Clinical and imaging outcomes after Intrathecal injection of umbilical cord tissue mesenchymal stem cells in cerebral palsy: a randomized double-blind sham-controlled clinical trial Study Below:

Click Here for the Link to the Study

Abstract

Background: This study assessed the safety and efficacy of intrathecal injection of umbilical cord tissue mesenchymal stem cells (UCT-MSC) in individuals with cerebral palsy (CP). The diffusion tensor imaging (DTI) was performed to evaluate the alterations in white-matter integrity. Methods: Participants (4–14 years old) with spastic CP were assigned in 1:1 ratio to receive either UCT-MSC or sham procedure. Single-dose (2 × 107) cells were administered in the experimental group. Small needle pricks to the lower back were performed in the sham-control arm. All individuals were sedated to prevent awareness. The primary endpoints were the mean changes in gross motor function measure (GMFM)-66 from baseline to 12 months after procedures. The mean changes in the modified Ashworth scale (MAS), pediatric evaluation of disability inventory(PEDI), and CP quality of life (CP-QoL) were also assessed. Secondary endpoints were the mean changes in fractional anisotropy (FA) and mean diffusivity (MD) of corticospinal tract (CST) and posterior thalamic radiation (PTR).

can also be useful to better understand the underlying mechanisms of stem cells in neuronal repair.

Conclusions

The intrathecal injection of the UCT-MSC may be safe in children diagnosed with CP and improve the clinical and imaging outcomes.

Results: There were 36 participants in each group. The mean GMFM-66 scores after 12 months of intervention were significantly higher in the UCT-MSC group compared to baseline (10.65; 95%CI 5.39, 15.91) and control (β 8.07; 95%CI1.62, 14.52; Cohen’s d 0.92). The increase was also seen in total PEDI scores (vs baseline 8.53; 95%CI 4.98, 12.08; vs control: β 6.87; 95%CI 1.52, 12.21; Cohen’s d 0.70). The mean change in MAS scores after 12 months of cell injection reduced compared to baseline (−1.0; 95%CI −1.31, −0.69) and control (β −0.72; 95%CI −1.18, −0.26; Cohen’s d 0.76).Regarding CP-QoL, mean changes in domains including friends and family, participation in activities, and communication were higher than the control group with a large effect size. The DTI analysis in the experimental group showed that mean FA increased (CST 0.032; 95%CI 0.02, 0.03. PTR 0.024; 95%CI 0.020, 0.028) and MD decreased (CST −0.035 × 10-3; 95%CI −0.04 × 10-3, −0.02 × 10-3. PTR −0.045 × 10-3; 95%CI −0.05 × 10-3, −0.03 × 10-3); compared tobaseline. The mean changes were significantly higher than the control group.

Conclusions: The UCT-MSC transplantation was safe and may improve the clinical and imaging outcomes.

Trial registration: The study was registered with ClinicalTrials.gov (NCT03795974).

Keywords: Cerebral palsy, Stem cell, Diffusion tensor imaging, Gross motor function, Children

