Skip to main content
Advanced Biomedical Research logoLink to Advanced Biomedical Research
. 2016 Mar 16;5:46. doi: 10.4103/2277-9175.178791

The role of stem cell therapy in multiple sclerosis: An overview of the current status of the clinical studies

Rokhsareh Meamar 1,2, Shahrzad Nematollahi 3, Leila Dehghani 1,2, Omid Mirmosayyeb 2, Vahid Shayegannejad 2,4, Keivan Basiri 2,4, Amir Pouya Tanhaei 2,
PMCID: PMC4817403  PMID: 27110543

Abstract

The complexity of multiple sclerosis (MS) and the incompetence of a large number of promised treatments for MS urge us to plan new and more effective therapeutic approaches that aim to suppress ongoing autoimmune responses and induction of local endogenous regeneration. Emerging data propose that hematopoietic, mesenchymal, and neural stem cells have the potential to restore self-tolerance, provide in situ immunomodulation and neuroprotection, as well as promote regeneration. Thus, in this article, we will first provide an overview of the cell sources for proposed mechanisms that contribute to the beneficial effects of stem cell transplantation, the ideal route and/or timing of stem cell-based therapies for each main stem cell group, and finally, an overview of the current status of stem cell research in clinical trial stages in MS by comparable and healthy therapeutic effects of different stem cell therapies for MS patients.

Keywords: Cell therapy and transplantation, clinical trial, hematopoietic stem cells, mesenchymal stem cells, multiple sclerosis, neural stem/precursor cells, stem cells

INTRODUCTION

Multiple Sclerosis (MS) is an autoimmune and neurodegenerative disease of the central nervous system (CNS). In autoimmune etiology, there is a prevailing theory in which oligodendrocytes are believed to be permanently damaged by CD4+ T-cells, CD8+ T-cells, and macrophages.[1] In MS patients, autoreactive CD4 T-cell penetration of the CNS leads to myelin injury and inflammatory responses and scarring of white matter, which can lead to severe disability and neurological defects.[1] MS progression following demyelination typically pursues one of four courses: Relapsing-remitting MS (RRMS), secondary progressive MS (SPMS), primary progressive MS (PPMS) and progressive-relapsing MS (PRMS). To date, the effectiveness of disease-modifying drugs has been approved only in a limited number of MS patients, especially in the relapsing forms of PRMS,[2,3] and the apparent repair-promoting activity of these drugs has not yet been reported, due to partial inhibitory effect on disease progression.

Stem cells (SCs) have uncovered a new perspective as therapeutic tools in neurological disorders such as MS. These cells are pluripotent cells with a capacity to give rise to different cell types,[4,5,6,7] that are infinite sources of neurons and glia for therapies aimed at cell replacement or neuroprotection in disorders affecting the brain and spinal cord like MS.[8,9,10,11] The two main stem cell types are embryonic stem cells (ESCs) and adult stem cells. ESCs are formed four to five days after fertilization from the inner cell mass of the blastocytes with an ability for unlimited growth in culture that could be related to a high risk of teratoma formation. Adult stem cells are specialized cells including hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs) and neural stem cells (NSCs).[8] In the present article, we aim to review the literatures regarding major kinds of adult stem cells, which we have transplanted in clinical trial studies into MS patients.

Mesenchymal stem cell and sources

Mesenchymal stem cells are self-replicating cells that are capable of differentiating in multidirectional pathways, such as, osteoblasts, chondrocytes, myocytes, marrow stromal cells, tendon-ligament fibroblasts, adipocytes, and neural cells.[12,13,14] While a routine source of human MSCs is the bone marrow, they have been also derived from multiple adult tissues comprising of adipose tissue, umbilical cord blood, placenta, thymus, and dental pulp.[15,16,17,18]

The application of bone marrow-derived MSCs consists of several practical advantages: First, MSCs can be gained readily and safely from adult bone marrow, even from patients with progressive disease; second, MSCs, which are normally present in small concentrations in the bone marrow section, can be enriched and greatly prolonged by in vitro culturing; third, autologous MSCs can be administered securely without the need for immunosuppressive treatment, to avoid rejection; and finally, adult MSCs have been shown to be less prone to genetic defects and malignant transformation during multiple routes in vitro, implying a low risk for induction of treatment–related malignant neoplasm.[19]

Mechanisms of beneficial effects of mesenchymal stem cells

Transplanted MSCs not only directly differentiate into neurons and endothelial cells after induction, but also, it is surely believed, that their secretome can mediate their valuable actions. Several autocrine/paracrine factors could be secreted from MSCs that have many benefits.[20,21,22]

A broad range of these neuroregulatory secretions could be released both in vivo and in vitro that yield a summation in neurogenesis, inhibition of apoptosis, chemoattraction, glial scar formation, immunomodulation, angiogenesis, neuronal and glial cell survival, expansion of endogenous axonal and myelin repair processes, neurotrophic and neuroprotective actions, and integration and improvement of local progenitor cells.[23]

Several inhibitory responses of immune system are accountable for these protective effects, as well as anti-inflammatory responses by decrements in peripheral T-cells, B cells, regulatory T cells (Tregs) and natural killer (NK) cells. On the other hand, they inhibit the maturation and function of antigen-presenting cells and reduce pro-inflammatory cytokines.[24,25]

Bone marrow MSCs can transdifferentiate into neuron-like cells in vitro under specific-induced culture situations and, therefore, might also deliver cell substitutes to the injured CNS. However, the mechanism of the bone marrow stem cells’ transformation to neuro-ectodermal lineage is still uncertain.[26]

