Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: an open-label phase 2a proof-of-concept study

Peter Connick, Madhan Kolappan, Charles Crawley, Daniel J Webber, Rickie Patani, Andrew W Michell, Ming-Qing Du, Shi-Lu Luan, Daniel R Altmann, Alan J Thompson, Alastair Compston, Michael A Scott, David H Miller, Siddharthan Chandran, Peter Connick, Madhan Kolappan, Charles Crawley, Daniel J Webber, Rickie Patani, Andrew W Michell, Ming-Qing Du, Shi-Lu Luan, Daniel R Altmann, Alan J Thompson, Alastair Compston, Michael A Scott, David H Miller, Siddharthan Chandran

Abstract

Background: More than half of patients with multiple sclerosis have progressive disease characterised by accumulating disability. The absence of treatments for progressive multiple sclerosis represents a major unmet clinical need. On the basis of evidence that mesenchymal stem cells have a beneficial effect in acute and chronic animal models of multiple sclerosis, we aimed to assess the safety and efficacy of these cells as a potential neuroprotective treatment for secondary progressive multiple sclerosis.

Methods: Patients with secondary progressive multiple sclerosis involving the visual pathways (expanded disability status score 5·5-6·5) were recruited from the East Anglia and north London regions of the UK. Participants received intravenous infusion of autologous bone-marrow-derived mesenchymal stem cells in this open-label study. Our primary objective was to assess feasibility and safety; we compared adverse events from up to 20 months before treatment until up to 10 months after the infusion. As a secondary objective, we chose efficacy outcomes to assess the anterior visual pathway as a model of wider disease. Masked endpoint analyses was used for electrophysiological and selected imaging outcomes. We used piecewise linear mixed models to assess the change in gradients over time at the point of intervention. This trial is registered with ClinicalTrials.gov, number NCT00395200.

Findings: We isolated, expanded, characterised, and administered mesenchymal stem cells in ten patients. The mean dose was 1·6×10(6) cells per kg bodyweight (range 1·1-2·0). One patient developed a transient rash shortly after treatment; two patients had self-limiting bacterial infections 3-4 weeks after treatment. We did not identify any serious adverse events. We noted improvement after treatment in visual acuity (difference in monthly rates of change -0·02 logMAR units, 95% CI -0·03 to -0·01; p=0·003) and visual evoked response latency (-1·33 ms, -2·44 to -0·21; p=0·020), with an increase in optic nerve area (difference in monthly rates of change 0·13 mm(2), 0·04 to 0·22; p=0·006). We did not identify any significant effects on colour vision, visual fields, macular volume, retinal nerve fibre layer thickness, or optic nerve magnetisation transfer ratio.

Interpretation: Autologous mesenchymal stem cells were safely given to patients with secondary progressive multiple sclerosis in our study. The evidence of structural, functional, and physiological improvement after treatment in some visual endpoints is suggestive of neuroprotection.

Funding: Medical Research Council, Multiple Sclerosis Society of Great Britain and Northern Ireland, Evelyn Trust, NHS National Institute for Health Research, Cambridge and UCLH Biomedical Research Centres, Wellcome Trust, Raymond and Beverly Sackler Foundation, and Sir David and Isobel Walker Trust.

Copyright © 2012 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Study profile
Figure 2
Figure 2
Change in visual function, visual evoked response amplitude, and optic nerve area Paired monthly estimated rates of change in log of minimum angle of resolution (logMAR) visual acuity, whole-field visual evoked response latency, and optic nerve area are shown for individual patients before and after treatment connected by solid lines. Pretreatment and post-treatment mean rates of change are also shown connected with a dashed line. Significance tests are shown for a difference between mean rates of change before and after treatment.

