta/alpha band power ratio (DAR) was obtained from five brain regions including the frontal, central, temporal, parietal, and occipital cortices, and their differences from pre-treatment to 12 months post-treatment were determined (Additional file 5).

Cytokines

Cytokines were analyzed using blood samples collected at 4 days before UCB infusion (D− 4), at the day of UCB injection prior to infusion (D− 0), and at 3 days, 10 days, and 30 days after UCB infusion (D+ 3, D+ 10, and D+ 30). Plasma levels of PTX-3, IL-8, TNF-α, and IL-1β were measured by an enzyme-linked immunosorbent assay and mRNA expression of the corresponding cytokines was measured by the reverse transcription polymerase chain reaction (Additional file 6) [28].

Statistical analyses

Statistical analyses were performed using SPSS version 21.0 software (SPSS, Inc., Chicago, IL, USA) and Prism 5.0 software (GraphPad, Inc., San Diego, CA, USA). Categorical variables were analyzed by the Fisher’s exact test. Functional outcomes and the FA values from DTIs were compared by Kruskal-Wallis test with post hoc analyses and Mann-Whitney U test appropriately. As for primary outcomes (GMPM and GMFM), changes in raw scores from baseline were compared among four groups at each time point (1, 3, 6, and 12 months). Then, the changed values between baseline and each time point were divided by baseline values, expressed as GMPM or GMFM change ratio in order to adjust the baseline function. Ratio values were also compared as changes of raw scores.

Analysis of variance (ANOVA) and the paired t test were used to evaluate regional brain glucose metabolism. EEG data were analyzed with the iSyncBrain® software version 2.0 (iMediSync, Inc., Seoul, Korea). Average DARs were calculated from five brain regions and the Mann-Whitney U test was used. Data were locked on March 27, 2018, and all statistical analyses were confirmed by a statistician. Missing data were filled in by the last observational carried forward imputation.

Results

From December 2013 to May 2016, 124 children with CP were enrolled, and 32 were excluded. Ninety-two subjects were randomly assigned to each group and four subjects withdrew their participation after the randomization. Eighty-eight participants (3.05 ± 1.22 years) were finally included: group A (UCB+EPO, n = 22), group B (UCB+P-EPO, n = 24), group C (P-UCB+EPO, n = 20), and group D (P-UCB+P-EPO, n = 22) (Fig. 1, Additional file 7). The demographic data revealed no significant differences in baseline variables among the groups (Table 1).

Table 1 Demographic and baseline participant characteristics (n = 88)

Adverse events

In groups A and C who were administered true EPO, the levels of hemoglobin, hematocrit, and red blood cells increased to the upper reference limits at 1 month post-therapy and then returned to the baseline levels (Additional file 8). All other laboratory data were within the reference ranges during the study period.

Eleven serious AEs were reported in the safety set. The distributions of serious AEs and non-serious AEs did not differ among the four groups, and all subjects recovered (Additional file 9).

Functional outcomes

There were no significant differences in baseline measurements among the four groups. All groups showed improvements in primary outcomes except for GMPM in group D during 1 year. Group A showed a greater improvement in the GMPM score at 1 month (2.30) and 12 months (6.85) post-treatment compared to group D (0.71 and 2.34) (P = 0.025 and P = 0.018, respectively) (Fig. 2A (a), Additional file 10). Randomization was stratified according to motor severity and age at the baseline, likely explaining the reason of the functional status that did not differ among the four groups. Despite performing a stratified randomization to ensure an even distribution, more participants in group C tended to have better motor function. Thus, we also calculated GMPM change ratios as (\( \frac{\left(\mathrm{score}\ \mathrm{at}\ \mathrm{the}\ \mathrm{time}\ \mathrm{point}-\mathrm{score}\ \mathrm{at}\ \mathrm{baseline}\ \right)}{\mathrm{score}\ \mathrm{at}\ \mathrm{baseline}\ } \)) for outcome comparisons to adjust motor function at the baseline. Group A showed a larger improvement in the GMPM change ratio at 1 month (0.11) and 12 months (0.33) post-treatment than group D (0.02 and 0.07) (P = 0.023 and P = 0.016, respectively) (Fig. 2A (c), Additional file 11). At 12 months post-treatment, the GMPM change ratios were in the order of groups A, B, C, and D, with changes in the GMFM ratio showing the same order (Fig. 2A (b, c)). These results indicate synergistic effect from UCB and EPO combination according to the changed score value in comparison with those values in individual therapies. The improved GMPM score (Δ 6.85) of group A is higher than those of group B (Δ 5.58) or C (Δ 3.67) at 12 months post-treatment.