Thus, MSC–based treatments have the potential to be an advanced and reasonable treatment to repair inflamed and impaired tissues.[20]

Route of administration of mesenchymal stem cells

Intrathecal injection of MSCs does not affect cytokine dissimilarity in peripheral blood.[13] The safety and possibility of autologous intravenous (IV) MSC therapy in MS has been established.[27] Intrathecal injection is a route of drug administration, which is performed by injection into the spinal canal, more specifically into the subarachnoid space, to reach the cerebrospinal fluid (CSF). The rationale for intrathecal management is transportation of cells directly into the CNS and overcoming the restricted amount of cell engrafting upon IV administration and enhancing the total yield at position of damage. However, local delivery may increase MSC ability to promote repair by secreting neurotrophic factors, such as brain-derived neurotrophic factor (BDNF) and antioxidant molecules.[15]

In the animal model of MS, experimental autoimmune encephalomyelitis, IV injection of MSCs into C57BL/6J mice was shown to down regulate the clinical harshness of the disease with a parallel suppression of CNS inflammation through induction of T-cell and decrease of demyelination.[19]

It is claimed that intrathecal injection is less invasive compared to direct injection into lesions, demyelinating lesions especially. However, the pathological heterogeneity and multifocality of MS lesions could limit the efficacy of such a method.[28] Also, intrathecal administration in humans may lead to meningeal irritation. In one case, temporary acute encephalopathy with seizures, likely related to CNS inflammation, was reported in a subject who had received a high dose of MSCs intrathecally. Based on this evidence, IV administration of MSCs should be considered the preferable method in comparison with intrathecal delivery.[15,29]

Hematopoietic stem cell and sources

Hematopoietic stem cells are found chiefly within bone marrow in niches created by surrounding stromal cells. HSCs have the potential to differentiate into the main hemato- and lymphopoietic precursors, which then differentiate into mature cells. They are generated in large numbers throughout human life and continually repopulate blood and immune systems.[30]

Hematopoietic stem cells have some advantages including self-renewal, differentiation to a variety of specialized cells, mobilization out of the bone marrow into circulating blood, and undergone apoptosis. There appears to be two kinds of HSCs; long-term stem cells that are capable of self-renewal and short-term stem cells that can immediately regenerate all the different types of blood cells, but under normal circumstances cannot renew themselves over the long term. Short-term stem cells are capable of proliferating, but have a limited capacity to differentiate into more than one cell type.[31]

The sources of HSCs are bone marrow, peripheral blood, umbilical cord blood, fetal hematopoietic system, ESCs and embryonic germ cells.[31]

Avoidance of anaesthesia, the lack of need for hospitalization or blood transfusion, and low risk of serious adverse events are major advantages of the peripheral SCs, which make them the favourable source of SCs worldwide.[31]

Mechanisms of beneficial effects of hematopoietic stem cells

Hematopoietic stem cell transplantation (HSCT) is unique among stem cell-related treatments as it does not primarily aim at neuroregeneration; it rather aims at replacement/resetting of the whole immune system. All the series of immune cells from progenitor HSCs are regenerated following the destruction of the ’old’ immune system by radical immunosuppression. In addition, HSCs can transdifferentiate into cells from the neuronal lineage and show their neuroprotective/neurotrophic effects.[32,33,34]

Route of administration of hematopoietic stem cells

Hematopoietic stem cells may be collected directly from the bone marrow through multiple aspirations performed under regional or general anesthesia. For transplantation purpose, HSCs could be mobilized from the bone marrow into the circulation intravenously using chemotherapy and/or hematopoietic growth factors and then collected by leukopheresis. The product can be processed to remove contaminating immune cells and then cryopreserved or it can be cryopreserved without further manipulation.[35]

Hematopoietic stem cells are transplanted in two approaches — autologous and allogeneic. In autologous HSCT (AHSCT), the immune system is wiped out by reinfusion of the patient's own HSCs. This approach has originated from the idea that by immune re-formation the status of tolerance to self-proteins can be re-established and the freshly developing, ’re-educated’ lymphocytes stop their role as the carriers of the immunological memory of previous autoimmunity.[36]

Neural stem/precursor cells and sources

Neural stem cells are defined as self-renewing multipotent progenitors existing in the developing and adult CNS. Generally, they are considered by their capacity to symmetrically self-renew and their ability to discriminate into neurons, oligodendrocytes, and astrocytes through asymmetrical fate-committed division.[37]

Neural stem or progenitor cells can be obtained in different approaches and one way is the direct isolation of these cells from embryonic or adult brain tissue.[38,39,40] The application of NSC therapy will be realized once their purification, mass generation, and safety are guaranteed.[41]

An overview of the applicable cells in MS has shown that NSCs could provide a source of remyelinating cells with capabilities to structurally repair the CNS.[42]

Although the adult brain has long been considered to have no regenerative potential, NSCs were identified in three specific neural stem cell niches: The subventricular zone (SVZ), the external germinal layer of the cerebellum, and the subgranular zone of the dentate gyrus (SGZ). Over two decades, many research groups have reported on the successful isolation and ex vivo culture of these cells from early embryonic or adult SVZ brain tissue[38,39,40] and they can be safely expanded in chemically defined culture media for a prolonged period.[43]

