Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biol Pharm Bull ; 43(5): 757-761, 2020 May 01.
Article in English | MEDLINE | ID: mdl-32132342

ABSTRACT

In this study, a new and sensitive enzyme-linked immunosorbent assay (ELISA) was developed by introducing a polymer as a reaction carrier. The results suggest that the newly developed ELISA method is more convenient than the existing paper-based ELISA method and applicable to a wider range of environments. In addition, the sensitivity of the new method is much higher than that of the existing paper-based ELISA method and even higher than that of the traditional ELISA method.


Subject(s)
Dendrimers/chemistry , Enzyme-Linked Immunosorbent Assay/methods , Polyethylene Glycols/chemistry , Antibodies/immunology , Glutathione Transferase/immunology , Humans , Nogo Proteins/blood , Nogo Proteins/immunology , Sensitivity and Specificity
2.
Neural Dev ; 15(1): 1, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31918754

ABSTRACT

As humans, we cannot regenerate axons within the central nervous system (CNS), therefore, making any damage to it permanent. This leads to the loss of sensory and motor function below the site of injury and can be crippling to a person's health. Spontaneous recovery can occur from plastic changes, but it is minimal. The absence of regeneration is due to the inhibitory environment of the CNS as well as the inherent inability of CNS axons to form growth cones. Amongst many factors, one of the major inhibitory signals of the CNS environment is the myelin-associated Nogo pathway. Nogo-A, Nogo-B and Nogo-C (Nogo), stimulate the Nogo receptor, inhibiting neurite outgrowth by causing growth cones to collapse through activation of Rho Kinase (ROCK). Antibodies can be used to target this signalling pathway by binding to Nogo and thus promote the outgrowth of neuronal axons in the CNS. This use of anti-Nogo antibodies has been shown to upregulate CNS regeneration as well as drastically improve sensory and motor function in both rats and primates when coupled with adequate training. Here, we evaluate whether the experimental success of anti-Nogo at improving CNS regeneration can be carried over into the clinical setting to treat spinal cord injuries (SCI) and their symptoms successfully. Furthermore, we also discuss potential methods to improve the current treatment and any developmental obstacles.


Subject(s)
Immunotherapy/methods , Nerve Regeneration , Nogo Proteins/antagonists & inhibitors , Nogo Proteins/immunology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/physiopathology , Animals , Antibodies/administration & dosage , Growth Cones/drug effects , Growth Cones/physiology , Humans , Neurons/drug effects , Neurons/physiology , Signal Transduction/drug effects , Treatment Outcome
3.
Sci Rep ; 9(1): 20040, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882970

ABSTRACT

Angiogenesis is a key restorative process following stroke but has also been linked to increased vascular permeability and blood brain barrier (BBB) disruption. Previous pre-clinical approaches primarily focused on the administration of vascular endothelial growth factor (VEGF) to promote vascular repair after stroke. Although shown to improve angiogenesis and functional recovery from stroke, VEGF increased the risk of blood brain barrier disruption and bleedings to such an extent that its clinical use is contraindicated. As an alternative strategy, antibodies against the neurite growth inhibitory factor Nogo-A have recently been shown to enhance vascular regeneration in the ischemic central nervous system (CNS); however, their effect on vascular permeability is unknown. Here, we demonstrate that antibody-mediated Nogo-A neutralization following stroke has strong pro-angiogenic effects but does not increase vascular permeability as opposed to VEGF. Moreover, VEGF-induced vascular permeability was partially prevented when VEGF was co-administered with anti-Nogo-A antibodies. This study may provide a novel therapeutic strategy for vascular repair and maturation in the ischemic brain.


Subject(s)
Angiogenesis Inducing Agents/immunology , Autoantibodies/immunology , Capillary Permeability/immunology , Nogo Proteins/immunology , Stroke/physiopathology , Animals , Disease Models, Animal , Humans , Neovascularization, Pathologic , Vascular Endothelial Growth Factors/administration & dosage
4.
Cell Rep ; 29(3): 671-684.e6, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31618635