Fig. 2
figure 2

Changes in gross motor outcome. A Changes in (a) GMPM, (b) GMFM change ratio, and (c) GMPM change ratio from baseline to 1, 3, 6, and 12 months post-treatment among group A, B, C, and D. GMPM and GMFM change ratios were calculated as \( \frac{\left(\mathrm{GMPM}\ \mathrm{at}\ \mathrm{the}\ \mathrm{time}\ \mathrm{point}-\mathrm{GMPM}\ \mathrm{at}\ \mathrm{baseline}\ \right)}{\mathrm{GMPM}\ \mathrm{at}\ \mathrm{baseline}\ } \) and \( \frac{\left(\mathrm{GMFM}\ \mathrm{at}\ \mathrm{the}\ \mathrm{time}\ \mathrm{point}-\mathrm{GMFM}\ \mathrm{at}\ \mathrm{baseline}\ \right)}{\mathrm{GMFM}\ \mathrm{at}\ \mathrm{baseline}\ } \), respectively. Group A (n = 22) received umbilical cord blood (UCB) with erythropoietin (EPO), group B (n = 24) received UCB with placebo EPO (P-EPO), group C (n = 20) received placebo UCB (P-UCB) and EPO, and group D (n = 22) received P-UCB and P-EPO. Data are shown in violin plots where dots represent each value, bold dotted lines represent the median and fine dotted lines represent lower and upper quartiles. Asterisk indicates significant difference in outcome scores between two groups based on post hoc analyses (P < 0.05) (Dunn’s multiple comparison test) following Kruskal-Wallis test. B Changes in GMPM change ratio according to (a) cell dose and (b) HLA disparity in group A. Subgroups with lower and higher TNC were categorized according to the median value of TNC in groups A and B. Subgroup from group A with higher TNC showed significant improvement in GMPM change ratio compared to group D after 12 months post-intervention. Data are also shown in violin plots where dots represent each value, bold dotted lines represent the median and fine dotted lines represent lower and upper quartiles. Asterisk indicates significant difference in outcome scores between two groups based on post hoc analyses (P < 0.05) (Dunn’s multiple comparison test) following Kruskal-Wallis test. The impact of HLA incompatibility was analyzed between HLA full-matched or 1 mis-matched and HLA 2 mis-matched cases in group A and B. In group A, variances of GMFM during baseline to 1 month (P = 0.036) and to 3 months (P = 0.05) were larger among the subjects who received more HLA-compatible UCB (n = 10) than those treated with HLA 2-mismatched UCB (n = 12). Asterisk indicates significant difference in outcome scores between two groups based on Mann-Whitney U test. EPO, erythropoietin; GMFM, gross motor function measure; GMPM, gross motor performance measure; HLA, human leukocyte antigen; TNC, total nucleated cell; UCB, umbilical cord blood

Efficacy factor analysis for UCB conditions revealed two significant findings (Additional file 12). When participants in groups A and B were divided into 2 subgroups by the median TNC value per body weight of each groups, the higher TNC subgroup in group A than in group D resulted in greater improvement in the GMPM change ratio at 12 months post-treatment (Fig. 2B (a)). Additionally, subjects administered higher matched units (HLA full-matched or 1 mis-matched; n = 10) showed greater increases in the GMFM score than those administered with the HLA 2 mis-matched units (n = 12) in group A at 1 month and 3 months post-treatment (P = 0.036 and P = 0.05, respectively) (Fig. 2B (b)). The changes of BSID-II raw scores in four groups were not different during the study period. Other secondary outcomes also did not differ among four groups.

Survey of parent perception of the intervention

The survey among the caregivers showed significantly higher satisfaction for language improvement in group A (P = 0.05) and for mental improvement in group B (P = 0.015) compared to those in group D (Additional file 13).

Subgroup analyses

Mild vs severe impairment

In the severe impairment subgroup (n = 55), group A showed a greater improvement in the GMPM change ratio compared to groups C and D, whereas comparison in the mild impairment group (n = 33) did not show a different outcome (panel A in Additional file 14).

Term vs preterm

In the term birth subgroup (n = 23), groups A and B showed a greater improvement in the GMPM change ratio compared to that in the groups C and D. There were no significant differences among 4 groups in preterm birth subgroup (n = 65) (panel B in Additional file 14).

Younger vs older age

There were no significant differences on any outcome measures in neither younger (n = 37) or older (n = 51) subgroups.

Structural changes in DTI

DTI data were obtained from 80 patients. No significant differences were observed in the FA change ratios calculated as (\( \frac{\left(\mathrm{FA}\ \mathrm{at}\ \mathrm{the}\ \mathrm{time}\ \mathrm{point}-\mathrm{FA}\ \mathrm{at}\ \mathrm{baseline}\ \right)}{\mathrm{FA}\ \mathrm{at}\ \mathrm{baseline}\ } \)) in 19 regions of interest among the 4 groups. However, in subpopulations of > 3 years, group A displayed the higher increment in the FA change ratio at the right ATR than group D (P < 0.05) (Additional file 15).

Metabolic changes in PET/CT

PET/CT data from 71 patients were available for analysis. Increased glucose metabolism was observed at the bilateral cerebellar hemisphere in group B, whereas it was increased at the midbrain and the thalamus in group D (Additional file 16).

EEG mapping of band power

EEG data from 78 patients were available for analysis. The relative value of average DAR showed a decreasing trend after treatment in groups A, B, and C, particularly at the posterior parietal and the occipital regions compared to that in group D. However, only group C showed a significant change between the baseline and post-treatment DAR (Fig. 3).

Fig. 3
figure 3

Electroencephalogram mapping before and after UCB injection. Average delta/alpha band power ratio (DAR) from electroencephalogram (EEG) is depicted on the 2D brain topomap. DAR from EEG taken before treatment, 12 months after treatment, and their difference (post-treatment–pre-treatment) are shown from left towards right. a Taken from group A (n = 20, mean age of pre-treatment EEG was 2.95 ± 1.20 years), b from group B (n = 20, mean age of pre-treatment EEG was 2.71 ± 1.27 years), c from group C (n = 20, mean age of pre-treatment EEG was 3.28 ± 1.27 years), and d from group D (n = 19, mean age of pre-treatment EEG was 4.17 ± 1.41 years). Among the total 88 participants, only 79 EEG data at baseline and 12 months post-treatment were able to be appropriately processed. Six participants lacked follow-up study, and 3 files were invalid on the analyzing program. *P < 0.05 by Mann-Whitney U test comparing the difference between pre- and post-treatment DAR. DAR, delta/alpha ratio; EEG, electroencephalogram; UCB, umbilical cord blood

Changes in mRNA expressions and cytokines associated with inflammation and innate immunity

Blood samples from 32 patients were available for analysis. They were re-grouped into 2 subgroups: those showing improvements in GMFCS (n = 12) or without improvements (n = 20) at 12 months post-treatment. The mRNA levels of IL-1β showed a greater increase at 3 days (P = 0.036) and 10 days (P = 0.013) post-treatment from baseline in the improved subgroup (Additional file 17).