Also, recently, there has been identified a novel population of cells in the subventricular zone (SVZ) of the mammalian brain that expresses Tubulin beta-4B chain and has properties of primitive neuroectodermal cells. Tubulin beta-4B chains are significantly increased in the SVZs bordering demyelinated white matter in MS brains. Such rapid and extensive mature CNS cell generation by a rather small number of transplanted cells provides in vivo support for the therapeutic potential of this population of cells, which is reproducing Tubulin beta-4B chain in the SVZ.[44]

Mechanisms of the beneficial effects of neural stem cells

In view of the multiple mechanisms by which neural precursor cells could induce beneficial effects in MS, including their regenerative potential, their trophic, immunomodulatory, and neuroprotective properties, they seem to be an excellent candidate for cell therapy. Specifically, they may have a benefit on committed myelin–forming cells that might not possess other stem cell properties, and on non-neural cells that cannot perform remyelination.[41]

The remyelinating effect of these cells may be via one or more mechanisms, including as an immunomodulator by generating soluble factors, direct cell replacement by differentiating into neural and glial cells in the lesion; and finally, indirect action by stimulating neural and glial differentiation of endogenous cells.[3] However, this regenerative function is inadequate in chronic MS in which progenitor cells either fail to be recruited into lesion sites or they encounter some difficulties with respect to differentiation.[45]

One of the major outcomes of the neuroprotective effects of transplanted NPCs is the significant rise in survival and function of endogenous glial and neuronal progenitors escaping from primary insults. This phenomenon has broad implications and is usually accompanied by increased availability of a milieu of molecules, such as neurotrophins and growth factors, immune modulatory molecules, and developmental stem cell regulators.[46]

Neural stem and progenitor cells decrease the acute deleterious inflammatory process and induce a permissive environment for axonal regeneration after spinal cord injury.[41,47] On the other hand, there is enough correlative evidence for envisaging that the injury (inflammatory) microenvironment is likely to play a critical component to impact the establishment of atypical ectopic niches and in turn to maintain cell-to-cell communication between transplanted NPCs and endogenous cells.[48]

Route of administration of neural stem cells

Several animal model of multiple sclerosis studies[47] showed that IV administration of NSCs presently has limited or without any therapeutic potential for neuroinflammatory disease in mice, and probably also for human MS.[49]

Interestingly, intrathecal or intraventricular injections, which bypass the blood–brain barrier (BBB) by placing cells in the cerebrospinal fluid (CSF) compartment are sufficient to produce benefit in various animal models of neurological diseases such as MS.[50]

A major rationale for intracerebroventricular (ICV) route of cell delivery is that most white matter tracts that are involved in MS are in close proximity to the ventricular and spinal subarachnoid spaces. Following ICV injection, transplanted neural precursors may disseminate throughout the ventricular and subarachnoid space, enabling their inflammation-induced targeted migration into the white matter and may get the remyelinating cells closest to the multiple foci of disease in MS without a separating barrier.[41]

Clinical trial studies of stem cell therapy in multiple sclerosis

There has been a rapid surge in clinical trials involving stem cell therapies recently and those trials are establishing the clinical pathways for an emergent new medicine. There are many studies involving autologous therapies based on the recovery of mobilized bone marrow cells, including mesenchymal and hematopoietic stem cells, which are used widely in the treatment of multiple sclerosis.[51] The characteristics of the trials conducted to test the safety and validity of MSC treatment for MS are presented in Table 1. In summary, most of the trials which are in phase 2 (to examine safety and tolerability of the stem cell treatment) had patients with median Expanded Disability Status Score (EDSS)\with median follow up between three and twenty-six months. All these trials provide evidence of safety and effectiveness of MSCs without using any conditioning regimen. Based on Table 1, neither death nor major side events have occurred throughout the study courses. In a case-report by Hou et al., it is also shown that repeated injections of bone marrow–derived MSCs followed by frequent injections of umbilical cord MSCs (both intravenously) would improve one point on patient's EDSS score and diminish many magnetic resonance imaging (MRI) lesions.[58]

Table 1.

Summary of published clinical trials for mesenchymal stem cell transplantation in multiple sclerosis

graphic file with name ABR-5-46-g001.jpg

Notwithstanding the promising results of MSCs therapy, it induced some major adverse events in one case report. Alderazi, et al. described an MS patient, who received repeated intrathecal doses of allogenic CD 34+ MSCs derived from umbilical cord blood, as well as infusions of autologous adipose-derived stem cells obtained by liposuction. The authors reported severe meningoencephalomyelitis, which was probably due to stem cell transplantation.[59]

The characteristics of clinical trials in the role of Autologous Hematopoietic Stem Cell Transplantation (AHSCT) in the treatment of MS are presented in Table 2. Most trials have been conducted in small phase 1 (to determine toxicity and major side effects of the treatment) or 2, with SPMS participants who had a mean EDSS score baseline between 3 and 9.5 and a median follow-up between 5 months and 15 years. There are some adverse events reported including breakdown in task performance, bacterial infections, or sepsis; However, fever is the most frequent adverse event reported.[71] It is also shown that AHSCT could result in significant improvement of patient's quality of life.[63] The Progression–Free Survival (PFS) ranged between 82% in 100 days and 25% in 15 years after transplantation. In addition, BCNU (bis-chloroethylnitrosourea) -etoposide-cytarabine -melphalan (BEAM) regimen (BCNU at a dosage of 300 mg/m2; etoposide at a dosage of 200 mg/m2; cytarabine at a dosage of 200mg/m2; melphalan at a dosage of 140 mg/m2)[66] is the most used immunosuppressive conditioning, which is found to be more appropriate for patients with progressive MS undergoing transplantation.