ABSTRACT

Precisely controlling the excitatory and inhibitory balance is crucial for the stability and information-processing ability of neuronal networks. However, the molecular mechanisms maintaining this balance during ongoing sensory experiences are largely unclear. We show that Nogo-A signaling reciprocally regulates excitatory and inhibitory transmission. Loss of function for Nogo-A signaling through S1PR2 rapidly increases GABAAR diffusion, thereby decreasing their number at synaptic sites and the amplitude of GABAergic mIPSCs at CA3 hippocampal neurons. This increase in GABAAR diffusion rate is correlated with an increase in Ca2+ influx and requires the calcineurin-mediated dephosphorylation of the γ2 subunit at serine 327. These results suggest that Nogo-A signaling rapidly strengthens inhibitory GABAergic transmission by restricting the diffusion dynamics of GABAARs. Together with the observation that Nogo-A signaling regulates excitatory transmission in an opposite manner, these results suggest a crucial role for Nogo-A signaling in modulating the excitation and inhibition balance to restrict synaptic plasticity.


Subject(s)
Nogo Proteins/metabolism , Receptors, GABA-A/metabolism , Animals , Antibodies, Blocking/immunology , Calcineurin/metabolism , Calcium/metabolism , Cells, Cultured , Female , Hippocampus/cytology , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Nogo Proteins/immunology , Patch-Clamp Techniques , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Rats, Wistar , Receptors, GABA-A/genetics , Signal Transduction , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Sphingosine-1-Phosphate Receptors/metabolism , Synapses/metabolism , Synaptic Transmission
5.
Proc Natl Acad Sci U S A ; 116(28): 14270-14279, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31235580

ABSTRACT

Stroke is a major cause of serious disability due to the brain's limited capacity to regenerate damaged tissue and neuronal circuits. After ischemic injury, a multiphasic degenerative and inflammatory response is coupled with severely restricted vascular and neuronal repair, resulting in permanent functional deficits. Although clinical evidence indicates that revascularization of the ischemic brain regions is crucial for functional recovery, no therapeutics that promote angiogenesis after cerebral stroke are currently available. Besides vascular growth factors, guidance molecules have been identified to regulate aspects of angiogenesis in the central nervous system (CNS) and may provide targets for therapeutic angiogenesis. In this study, we demonstrate that genetic deletion of the neurite outgrowth inhibitor Nogo-A or one of its corresponding receptors, S1PR2, improves vascular sprouting and repair and reduces neurological deficits after cerebral ischemia in mice. These findings were reproduced in a therapeutic approach using intrathecal anti-Nogo-A antibodies; such a therapy is currently in clinical testing for spinal cord injury. These results provide a basis for a therapeutic blockage of inhibitory guidance molecules to improve vascular and neural repair after ischemic CNS injuries.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Brain Ischemia/drug therapy , Nogo Proteins/genetics , Sphingosine-1-Phosphate Receptors/genetics , Stroke/drug therapy , Animals , Brain/drug effects , Brain/pathology , Brain Ischemia/genetics , Brain Ischemia/immunology , Brain Ischemia/pathology , Central Nervous System/drug effects , Central Nervous System/pathology , Disease Models, Animal , Humans , Mice , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/immunology , Neurons/drug effects , Neurons/pathology , Nogo Proteins/antagonists & inhibitors , Nogo Proteins/immunology , Pyramidal Tracts/drug effects , Pyramidal Tracts/pathology , Recovery of Function/genetics , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Sphingosine-1-Phosphate Receptors/immunology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/immunology , Spinal Cord Injuries/pathology , Stroke/genetics , Stroke/immunology , Stroke/pathology
6.
Med Sci (Paris) ; 35(12): 1005-1007, 2019 Dec.
Article in French | MEDLINE | ID: mdl-31903908

ABSTRACT

Therapeutic antibodies have been successfully developed in neurology. However, their efficacy needs to overcome a main hurdle that is their limited access to the brain. Moreover, the multifactorial characteristics of many neurological diseases complicate the identification of a specific target. In this review, we present the neuroroprotective effect of some monoclonal antibodies and we summarize how they can interact with neurodegenerative and inflammatory processes. Monoclonal antibodies, such as natalizumab, that represent a major achievement in multiple sclerosis, have been approved in this indication and used in the routine practice. Also, they are still in a phase of development in degenerative diseases and mainly aim to counteract the aggregation of misfolded proteins such as amyloid beta or tau proteins. Another strategy in development of new monoclonal antibodies is to block physiological inhibitors of axonal growth and myelination such as Nogo-A or its co-transporter LINGO-1. Finally, the recent approbation by the FDA of monoclonal antibodies against CGRP for migraine treatment has been a major breakthrough that expanded the potential fields of application of antibodies in neurology. Thus, a lot of research efforts are now devoted to improve the galenic forms, the routes of administration and to extend these various approaches to other targets.