Introduction

Stem cells are defined as pluripotent cells with the ability of self-renewal and the capacity of differentiation into the other cell types. There has been greater interest in the use of stem cell therapy in recent years; especially for the treatment of neurological disorders [1]. The central nervous system (CNS) is unable to regenerate new cells and damages to CNS can be permanent. Several studies assessed the safety and efficacy of different stem cells in the treatment of individuals diagnosed with stroke [2], multiple sclerosis [3], Parkinson’s disease[4], Huntington’s disease [5], and spinal cord injury [6].To date, many aspects of cell-based therapy remained unknown. The optimal dose, the most appropriate type of cell, and the best route of cell administration should be identified to provide safe and effective protocols without raising ethical concerns. Different underlying mechanisms of action have been described to justify the potential efficacy of stem cell therapy. Regarding mesenchymal stem cells (MSCs), it is believed that paracrine signaling and immunomodulation have the most critical effects. These cells can release neurotrophic factors, anti-oxidant molecules, angiogenic, anti-inflammatory, anti-fibrotic, and anti-apoptotic agents that enhance tissue repair after injury [7, 8]. The capacity of MSCs to regenerate and differentiate to new cells is another proposed mechanism [9] but studies reported its limited efficacy and showed that migration of cells to the site of injury is not necessary [7]. It is now clear that the mechanism of action for many stem cells is not a consequence of differentiation [10].The umbilical cord derives from the yolk sac and contains two arteries, one vein, and a gelatinous substance composed of sulfated proteoglycans with collagenous fibers; known as Wharton’s jelly [11]. The umbilical cord was found to have great proportions of stem cells. The first successful stem cell transplant was from umbilical cord blood cells on a 6-year-old-boy with Fanconi anemia in 1988 [12]. Low immunogenicity, low risk of graft versus host disease, and ease of cell collection are major advantages of using cells derived from the umbilical cords [11]. Umbilical cord tissue mesenchymal stem cell (UCT-MSC) has been used in recent studies to determine their safety and clinical efficacy [1315].Cerebral palsy (CP) is the leading cause of physical disability in children and is known as a group of non-progressive permanent CNS disorders that affected movements, muscle tone, and coordination [16]. The global prevalence of CP was estimated to be up to 3 per1000 individuals [17]. Preclinical studies reported that some types of stem cells (e.g., MSC) had neuro protective effects on animal models of neonatal hypoxia-ischemia[1820]. Few randomized trials demonstrated the promising clinical effects of stem cell therapy in children with CP (reviewed in [2123]). We conducted this randomized double-blind sham-controlled trial to assess the safety and clinical effects of intrathecal injection of UCT-MSC in CP. To assess the impact of cell therapy on the alteration of white matter integrity, we performed quantitative diffusion tensor imaging (DTI) before and after treatment. DTI is a non-invasive imaging method that can characterize the micro-structural changes in white matter tracts based on the diffusion of water molecules. We hypothesized that the UCT-MSC could significantly improve clinical and imaging outcomes compared to the control group (superiority trial).

Abbreviations

ANOVA: Analysis of variance; CI: Confidence interval; CNS: Central nervoussystem; CP: Cerebral palsy; CP-QoL: Cerebral palsy quality of life;CST: Corticospinal tract; DMEM: Dulbecco’s modified Eagle’s medium;DTI: Diffusion tensor imaging; EEG: Electro-encephalography; FA: Fractionalanisotropy; GEE: Generalized estimating equations; GMFCS: Gross motorfunction classification system; GMFM: Gross motor function measure;LAL: Limulus amebocyte lysate; MD: Mean diffusivity; MAS: ModifiedAshworth scale; MRI: Magnetic resonance imaging; PEDI: Pediatric evaluationof disability inventory; PTR: Posterior thalamic radiation; ROI: Region of interest; SD: Standard deviation; SEM: Standard error of the mean; UCT-MSC: Umbilical cord tissue mesenchymal stem cellSupplementary information The online version contains supplementary material available at https://doi.org/10.1186/s13287-021-02513-4.

Additional file 1: Supplement 1. Levels of gross motor functionclassification system (GMFCS).

Additional file 2: Supplement 2. Modified Ashworth scale

Additional file 3: Supplement 3. Sample size calculation

Additional file 4: Supplement 4. CONSORT 2010 checklist of information to include when reporting a randomised trial*

Acknowledgements The authors are grateful to all parents and patients for their participation inthis study. Our special thanks to Mr. Abolfazl Abdi, Mrs. Asa Fazilat, Dr. SamiraRaminfard, Dr. Houman Alizadeh, and Dr. Neda Pak for analyses of DTI data.