Table 2.

Summary of the published clinical trials for Hematopoeitic stem cells transplantation in multiple sclerosis

graphic file with name ABR-5-46-g002.jpg

The broad spectrum of PFS seen in Table 2 could be a result of various immunosuppressive therapies and long duration of follow-up in some studies (up to 15 years). A recent meta-regression of AHSCT (2011) found that intermediate-intensity regimens were associated with a significantly higher PFS compared to high-intensity regimens like total body irradiation (TBI).[71] A possible explanation for this finding is that the neurotoxicity of the high-intensity TBI plus cyclophosphamide (CY) regimen may lead to increased axonal damage and degeneration in patients with progressive MS, thus contributing to disease progression.[72] However, it is shown that in severely disabled patients with malignant MS, pulsed CY may improve functional status and permit successful delivery of AHSCT.[73]

Authors of another systematic review (2008) concluded that intermediate-intensity immunosuppressive therapy (such as BEAM) is more favorable as a conditioning regimen since it is assumed to be related to a lower risk of treatment-related toxicity.[74] Moreover, in this work it is found that deaths due to MS-related complications are prominent while it is reported that pneumonia is the major cause of death after AHSCT.[71] Overall, it is shown that AHSCT can be regarded as a potential therapeutic procedure for MS patients, particularly those in the early stages of the disease.

We found no clinical trial regarding the use of NSC in the treatment of MS up to now.[51] It may refer to controversial results in preclinical studies. Unfortunately, most previous observations lack detailed cell graft survival and/or glial reactivity analysis at early and late time-points post-grafting.,[75] although it is important to note that early NSC graft mortality and subsequent glial cell responses themselves might be responsible for many of the observed beneficial effects following cell transplantation in CNS disorders.[75] Moreover, NSCs have been shown to have a low immunogenic potential that is nevertheless capable of activating peripheral lymphocytes.[76] Thus more extensive characterization and pre-clinical studies are necessary before neural stem cell-based therapies are used in a clinical setting.

CONCLUSION

On the basis of the published evidence, to date, SC transplantation can be regarded as a potential source of treatment for MS. According to our review in the first part of this article for the stem cell sources and their mechanisms, focused on pre-clinical studies, it can be concluded that the availability of HSCs and MSCs are more than NSCs, but still NSCs which have the unique feature of beneficial effects with remyelination make it attractive for further studies in clinical stages to see whether they show this benefit in practice, particularly in the progressive stages of MS.

The inclusion criteria for patients in the limited clinical trial studies usually consist of patients who are refractory to conventional medical therapy, which decreases the likelihood to remain progression-free in the long period. In addition, in the absence of randomized trials, the probability that the included studies might differ from each other with respect to unknown prognostic factors increases. In addition, one of the major outcomes of the reports is EDSS, which is proved to have poor repeatability between different raters; therefore, conservative judgments regarding observed discrepancy in terms of EDSS is warranted.[77] Because most of the patients had SPMS, and relatively few patients with other types of MS (PPMS, PRMS, and RRMS) have so far received stem cell transplants, it is not yet possible to determine whether patients with these subtypes of MS have better outcomes.

In summary, transplantation of stem cells from either cell source could be a safe and effective therapy for MS. However, since up to now there is no controlled studies (randomized or non-randomized) comparing stem cell therapy, finding a consistent answer regarding the safety and efficacy of this type of therapy for MS patients needs future comprehensive research with large group of patients.

Footnotes

Source of Support: Nil

Conflict of Interest: None declared.