In group B, when the cytokine change ratios calculated as (\( \frac{\left(\mathrm{value}\ \mathrm{at}\ \mathrm{the}\ \mathrm{time}\ \mathrm{point}-\mathrm{value}\ \mathrm{at}\ \mathrm{baseline}\ \right)}{\mathrm{value}\ \mathrm{at}\ \mathrm{baseline}\ } \)) were compared between “more-improved” and “less-improved” changes in the median GMPM score over 12 months, those of IL-8 and PTX3 were higher at 10 days post-treatment in the “more-improved” group than in the “less-improved” group (P = 0.039 and P = 0.031 respectively) (Additional file 18).

Discussion

This study aimed to verify the results of our previous clinical trial regarding the efficacy and safety of UCB therapy potentiated with EPO in children with CP [27]. The therapeutic effect of allogeneic UCB combined with EPO on motor function was reproduced. Our two published trials of UCB plus EPO and UCB alone were incomplete to assess the efficacy of allogeneic UCB and/or EPO under the same controlled conditions. This is the first study to analyze the contributions of combined or individual UCB and EPO in children with CP among four groups.

We observed no harmful effects related to UCB, EPO, or their combination which were decided according to the statistical analysis, the period of the occurrence, likely risk of each serious event depending on the treatment the patients received. Immunosuppressant administration for 16 days did not increase the occurrence of AEs in groups A and B. In the previous trial, pneumonia and irritability occurred more in the UCB- and the EPO-administered groups, possibly because of the long 1-month duration of immunosuppression treatment [27]. In this study, pneumonia listed in non-serious adverse events seemed to be more noticeable in group C (4 cases) than the other groups (P = 0.058). However, they were decided to be unlikely related to the intervention, according to clinical context. Furthermore, among the four, two patients were reported to have pneumonia at the baseline screening period and not after the intervention. All participants could be followed for 3–5 years, and there were no reports of serious AEs suspected to be related to the treatment. As hypothesized, the levels of hemoglobin, hematocrit, and red blood cell counts were increased by EPO administration, which returned to baseline within 1 year; no thromboembolic events were observed.

In the analyses of efficacy, group A showed better outcomes in the GMPM and GMPM change ratio than group D (Δ6.85 vs. Δ2.34, and 0.33 vs. 0.04, respectively), at 12 months post-treatment (Fig. 2A; Additional files 10, 11). Additionally, the GMFM change ratio showed a similar trend without statistical significance. The GMFM and the GMPM are specific tools for evaluating gross motor ability in children with CP. Typically, the scores of GMFM and GMPM are highly correlated [34]. The GMFM represents motor function related to ambulatory ability, whereas GMPM assesses the quality of movement which is specifically applied to CP [31, 50]. Thus, UCB and EPO combination therapy improved gross motor ability without reaching alteration of ambulatory function. As shown in Fig. 2A, UCB mono- (group B) or EPO mono- (group C) therapy groups also showed the trend of better motor recovery than control group. However, only merging treatment of UCB and EPO (group A) demonstrated noteworthy improvement after 1 year. Considering the difficulties in gaining function and frequent occurrence of motor deterioration in CP, this finding may be clinically applicable [3]. Therefore, in future trials, repeated treatments with UCB and EPO combination may lead to greater functional improvements. For BSID-II, there were no differences between groups whereas it showed significant difference in our previous trial [27]. The reason could be thought as follows: First, the means of cell dose were lower (in this study, A group 4.8 × 107, B group 5.0 × 107) than that of our previous study (8.33 × 107). Second, BSID-II may not have been able to reflect the changes because of its modest stability as a development assessment tool [51]. BSID-II has concerns on having limited floors and ceilings with selecting item sets. Third, there could be differences in clinical characteristics including typology and severity between the trials. Another prior clinical study also did not show significant findings in BSID-II [28]. However, evaluation tools seem to show different sensitivities according to slightly different typology and severity of study populations each time, and the tools which showed significance at 0–1 month seem to show consistent significance at longer terms [27, 28].

The differences in the number of introduced UCB cells in the clinical studies seemed to bring different results. In our previous trial, which used higher dose of the cells, GMPM score showed significant improvement by UCB and EPO therapy from 3 months [27], while it showed significance only at 12 months in this trial with small number of cells. Amount of cell dose is thought to be an important factor in administrating UCB as it was appeared in the previous clinical researches [17, 27, 28] and also an animal study showed a dose-response relationship [52]. Shorter duration of immunosuppression (16 days) in this trial than in the previous one (28 days) could be another factor. However, SAE and AE that might relate to use of cyclosporin was not reported this time. To enhance efficacy, administration of higher cell dosage and also repeated cell delivery could be suggested referring the other clinical trials [53, 54].

According to subgroup analyses, this therapy may be more effective in the severely motor impaired and term birth subgroup with a high risk of postnatal asphyxia. A higher cell dose and higher histocompatibility were reportedly found to be related to efficacy [27, 28]. First, TNC affected the treatment outcomes by increasing the GMPM score variance ratio in the higher TNC subgroup in group A, leading to better outcomes compared to those in groups C and D. Second, in terms of histocompatibility, fully matched and HLA 1-mismatched units administered to the subjects yielded better motor outcomes than those in the HLA 2-mismatched group. Thus, autologous UCB, although not available in most cases, may have superior clinical results in CP [17, 55].

Along with positive results upon functional measurements, brain imaging and EEG yielded certain significant findings. Analyses of DTI revealed the largest increase in the FA value in the right ATR in group A among patients of older ages (≥ 3 years), demonstrating improvement in the integrity of the white matter tract including myelination [56]. Changes in ATR indicate that facilitated reorganization occurred at the ascending somatosensory tracts [57]. The average age of group A was 3 years; at this age, the FA value does not typically increase, as the DTI values start plateauing at age 24 months [58, 59]. According to 18F-fluorodeoxyglucose-PET analysis, specific changes were not observed. In our previous studies with 2-week interval follow-up, inflammation was ameliorated in the posterior white matter [27, 28]. The large difference in the evaluation interval, which was 1 year in this study, appeared to give different results.

