Skip to main content

Advertisement

Log in

An open-label phase 1 clinical trial of the allogeneic side population adipose-derived mesenchymal stem cells in SMA type 1 patients

  • Original Article
  • Published:
Neurological Sciences Aims and scope Submit manuscript

Abstract

Introduction

Spinal muscular atrophy (SMA), an autosomal recessive neurodegenerative disorder of alpha motor neurons of spinal cord associated with progressive muscle weakness and hypotonia, is the most common genetic cause of infant mortality. Although there is few promising treatment for SMA, but the field of translational research is active in it, and stem cell-based therapy clinical trials or case studies are ongoing. Combination of different therapeutic approaches for noncurative treatments may increase their effectiveness and compliance of patients. We present a phase 1 clinical trial in patients with SMA1 who received side population adipose-derived mesenchymal stem cells (SPADMSCs).

Methods

The intervention group received three intrathecal administrations of escalating doses of SPADMSCs and followed until 24 months or the survival time. The safety analysis was assessed by controlling the side effects and efficacy evaluations performed by the Hammersmith Infant Neurological Examination (HINE), Ballard score, and electrodiagnostic (EDX) evaluation. These evaluations were performed before intervention and at the end of the follow-up.

Results

The treatment was safe and well tolerated, without any adverse event related to the stem cell administration. One of the patients in the intervention group was alive after 24 months of study follow-up. He is a non-sitter 62-month-old boy with appropriate weight gain and need for noninvasive ventilation (NIV) for about 8 h per day. Clinical scores, need for supportive ventilation, and number of hospitalizations were not meaningful parameters in the response of patients in the intervention and control groups. All five patients in the intervention group showed significant improvement in the motor amplitude response of the tibial nerve (0.56mV; p: 0.029).

Conclusion

This study showed that SPADMSCs therapy is tolerable and safe with promising efficacy in SMA I. Probably same as other treatment strategies, early intervention will increase its efficacy and prepare time for more injections. We suggest EDX evaluation for the follow-up of treatment efficacy.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Data availability

All of the data and material is available in the Pediatrics Cell Therapy Research Center of the Tehran University of Medical Sciences. The data will be available on the ClinicalTrials.gov with the identifier no.: NCT02855112.

References

  1. Cobben JM, Lemmink HH, Snoeck I, Barth PA, van der Lee JH, de Visser M (2008) Survival in SMA type I: a prospective analysis of 34 consecutive cases. Neuromuscul Disord 18(7):541–544

    Article  CAS  Google Scholar 

  2. Lefebvre S, Bürglen L, Reboullet S, Clermont O, Burlet P, Viollet L, Benichou B, Cruaud C, Millasseau P, Zeviani M et al (1995) Identification and characterization of a spinal muscular atrophy-determining gene. Cell. 80(1):155–165

    Article  CAS  Google Scholar 

  3. Gawel M, Kostera-Pruszczyk A, Lusakowska A, Jedrzejowska M, Ryniewicz B, Lipowska M, Gawel D, Kaminska A (2015) Motor unit loss estimation by the multipoint incremental MUNE method in children with spinal muscular atrophy-a preliminary study. Neuromuscul Disord 25(3):216–221

    Article  Google Scholar 

  4. Mohseni R, Ashrafi MR, Ai J, Nikougoftar M, Mohammadi M, Ghahvechi-Akbari M, Shoae-Hassani A, Hamidieh AA (2019) Overexpression of SMN2 gene in motoneuron-like cells differentiated from adipose-derived mesenchymal stem cells by ponasterone A. J Mol Neurosci 67(2):247–257

    Article  CAS  Google Scholar 

  5. Wirth B (2000) An update of the mutation spectrum of the survival motor neuron gene (SMN1) in autosomal recessive spinal muscular atrophy (SMA). Hum Mutat 15(3):228–237

    Article  CAS  Google Scholar 

  6. Yeo CJJ, Darras BT (2020) Overturning the paradigm of spinal muscular atrophy as just a motor neuron disease. Pediatr Neurol 109:12–19

    Article  Google Scholar 

  7. Keinath MC, Prior DE, Prior TW (2021) Spinal muscular atrophy: mutations, testing, and clinical relevance. Appl Clin Genet 25(14):11–25. https://doi.org/10.2147/TACG.S239603