Authors contributionsMA, ARM, and MRA conducted the study design. SN and MGA performedthe rehabilitation therapy. HoM, HaM,AM, MZ, MN, RSB, AAH, and MV performed data acquisition. OG, FZ, and MH conducted the brain imaging of participants. MA and ART performed the interpretation of data and statisticalanalysis. MA and MS prepared the draft of the manuscript. AF and AGrevised the paper before submission. The author(s) read and approved thefinal manuscript.

Funding The Research Deputy of Tehran University of Medical Sciences providedfinancial and logistic support for this trial but had no role in study design,the collection, analysis, and interpretation of data, in the writing of thereport, or in the decision to submit the article for publication.

Availability of data and materials The datasets generated and/or analyzed during the current study are notpublicly available but are available from the corresponding author onreasonable request.

DeclarationsEthics approval and consent to participate The procedures were fully explained to the participants and their parents.Ethics committee of Tehran University of Medical Sciences approved thefinal methods (Number: IR.TUMS.VCRREC.1996.2506). Parents of participantshad access to all information. They were informed that participation wasoptional and withdrawal was possible whenever they asked for.Consent for publicationNot applicable.

Competing interests The authors declare that they have no competing interests.

Author details1Department of Science and Research Branch, AJA University of MedicalSciences, Tehran, Iran. 2Pediatrics Center of Excellence, Department of Pediatric Neurology, Children’s Medical Center, Growth and DevelopmentResearch Center, Tehran University of Medical Sciences, Tehran, Iran.3Department of Regenerative Medicine, Cell Science Research Center, RoyanInstitute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.4Department of Pediatric Neurology, Clinical Research Development Centerof Children Hospital, Hormozgan University of Medical Sciences, BandarAbass, Iran. 5Pediatrics Center of Excellence, Department of Radiology,Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran.6Clinical Biomechanics and Ergonomics Research Center, Department of Physical Medicine and Rehabilitation, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran. 7Pediatrics Center of Excellence PediatricHematology, Oncology and Stem Cell Transplantation Department, Children’sMedical Center, Tehran University of Medical Sciences, Tehran, Iran.8Psychiatry and Psychology Research Center, Tehran University of MedicalSciences, Tehran, Iran. 9Department of Pediatrics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran. 10Departmentof Pediatric Neurology, AJA University of Medical Sciences, Tehran, Iran.11Moser Center for Leukodystrophies, Kennedy Krieger Institute, Baltimore,MD 21205, USA. 12Department of Neurology and Pediatrics, Johns HopkinsUniversity School of Medicine, Baltimore, MD 21287, USA. 13Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA. 14Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.Received: 15 December 2020 Accepted: 8 June 2021