Brain wave analysis revealed a decreasing trend in DAR in groups A–C in the posterior cerebral cortices, whereas group D did not show this trend. While the delta band decrease starts from 6 months to 15 months [60] concurrent with the decreasing delta/theta band power ratio [61], alpha bands consistently increase with age showing over 80% dominant frequency within the alpha range by 3 years [62]. Therefore, the decreased DAR can be interpreted as the emergence of a more mature type of cortical sleep rhythms in the EEG.

Our previous study revealed increased PTX3 and IL-8 plasma levels within 2 weeks, which were correlated with functional outcomes in children with CP treated with UCB [28]. The current study also demonstrated the same results as plasma PTX3 and IL-8 levels were elevated in the more-improved subgroup only in group B. Thus, PTX3 and IL-8 appear to be related to the efficacy of UCB monotherapy. IL-8-mediated angiogenic pathway was known to be stimulated by UCB mononuclear cells [63]. Furthermore, IL-1β gene expression was elevated at 3 days and 10 days post-treatment in patients showing a definite improvement in the ambulatory level in groups A and B. IL-1β was known to be pro-inflammatory, but its neuroprotective characteristics in the injured brain was reported [64]. The core mechanism of the synergistic effect of UCB and EPO remains to be solved and the common pathway of UCB and EPO has not been investigated. IL-1β was increased in the subjects who are presumed to be responders in both groups A and B. And this new finding suggests potential role of IL-1β in neuroprotective mechanism of UCB and/or EPO treatment. Further studies will be required to determine other mechanisms other than those by PTX3 and IL-8.

This trial had some limitations. First, the ratio of outcome variance from baseline was adopted because subjects in group C tended to have better motor function. Although differences in baseline were not statistically significant, only a little difference may have affected their outcomes since it is more difficult to gain motor improvement from their palsied status in case of more severely impaired subjects [3, 4]. Therefore, we adopted changed ratios in the scores to minimize influence in their outcome by the baseline function. The efficacy showing time points in GMPM were 1 month and 12 months post-treatment, which differed from those in our previous studies, 3 and 6 months [27, 28]. This may be related to the lower cell number and shorter duration of immunosuppression in this study. Additionally, the ceiling effect may have also led to the negative results in Bayley scales. Lastly, the results subgroup analysis about cell dose and HLA disparity should be interpreted with deliberation due to small patient numbers.

Conclusion

In conclusion, these results suggest that allogeneic UCB infusion therapy with EPO is safe and UCB plus EPO can be synergistically effective than single treatment of each for children with CP. More compatible and greater numbers of cells may lead to better outcomes. Further studies are necessary to reveal the core pathway related to neuronal recovery and means for potentiating this efficacy.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

AE:

Adverse event

ATR:

Anterior thalamic radiation

BSID-II:

Bayley Scales for Infant Development-II

CP:

Cerebral palsy

DAR:

Delta/alpha band power ratio

EEG:

Electroencephalography

EPO:

Erythropoietin

FA:

Fractional anisotropy

18F-PET/CT:

18F-fluorodeoxyglucose positron emission tomography/computed tomography

GMFCS:

Gross motor function classification system

GMFM:

Gross motor functional measure

GMPM:

Gross motor performance measure

HLA:

Human leukocyte antigen

IL:

Interleukin

MRI:

Magnetic resonance imaging

PTX3:

Pentraxin 3

TNC:

Total nucleated cell

UCB:

Umbilical cord blood

References

  1. Colver A, Fairhurst C, Pharoah PO. Cerebral palsy. Lancet (London). 2014;383(9924):1240–9.

    Article  Google Scholar 

  2. Aisen ML, Kerkovich D, Mast J, Mulroy S, Wren TA, Kay RM, Rethlefsen SA. Cerebral palsy: clinical care and neurological rehabilitation. Lancet Neurol. 2011;10(9):844–52.

    Article  PubMed  Google Scholar 

  3. Rosenbaum PL, Walter SD, Hanna SE, Palisano RJ, Russell DJ, Raina P, Wood E, Bartlett DJ, Galuppi BE. Prognosis for gross motor function in cerebral palsy: creation of motor development curves. JAMA. 2002;288(11):1357–63.

    Article  PubMed  Google Scholar 

  4. Hanna SE, Rosenbaum PL, Bartlett DJ, Palisano RJ, Walter SD, Avery L, Russell DJ. Stability and decline in gross motor function among children and youth with cerebral palsy aged 2 to 21 years. Dev Med Child Neurol. 2009;51(4):295–302.

    Article  PubMed  Google Scholar 

  5. Dammann O. Persistent neuro-inflammation in cerebral palsy: a therapeutic window of opportunity? Acta Paediatr. 2007;96(1):6–7.

    Article  PubMed  Google Scholar 

  6. Fleiss B, Gressens P. Tertiary mechanisms of brain damage: a new hope for treatment of cerebral palsy? Lancet Neurol. 2012;11(6):556–66.

    Article  PubMed  Google Scholar 

  7. Stavsky M, Mor O, Mastrolia SA, Greenbaum S, Than NG, Erez O. Cerebral palsy-trends in epidemiology and recent development in prenatal mechanisms of disease, treatment, and prevention. Front Pediatr. 2017;5:21.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Dan B. Stem cell therapy for cerebral palsy. Dev Med Child Neurol. 2016;58(5):424.