    Article  Google Scholar 

  8. Lunn MR, Wang CH (2008) Spinal muscular atrophy. Lancet 371(9630):2120–2133

    Article  Google Scholar 

  9. Gioia D et al (2020) Neural stem cell transplantation for neurodegenerative diseases. Int J Mol Sci 21(9):3103

    Article  Google Scholar 

  10. Noureddini M, Verdi J, Mortazavi-Tabatabaei SA, Sharif S, Azimi A, Keyhanvar P, Shoae-Hassani A (2012) Human endometrial stem cell neurogenesis in response to NGF and bFGF. Cell Biol Int 36(10):961–966

    Article  CAS  Google Scholar 

  11. Uccelli A (2013) Mesenchymal stem cells exert a remarkable regenerative effect requiring minimal CNS integration: commentary on: “Mesenchymal stem cells protect CNS neurons against glutamate excitotoxicity by inhibiting glutamate receptor expression and function” by Voulgari-Kokota et al. Exp Neurol 247:292–295

    Article  Google Scholar 

  12. O’Hare E, Young PJ (2009) Childhood spinal muscular atrophy and stem cell research: is cellular replacement therapy the answer? (Review). Mol Med Rep 2(1):3–5

    PubMed  Google Scholar 

  13. Abati E, Bresolin N, Comi GP, Corti S (2019) Preconditioning and cellular engineering to increase the survival of transplanted neural stem cells for motor neuron disease therapy. Mol Neurobiol 56(5):3356–3367

    Article  CAS  Google Scholar 

  14. Li T, Ma H, Ma H, Ma Z, Qiang L, Yang Z, Yang X, Zhou X, Dai K, Wang J (2019) Mussel-inspired nanostructures potentiate the immunomodulatory properties and angiogenesis of mesenchymal stem cells. ACS Appl Mater Interfaces 11(19):17134–17146. https://doi.org/10.1021/acsami.8b22017

    Article  CAS  PubMed  Google Scholar 

  15. Urrutia DN, Caviedes P, Mardones R, Minguell JJ, Vega-Letter AM, Jofre CM (2019) Comparative study of the neural differentiation capacity of mesenchymal stromal cells from different tissue sources: an approach for their use in neural regeneration therapies. PLoS One 14(3):e0213032. https://doi.org/10.1371/journal.pone.0213032

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Jin J, Shi Y, Gong J, Zhao L, Li Y, He Q, Huang H (2019) Exosome secreted from adipose-derived stem cells attenuates diabetic nephropathy by promoting autophagy flux and inhibiting apoptosis in podocyte. Stem Cell Res Ther 10(1):95. https://doi.org/10.1186/s13287-019-1177-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Carrozzi M, Amaddeo A, Biondi A, Zanus C, Monti F, Alessandro V (2012) Stem cells in severe infantile spinal muscular atrophy (SMA1). Neuromuscul Disord 22(11):1032–1034

    Article  Google Scholar 

  18. Villanova M, Bach JR (2015) Allogeneic mesenchymal stem cell therapy outcomes for three patients with spinal muscular atrophy type 1. Am J Phys Med Rehabil 94(5):410–415

    Article  Google Scholar 

  19. El-Badawy A, Amer M, Abdelbaset R, Sherif SN, Abo-Elela M, Ghallab YH, Abdelhamid H, Ismail Y, El-Badri N (2016) Adipose stem cells display higher regenerative capacities and more adaptable electro-kinetic properties compared to bone marrow-derived mesenchymal stromal cells. Sci Rep 6:37801

    Article  CAS  Google Scholar 

  20. Hirano A, Sano M, Urushihata N, Tanemura H, Oki K, Suzaki E (2018) Assessment of safety and feasibility of human allogeneic adipose-derived mesenchymal stem cells in a pediatric patient. Pediatr Res 84(5):575–577

