Library - Amyotrophic lateral sclerosis

Neuroscience. 2013 Sep 17;248:333-43. doi: 10.1016/j.neuroscience.2013.05.034. Epub 2013 May 28.

Systemic treatment with adipose-derived mesenchymal stem cells ameliorates clinical and pathological features in the amyotrophic lateral sclerosis murine model.

Marconi S1Bonaconsa M1Scambi I1Squintani GM2Rui W1Turano E1Ungaro D3D’Agostino S1Barbieri F1Angiari S4Farinazzo A1Constantin G4,Del Carro U3Bonetti B1Mariotti R5.

Author information

1Department of Neurological Sciences and Movement, University of Verona, Verona, Italy.

2University Hospital of Verona, Verona, Italy.

3San Raffaele Hospital, Neurophysiology Unit, Milan, Italy.

4Department of Pathology and Diagnostics, University of Verona, Verona, Italy.

5Department of Neurological Sciences and Movement, University of Verona, Verona, Italy.

Abstract

Therapeutic strategies for the fatal neurodegenerative disease amyotrophic lateral sclerosis (ALS) are actually minimally effective on patients’ survival and quality of life. Although stem cell therapy has raised great expectations, information on the involved molecular mechanisms is still limited. Here we assessed the efficacy of the systemic administration of adipose-derived mesenchymal stem cells (ASC), a previously untestedstem cell population, in superoxide-dismutase 1 (SOD1)-mutant transgenic mice, the animal model of familial ALS. The administration of ASC to SOD1-mutant mice at the clinical onset significantly delayed motor deterioration for 4-6 weeks, as shown by clinical and neurophysiological tests. Neuropathological examination of ASC-treated SOD1-mutant mice at day 100 (i.e. the time of their best motor performance) revealed a higher number of lumbar motorneurons than in phosphate-buffered saline-treated SOD1-mutant mice and a restricted number of undifferentiated green fluorescent protein-labeled ASC in the spinal cord. By examining the spinal cord tissue factors that may prolong neuronal survival, we found a significant up-regulation in levels of glial-derived neurotrophic factor (GDNF) and basic fibroblast growth factor (bFGF) after ASC treatment. Considering that ASC produce bFGF but not GDNF, these findings indicate that ASC may promote neuroprotection either directly and/or by modulating the secretome of local glial cells toward a neuroprotective phenotype. Such neuroprotection resulted in a strong and long-lasting effect on motor performance and encourages the use of ASC in human pathologies, in which current therapies are not able to maintain a satisfying neurological functional status.

KEYWORDS:

GDNF; amyotrophic lateral sclerosis; motorneuron disease; neuroprotection; neurotrophins

JAMA Neurol. 2016 Mar;73(3):337-44. doi: 10.1001/jamaneurol.2015.4321.

Safety and Clinical Effects of Mesenchymal Stem Cells Secreting Neurotrophic Factor Transplantation in Patients With Amyotrophic Lateral Sclerosis: Results of Phase 1/2 and 2a Clinical Trials.

Petrou P1Gothelf Y2Argov Z1Gotkine M1Levy YS2Kassis I1Vaknin-Dembinsky A1Ben-Hur T1Offen D3Abramsky O1Melamed E4Karussis D1.

Author information

1Neurology Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.

2BrainStorm Cell Therapeutics, Petach Tikva, Israel.

3BrainStorm Cell Therapeutics, Petach Tikva, Israel3Felsenstein Medical Research Centre, Tel Aviv University, Tel Aviv, Israel.

4BrainStorm Cell Therapeutics, Petach Tikva, Israel4Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.

Abstract

IMPORTANCE:

Preclinical studies have shown that neurotrophic growth factors (NTFs) extend the survival of motor neurons in amyotrophic lateral sclerosis (ALS) and that the combined delivery of these neurotrophic factors has a strong synergistic effect. We have developed a culture-based method for inducing mesenchymal stem cells (MSCs) to secrete neurotrophic factors. These MSC-NTF cells have been shown to be protective in several animal models of neurodegenerative diseases.

OBJECTIVE:

To determine the safety and possible clinical efficacy of autologous MSC-NTF cells transplantation in patients with ALS.