    Article  PubMed  Google Scholar 

  9. Novak I, Walker K, Hunt RW, Wallace EM, Fahey M, Badawi N. Concise review: stem cell interventions for people with cerebral palsy: systematic review with meta-analysis. Stem Cells Transl Med. 2016;5(8):1014–25.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Kurtzberg J. A history of cord blood banking and transplantation. Stem Cells Transl Med. 2017;6(5):1309–11.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Sun J, Allison J, McLaughlin C, Sledge L, Waters-Pick B, Wease S, Kurtzberg J. Differences in quality between privately and publicly banked umbilical cord blood units: a pilot study of autologous cord blood infusion in children with acquired neurologic disorders. Transfusion. 2010;50(9):1980–7.

    Article  PubMed  Google Scholar 

  12. Mayani H, Wagner JE, Broxmeyer HE. Cord blood research, banking, and transplantation: achievements, challenges, and perspectives. Bone Marrow Transplant. 2020;55(1):14-61.

  13. Dessels C, Alessandrini M, Pepper MS. Factors influencing the umbilical cord blood stem cell industry: an evolving treatment landscape. Stem Cells Transl Med. 2018;7(9):643–50.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Kurtzberg J. Update on umbilical cord blood transplantation. Curr Opin Pediatr. 2009;21(1):22–9.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Liu WS, Chen CT, Foo NH, Huang HR, Wang JJ, Chen SH, Chen TJ. Human umbilical cord blood cells protect against hypothalamic apoptosis and systemic inflammation response during heatstroke in rats. Pediatr Neonatol. 2009;50(5):208–16.

    Article  PubMed  Google Scholar 

  16. Lee YH, Choi KV, Moon JH, Jun HJ, Kang HR, Oh SI, Kim HS, Um JS, Kim MJ, Choi YY, et al. Safety and feasibility of countering neurological impairment by intravenous administration of autologous cord blood in cerebral palsy. J Transl Med. 2012;10:58.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Sun JM, Song AW, Case LE, Mikati MA, Gustafson KE, Simmons R, Goldstein R, Petry J, McLaughlin C, Waters-Pick B, et al. Effect of autologous cord blood infusion on motor function and brain connectivity in young children with cerebral palsy: a randomized, placebo-controlled trial. Stem Cells Transl Med. 2017;6(12):2071–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Riordan NH, Chan K, Marleau AM, Ichim TE. Cord blood in regenerative medicine: do we need immune suppression? J Transl Med. 2007;5:8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Jing M, Shingo T, Yasuhara T, Kondo A, Morimoto T, Wang F, Baba T, Yuan WJ, Tajiri N, Uozumi T, et al. The combined therapy of intrahippocampal transplantation of adult neural stem cells and intraventricular erythropoietin-infusion ameliorates spontaneous recurrent seizures by suppression of abnormal mossy fiber sprouting. Brain Res. 2009;1295:203–17.

    Article  CAS  PubMed  Google Scholar 

  20. Rah WJ, Lee YH, Moon JH, Jun HJ, Kang HR, Koh H, Eom HJ, Lee JY, Lee YJ, Kim JY, et al. Neuroregenerative potential of intravenous G-CSF and autologous peripheral blood stem cells in children with cerebral palsy: a randomized, double-blind, cross-over study. J Transl Med. 2017;15(1):16.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Liu W, Shen Y, Plane JM, Pleasure DE, Deng W. Neuroprotective potential of erythropoietin and its derivative carbamylated erythropoietin in periventricular leukomalacia. Exp Neurol. 2011;230(2):227–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hwang S, Choi J, Kim M. Combining human umbilical cord blood cells with erythropoietin enhances angiogenesis/neurogenesis and behavioral recovery after stroke. Front Neurol. 2019;10:357.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Dasari VR, Veeravalli KK, Saving KL, Gujrati M, Fassett D, Klopfenstein JD, Dinh DH, Rao JS. Neuroprotection by cord blood stem cells against glutamate-induced apoptosis is mediated by Akt pathway. Neurobiol Dis. 2008;32(3):486–98.

    Article  CAS  PubMed  Google Scholar 

  24. van der Kooij MA, Groenendaal F, Kavelaars A, Heijnen CJ, van Bel F. Neuroprotective properties and mechanisms of erythropoietin in in vitro and in vivo experimental models for hypoxia/ischemia. Brain Res Rev. 2008;59(1):22–33.

    Article  PubMed  CAS  Google Scholar 

  25. Leuchter RH, Gui L, Poncet A, Hagmann C, Lodygensky GA, Martin E, Koller B, Darque A, Bucher HU, Huppi PS. Association between early administration of high-dose erythropoietin in preterm infants and brain MRI abnormality at term-equivalent age. JAMA. 2014;312(8):817–24.

    Article  PubMed  CAS  Google Scholar 

  26. Cho KH, Min K, Lee SH, Lee S, An SA, Kim M. Clinical trial of erythropoietin in young children with cerebral palsy. J Child Neurol. 2016;31(10):1227–34.

    Article  PubMed  Google Scholar 

  27. Min K, Song J, Kang JY, Ko J, Ryu JS, Kang MS, Jang SJ, Kim SH, Oh D, Kim MK, et al. Umbilical cord blood therapy potentiated with erythropoietin for children with cerebral palsy: a double-blind, randomized, placebo-controlled trial. Stem cells (Dayton). 2013;31(3):581–91.

    Article  CAS  Google Scholar 

  28. Kang M, Min K, Jang J, Kim SC, Kang MS, Jang SJ, Lee JY, Kim SH, Kim MK, An SA, et al. Involvement of immune responses in the efficacy of cord blood cell therapy for cerebral palsy. Stem Cells Dev. 2015;24(19):2259–68.

    Article  CAS  PubMed  Google Scholar 

  29. Cho KH, Min K, Lee SH, Kim M. Safety and efficacy of allogeneic umbilical cord blood therapy combined with erythropoietin in children with cerebral palsy: study protocol for a double-blind, randomized, placebo-controlled trial. Asia Pac J Clin Trials. 2017;2(4):129.