REFERENCES

  • 1.Connick P, Kolappan M, Crawley C, Webber DJ, Patani R, Michell AW, et al. Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: An open-label phase 2a proof-of-concept study. Lancet Neurol. 2012;11:150–6. doi: 10.1016/S1474-4422(11)70305-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Compston A, Coles A. Multiple sclerosis. Lancet. 2008;372:1502–17. doi: 10.1016/S0140-6736(08)61620-7. [DOI] [PubMed] [Google Scholar]
  • 3.Giacomini PS, Darlington PJ, Bar-Or A. Emerging multiple sclerosis disease-modifying therapies. Curr Opin Neurol. 2009;22:226–32. doi: 10.1097/wco.0b013e32832b4ca1. [DOI] [PubMed] [Google Scholar]
  • 4.Meamar R, Karamali F, Sadeghi HM, Etebari M, Nasr-Esfahani MH, Baharvand H. Toxicity of ecstasy (MDMA) towards embryonic stem cell-derived cardiac and neural cells. Toxicol In Vitro. 2010;24:1133–8. doi: 10.1016/j.tiv.2010.03.005. [DOI] [PubMed] [Google Scholar]
  • 5.Meamar R, Dehghani L, Karamali F. Toxicity effects of methamphetamine on embryonic stem cell-derived neuron. J Res Med Sci. 2012;17:470–4. [PMC free article] [PubMed] [Google Scholar]
  • 6.Dehghani L, Farokhpour M, Karbalaie K, Nematollahi M, Tanhaie S, Hayati-Rodbari N, et al. The influence of dexamethasone administration on the protection against doxorubicin-induced cardiotoxicity in purified embryonic stem cell-derived cardiomyocytes. Tissue Cell. 2013;45:101–6. doi: 10.1016/j.tice.2012.09.009. [DOI] [PubMed] [Google Scholar]
  • 7.Dehghani L, Hashemi-Beni B, Poorazizi E, Khorvash F, Shaygannejad V, Sedghi M, et al. Evaluation of neural gene expression in serum treated embryonic stem cells in Alzheimer's patients. J Res Med Sci. 2013;18(Suppl 1):S20–3. [PMC free article] [PubMed] [Google Scholar]
  • 8.Meamar R, Nasr-Esfahani MH, Mousavi SA, Basiri K. Stem cell therapy in amyotrophic lateral sclerosis. J Clin Neurosci. 2013;20:1659–63. doi: 10.1016/j.jocn.2013.04.024. [DOI] [PubMed] [Google Scholar]
  • 9.Meamar R, Dehghani L, Ghasemi M, Khorvash F, Shaygannejad V. Stem cell therapy in stroke: A review literature. Int J Prev Med. 2013;4(Suppl 2):S139–46. [PMC free article] [PubMed] [Google Scholar]
  • 10.Gögel S, Gubernator M, Minger SL. Progress and prospects: Stem cells and neurological diseases. Gene Ther. 2011;18:1–6. doi: 10.1038/gt.2010.130. [DOI] [PubMed] [Google Scholar]
  • 11.Lindvall O, Kokaia Z. Stem cells for the treatment of neurological disorders. Nature. 2006;441:1094–6. doi: 10.1038/nature04960. [DOI] [PubMed] [Google Scholar]
  • 12.Cohen JA. Mesenchymal stem cell transplantation in multiple sclerosis. J Neurol Sci. 2013;333:43–9. doi: 10.1016/j.jns.2012.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Mohyeddin Bonab M, Mohajeri M, Sahraian MA, Yazdanifar M, Aghsaie A, Farazmand A, et al. Evaluation of cytokines in multiple sclerosis patients treated with mesenchymal stem cells. Arch Med Res. 2013;44:266–72. doi: 10.1016/j.arcmed.2013.03.007. [DOI] [PubMed] [Google Scholar]
  • 14.Yamout B, Hourani R, Salti H, Barada W, El-Hajj T, Al-Kutoubi A, et al. Bone marrow mesenchymal stem cell transplantation in patients with multiple sclerosis: A pilot study. J Neuroimmunol. 2010;227:185–9. doi: 10.1016/j.jneuroim.2010.07.013. [DOI] [PubMed] [Google Scholar]
  • 15.Uccelli A, Laroni A, Freedman MS. Mesenchymal stem cells as treatment for MS-progress to date. Mult Scler. 2013;19:515–9. doi: 10.1177/1352458512464686. [DOI] [PubMed] [Google Scholar]
  • 16.Harris VK, Yan QJ, Vyshkina T, Sahabi S, Liu X, Sadiq SA. Clinical and pathological effects of intrathecal injection of mesenchymal stem cell-derived neural progenitors in an experimental model of multiple sclerosis. J Neurol Sci. 2012;313:167–77. doi: 10.1016/j.jns.2011.08.036. [DOI] [PubMed] [Google Scholar]
  • 17.Fisher-Shoval Y, Barhum Y, Sadan O, Yust-Katz S, Ben-Zur T, Lev N, et al. Transplantation of placenta-derived mesenchymal stem cells in the EAE mouse model of MS. J Mol Neurosci. 2012;48:176–84. doi: 10.1007/s12031-012-9805-6. [DOI] [PubMed] [Google Scholar]
  • 18.Auletta JJ, Bartholomew AM, Maziarz RT, Deans RJ, Miller RH, Lazarus HM, et al. The potential of mesenchymal stromal cells as a novel cellular therapy for multiple sclerosis. Immunotherapy. 2012;4:529–47. doi: 10.2217/imt.12.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Karussis D, Karageorgiou C, Vaknin-Dembinsky A, Gowda-Kurkalli B, Gomori JM, Kassis I, et al. Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyotrophic lateral sclerosis. Arch Neurol. 2010;67:1187–94. doi: 10.1001/archneurol.2010.248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.de Almeida DC, Donizetti-Oliveira C, Barbosa-Costa P, Origassa CS, Câmara NO. In search of mechanisms associated with mesenchymal stem cell-based therapies for acute kidney injury. The Clin Biochem Rev. 2013;34:131–44. [PMC free article] [PubMed] [Google Scholar]