TITLE: Les anticorps monoclonaux en neurologie. ABSTRACT: L'utilisation des anticorps thérapeutiques commence à se développer avec succès en neurologie. Leur efficacité est conditionnée par la capacité à contourner la principale limite à l'utilisation de ces molécules dans ce type d'indication qu'est l'accessibilité au cerveau. Le caractère multifactoriel de ces pathologies neurologiques rend également complexe l'identification d'une cible spécifique. Nous exposons dans cette revue les effets neuroprotecteurs des anticorps monoclonaux et résumons leurs activités sur les mécanismes neurodégénératifs et inflammatoires. Les anticorps monoclonaux, tels que le natalizumab, représentent une avancée importante dans le traitement de la sclérose en plaque (SEP) et sont désormais utilisés en routine. Ce type de thérapeutique est toujours en développement dans les maladies neurodégénératives, en agissant principalement sur l'agrégation des protéines mal repliées, telles que la protéine béta-amyloïde et la protéine tau. Une autre stratégie en développement consiste à bloquer les inhibiteurs physiologiques de croissance axonale et de myélinisation, tels que Nogo-A et son co-transporteur LINGO-1. L'autorisation récente par la food and drug administration américaine des anticorps monoclonaux anti-calcitonin gene-related peptide (CGRP) dans la migraine a soulevé un regain d'intérêt pour ces thérapeutiques en neurologie. La recherche est de ce fait très active pour améliorer les formes galéniques et les voies d'administration et pour étendre ces thérapeutiques à d'autres cibles.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Nervous System Diseases/drug therapy , Neurology/trends , Humans , Membrane Proteins/immunology , Multiple Sclerosis/drug therapy , Natalizumab/therapeutic use , Nerve Tissue Proteins/immunology , Nervous System Diseases/immunology , Neurology/methods , Neuroprotective Agents/therapeutic use , Nogo Proteins/immunology
7.
Eur J Neurosci ; 48(4): 2050-2070, 2018 08.
Article in English | MEDLINE | ID: mdl-30019432

ABSTRACT

Functional recovery from central nervous system injury is likely to be partly due to a rearrangement of neural circuits. In this context, the corticobulbar (corticoreticular) motor projections onto different nuclei of the ponto-medullary reticular formation (PMRF) were investigated in 13 adult macaque monkeys after either, primary motor cortex injury (MCI) in the hand area, or spinal cord injury (SCI) or Parkinson's disease-like lesions of the nigro-striatal dopaminergic system (PD). A subgroup of animals in both MCI and SCI groups was treated with neurite growth promoting anti-Nogo-A antibodies, whereas all PD animals were treated with autologous neural cell ecosystems (ANCE). The anterograde tracer BDA was injected either in the premotor cortex (PM) or in the primary motor cortex (M1) to label and quantify corticobulbar axonal boutons terminaux and en passant in PMRF. As compared to intact animals, after MCI the density of corticobulbar projections from PM was strongly reduced but maintained their laterality dominance (ipsilateral), both in the presence or absence of anti-Nogo-A antibody treatment. In contrast, the density of corticobulbar projections from M1 was increased following opposite hemi-section of the cervical cord (at C7 level) and anti-Nogo-A antibody treatment, with maintenance of contralateral laterality bias. In PD monkeys, the density of corticobulbar projections from PM was strongly reduced, as well as that from M1, but to a lesser extent. In conclusion, the densities of corticobulbar projections from PM or M1 were affected in a variable manner, depending on the type of lesion/pathology and the treatment aimed to enhance functional recovery.