DESIGN, SETTING, AND PARTICIPANTS:

In these open-label proof-of-concept studies, patients with ALS were enrolled between June 2011 and October 2014 at the Hadassah Medical Center in Jerusalem, Israel. All patients were followed up for 3 months before transplantation and 6 months after transplantation. In the phase 1/2 part of the trial, 6 patients with early-stage ALS were injected intramuscularly (IM) and 6 patients with more advanced disease were transplanted intrathecally (IT). In the second stage, a phase 2a dose-escalating study, 14 patients with early-stage ALS received a combined IM and IT transplantation of autologous MSC-NTF cells.

INTERVENTIONS:

Patients were administered a single dose of MSC-NTF cells.

MAIN OUTCOMES AND MEASURES:

The primary end points of the studies were safety and tolerability of this cell therapy. Secondary end points included the effects of the treatment on various clinical parameters, such as the ALS Functional Rating Scale-Revised score and the respiratory function.

RESULTS:

Among the 12 patients in the phase 1/2 trial and the 14 patients in the phase 2a trial aged 20 and 75 years, the treatment was found to be safe and well tolerated over the study follow-up period. Most of the adverse effects were mild and transient, not including any treatment-related serious adverse event. The rate of progression of the forced vital capacity and of the ALS Functional Rating Scale-Revised score in the IT (or IT+IM)-treated patients was reduced (from -5.1% to -1.2%/month percentage predicted forced vital capacity, P < .04 and from -1.2 to 0.6 ALS Functional Rating Scale-Revised points/month, P = .052) during the 6 months following MSC-NTF cell transplantation vs the pretreatment period. Of these patients, 13 (87%) were defined as responders to either ALS Functional Rating Scale-Revised or forced vital capacity, having at least 25% improvement at 6 months after treatment in the slope of progression.

CONCLUSIONS AND RELEVANCE:

The results suggest that IT and IM administration of MSC-NTF cells in patients with ALS is safe and provide indications of possible clinical benefits, to be confirmed in upcoming clinical trials.

TRIAL REGISTRATION:

clinicaltrials.gov Identifiers: NCT01051882 and NCT01777646.

Stem Cells. 2014 Oct;32(10):2724-31. doi: 10.1002/stem.1770.

Biological markers of mesenchymal stromal cells as predictors of response to autologous stem cell transplantation in patients with amyotrophic lateral sclerosis: an investigator-initiated trial and in vivo study.

Kim HY1Kim HOh KWOh SIKoh SHBaik WNoh MYKim KSKim SH.

Author information

1Department of Neurology, College of Medicine, Hanyang University, Seoul, South Korea.

Abstract

Bone marrow mesenchymal stromal cells (MSCs) can modify disease progression in amyotrophic lateral sclerosis (ALS) model. However, there are currently no accurate biological markers for predicting the efficacy of autologous MSC transplants in ALS patients. This open-label, single-arm, investigator-initiated clinical study was designed to identify markers of MSCs that could be used as potential predictors of response to autologous MSC therapy in patients with ALS. We enrolled 37 patients with ALS who received autologous MSCs via intrathecal injection in two monthly doses. After a 6-month follow-up period, the patients were categorized as responders and non-responders based on their scores on the revised ALS Functional Rating Scale (ALSFRS-R). Biological markers including β-fibroblast growth factor-2, stromal cell-derived factor-1α, vascular endothelial growth factor (VEGF), insulin-like growth factor-1, brain-derived neurotrophic factor, angiogenin (ANG), interleukin (IL)-4, IL-10, and transforming growth factor-β (TGF-β) were measured in the MSC cultures and their levels were compared between the responders and nonresponders. To confirm the markers’ predictive ability, MSCs isolated from one patient in each group were transplanted into the cisterna magna of mutant SOD1(G93A) transgenic mice to measure their lifespans, locomotor activity, and motor neuron numbers. The levels of VEGF, ANG, and TGF-β were significantly higher in responders than in nonresponders. In the mouse model, the recipients of responder MSCs had a significantly slower onset of symptoms and a significantly longer lifespan than the recipients of nonresponders or controls. Our data suggest that VEGF, ANG, and TGF-β levels in MSCs could be used as potential biological markers to predict the effectiveness of autologous MSC therapy and to identify those patients who could optimally benefit from MSC treatment.

KEYWORDS: Amyotrophic lateral sclerosis; Biological marker; In vivo study; Investigator-initiated trial; Mesenchymal stromal cell