  • 21.Lee T. Stem cell therapy independent of stemness. World J Stem Cells. 2012;4:120–4. doi: 10.4252/wjsc.v4.i12.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hsieh JY, Wang HW, Chang SJ, Liao KH, Lee IH, Lin WS, et al. Mesenchymal stem cells from human umbilical cord express preferentially secreted factors related to neuroprotection, neurogenesis, and angiogenesis. PloS One. 2013;8:e72604. doi: 10.1371/journal.pone.0072604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Teixeira FG, Carvalho MM, Sousa N, Salgado AJ. Mesenchymal stem cells secretome: A new paradigm for central nervous system regeneration? Cell Mol Life Sci. 2013;70:3871–82. doi: 10.1007/s00018-013-1290-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Payne NL, Sun G, McDonald C, Moussa L, Emerson-Webber A, Loisel-Meyer S, et al. Human adipose-derived mesenchymal stem cells engineered to secrete IL-10 inhibit APC function and limit CNS autoimmunity. Brain Behav Immun. 2013;30:103–14. doi: 10.1016/j.bbi.2013.01.079. [DOI] [PubMed] [Google Scholar]
  • 25.Cobo M, Anderson P, Benabdellah K, Toscano MG, Muñoz P, García-Pérez A, et al. Mesenchymal stem cells expressing vasoactive intestinal peptide ameliorate symptoms in a model of chronic multiple sclerosis. Cell transplant. 2013;22:839–54. doi: 10.3727/096368912X657404. [DOI] [PubMed] [Google Scholar]
  • 26.Wu R, Tang Y, Zang W, Wang Y, Li M, Du Y, et al. MicroRNA-128 regulates the differentiation of rat bone mesenchymal stem cells into neuron-like cells by Wnt signaling. Mol Cell Biochem. 2014;387:151–8. doi: 10.1007/s11010-013-1880-7. [DOI] [PubMed] [Google Scholar]
  • 27.Connick P, Kolappan M, Patani R, Scott MA, Crawley C, He XL, et al. 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. doi: 10.1186/1745-6215-12-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Martino G, Franklin RJ, Baron Van Evercooren A, Kerr DA Stem Cells in Multiple Sclerosis (STEMS) Consensus Group. Stem cell transplantation in multiple sclerosis: Current status and future prospects. Nat Rev Neurol. 2010;6:247–55. doi: 10.1038/nrneurol.2010.35. [DOI] [PubMed] [Google Scholar]
  • 29.Rice CM, Cottrell D, Wilkins A, Scolding NJ. Primary progressive multiple sclerosis: Progress and challenges. J Neurol Neurosurg Psychiatry. 2013;84:1100–6. doi: 10.1136/jnnp-2012-304140. [DOI] [PubMed] [Google Scholar]
  • 30.Holloman JP, Ho CC, Hukki A, Huntley JL, Gallicano GI. The development of hematopoietic and mesenchymal stem cell transplantation as an effective treatment for multiple sclerosis. Am J Stem Cells. 2013;2:95–107. [PMC free article] [PubMed] [Google Scholar]
  • 31.Bethesda MD. USA: National Institutes of Health, U.S. Department of Health and Human Services; [Last accessed on 2001/6/1]. Stem Cell Information. Available from: http://www.stemcells.nih.gov/info/scireport/pages/chapter 5.aspx . [Google Scholar]
  • 32.Mezey E, Chandross KJ, Harta G, Maki RA, McKercher SR. Turning blood into brain: Cells bearing neuronal antigens generated in vivo from bone marrow. Science. 2000;290:1779–82. doi: 10.1126/science.290.5497.1779. [DOI] [PubMed] [Google Scholar]
  • 33.Cogle CR, Yachnis AT, Laywell ED, Zander DS, Wingard JR, Steindler DA, et al. Bone marrow transdifferentiation in brain after transplantation: A retrospective study. Lancet. 2004;363:1432–7. doi: 10.1016/S0140-6736(04)16102-3. [DOI] [PubMed] [Google Scholar]
  • 34.Cabanes C, Bonilla S, Tabares L, Martínez S. Neuroprotective effect of adult hematopoietic stem cells in a mouse model of motoneuron degeneration. Neurobiol Dis. 2007;26:408–18. doi: 10.1016/j.nbd.2007.01.008. [DOI] [PubMed] [Google Scholar]
  • 35.Atkins HL, Freedman MS. Hematopoietic stem cell therapy for multiple sclerosis: Top 10 lessons learned. Neurotherapeutics. 2013;10:68–76. doi: 10.1007/s13311-012-0162-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Karussis D, Petrou P, Vourka-Karussis U, Kassis I. Hematopoietic stem cell transplantation in multiple sclerosis. Expert Rev Neurother. 2013;13:567–78. doi: 10.1586/ern.13.36. [DOI] [PubMed] [Google Scholar]
  • 37.Reekmans K, Praet J, Daans J, Reumers V, Pauwels P, Van der Linden A, et al. Current challenges for the advancement of neural stem cell biology and transplantation research. Stem Cell Rev. 2012;8:262–78. doi: 10.1007/s12015-011-9266-2. [DOI] [PubMed] [Google Scholar]
  • 38.Richards LJ, Kilpatrick TJ, Bartlett PF. De novo generation of neuronal cells from the adult mouse brain. Proc Natl Acad Sci U S A. 1992;89:8591–5. doi: 10.1073/pnas.89.18.8591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Reynolds BA, Tetzlaff W, Weiss S. A multipotent EGF-responsive striatal embryonic progenitor cell produces neurons and astrocytes. J Neurosci. 1992;12:4565–74. doi: 10.1523/JNEUROSCI.12-11-04565.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Reynolds BA, Weiss S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science. 1992;255:1707–10. doi: 10.1126/science.1553558. [DOI] [PubMed] [Google Scholar]