    Article  Google Scholar 

  30. Rubinstein P, Dobrila L, Rosenfield RE, Adamson JW, Migliaccio G, Migliaccio AR, Taylor PE, Stevens CE. Processing and cryopreservation of placental/umbilical cord blood for unrelated bone marrow reconstitution. Proc Natl Acad Sci U S A. 1995;92(22):10119–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Boyce WF, Gowland C, Rosenbaum PL, Lane M, Plews N, Goldsmith CH, Russell DJ, Wright V, Potter S, Harding D. The gross motor performance measure: validity and responsiveness of a measure of quality of movement. Phys Ther. 1995;75(7):603–13.

    Article  CAS  PubMed  Google Scholar 

  32. Russell DJ, Rosenbaum PL, Avery LM, Lane M. Gross motor function measure (GMFM-66 & GMFM-88) user's manual. London: Mac Keith Press; 2002.

    Google Scholar 

  33. Bayley N. Bayley scales of infant development, second edition edn. San Antonio: The Psychological Corporation; 1993.

    Google Scholar 

  34. Ko J, Kim M. Inter-rater reliability of the K-GMFM-88 and the GMPM for children with cerebral palsy. Ann Rehabil Med. 2012;36(2):233–9.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Ko J, Kim M. Reliability and responsiveness of the gross motor function measure-88 in children with cerebral palsy. Phys Ther. 2013;93(3):393–400.

    Article  PubMed  Google Scholar 

  36. Lee JH, Lim HK, Park E, Song J, Lee HS, Ko J, Kim M. Reliability and applicability of the Bayley scale of infant development-II for children with cerebral palsy. Ann Rehabil Med. 2013;37(2):167–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Palisano R, Rosenbaum P, Walter S, Russell D, Wood E, Galuppi B. Development and reliability of a system to classify gross motor function in children with cerebral palsy. Dev Med Child Neurol. 1997;39(4):214–23.

    Article  CAS  PubMed  Google Scholar 

  38. Haley SM, Coster WJ, Ludlow LH, Haltiwanger JT, Andrellos PJ. Pediatric Evaluation of Disability Inventory (PEDI). Boston: PEDI Research Group; 1992.

    Google Scholar 

  39. Uniform Data System for Medical Rehabilitation. Guide for the Uniform Data Set for Medical Rehabilitation for Children (WeeFIM). In.: State University of New York at Buffalo; 1993.

  40. Paternostro-Sluga T, Grim-Stieger M, Posch M, Schuhfried O, Vacariu G, Mittermaier C, Bittner C, Fialka-Moser V. Reliability and validity of the Medical Research Council (MRC) scale and a modified scale for testing muscle strength in patients with radial palsy. J Rehabil Med. 2008;40(8):665–71.

    Article  PubMed  Google Scholar 

  41. Beery KE, Buktenica NA, Beery NA. The Beery-Buktenica Developmental Test of Visual-Motor Integration : for children and adults (Beery VMI). Parsippany: Pearson; 2010.

    Google Scholar 

  42. Fowler EG, Staudt LA, Greenberg MB, Oppenheim WL. Selective Control Assessment of the Lower Extremity (SCALE): development, validation, and interrater reliability of a clinical tool for patients with cerebral palsy. Dev Med Child Neurol. 2009;51(8):607–14.

    Article  PubMed  Google Scholar 

  43. Yam WK, Leung MS. Interrater reliability of Modified Ashworth Scale and Modified Tardieu Scale in children with spastic cerebral palsy. J Child Neurol. 2006;21(12):1031–5.

    Article  PubMed  Google Scholar 

  44. Boyd RN, Graham HK. Objective measurement of clinical findings in the use of botulinum toxin type A for the management of children with cerebral palsy. Eur J Neurol. 1999;6(S4):s23–35.

    Article  Google Scholar 

  45. DeMatteo C, Law M, Russell D, Pollock N, Rosenbaum P, Walter S. QUEST: quality of upper extremity skills test manual. In. Hamilton: Neurodevelopmental Research Unit, Chedoke Campus, Chedoke-McMasters Hospital; 1992.

    Google Scholar 

  46. Merisaari H, Tuulari JJ, Karlsson L, Scheinin NM, Parkkola R, Saunavaara J, Lahdesmaki T, Lehtola SJ, Keskinen M, Lewis JD, et al. Test-retest reliability of diffusion tensor imaging metrics in neonates. NeuroImage. 2019;197:598–607.

    Article  PubMed  Google Scholar 

  47. Feng K, Rowell AC, Andres A, Bellando BJ, Lou X, Glasier CM, Ramakrishnaiah RH, Badger TM, Ou X. Diffusion tensor MRI of white matter of healthy full-term newborns: relationship to neurodevelopmental outcomes. Radiology. 2019;292(1):179–87.

    Article  PubMed  Google Scholar 

  48. Wakana S, Jiang H, Nagae-Poetscher LM, van Zijl PC, Mori S. Fiber tract-based atlas of human white matter anatomy. Radiology. 2004;230(1):77–87.

    Article  PubMed  Google Scholar 

  49. Ashwal S, Russman BS, Blasco PA, Miller G, Sandler A, Shevell M, Stevenson R. Quality Standards Subcommittee of the American Academy of N, Practice Committee of the Child Neurology S: Practice parameter: diagnostic assessment of the child with cerebral palsy: report of the Quality Standards Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society. Neurology. 2004;62(6):851–63.

    Article  CAS  PubMed  Google Scholar 

  50. Wright FV, Rosenbaum P, Fehlings D, Mesterman R, Breuer U, Kim M. The quality function measure: reliability and discriminant validity of a new measure of quality of gross motor movement in ambulatory children with cerebral palsy. Dev Med Child Neurol. 2014;56(8):770–8.