Subject(s)
Brain Injuries/pathology , Motor Cortex/injuries , Motor Cortex/pathology , Parkinson Disease/pathology , Pyramidal Tracts/pathology , Reticular Formation/pathology , Rhombencephalon/pathology , Spinal Cord Injuries/pathology , Animals , Antibodies, Blocking/administration & dosage , Brain Injuries/therapy , Cell Transplantation , Disease Models, Animal , Female , Hand/pathology , Macaca fascicularis , Male , Neuroanatomical Tract-Tracing Techniques , Nogo Proteins/immunology , Parkinson Disease/therapy , Spinal Cord Injuries/therapy , Transplantation, Autologous
8.
Glia ; 66(10): 2079-2093, 2018 10.
Article in English | MEDLINE | ID: mdl-30051920

ABSTRACT

Nogo-A is a potent glial-derived inhibitor of axon growth in the injured CNS and acts as a negative regulator of developmental angiogenesis by inhibiting vascular endothelial cell migration. However, its function in pathological angiogenesis has never been studied after ischemic injury in the CNS. Using the mouse model of oxygen-induced retinopathy (OIR) which yields defined zones of retinal ischemia, our goal was to investigate the role of Nogo-A in vascular regeneration. We demonstrate a marked upregulation of the Nogo-A receptor sphingosine 1-phosphate receptor 2 in blood vessels following OIR, while Nogo-A is abundantly expressed in surrounding glial cells. Acute inhibition of Nogo-A with function-blocking antibody 11C7 significantly improved vascular regeneration and consequently prevented pathological pre-retinal angiogenesis. Ultimately, inhibition of Nogo-A led to restoration of retinal function as determined by electrophysiological response of retinal cells to light stimulation. Our data suggest that anti-Nogo-A antibody may protect neuronal cells from ischemic damage by accelerating blood vessel repair in the CNS. Targeting Nogo-A by immunotherapy may improve CNS perfusion after vascular injuries.


Subject(s)
Ischemia/metabolism , Neovascularization, Physiologic/physiology , Nogo Proteins/metabolism , Regeneration/physiology , Retinal Diseases/metabolism , Retinal Vessels/metabolism , Angiogenesis Inducing Agents/pharmacology , Animals , Disease Models, Animal , Ischemia/drug therapy , Ischemia/pathology , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Nogo Proteins/antagonists & inhibitors , Nogo Proteins/immunology , Receptors, Lysosphingolipid/metabolism , Regeneration/drug effects , Retinal Diseases/drug therapy , Retinal Diseases/pathology , Retinal Vessels/drug effects , Retinal Vessels/pathology , Sphingosine-1-Phosphate Receptors , Vision, Ocular/drug effects , Vision, Ocular/physiology
9.
Neurorehabil Neural Repair ; 32(6-7): 578-589, 2018 06.
Article in English | MEDLINE | ID: mdl-29869587

ABSTRACT

BACKGROUND: Neutralization of central nervous system neurite growth inhibitory factors, for example, Nogo-A, is a promising approach to improving recovery following spinal cord injury (SCI). In animal SCI models, intrathecal delivery of anti-Nogo-A antibodies promoted regenerative neurite growth and functional recovery. OBJECTIVE: This first-in-man study assessed the feasibility, safety, tolerability, pharmacokinetics, and preliminary efficacy of the human anti-Nogo-A antibody ATI355 following intrathecal administration in patients with acute, complete traumatic paraplegia and tetraplegia. METHODS: Patients (N = 52) started treatment 4 to 60 days postinjury. Four consecutive dose-escalation cohorts received 5 to 30 mg/2.5 mL/day continuous intrathecal ATI355 infusion over 24 hours to 28 days. Following pharmacokinetic evaluation, 2 further cohorts received a bolus regimen (6 intrathecal injections of 22.5 and 45 mg/3 mL, respectively, over 4 weeks). RESULTS: ATI355 was well tolerated up to 1-year follow-up. All patients experienced ≥1 adverse events (AEs). The 581 reported AEs were mostly mild and to be expected following acute SCI. Fifteen patients reported 16 serious AEs, none related to ATI355; one bacterial meningitis case was considered related to intrathecal administration. ATI355 serum levels showed dose-dependency, and intersubject cerebrospinal fluid levels were highly variable after infusion and bolus injection. In 1 paraplegic patient, motor scores improved by 8 points. In tetraplegic patients, mean total motor scores increased, with 3/19 gaining >10 points, and 1/19 27 points at Week 48. Conversion from complete to incomplete SCI occurred in 7/19 patients with tetraplegia. CONCLUSIONS: ATI335 was well tolerated in humans; efficacy trials using intrathecal antibody administration may be considered in acute SCI.