    Article  Google Scholar 

  21. Gimeno ML, Fuertes F, Barcala Tabarrozzi AE, Attorressi AI, Cucchiani R, Corrales L, Oliveira TC, Sogayar MC, Labriola L, Dewey RA, Perone MJ (2017) Pluripotent nontumorigenic adipose tissue-derived muse cells have immunomodulatory capacity mediated by transforming growth factor-beta1. Stem Cells Transl Med 6(1):161–173

    Article  CAS  Google Scholar 

  22. Xu Y, Sun P, Wang JY, Li ZZ, Gao RL, Wang XZ, Phillips WD, Liang SX (2019) Differentiation of CD45/CD31+ lung side population cells into endothelial and smooth muscle cells in vitro. Int J Mol Med 43(3):1128–1138

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhou S, Schuetz JD, Bunting KD, Colapietro AM, Sampath J, Morris JJ, Lagutina I, Grosveld GC, Osawa M, Nakauchi H, Sorrentino BP (2001) The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat Med 7(9):1028–1034

    Article  CAS  Google Scholar 

  24. Liang SX, Summer R, Sun X, Fine A (2005) Gene expression profiling and localization of Hoechst-effluxing CD45- and CD45+ cells in the embryonic mouse lung. Physiol Genomics 23(2):172–181

    Article  CAS  Google Scholar 

  25. Amirkhani MA, Shoae-Hassani A, Soleimani M, Hejazi S, Ghalichi L, Nilforoushzadeh MA (2016) Rejuvenation of facial skin and improvement in the dermal architecture by transplantation of autologous stromal vascular fraction: a clinical study. Bioimpacts. 6(3):149–154

    Article  Google Scholar 

  26. Wang XZ, Gao RL, Sun P, Liu S, Xu Y, Liang DZ, Yin LM, Phillips WD, Liang SX (2017) Proliferation, differentiation and migration of SCA1(-)/CD31(-) cardiac side population cells in vitro and in vivo. Int J Cardiol 227:378–386

    Article  Google Scholar 

  27. Galvez P, Clares B, Bermejo M, Hmadcha A, Soria B (2014) Standard requirement of a microbiological quality control program for the manufacture of human mesenchymal stem cells for clinical use. Stem Cells Dev 23(10):1074–1083

    Article  CAS  Google Scholar 

  28. Zhao Y, Jiang H, Liu XW, Chen JT, Xiang LB, Zhou DP (2015) Neurogenic differentiation from adipose-derived stem cells and application for autologous transplantation in spinal cord injury. Cell Tissue Bank 16(3):335–342

    Article  CAS  Google Scholar 

  29. Gowing G, Svendsen CN (2011) Stem cell transplantation for motor neuron disease: current approaches and future perspectives. Neurotherapeutics. 8(4):591–606

    Article  Google Scholar 

  30. McDonald JW, Becker D, Holekamp TF, Howard M, Liu S, Lu A, Lu J, Platik MM, Qu Y, Stewart T, Vadivelu S (2004) Repair of the injured spinal cord and the potential of embryonic stem cell transplantation. J Neurotrauma 21(4):383–393

    Article  Google Scholar 

  31. Mezey E, Key S, Vogelsang G, Szalayova I, Lange GD, Crain B (2003) Transplanted bone marrow generates new neurons in human brains. Proc Natl Acad Sci 100(3):1364–1369

    Article  CAS  Google Scholar 

  32. Corti S, Nizzardo M, Nardini M, Donadoni C, Salani S, Ronchi D, Saladino F, Bordoni A, Fortunato F, Del Bo R, Papadimitriou D, Locatelli F et al (2008) Neural stem cells transplantation can ameliorate the phenotype of a mouse model of spinal muscular atrophy. J Clin Invest 118:3316–3330

    Article  CAS  Google Scholar 

  33. Rudnik-Schoneborn S, Goebel HH, Schlote W, Molaian S, Omran H, Ketelsen U, Korinthenberg R, Wenzel D, Lauffer H, Kreiss-Nachtsheim M, Wirth B, Zerres K (2003) Classical infantile spinal muscular atrophy with SMN deficiency causes sensory neuronopathy. Neurology. 60(6):983–987