    Article  PubMed  Google Scholar 

  51. Johnson S, Marlow N. Developmental screen or developmental testing? Early Hum Dev. 2006;82(3):173–83.

    Article  PubMed  Google Scholar 

  52. Misra V, Ritchie MM, Stone LL, Low WC, Janardhan V. Stem cell therapy in ischemic stroke: role of IV and intra-arterial therapy. Neurology. 2012;79(13 Suppl 1):S207–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Gu J, Huang L, Zhang C, Wang Y, Zhang R, Tu Z, Wang H, Zhou X, Xiao Z, Liu Z, et al. Therapeutic evidence of umbilical cord-derived mesenchymal stem cell transplantation for cerebral palsy: a randomized, controlled trial. Stem Cell Res Ther. 2020;11(1):43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Huang L, Zhang C, Gu J, Wu W, Shen Z, Zhou X, Lu H. A randomized, placebo-controlled trial of human umbilical cord blood Mesenchymal stem cell infusion for children with cerebral palsy. Cell Transplant. 2018;27(2):325–34.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Papadopoulos KI, Low SS, Aw TC, Chantarojanasiri T. Safety and feasibility of autologous umbilical cord blood transfusion in 2 toddlers with cerebral palsy and the role of low dose granulocyte-colony stimulating factor injections. Restor Neurol Neurosci. 2011;29(1):17–22.

    PubMed  Google Scholar 

  56. Chang MC, Jang SH, Yoe SS, Lee E, Kim S, Lee DG, Son SM. Diffusion tensor imaging demonstrated radiologic differences between diplegic and quadriplegic cerebral palsy. Neurosci Lett. 2012;512(1):53–8.

    Article  CAS  PubMed  Google Scholar 

  57. Staudt M, Braun C, Gerloff C, Erb M, Grodd W, Krageloh-Mann I. Developing somatosensory projections bypass periventricular brain lesions. Neurology. 2006;67(3):522–5.

    Article  CAS  PubMed  Google Scholar 

  58. Yoshida S, Oishi K, Faria AV, Mori S. Diffusion tensor imaging of normal brain development. Pediatr Radiol. 2013;43(1):15–27.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Tau GZ, Peterson BS. Normal development of brain circuits. Neuropsychopharmacology. 2010;35(1):147–68.

    Article  PubMed  Google Scholar 

  60. Chu CJ, Leahy J, Pathmanathan J, Kramer MA, Cash SS. The maturation of cortical sleep rhythms and networks over early development. Clin Neurophysiol. 2014;125(7):1360–70.

    Article  CAS  PubMed  Google Scholar 

  61. Olbrich E, Rusterholz T, LeBourgeois MK, Achermann P. Developmental changes in sleep oscillations during early childhood. Neural plasticity. 2017;2017:6160959.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Scher MS. Pediatric neurophysiologic evaluation. In: Swaiman KF, Ashwal S, Ferriero DM, Schor NF, Finkel RS, editors. Swaiman's pediatric neurology: principles and practice. 6th ed. Edinburgh: Elsevier; 2018. p. 229–30.

    Google Scholar 

  63. Cho KH, Choi JI, Kim JO, Jung JE, Kim DW, Kim M. Therapeutic mechanism of cord blood mononuclear cells via the IL-8-mediated angiogenic pathway in neonatal hypoxic-ischaemic brain injury. Sci Rep. 2020;10(1):4446.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Hewett SJ, Jackman NA, Claycomb RJ. Interleukin-1beta in central nervous system injury and repair. Eur J Neurodegener Dis. 2012;1(2):195–211.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Umbilical cord blood was supplied by the CHA Cord Blood Bank. We appreciate a statistician, Sung Soo Kim, PhD, for the advice of statistical analysis, and a clinical research coordinator of this trial, Sunhee Lee, RN.

LG Chem, Ltd. (Republic of Korea) supported this trial by supplying Espogen® and its placebo made of corresponding vehicle and ChongKunDang Pharm, Corp. (Republic of Korea) supplied cyclosporine placebo also made of corresponding vehicle. EEG analysis was supported by iMediSync Inc. (Republic of Korea), and iCReaT system including e-CRF was provided by Korea Centers for Disease Control and Prevention.

Funding

This research was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant numbers HI13C1204 and HI16C1559).

Author information

Authors and Affiliations

Authors

Contributions

Kyunghoon Min: Data analysis and interpretation and manuscript writing. Mi Ri Suh: Data analysis and interpretation and manuscript writing. Kye Hee Cho: Collection and/or assembly of data and administrative support. Wookyung Park: Administrative support. Myung Seo Kang: Collection and/or assembly of data. Su Jin Jang: Data analysis and interpretation. Sang Heum Kim: Collection and/or assembly of data. Seonkyeong Rhie: Data analysis and interpretation. Jee In Choi: Collection and/or assembly of data. Hyun-Jin Kim: Collection and/or assembly of data. Kwang Yul Cha: Provision of study material and interpretation of results. MinYoung Kim: Conception and design, conduction of clinical trial as PI, manuscript writing, and final approval of manuscript. The authors read and approved the final manuscript.

Corresponding author

Correspondence to MinYoung Kim.

Ethics declarations

Ethics approval and consent to participate

The protocol was approved by the institutional review board (No. 2013-04-41) of CHA Bundang Medical Center and the Korean Ministry of Food and Drug Safety (No. 12515). Parents or representatives of all participant (age under 6 years or below) provided written informed consent to participate in the study.

Consent for publication

Consent for publication is not applicable for this manuscript.

Competing interests

The authors declare no potential conflicts of 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.

Primary functional outcome measurements.

Additional file 2.

Secondary functional outcome measurements and survey of parent perception of the intervention.