References1. Lindvall O, Kokaia Z. Stem cells for the treatment of neurological disorders.Nature. 2006;441(7097):1094–6. https://doi.org/10.1038/nature04960.2. Prasad K, Sharma A, Garg A, Mohanty S, Bhatnagar S, Johri S, et al.Intravenous autologous bone marrow mononuclear stem cell therapy forischemic stroke: a multicentric, randomized trial. Stroke. 2014;45(12):3618–24. https://doi.org/10.1161/STROKEAHA.114.007028.3. Mancardi GL, Sormani MP, Gualandi F, Saiz A, Carreras E, Merelli E, et al.Autologous hematopoietic stem cell transplantation in multiple sclerosis: aphase II trial. Neurology. 2015;84(10):981–8. https://doi.org/10.1212/WNL.0000000000001329.4. Venkataramana NK, Kumar SK, Balaraju S, Radhakrishnan RC, Bansal A, DixitA, et al. Open-labeled study of unilateral autologous bone-marrow-derivedmesenchymal stem cell transplantation in Parkinson’s disease. Transl Res.2010;155(2):62–70. https://doi.org/10.1016/j.trsl.2009.07.006.5. Bachoud-Lévi AC. Multicentric Intracerebral Grafting in Huntington’s,Schramm C, Remy P, Aubin G, Blond S, Bocket L, Brugières P, Calvas F,Calvier E, Cassim F. Human Fetal cell therapy in Huntington’s disease: arandomized, multicenter, phase ii trial. Mov Disord. 2020;35(8):1323–35.https://doi.org/10.1002/mds.28201.6. Zhu H, Poon W, Liu Y, Leung GK, Wong Y, Feng Y, et al. Phase I–II clinicaltrial assessing safety and efficacy of umbilical cord blood mononuclear celltransplant therapy of chronic complete spinal cord injury. Cell Transplant.2016;25(11):1925–43. https://doi.org/10.3727/096368916X691411.7. Baraniak PR, McDevitt TC. Stem cell paracrine actions and tissueregeneration. Regen Med. 2010;5(1):121–43. https://doi.org/10.2217/rme.09.74.8. Kusuma GD, Carthew J, Lim R, Frith JE. Effect of the microenvironment onmesenchymal stem cell paracrine signaling: opportunities to engineer thetherapeutic effect. Stem Cells Dev. 2017;26(9):617–31. https://doi.org/10.1089/scd.2016.0349.9. Giuliani N, Lisignoli G, Magnani M, Racano C, Bolzoni M, Dalla Palma B, et al.New insights into osteogenic and chondrogenic differentiation of humanbone marrow mesenchymal stem cells and their potential clinicalapplications for bone regeneration in pediatric orthopaedics. Stem Cells Int.2013;2013:1–11. https://doi.org/10.1155/2013/312501.Amanat et al. Stem Cell Research & Therapy (2021) 12:439Page 13 of 15