  • 41.Ben-Hur T. Cell therapy for multiple sclerosis. Neurotherapeutics. 2011;8:625–42. doi: 10.1007/s13311-011-0073-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Blakemore WF. Regeneration and repair in multiple sclerosis: The view of experimental pathology. J Neurol Sci. 2008;265:1–4. doi: 10.1016/j.jns.2007.03.006. [DOI] [PubMed] [Google Scholar]
  • 43.Einstein O, Ben-Hur T. The changing face of neural stem cell therapy in neurologic diseases. Arch Neurol. 2008;65:452–6. doi: 10.1001/archneur.65.4.452. [DOI] [PubMed] [Google Scholar]
  • 44.Wu C, Chang A, Smith MC, Won R, Yin X, Staugaitis SM, et al. Beta4 tubulin identifies a primitive cell source for oligodendrocytes in the mammalian brain. J Neurosci. 2009;29:7649–57. doi: 10.1523/JNEUROSCI.1027-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Jadasz JJ, Aigner L, Rivera FJ, Küry P. The remyelination Philosopher's Stone: Stem and progenitor cell therapies for multiple sclerosis. Cell Tissue Res. 2012;349:331–47. doi: 10.1007/s00441-012-1331-x. [DOI] [PubMed] [Google Scholar]
  • 46.Martino G, Pluchino S. The therapeutic potential of neural stem cells. Nat Rev Neurosci. 2006;7:395–406. doi: 10.1038/nrn1908. [DOI] [PubMed] [Google Scholar]
  • 47.Taupin P. Adult neural stem cells for the treatment of neuroinflammation. Fondazione Centro San Raffaele del Monte Tabor: WO2007015173. Expert Opin Ther Pat. 2009;19:373–6. doi: 10.1517/13543770802681649. [DOI] [PubMed] [Google Scholar]
  • 48.Martino G, Pluchino S, Bonfanti L, Schwartz M. Brain regeneration in physiology and pathology: The immune signature driving therapeutic plasticity of neural stem cells. Physiol Rev. 2011;91:1281–304. doi: 10.1152/physrev.00032.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Reekmans KP, Praet J, De Vocht N, Tambuyzer BR, Bergwerf I, Daans J, et al. Clinical potential of intravenous neural stem cell delivery for treatment of neuroinflammatory disease in mice? Cell Transplant. 2011;20:851–69. doi: 10.3727/096368910X543411. [DOI] [PubMed] [Google Scholar]
  • 50.Hess DC, Borlongan CV. Stem cells and neurological diseases. Cell prolif. 2008;41(Suppl 1):94–114. doi: 10.1111/j.1365-2184.2008.00486.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Trounson A, Thakar RG, Lomax G, Gibbons D. Clinical trials for stem cell therapies. BMC Med. 2011;9:52. doi: 10.1186/1741-7015-9-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Mohyeddin-Bonab M, Mohajeri M, Sahraian MA, Yazdanifar M, Aghsaie A, Farazmand A, et al. Evaluation of cytokines in multiple sclerosis patients treated with mesenchymal stem cells. Arch Med Res. 2013;44:266–72. doi: 10.1016/j.arcmed.2013.03.007. [DOI] [PubMed] [Google Scholar]
  • 53.Connick P, Kolappan M, Crawley C, Webber DJ, Patani R, Michell AW, et al. Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: An open-label phase 2a proof-of-concept study. Lancet Neurol. 2012;11:150–6. doi: 10.1016/S1474-4422(11)70305-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Odinak MM, Bisaga GN, Novitskii AV, Tyrenko VV, Fominykh MS, Bilibina AA, et al. Transplantation of mesenchymal stem cells in multiple sclerosis. Neurosci Behav Physiol. 2012;42:516–20. [PubMed] [Google Scholar]
  • 55.Mohajeri M, Farazmand A, Mohyeddin-Bonab M, Nikbin B, Minagar A. FOXP3 gene expression in multiple sclerosis patients pre- and post mesenchymal stem cell therapy. Iran J Allergy Asthma Immunol. 2011;10:155–61. [PubMed] [Google Scholar]
  • 56.Karussis D, Karageorgiou C, Vaknin-Dembinsky A, Gowda-Kurkalli B, Gomori JM, Kassis I, et al. Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyotrophic lateral sclerosis. Arch Neurol. 2010;67:1187–94. doi: 10.1001/archneurol.2010.248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Mohyeddin Bonab M, Yazdanbakhsh S, Lotfi J, Alimoghaddom K, Talebian F, Hooshmand F, et al. Does mesenchymal stem cell therapy help multiple sclerosis patients? Report of a pilot study. Iran J Immunol. 2007;4:50–7. [PubMed] [Google Scholar]
  • 58.Hou ZL, Liu Y, Mao XH, Wei CY, Meng MY, Liu YH, et al. Transplantation of umbilical cord and bone marrow-derived mesenchymal stem cells in a patient with relapsing-remitting multiple sclerosis. Cell Adh Migr. 2013;7:404–7. doi: 10.4161/cam.26941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Alderazi Y, Coons SW, Chapman K. Catastrophic demyelinating encephalomyelitis after intrathecal and intravenous stem cell transplantation in a patient with multiple sclerosis. J Child Neurol. 2011;27:632–5. doi: 10.1177/0883073811422831. [DOI] [PubMed] [Google Scholar]
  • 60.Berard JA, Bowmanb M, Atkins HL, Freedman MS, Walker LA. Cognitive fatigue in individuals with multiple sclerosis undergoing immunoablative therapy and hematopoietic stem cell transplantation. J Neurol Sci. 2014;336:132–7. doi: 10.1016/j.jns.2013.10.023. [DOI] [PubMed] [Google Scholar]