    Article  CAS  Google Scholar 

  34. Mercuri E, Bertini E (2012) Stem cells in severe infantile spinal muscular atrophy. Neuromuscul Disord 22(12):1105

    Article  Google Scholar 

  35. Finkel RS (2012) Stem cells in severe infantile spinal muscular atrophy (SMA1). Neuromuscul Disord 22(12):1105–1106

    Article  Google Scholar 

  36. Cohn RD (2012) Stem cells in severe infantile spinal muscular atrophy (SMA1). Neuromuscul Disord 22(12):1106–1107

    Article  Google Scholar 

  37. Barbash IM, Chouraqui P, Baron J, Feinberg MS, Etzion S, Tessone A, Miller L, Guetta E, Zipori D, Kedes LH, Kloner RA, Leor J (2003) Systemic delivery of bone marrow-derived mesenchymal stem cells to the infarcted myocardium: feasibility, cell migration, and body distribution. Circulation. 108(7):863–868

    Article  Google Scholar 

  38. Togel F, Yang Y, Zhang P, Hu Z, Westenfelder C (2008) Bioluminescence imaging to monitor the in vivo distribution of administered mesenchymal stem cells in acute kidney injury. Am J Physiol Ren Physiol 295(1):F315–F321

    Article  CAS  Google Scholar 

  39. Markowitz JA, Singh P, Darras BT (2012) Spinal muscular atrophy: a clinical and research update. Pediatr Neurol 46(1):1–12

    Article  Google Scholar 

  40. Hausmanowa-Petrusewicz I, Karwanska A (1991) Electromyographic findings in different forms of infantile and juvenile proximal spinal muscular atrophy. Muscle Nerve 14:585–597

    Article  Google Scholar 

  41. Drago D, Cossetti C, Iraci N, Gaude E, Musco G, Bachi A, Pluchino S (2013) The stem cell secretome and its role in brain repair. Biochimie. 95(12):2271–2285

    Article  CAS  Google Scholar 

  42. Cossetti C, Smith JA, Iraci N, Leonardi T, Alfaro-Cervello C, Pluchino S (2012) Extracellular membrane vesicles and immune regulation in the brain. Front Physiol 3:117

    Article  CAS  Google Scholar 

  43. Xin H, Li Y, Cui Y, Yang JJ, Zhang ZG, Chopp M (2013) Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J Cereb Blood Flow Metab 33(11):1711–1715

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank the anonymous referees for their time.

Funding

This project was completely supported by the Tehran University of Medical Sciences by the Grant No: 94-01-87-28524 for the clinical trial program in SMA1 (Werdnig Hoffman) patients.

Author information

Authors and Affiliations

Authors

Contributions

RM carried out cell harvest, culture, and characterization, AH participated in the design and conduction of the study, AS participated in quality control test and final approval of the stem cell products, MG carried out EMG and NCV, AM selected the patients according to the criteria, MM visited all patients, JA wrote the manuscript draft, MN carried out immunophenotyping, RS participated in visiting patients, HM participated in stem cell administration to patients, and MRA participated in its design, patient selection, and coordination and helped to draft the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Amir Ali Hamidieh or Mahmoud Reza Ashrafi.

Ethics declarations

Competing interest

The authors declare no competing interests.

Ethical approval

This study approved by the ethical committee of the Tehran University of Medical Sciences and the Iranian Ministry of Health and Medical Education (MOHME) Ethics board, Tehran, Iran.

Informed consent

The informed consent forms were filled by the legal guardian (parents) of the all participants in our intervention. All donors entering the program agreed to and signed an institutional review board-approved statement of informed consent.

Consent for publication

All the consent forms are signed by the parents of the patients or their legal representatives. A copy of the signed consent forms is available in the supplementary file section.

Statement of human and animal rights

All of the experimental procedures involving animals were conducted in accordance with the institutional animal care guidelines of Tehran University of Medical Sciences and approved by the administration committee of Experimental Animals, Tehran, Iran.

Additional information

Publisher’s note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mohseni, R., Hamidieh, A.A., Shoae-Hassani, A. et al. An open-label phase 1 clinical trial of the allogeneic side population adipose-derived mesenchymal stem cells in SMA type 1 patients. Neurol Sci 43, 399–410 (2022). https://doi.org/10.1007/s10072-021-05291-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10072-021-05291-2

Keywords

Navigation