 10. Veneruso V, Rossi F, Villella A, Bena A, Forloni G, Veglianese P. Stem cellparacrine effect and delivery strategies for spinal cord injury regeneration. JControl Release. 2019;300:141–53. https://doi.org/10.1016/j.jconrel.2019.02.038.11. Azzopardi JI, Blundell R. Umbilical cord stem cells. Stem Cell Discov. 2018;8(01):1–11. https://doi.org/10.4236/scd.2018.81001.12. Gluckman E, Broxmeyer HE, Auerbach AD. Hematopoietic reconstitution in apatient with Fanconi’s anemia by means of umbilical cord blood from anHLA-identical sibling. N Engl J Med. 1989;321(17):1174–8. https://doi.org/10.1056/NEJM198910263211707.13. Riordan NH, Morales I, Fernández G, Allen N, Fearnot NE, Leckrone ME, et al.Clinical feasibility of umbilical cord tissue-derived mesenchymal stem cellsin the treatment of multiple sclerosis. J Transl Med. 2018;16(1):57. https:// doi.org/10.1186/s12967-018-1433-7.14. Bartolucci J, Verdugo FJ, González PL, Larrea RE, Abarzua E, Goset C, et al.Safety and efficacy of the intravenous infusion of umbilical cordmesenchymal stem cells in patients with heart failure: a phase 1/2randomized controlled trial (RIMECARD trial [randomized clinical trial of intravenous infusion umbilical cord mesenchymal stem cells oncardiopathy]). Circ Res. 2017;121(10):1192–204. https://doi.org/10.1161/ CIRCRESAHA.117.310712.15. Meng F, Xu R, Wang S, Xu Z, Zhang C, Li Y, et al. Human umbilical cord-derived mesenchymal stem cell therapy in patients with COVID-19: a phase1 clinical trial. Signal Transduct Target Ther. 2020;5(1):172. https://doi.org/10.1038/s41392-020-00286-5.16. Rosenbaum P. A report: the definition and classification of cerebral palsy.Dev Med Child Neurol. 2007;49(6):480.17. Toyokawa S, Maeda E, Kobayashi Y. Estimation of the number of children withcerebral palsy using nationwide health insurance claims data in Japan. DevMed Child Neurol. 2017;59(3):317–21. https://doi.org/10.1111/dmcn.13278.18. Zhang J, Yang C, Chen J, Luo M, Qu Y, Mu D, et al. Umbilical cordmesenchymal stem cells and umbilical cord blood mononuclear cellsimprove neonatal rat memory after hypoxia-ischemia. Behav Brain Res.2019;362:56–63. https://doi.org/10.1016/j.bbr.2019.01.012.19. Serrenho I, Rosado M, Dinis A, MCardoso C, Grãos M, Manadas B, et al. Stemcell therapy for neonatal hypoxic-ischemic encephalopathy: a systematicreview of preclinical studies. Int J Mol Sci. 2021;22(6):3142.20. Phillips AW, Johnston MV, Fatemi A. The potential for cell-based therapy inperinatal brain injuries. Transl Stroke Res. 2013;4(2):137–48. https://doi.org/10.1007/s12975-013-0254-5.21. Novak I, Walker K, Hunt RW, Wallace EM, Fahey M, Badawi N. Concisereview: Stem cell interventions for people with cerebral palsy: systematicreview with meta-analysis. Stem Cells Transl Med. 2016;5(8):1014–25. https:// doi.org/10.5966/sctm.2015-0372.22. Jantzie LL, Scafidi J, Robinson S. Stem cells and cell-based therapies forcerebral palsy: a call for rigor. Pediatr Res. 2018;83(1):345–55. https://doi.org/10.1038/pr.2017.233.23. Eggenberger S, Boucard C, Schoeberlein A, Guzman R, Limacher A, Surbek D, et al. Stem cell treatment and cerebral palsy: systemic review and meta-analysis. World J Stem Cells. 2019;11(10):891–903. https://doi.org/10.4252/ wjsc.v11.i10.891.24. Bobath K, Bobath B. The neurodevelopmental treatment. In: Scrutton D,editor. Management of the motor disorders of children with cerebral palsy.Oxford: Blackwell Scientific Publications Ltd; 1984. p. 6–18.25. Knox V, Evans AL. Evaluation of the functional effects of a course of Bobaththerapy in children with cerebral palsy: a preliminary study. Dev Med ChildNeurol. 