Subject(s)
Immunoglobulin G/therapeutic use , Nerve Regeneration/drug effects , Neurites/drug effects , Nogo Proteins/immunology , Paraplegia/drug therapy , Quadriplegia/drug therapy , Spinal Cord Injuries/drug therapy , Adolescent , Adult , Dose-Response Relationship, Drug , Feasibility Studies , Female , Humans , Immunoglobulin G/administration & dosage , Injections, Spinal , Male , Middle Aged , Paraplegia/etiology , Quadriplegia/etiology , Recovery of Function/drug effects , Treatment Outcome , Young Adult
10.
J Neuropathol Exp Neurol ; 76(8): 683-696, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28789474

ABSTRACT

Ischemic stroke is a leading cause of adult disability with no pharmacological treatments to promote the recovery of lost function. Neutralizing antibodies against the neurite outgrowth inhibitor Nogo-A have emerged as a promising treatment for subacute and chronic stroke in animal models; however, whether anti-Nogo-A treatment affects poststroke neurogenesis remains poorly understood. In this study, we confirmed expression of Nogo-A by neuroblasts in the adult rat subventricular zone (SVZ), a major neurogenic niche; however, we found no evidence that Nogo-A was expressed at the surface of these cells. In vitro migration assays demonstrated that Nogo-A signaling induced a modest reduction in neuroblast migration speed, while anti-Nogo-A antibodies had no effect on motility properties. Using a permanent distal middle cerebral artery occlusion model of cortical stroke, we found that the number of proliferating cells in the SVZ was unaffected in response to stroke, while neuroblast mobilization from the SVZ toward the stroke lesion correlated positively with lesion size. However, we found no evidence that proliferation or neuroblast mobilization were affected by anti-Nogo-A antibody treatment. Our results suggest that the SVZ is not a therapeutic target of anti-Nogo-A immunotherapy, and contribute to our understanding of the SVZ response to cortical stroke.


Subject(s)
Antibodies/pharmacology , Antibodies/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Lateral Ventricles/drug effects , Nogo Proteins/immunology , Animals , Bromodeoxyuridine/metabolism , Cell Movement/drug effects , Cyclosporine/immunology , Disease Models, Animal , Functional Laterality , In Vitro Techniques , Infusions, Intraventricular , Lateral Ventricles/cytology , Male , Nerve Tissue Proteins/metabolism , Nogo Proteins/metabolism , Nogo Receptor 1/metabolism , Organ Culture Techniques , Rats , Rats, Long-Evans , Receptors, Lysosphingolipid/metabolism , Sphingosine-1-Phosphate Receptors , Time Factors
11.
Acta Neuropathol ; 134(3): 423-440, 2017 09.
Article in English | MEDLINE | ID: mdl-28646336

ABSTRACT

Two hallmarks of chronic multiple sclerosis lesions are the absence of significant spontaneous remyelination and primary as well as secondary neurodegeneration. Both characteristics may be influenced by the presence of inhibitory factors preventing myelin and neuronal repair. We investigated the potential of antibodies against Nogo-A, a well-known inhibitory protein for neuronal growth and plasticity, to enhance neuronal regeneration and remyelination in two animal models of multiple sclerosis. We induced a targeted experimental autoimmune encephalomyelitis (EAE) lesion in the dorsal funiculus of the cervical spinal cord of adult rats resulting in a large drop of skilled forelimb motor functions. We subsequently observed improved recovery of forelimb function after anti-Nogo-A treatment. Anterograde tracing of the corticospinal tract revealed enhanced axonal sprouting and arborisation within the spinal cord gray matter preferentially targeting pre-motor and motor spinal cord laminae on lesion level and above in the anti-Nogo-A-treated animals. An important additional effect of Nogo-A-neutralization was enhanced remyelination observed after lysolecithin-induced demyelination of spinal tracts. Whereas remyelinated fiber numbers in the lesion site were increased several fold, no effect of Nogo-A-inhibition was observed on oligodendrocyte precursor proliferation, migration, or differentiation. Enhancing remyelination and promoting axonal regeneration and plasticity represent important unmet medical needs in multiple sclerosis. Anti-Nogo-A antibodies hold promise as a potential new therapy for multiple sclerosis, in particular during the chronic phase of the disease when neurodegeneration and remyelination failure determine disability evolution.


