101
|
Ciampi E, Pareto D, Sastre-Garriga J, Vidal-Jordana A, Tur C, Río J, Tintoré M, Auger C, Rovira A, Montalban X. Grey matter atrophy is associated with disability increase in natalizumab-treated patients. Mult Scler 2016; 23:556-566. [DOI: 10.1177/1352458516656808] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Brain volume loss (BVL) is a key outcome in multiple sclerosis (MS) trials. Natalizumab is highly effective on inflammation with moderate impact on atrophy. Objective: To explore BVL in patients receiving natalizumab with an emphasis on grey matter (GM). Methods: We performed a retrospective post hoc analysis of BVL in 38 patients receiving natalizumab for 3 years using longitudinal voxel-based morphometry (VBM) and FreeSurfer. Results: Significant BVL was observed during first year: brain parenchymal fraction (BPF): −1.12% ( p < 0.001); white matter fraction (WMF): −0.9% ( p = 0.001); grey matter fraction (GMF): −1.28% ( p = 0.002). GM loss was found using VBM in bilateral cerebellum, cingulum, left > right fronto-parietal cortex, right > left hippocampus and left caudate. FreeSurfer showed significant volume losses in subcortical GM, brainstem and cerebellum, and cortical thinning in the left insula. In the second year, only WMF decrease (−0.6%; p = 0.015) was observed with no VBM changes, although FreeSurfer detected significant volume loss in thalamus, hippocampus and cerebellum. Baseline gadolinium enhancement influenced WMF and BPF changes during the first year, but not GMF. Patients with confirmed Expanded Disability Status Scale (EDSS) worsening at 3 years had lower baseline GMF and left thalamus volume and greater BVL over follow-up. Conclusion: BVL develops mainly during the first year of natalizumab therapy. GM changes are independent of baseline inflammation and correlate with disability.
Collapse
Affiliation(s)
- Ethel Ciampi
- Servei de Neurologia/Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Deborah Pareto
- Servei de Radiologia, Unitat de Ressonància Magnètica, Institut de Diagnòstic per la Imatge (IDI), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jaume Sastre-Garriga
- Servei de Neurologia/Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Angela Vidal-Jordana
- Servei de Neurologia/Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carmen Tur
- Servei de Neurologia/Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jordi Río
- Servei de Neurologia/Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mar Tintoré
- Servei de Neurologia/Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Auger
- Servei de Radiologia, Unitat de Ressonància Magnètica, Institut de Diagnòstic per la Imatge (IDI), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Rovira
- Servei de Radiologia, Unitat de Ressonància Magnètica, Institut de Diagnòstic per la Imatge (IDI), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Montalban
- Servei de Neurologia/Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
102
|
|
103
|
Khalid F, Tauhid S, Chua AS, Healy BC, Stankiewicz JM, Weiner HL, Bakshi R. A longitudinal uncontrolled study of cerebral gray matter volume in patients receiving natalizumab for multiple sclerosis. Int J Neurosci 2016; 127:396-403. [PMID: 27143245 DOI: 10.1080/00207454.2016.1185421] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Brain atrophy in multiple sclerosis (MS) selectively affects gray matter (GM), which is highly relevant to disability and cognitive impairment. We assessed cerebral GM volume (GMV) during one year of natalizumab therapy. DESIGN/METHODS Patients with relapsing-remitting (n = 18) or progressive (n = 2) MS had MRI ∼1 year apart during natalizumab treatment. At baseline, patients were on natalizumab for (mean ± SD) 16.6 ± 10.9 months with age 38.5 ± 7.4 and disease duration 9.7 ± 4.3 years. RESULTS At baseline, GMV was 664.0 ± 56.4 ml, Expanded Disability Status Scale (EDSS) score was 2.3 ± 2.0, timed 25-foot walk (T25FW) was 6.1±3.4 s; two patients (10%) had gadolinium (Gd)-enhancing lesions. At follow-up, GMV was 663.9 ± 60.2 mL; EDSS was 2.6 ± 2.1 and T25FW was 5.9 ± 2.9 s. One patient had a mild clinical relapse during the observation period (0.052 annualized relapse rate for the entire cohort). No patients had Gd-enhancing lesions at follow-up. Linear mixed-effect models showed no significant change in annualized GMV [estimated mean change per year 0.338 mL, 95% confidence interval -9.66, 10.34, p = 0.94)], GM fraction (p = 0.92), whole brain parenchymal fraction (p = 0.64), T2 lesion load (p = 0.64), EDSS (p = 0.26) or T25FW (p = 0.79). CONCLUSIONS This pilot study shows no GM atrophy during one year of natalizumab MS therapy. We also did not detect any loss of whole brain volume or progression of cerebral T2 hyperintense lesion volume during the observation period. These MRI results paralleled the lack of clinical worsening.