2002 Jul;44(7):447–60. https://doi.org/10.1017/s0012162201002353.26. Palisano R, Rosenbaum P, Walter S, Russell D, Wood E, Galuppi B.Development and reliability of a system to classify gross motorfunction in children with cerebral palsy. Dev Med Child Neurol. 1997;39(4):214–23. https://doi.org/10.1111/j.1469-8749.1997.tb07414.x.27. Rosenbaum PL, Palisano RJ, Bartlett DJ, Galuppi BE, Russell DJ. Developmentof the gross motor function classification system for cerebral palsy. DevMed Child Neurol. 2008;50(4):249–53. https://doi.org/10.1111/j.1469-8749.2008.02045.x.28. Becher JG, Pangalila RF, Vermeulen RJ, Barneveld TA, Raats CJI. Richtlijndiagnostiek en behandeling van kinderen met spastische Cerebrale Parese.Utrecht: Nederlandse Vereniging van Revalidatieartse; 2006.29. Russell DJ, Rosenbaum PL, Wright M, Avery LM. Gross motor functionmeasure (GMFM-66 & GMFM-88) User’s Manual. London: Mac KeithPress; 2013.30. Salehi R, Keshavarz A, Negahban H, Saeedi A, Shiravi A, Ghorbani S,et al. Development of the Persian version of gross motor functionmeasure-88 (GMFM-88): A study of reliability. Trends Med Res. 2015;10(3):69–74.31. Gracies JM. Pathophysiology of spastic paresis. I: paresis and soft tissuechanges. Muscle Nerve. 2005;31(5):535–51. https://doi.org/10.1002/mus.20284.32. Charalambous CP. Interrater reliability of a modified Ashworth scale of muscle spasticity. In: Classic papers in orthopaedics. London: Springer; 2014.p. 415–7.33. Fosang AL, Galea MP, McCoy AT, Reddihough DS, Story I. Measures of muscle and joint performance in the lower limb of children with cerebralpalsy. Dev Med Child Neurol. 2003;45(10):664–70. https://doi.org/10.1017/ s0012162203001245.34. Clopton N, Dutton J, Featherston T, Grigsby A, Mobley J, Melvin J. Interraterand intrarater reliability of the Modified Ashworth Scale in children withhypertonia. Pediatr Phys Ther. 2005;17(4):268–74. https://doi.org/10.1097/01.pep.0000186509.41238.1a.35. Ansari NN, Naghdi S, Arab TK, Jalaie S. The interrater and intrarater reliabilityof the Modified Ashworth Scale in the assessment of muscle spasticity: limband muscle group effect. NeuroRehabilitation. 2008;23(3):231–7. https://doi.org/10.3233/NRE-2008-23304.36. Moradi Abbasabadi M, Akbarfahimi N, Hosseini SA, Rezasoltani P. Reliabilityof the Persian version of the pediatric evaluation of disability inventory in 3to 9-year old children with cerebral palsy. J Mazandaran Univ Med Sci. 2015;25(130):129–37.37. Soleimani F, Vameghi R, Kazemnejad A, Fahimi NA, Nobakht Z,Rassafiani M. Psychometric properties of the persian version of cerebralpalsy quality of life questionnaire for children. Iran J Child Neurol. 2015;9(1):76–86.38. Leemans A, Jeurissen B, Sijbers J, Jones DK. ExploreDTI: a graphical toolboxfor processing, analyzing, and visualizing diffusion MR data. Hawaii, USA:17th Annual Meeting of Intl Soc Mag Reson Med; 2009. p. 3537.39. Liang KY, Zeger SL. Longitudinal data analysis using generalized linearmodels. Biometrika. 1986;73(1):13–22. https://doi.org/10.1093/biomet/73.1.13.40. Wang X, Hu H, Hua R, Yang J, Zheng P, Niu X, et al. Effect of umbilicalcord mesenchymal stromal cells on motor functions of identical twinswith cerebral palsy: pilot study on the correlation of efficacy andhereditary factors. Cytotherapy. 