Subject(s)
Axons/immunology , Brain/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Nogo Proteins/immunology , Remyelination/immunology , Animals , Antibodies/pharmacology , Axons/drug effects , Axons/pathology , Brain/drug effects , Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Inflammation/immunology , Inflammation/pathology , Rats , Rats, Inbred Lew , Recovery of Function/physiology , Remyelination/drug effects
12.
Eur Rev Med Pharmacol Sci ; 21(8): 1780-1786, 2017 04.
Article in English | MEDLINE | ID: mdl-28485802

ABSTRACT

OBJECTIVE: The use of an antibody to block the neurite outgrowth inhibitor Nogo-A has been of great interest for promoting axonal recovery as a treatment for peripheral nerve injuries. The present study aimed to investigate the signaling pathway of p75 neurotrophin receptor (NTR) and Nogo receptor (NgR) in a sciatic nerve transection (SNT) rat model and evaluate the underlining mechanisms. MATERIALS AND METHODS: Seventy-five Sprague-Dawley (SD) rats were randomly divided into 3 groups (n=25), namely the sham group, sciatic nerve transection (model) group and Nogo-A-pAb group. Following euthanasia, spinal cord and sciatic nerve of the operation site were harvested, fixed in formalin. Hematoxylin and eosin (HE) staining was used to evaluate the sciatic nerve pathology. The mRNA and protein expression levels of Nogo-A, NTR were assessed by Real-time polymerase chain reaction (RT-PCR) and Western blotting, respectively. RESULTS: Histology showed enhanced regeneration of spinal axon in the anti-Nogo-A antibody group. At 48 hours after operation, mRNA of Nogo-A and NTR were higher in model group compared with control group. mRNAs were at their highest levels at 1 week, while these were at normal levels after 4 weeks in Nogo-A-pAb group. The protein levels of Nogo-A and NTR were higher in model group compared with sham-operation group at 1-week after operation; Nogo-A-pAb could reduce these proteins levels. CONCLUSIONS: The results suggested Nogo-A antibody might represent a promising repair strategy to promote recovery following SNT.


Subject(s)
Neurons/metabolism , Nogo Proteins/immunology , Sciatic Nerve/pathology , Animals , Rats , Rats, Sprague-Dawley
13.
Exp Neurol ; 292: 135-144, 2017 06.
Article in English | MEDLINE | ID: mdl-28341461

ABSTRACT

Intense training is the most clinically successful treatment modality following incomplete spinal cord injuries (SCIs). With the advent of plasticity enhancing treatments, understanding how treatments might interact when delivered in combination becomes critical. Here, we investigated a rational approach to sequentially combine treadmill locomotor training with antibody mediated suppression of the fiber growth inhibitory protein Nogo-A. Following a large but incomplete thoracic lesion, rats were immediately treated with either anti-Nogo-A or control antibody (2weeks) and then either left untrained or step-trained starting 3weeks after injury for 8weeks. It was found that sequentially combined therapy improved step consistency and reduced toe dragging and climbing errors, as seen with training and anti-Nogo-A individually. Animals with sequential therapy also adopted a more parallel paw position during bipedal walking and showed greater overall quadrupedal locomotor recovery than individual treatments. Histologically, sequential therapy induced the greatest corticospinal tract sprouting caudally into the lumbar region and increased the number of serotonergic synapses onto lumbar motoneurons. Increased primary afferent sprouting and synapse formation onto lumbar motoneurons observed with anti-Nogo-A antibody were reduced by training. Animals with sequential therapy also showed the highest reduction of lumbar interneuronal activity associated with walking (c-fos expression). No treatment effects for thermal nociception, mechanical allodynia, or lesion volume were observed. The results demonstrate that sequential administration of anti-Nogo-A antibody followed in time with intensive locomotor training leads to superior recovery of lost locomotor functions, which is probably mediated by changes in the interaction between descending sprouting and local segmental networks after SCI.


Subject(s)
Antibodies/pharmacology , Locomotion/drug effects , Nerve Regeneration/drug effects , Pyramidal Tracts/drug effects , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Animals , Female , Motor Activity/drug effects , Motor Activity/physiology , Myelin Proteins/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Nogo Proteins/immunology , Nogo Proteins/metabolism , Physical Conditioning, Animal , Rats, Sprague-Dawley , Recovery of Function/physiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
14.
Lancet Neurol ; 16(3): 208-216, 2017 03.
Article in English | MEDLINE | ID: mdl-28139349