  • 61.Bowen JD, Kraft GH, Wundes A, Guan Q, Maravilla KR, Gooley TA, et al. Autologous hematopoietic cell transplantation following high-dose immunosuppressive therapy for advanced multiple sclerosis: Long-term results. Bone Marrow Transplant. 2012;47:946–51. doi: 10.1038/bmt.2011.208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Chen B, Zhou M, Ouyang J, Zhou R, Xu J, Zhang Q, et al. Long-term efficacy of autologous haematopoietic stem cell transplantation in multiple sclerosis at a single institution in China. Neurol Sci. 2012;33:881–6. doi: 10.1007/s10072-011-0859-y. [DOI] [PubMed] [Google Scholar]
  • 63.Shevchenko JL, Kuznetsova AN, Ionova TI, Melnichenko VY, Fedorenko DA, Kartashov KA, et al. Autologous hematopoietic stem cell transplantation with reduced-intensity conditioning in multiple sclerosis. Exp Hematol. 2012;40:892–8. doi: 10.1016/j.exphem.2012.07.003. [DOI] [PubMed] [Google Scholar]
  • 64.Mancardi GL, Sormani MP, Di Gioia M, Vuolo L, Gualandi F, Amato MP, et al. Autologous haematopoietic stem cell transplantation with an intermediate intensity conditioning regimen in multiple sclerosis: The Italian multi-centre experience. Mult Scler. 2012;18:835–42. doi: 10.1177/1352458511429320. [DOI] [PubMed] [Google Scholar]
  • 65.Fassas A, Kimiskidis VK, Sakellari I, Kapinas K, Anagnostopoulos A, Tsimourtou V, et al. Long-term results of stem cell transplantation for MS: A single-center experience. Neurology. 2011;76:1066–70. doi: 10.1212/WNL.0b013e318211c537. [DOI] [PubMed] [Google Scholar]
  • 66.Hamerschlak N, Rodrigues M, Moraes DA, Oliveira MC, Stracieri AB, Pieroni F, et al. Brazilian experience with two conditioning regimens in patients with multiple sclerosis: BEAM/horse ATG and CY/rabbit ATG. Bone Marrow Transplant. 2010;45:239–48. doi: 10.1038/bmt.2009.127. [DOI] [PubMed] [Google Scholar]
  • 67.Rice CM, Mallam EA, Whone AL, Walsh P, Brooks DJ, Kane N, et al. Safety and feasibility of autologous bone marrow cellular therapy in relapsing-progressive multiple sclerosis. Clin Pharmacol Ther. 2010;87:679–85. doi: 10.1038/clpt.2010.44. [DOI] [PubMed] [Google Scholar]
  • 68.Burt RK, Loh Y, Cohen B, Stefosky D, Balabanov R, Katsamakis G, et al. Autologous non-myeloablative haemopoietic stem cell transplantation in relapsing-remitting multiple sclerosis: A phase I/II study. Lancet Neurol. 2009;8:244–53. doi: 10.1016/S1474-4422(09)70017-1. [DOI] [PubMed] [Google Scholar]
  • 69.Fagius J, Lundgren J, Oberg G. Early highly aggressive MS successfully treated by hematopoietic stem cell transplantation. Mult Scler. 2009;15:229–37. doi: 10.1177/1352458508096875. [DOI] [PubMed] [Google Scholar]
  • 70.Samijn JP, te Boekhorst PA, Mondria T, van- Doorn PA, Flach HZ, van-der-Meché FG, et al. Intense T cell depletion followed by autologous bone marrow transplantation for severe multiple sclerosis. J Neurol Neurosurg Psychiatry. 2006;77:46–50. doi: 10.1136/jnnp.2005.063883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Reston JT, Uhl S, Treadwell JR, Nash RA, Schoelles K. Autologous hematopoietic cell transplantation for multiple sclerosis: A systematic review. Mult Scler. 2011;17:204–13. doi: 10.1177/1352458510383609. [DOI] [PubMed] [Google Scholar]
  • 72.Burt RK, Cohen BA, Russell E, Spero K, Joshi A, Oyama Y, et al. Hematopoietic stem cell transplantation for progressive multiple sclerosis: Failure of a total body irradiation-based conditioning regimen to prevent disease progression in patients with high disability scores. Blood. 2003;102:2373–8. doi: 10.1182/blood-2003-03-0877. [DOI] [PubMed] [Google Scholar]
  • 73.Alix JJ, Blackburn DJ, Sokhi D, Craven I, Sharrack B, Snowden JA. Autologous hematopoietic stem cell transplantation following pulsed cyclophosphamide in a severely disabled patient with malignant multiple sclerosis. J Neurol. 2013;260:914–6. doi: 10.1007/s00415-012-6796-8. [DOI] [PubMed] [Google Scholar]
  • 74.Burt RK, Loh Y, Pearce W, Beohar N, Barr WG, Craig R, et al. Clinical applications of blood-derived and marrow-derived stem cells for nonmalignant diseases. JAMA. 2008;299:925–36. doi: 10.1001/jama.299.8.925. [DOI] [PubMed] [Google Scholar]
  • 75.Reekmans K, De Vocht N, Praet J, Fransen E, Le Blon D, Hoornaert C, et al. Spatiotemporal evolution of early innate immune responses triggered by neural stem cell grafting. Stem Cell Res Ther. 2012;3:56. doi: 10.1186/scrt147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ubiali F, Nava S, Nessi V, Frigerio S, Parati E, Bernasconi P, et al. Allorecognition of human neural stem cells by peripheral blood lymphocytes despite low expression of MHC molecules: Role of TGF-beta in modulating proliferation. Int Immunol. 2007;19:1063–74. doi: 10.1093/intimm/dxm079. [DOI] [PubMed] [Google Scholar]
  • 77.Joy JE, Johnston RB, editors. Multiple Sclerosis: Current Status and Strategies for the Future. Washington (DC): National Academies Press (US); 2001. Institute of Medicine (US) Committee on Multiple Sclerosis: Current Status and Strategies for the Future; pp. 25–36. [PubMed] [Google Scholar]

Articles from Advanced Biomedical Research are provided here courtesy of Wolters Kluwer -- Medknow Publications

RESOURCES