2015;17(2):224–31. https://doi.org/10.1016/j.jcyt.2014.09.010.41. Liu X, Fu X, Dai G, Wang X, Zhang Z, Cheng H, et al. Comparativeanalysis of curative effect of bone marrow mesenchymal stem cell andbone marrow mononuclear cell transplantation for spastic cerebralpalsy. J Transl Med. 2017;15(1):48. https://doi.org/10.1186/s12967-017-1149-0.42. Chen G, Wang Y, Xu Z, Fang F, Xu R, Wang Y, et al. Neural stem cell-likecells derived from autologous bone mesenchymal stem cells for thetreatment of patients with cerebral palsy. J Transl Med. 2013;11(1):21.https://doi.org/10.1186/1479-5876-11-21.43. Gu J, Huang L, Zhang C, Wang Y, Zhang R, Tu Z, et al. Therapeutic evidenceof umbilical cord-derived mesenchymal stem cell transplantation forcerebral palsy: a randomized, controlled trial. Stem Cell Res Ther. 2020;11(1):43. https://doi.org/10.1186/s13287-019-1545-x.44. Rosenbaum PL, Walter SD, Hanna SE, Palisano RJ, Russell DJ, Raina P, et al.Prognosis for gross motor function in cerebral palsy: creation of motordevelopment curves. JAMA. 2002;288(11):1357–63. https://doi.org/10.1001/jama.288.11.1357.45. Sharma A, Sane H, Gokulchandran N, Kulkarni P, Gandhi S, Sundaram J, et al.A clinical study of autologous bone marrow mononuclear cells for cerebralpalsy patients: a new frontier. Stem Cells Int. 2015;2015:905874.46. Zali A, Arab L, Ashrafi F, Mardpour S, Niknejhadi M, Hedayati-Asl AA, et al.Intrathecal injection of CD133-positive enriched bone marrow progenitorcells in children with cerebral palsy: feasibility and safety. Cytotherapy. 2015;17(2):232–41. https://doi.org/10.1016/j.jcyt.2014.10.011.47. Wang X, Cheng H, Hua R, Yang J, Dai G, Zhang Z, et al. Effects of bonemarrow mesenchymal stromal cells on gross motor function measurescores of children with cerebral palsy: a preliminary clinical study.Cytotherapy. 2013;15(12):1549–62. https://doi.org/10.1016/j.jcyt.2013.06.001.48. Shroff G, Gupta A, Barthakur JK. Therapeutic potential of human embryonicstem cell transplantation in patients with cerebral palsy. J Transl Med. 2014;12(1):318. https://doi.org/10.1186/s12967-014-0318-7.Amanat et al. Stem Cell Research & Therapy  (2021) 12:439Page 14 of 15 49. Findikli N, Candan NZ, Kahraman S. Human embryonic stem cell culture:current limitations and novel strategies. Reprod Biomed Online. 2006;13(4):581–90. https://doi.org/10.1016/S1472-6483(10)60648-7.50. Luan Z, Liu W, Qu S, du K, He S, Wang Z, et al. Effects of neuroprogenitorcells transplantation in severe cerebral palsy. Cell Transplant. 2012;21(1_suppl):91–8. https://doi.org/10.3727/096368912X633806.51. Chen L, Huang H, Xi H, Xie Z, Liu R, Jiang Z, et al. Intracranial transplant of olfactory ensheathing cells in children and adolescents with cerebral palsy:a randomized controlled clinical trial. Cell Transplant. 2010;19(2):185–91.https://doi.org/10.3727/096368910X492652.52. Huang L, Zhang C, Gu J, Wu W, Shen Z, Zhou X, et al. A randomized,placebo-controlled trial of human umbilical cord blood mesenchymal stemcell infusion for children with cerebral palsy. Cell Transplant. 2018;27(2):325–34. https://doi.org/10.1177/0963689717729379.53. Kang M, Min K, Jang J, Kim SC, Kang MS, Jang SJ, et al. Involvement of immune responses in the efficacy of cord blood cell therapy forcerebral palsy. Stem Cells Dev. 2015;24(19):2259–68. https://doi.org/10.1089/scd.2015.0074.54. Min K, Song J, Kang JY, Ko J, Ryu JS, Kang MS, et al. Umbilical cord bloodtherapy potentiated with erythropoietin for children with cerebral palsy: adouble-blind, randomized, placebo-controlled trial. Stem Cells. 2013;31(3):581–91. https://doi.org/10.1002/stem.1304.55. Li Y, Chen J, Wang L, Lu M, Chopp M. Treatment of stroke in rat withintracarotid administration of marrow stromal cells. Neurology. 2001;56(12):1666–7. https://doi.org/10.1212/WNL.56.12.1666.56. Rodríguez-Frutos B, Otero-Ortega L, Gutiérrez-Fernández M, Fuentes B,Ramos-Cejudo J, Díez-Tejedor E. Stem cell therapy and administrationroutes after stroke. Transl Stroke Res. 2016;7(5):378–87. https://doi.org/10.1007/s12975-016-0482-6.57. Walczak P, Zhang J, Gilad AA, Kedziorek DA, Ruiz-Cabello J, Young RG, et al.Dual-modality monitoring of targeted intraarterial delivery of mesenchymalstem cells after transient ischemia. Stroke. 