ABSTRACT

BACKGROUND: Neurite outgrowth inhibitor A (Nogo-A) is thought to have a role in the pathophysiology of amyotrophic lateral sclerosis (ALS). A monoclonal antibody against Nogo-A showed a positive effect in the SOD1G93A mouse model of ALS, and a humanised form of this antibody (ozanezumab) was well tolerated in a first-in-human trial. Therefore, we aimed to assess the safety and efficacy of ozanezumab in patients with ALS. METHODS: This randomised, double-blind, placebo-controlled, phase 2 trial was done in 34 centres in 11 countries. Patients aged 18-80 years with a diagnosis of familial or sporadic ALS were randomly assigned (1:1), centrally according to a computer-generated allocation schedule, to receive ozanezumab (15 mg/kg) or placebo as intravenous infusions over 1 h every 2 weeks for 46 weeks, followed by assessments at week 48 and week 60. Patients and study personnel were masked to treatment assignment. The primary outcome was a joint-rank analysis of function (ALS Functional Rating Scale-Revised) and overall survival, analysed at 48 weeks in all patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov, number NCT01753076, and with GSK-ClinicalStudyRegister.com, NOG112264, and is completed. FINDINGS: Between Dec 20, 2012, and Nov 1, 2013, we recruited 307 patients, of whom 303 were randomly assigned to receive placebo (n=151) or ozanezumab (n=152). The adjusted mean of the joint-rank score was -14·9 (SE 13·5) for the ozanezumab group and 15·0 (13·6) for the placebo group, with a least squares mean difference of -30·0 (95% CI -67·9 to 7·9; p=0·12). Overall, reported adverse events, serious adverse events, and adverse events leading to permanent discontinuation of study drug or withdrawal from study were similar between the treatment groups, except for dyspepsia (ten [7%] in the ozanezumab group vs four [3%] in the placebo group), depression (11 [7%] vs five [3%]), and diarrhoea (25 [16%] vs 12 [8%]). Respiratory failure was the most common serious adverse event (12 [8%] vs seven [5%]). At week 60, the number of deaths was higher in the ozanezumab group (20 [13%]) than in the placebo group (16 [11%]), mainly as a result of respiratory failure (ten [7%] vs five [3%]). Two deaths were considered related to the study drug (bladder transitional cell carcinoma in the ozanezumab group and cerebrovascular accident in the placebo group). INTERPRETATION: Ozanezumab did not show efficacy compared with placebo in patients with ALS. Therefore, Nogo-A does not seem to be an effective therapeutic target in ALS. FUNDING: GlaxoSmithKline.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Immunologic Factors/therapeutic use , Nogo Proteins/immunology , Treatment Outcome , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/mortality , Double-Blind Method , Electrocardiography , Female , Humans , International Cooperation , Male , Middle Aged , Survival Analysis , Young Adult
15.
CNS Drugs ; 31(3): 187-198, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28105588

ABSTRACT

Most of the current therapies, as well as many of the clinical trials, for multiple sclerosis (MS) target the inflammatory autoimmune processes, but less than 20% of all clinical trials investigate potential therapies for the chronic progressive disease stage of MS. The latter is responsible for the steadily increasing disability in many patients, and there is an urgent need for novel therapies that protect nervous system tissue and enhance axonal growth and/or remyelination. As outlined in this review, solid pre-clinical data suggest neutralization of the neurite outgrowth inhibitor Nogo-A as a potential new way to achieve both axonal and myelin repair. Several phase I clinical studies with anti-Nogo-A antibodies have been conducted in different disease paradigms including MS and spinal cord injury. Data from spinal cord injury and amyotrophic lateral sclerosis (ALS) trials accredit a good safety profile of high doses of anti-Nogo-A antibodies administered intravenously or intrathecally. An antibody against a Nogo receptor subunit, leucine rich repeat and immunoglobulin-like domain-containing protein 1 (LINGO-1), was recently shown to improve outcome in patients with acute optic neuritis in a phase II study. Nogo-A-suppressing antibodies could be novel drug candidates for the relapsing as well as the progressive MS disease stage. In this review, we summarize the available pre-clinical and clinical evidence on Nogo-A and elucidate the potential of Nogo-A-antibodies as a therapy for progressive MS.


Subject(s)
Antibodies/therapeutic use , Immunologic Factors/therapeutic use , Multiple Sclerosis/drug therapy , Nogo Proteins/immunology , Animals , Antibodies/pharmacology , Humans , Immunologic Factors/pharmacology , Multiple Sclerosis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...