Additional file 3.

Brain MRI measurements and processing procedures.

Additional file 4.

PET/CT measurements and processing procedures.

Additional file 5.

EEG measurements and processing procedures.

Additional file 6.

Cytokine assay with ELISA and RT-PCR.

Additional file 7.

The number of missing data for study outcomes.

Additional file 8

Changes in the levels of hemoglobin (A), hematocrit (B), and red blood cell (C) during the study period of 1 year. Legends: According to the protocol, laboratory results were monitored at screening (V1), baseline (V2), 10 days (V5), 1-month (V7) and 1-year (V10) after EPO administration. Groups A and C administered with EPO showed higher levels of hemoglobin, hematocrit and red blood cell at 10 days and 1 month compared to groups B and D not treated with EPO (all P values < 0.001 by Kruskal-Wallis test). Bars represent SE. Abbreviations: EPO, erythropoietin; Hct, hematocrit; Hgb, hemoglobin; RBC, red blood cell; UCB, umbilical cord blood.

Additional file 9.

The distribution of adverse events during study period of 12 months.

Additional file 10.

Comparison of score changes in primary outcome measures.

Additional file 11.

Comparison of score changes in ratio to baseline in primary outcome measures.

Additional file 12.

Composition of allogeneic UCB units for groups A and B.

Additional file 13.

Survey of parent perception of the intervention. Legends: Satisfaction towards the intervention was surveyed among the caregivers after the patients completed the trial before notified of the group assignment, and caregivers of 63 patients completed the survey (response rate of 71.6%). All items were positive numbers. In comparison among four groups, the items of language and mental function were significantly different (P = 0.05, 0.015 respectively). In satisfaction of caregivers in aspect of language function (A), the caregivers of group A agreed more strongly that the language abilities of their children had improved compared to group D (P = 0.05). For the satisfaction of caregivers in aspect of mental function (B), the caregivers of group B agreed more strongly that the cognitive ability improved compared to group D (P = 0.015).

Additional file 14.

Subgroup comparisons of GMPM change ratios among 4 groups. Legends: Panel A shows subgroup analyses using GMPM change ratios according to (a) severe (GMFCS levels IV and V) vs. (b) mild (GMFCS levels I to III) impairment while Panel B shows GMPM change ratios according to (a) preterm (GA < 37 weeks) vs. (b) term (GA ≥ 37 weeks) birth. Among severely impaired subjects (n = 55, A: 14, B: 15, C: 10, D: 16), group A showed a larger improvement in the GMPM change ratio at 1 month and 12 months post-treatment than group D (P = 0.028 and P = 0.008, respectively) (Panel A-(a)). In term birth subgroup (n = 23, A: 6, B: 4, C: 4, D: 9), groups A and B showed significant improvement in the GMPM change ratio at 1, 6 month and 12 months post-treatment compared to groups C and D (P = 0.003, P = 0.029 and P = 0.011, respectively) (Panel B-(b)). Abbreviations: CP, cerebral palsy; EPO, erythropoietin; GA, gestational age; GMFCS, gross motor function classification system; UCB, umbilical cord blood.

Additional file 15.

Changes in FA value in children aged over 3 years/ Legends: In the subgroup analysis classified by median ages of four groups - younger aged subgroups (aged below 3 years; n = 49, median age 2.28y; A: 12, B: 14, C: 10, D: 13) vs. older subgroup (aged over 3 years; n = 39, median age 4.12y; A: 10, B: 10, C: 10, D: 9). Primary outcome measures did not show any significant differences between four groups. FA change ratio in right anterior thalamic radiation (ATR) between baseline and 12 months after intervention are depicted in this figure. Group A showed significant difference of FA change ratio in ATRR compared with group D. *P < 0.05 by post-hoc analysis after Kruskal-Wallis test comparing the difference among 4 groups. Abbreviations: ATR: anterior thalamic radiation; FA, fraction anisotropy.

Additional file 16.

Metabolic changes after UCB injection Legends: In comparison between pre- and post-intervention of PET/CT in each group, glucose metabolism of (A) bilateral cerebellar hemisphere increased in group B, (B) while the metabolic activity increased in midbrain and thalamus in group D. There were no meaningful changes in groups A and C. Data of 71 subjects were included in PET/CT analysis because, 7 did not undergo PET/CT on 12 months post-intervention, and 10 PET/CT imaging data were not appropriately processed due to anatomical distortion on SPM 12. Abbreviations: PET/CT, positron emission tomography/computed tomography; UCB, umbilical cord blood.

Additional file 17.

Gene expression of IL-1β among responders vs. non-responders in groups A and B. Legends: Group A (UCB + EPO) and group B (UCB + P-EPO) were re-grouped into 2 groups as responder subgroup and non-responder subgroup, where responder (n = 13) subgroup refers to those who showed improvements in GMFCS levels and non-responders (n = 20) refers to those who did not show improvements in GMFCS levels at 12 months post-intervention. Gene expression assay with RT-PCR showed bigger increment in IL1-β mRNA level in their relative values to the baseline level (D-4) at 3 d (D+3; P = 0.032) and 10 d (D+10; P = 0.013) post-intervention when comparing responder subgroup (dark-pink) with non-responder subgroup (light pink). *P < 0.05 by Mann-Whitney U test. Abbreviations: GMFCS, Gross Motor Functional Classification System; IL, interleukin; RT-PCR, reverse transcription polymerase chain reaction.

Additional file 18.

The cytokine analysis between responders and non-responders in group B.

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

Min, K., Suh, M.R., Cho, K.H. et al. Potentiation of cord blood cell therapy with erythropoietin for children with CP: a 2 × 2 factorial randomized placebo-controlled trial. Stem Cell Res Ther 11, 509 (2020). https://doi.org/10.1186/s13287-020-02020-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-020-02020-y

Keywords