2008;39(5):1569–74. https://doi.org/10.1161/STROKEAHA.107.502047.58. Dong H, Li G, Shang C, Yin H, Luo Y, Meng H, et al. Umbilical cordmesenchymal stem cell (UC-MSC) transplantations for cerebral palsy. Am J Transl Res. 2018;10(3):901–6.59. Feng J, Offerman E, Lin J, Fisher E, Planchon SM, Sakaie K, et al. ExploratoryMRI measures after intravenous autologous culture-expanded mesenchymalstem cell transplantation in multiple sclerosis. Mult Scler J Exp Transl Clin.2019;5(2):2055217319856035.60. Chen YY, Zhang X, Lin XF, Zhang F, Duan XH, Zheng CS, et al. DTI metricscan be used as biomarkers to determine the therapeutic effect of stem cellsin acute peripheral nerve injury. J Magn Reson Imaging. 2017;45(3):855–62.https://doi.org/10.1002/jmri.25395.61. Jirjis MB, Valdez C, Vedantam A, Schmit BD, Kurpad SN. Diffusion tensorimaging as a biomarker for assessing neuronal stem cell treatmentsaffecting areas distal to the site of spinal cord injury. J Neurosurg Spine.2017;26(2):243–51. https://doi.org/10.3171/2016.5.SPINE151319.62. Yoshida S, Hayakawa K, Yamamoto A, Okano S, Kanda T, Yamori Y, et al.Quantitative diffusion tensor tractography of the motor and sensory tract inchildren with cerebral palsy. Dev Med Child Neurol. 2010;52(10):935–40.https://doi.org/10.1111/j.1469-8749.2010.03669.x.63. Thomas B, Eyssen M, Peeters R, Molenaers G, Van Hecke P, De Cock P, et al.Quantitative diffusion tensor imaging in cerebral palsy due toperiventricular white matter injury. Brain. 2005;128(11):2562–77. https://doi.org/10.1093/brain/awh600.64. Jiang H, Li X, Jin C, Wang M, Liu C, Chan KC, et al. Early diagnosis of spasticcerebral palsy in infants with periventricular white matter injury usingdiffusion tensor imaging. Am J Neuroradiol. 2019;40(1):162–8. https://doi.org/10.3174/ajnr.A5914.65. Wang S, Fan GG, Xu K, Wang C. Altered microstructural connectivity of thesuperior and middle cerebellar peduncles are related to motor dysfunctionin children with diffuse periventricular leucomalacia born preterm: a DTItractography study. Eur J Radiol. 2014;83(6):997–1004. https://doi.org/10.1016/j.ejrad.2014.03.010.66. Scheck SM, Boyd RN, Rose SE. New insights into the pathology of whitematter tracts in cerebral palsy from diffusion magnetic resonance imaging:a systematic review. Dev Med Child Neurol. 2012;54(8):684–96. https://doi.org/10.1111/j.1469-8749.2012.04332.x.67. Trivedi R, Gupta RK, Shah V, Tripathi M, Rathore RK, Kumar M, et al. Treatment-induced plasticity in cerebral palsy: a diffusion tensor imagingstudy. Pediatr Neurol. 2008;39(5):341–9. https://doi.org/10.1016/j.pediatrneurol.2008.07.012.68. Jiang H, Liu H, He H, Yang J, Liu Z, Huang T, et al. Specific white matterlesions related to motor dysfunction in spastic cerebral palsy: a meta-analysis of diffusion tensor imaging studies. J Child Neurol. 2020;35(2):146–54. https://doi.org/10.1177/0883073819879844.69. Arrigoni F, Peruzzo D, Gagliardi C, Maghini C, Colombo P, Iammarrone FS,et al. Whole-brain DTI assessment of white matter damage in children withbilateral cerebral palsy: evidence of involvement beyond the primary targetof the anoxic insult. Am J Neuroradiol. 2016;37(7):1347–53. https://doi.org/10.3174/ajnr.A4717.Publishers NoteSpringer Nature remains neutral with regard to jurisdictional claims inpublished maps and institutional affiliations.Amanat et al. Stem Cell Research & Therapy  (2021) 12:439Page 15 of 15