References

    1. Pugliatti M, Rosati G, Carton H. The epidemiology of multiple sclerosis in Europe. Eur J Neurol. 2006;13:700–722.
    1. Compston A, Coles A. Multiple sclerosis. Lancet. 2008;372:1502–1517.
    1. Coles AJ, Compston DAS, Selmaj KW. Alemtuzumab vs interferon beta-1a in early multiple sclerosis. N Engl J Med. 2008;359:1786–1801.
    1. Kappos L, Freedman MS, Polman CH. Effect of early versus delayed interferon beta-1b treatment on disability after a first clinical event suggestive of multiple sclerosis: a 3-year follow-up analysis of the BENEFIT study. Lancet. 2007;370:389–397.
    1. Coles A. The curious incident of disability in multiple sclerosis trials. Lancet Neurol. 2006;5:899–900.
    1. Pittenger MF, Mackay AM, Beck SC. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–147.
    1. Arthur A, Zannettino A, Gronthos S. The therapeutic applications of multipotential mesenchymal/stromal stem cells in skeletal tissue repair. J Cell Physiol. 2009;218:237–245.
    1. Uccelli A, Laroni A, Freedman MS. Mesenchymal stem cells for the treatment of multiple sclerosis and other neurological diseases. Lancet Neurol. 2011;10:649–656.
    1. Payne N, Siatskas C, Barnard A, Bernard CCA. The prospect of stem cells as multi-faceted purveyors of immune modulation, repair and regeneration in multiple sclerosis. Curr Stem Cell Res Ther. 2011;6:50–62.
    1. Zappia E, Casazza S, Pedemonte E. Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy. Blood. 2005;106:1755–1761.
    1. Zhang J, Li Y, Lu M. Bone marrow stromal cells reduce axonal loss in experimental autoimmune encephalomyelitis mice. J Neurosci Res. 2006;84:587–595.
    1. Bai L, Lennon DP, Eaton V. Human bone marrow-derived mesenchymal stem cells induce Th2-polarized immune response and promote endogenous repair in animal models of multiple sclerosis. Glia. 2009;57:1192–1203.
    1. Gerdoni E, Gallo B, Casazza S. Mesenchymal stem cells effectively modulate pathogenic immune response in experimental autoimmune encephalomyelitis. Ann Neurol. 2007;61:219–227.
    1. Kassis I, Grigoriadis N, Gowda-Kurkalli B. Neuroprotection and immunomodulation with mesenchymal stem cells in chronic experimental autoimmune encephalomyelitis. Arch Neurol. 2008;65:753–761.
    1. Le Blanc K, Frassoni F, Ball L. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet. 2008;371:1579–1586.
    1. Sun L, Akiyama K, Zhang H. Mesenchymal stem cell transplantation reverses multiorgan dysfunction in systemic lupus erythematosus mice and humans. Stem Cells. 2009;27:1421–1432.
    1. Carrion F, Nova E, Ruiz C. Autologous mesenchymal stem cell treatment increased T regulatory cells with no effect on disease activity in two systemic lupus erythematosus patients. Lupus. 2010;19:317–322.
    1. Mohyeddin Bonab M, Yazdanbakhsh S, Lotfi J. Does mesenchymal stem cell therapy help multiple sclerosis patients? Report of a pilot study. Iran J Immunol. 2007;4:50–57.
    1. Yamout B, Hourani R, Salti H. Bone marrow mesenchymal stem cell transplantation in patients with multiple sclerosis: a pilot study. J Neuroimmunol. 2010;227:185–189.
    1. Karussis D, Karageorgiou C, Vaknin-Dembinsky A. Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyotrophic lateral sclerosis. Arch Neurol. 2010;67:1187–1194.
    1. Mehta LR, Schwid SR, Arnold DL. Proof of concept studies for tissue-protective agents in multiple sclerosis. Mult Scler. 2009;15:542–546.
    1. Kolappan M, Henderson APD, Jenkins TM. Assessing structure and function of the afferent visual pathway in multiple sclerosis and associated optic neuritis. J Neurol. 2009;256:305–319.
    1. Connick P, Kolappan M, Patani R. The mesenchymal stem cells in multiple sclerosis (MSCIMS) trial protocol and baseline cohort characteristics: an open-label pre-test: post-test study with blinded outcome assessments. Trials. 2011;12:62.
    1. Poser CM, Paty DW, Scheinberg L. New diagnostic criteria for multiple sclerosis: guidelines for research protocols. Ann Neurol. 1983;13:227–231.
    1. Dominici M, Le Blanc K, Mueller I. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–317.
    1. Johnson NA, Hamoudi RA, Ichimura K. Application of array CGH on archival formalin-fixed paraffin-embedded tissues including small numbers of microdissected cells. Lab Invest. 2006;86:968–978.
    1. Oberg AL, Mahoney DW. Linear mixed effects models. Methods Mol Biol. 2007;404:213–234.
    1. Lublin FD, Reingold SC. Defining the clinical course of multiple sclerosis: results of an international survey. National Multiple Sclerosis Society (USA) Advisory Committee on Clinical Trials of New Agents in Multiple Sclerosis. Neurology. 1996;46:907–911.
    1. Barkhof F, Bruck W, De Groot CJA. Remyelinated lesions in multiple sclerosis: magnetic resonance image appearance. Arch Neurol. 2003;60:1073–1081.
    1. Schmierer K, Scaravilli F, Altmann DR, Barker GJ, Miller DH. Magnetization transfer ratio and myelin in postmortem multiple sclerosis brain. Ann Neurol. 2004;56:407–415.
    1. Franklin RJM, Ffrench-Constant C. Remyelination in the CNS: from biology to therapy. Nat Rev Neurosci. 2008;9:839–855.
    1. Rivera FJ, Couillard-Despres S, Pedre X. Mesenchymal stem cells instruct oligodendrogenic fate decision on adult neural stem cells. Stem Cells. 2006;24:2209–2219.
    1. Bai L, Caplan A, Lennon D, Miller RH. Human mesenchymal stem cells signals regulate neural stem cell fate. Neurochem Res. 2007;32:353–362.

Source: PubMed

3
購読する