Collapse
Affiliation(s)
- Fariha Khalid
- a a Laboratory for Neuroimaging Research, Department of Neurology, Brigham and Women's Hospital, Partners MS Center, Harvard Medical School , Boston, MA , USA
| | - Shahamat Tauhid
- a a Laboratory for Neuroimaging Research, Department of Neurology, Brigham and Women's Hospital, Partners MS Center, Harvard Medical School , Boston, MA , USA
| | - Alicia S Chua
- a a Laboratory for Neuroimaging Research, Department of Neurology, Brigham and Women's Hospital, Partners MS Center, Harvard Medical School , Boston, MA , USA
| | - Brian C Healy
- a a Laboratory for Neuroimaging Research, Department of Neurology, Brigham and Women's Hospital, Partners MS Center, Harvard Medical School , Boston, MA , USA.,c c Biostatistics Center, Massachusetts General Hospital, Harvard Medical School , Boston, MA , USA
| | - James M Stankiewicz
- a a Laboratory for Neuroimaging Research, Department of Neurology, Brigham and Women's Hospital, Partners MS Center, Harvard Medical School , Boston, MA , USA
| | - Howard L Weiner
- a a Laboratory for Neuroimaging Research, Department of Neurology, Brigham and Women's Hospital, Partners MS Center, Harvard Medical School , Boston, MA , USA
| | - Rohit Bakshi
- a a Laboratory for Neuroimaging Research, Department of Neurology, Brigham and Women's Hospital, Partners MS Center, Harvard Medical School , Boston, MA , USA.,b b Laboratory for Neuroimaging Research, Department of Radiology, Brigham and Women's Hospital, Partners MS Center, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
104
|
Zivadinov R, Hojnacki D, Bergsland N, Kennedy C, Hagemeier J, Melia R, Ramasamy DP, Durfee J, Carl E, Dwyer MG, Weinstock-Guttman B. Effect of natalizumab on brain atrophy and disability progression in multiple sclerosis patients over 5 years. Eur J Neurol 2016; 23:1101-9. [DOI: 10.1111/ene.12992] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/02/2016] [Indexed: 01/21/2023]
Affiliation(s)
- R. Zivadinov
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
- MR Imaging Clinical Translational Research Center; School of Medicine and Biomedical Sciences; University at Buffalo; State University of New York; Buffalo NY USA
| | - D. Hojnacki
- Jacobs MS Center; Department of Neurology; University at Buffalo; State University of New York; Buffalo NY USA
| | - N. Bergsland
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
- Magnetic Resonance Laboratory; IRCCS Don Gnocchi Foundation; Milan Italy
| | - C. Kennedy
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - J. Hagemeier
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - R. Melia
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - D. P. Ramasamy
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - J. Durfee
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - E. Carl
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - M. G. Dwyer
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - B. Weinstock-Guttman
- Jacobs MS Center; Department of Neurology; University at Buffalo; State University of New York; Buffalo NY USA
| |
Collapse
|
105
|
Affiliation(s)
- Benjamin M Segal
- Department of Neurology, University of Michigan, MI, USA; Neurology Section, VA Ann Arbor Health Care Systems, Ann Arbor, MI, USA
| | - Olaf Stüve
- Neurology Section, VA North Texas Health Care System, Medical Service Dallas, VA Medical Center, Dallas, TX, 75216, USA; Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA; Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
| |
Collapse
|
106
|
Wiebenga OT, Schoonheim MM, Hulst HE, Nagtegaal GJA, Strijbis EMM, Steenwijk MD, Polman CH, Pouwels PJW, Barkhof F, Geurts JJG. White Matter Diffusion Changes during the First Year of Natalizumab Treatment in Relapsing-Remitting Multiple Sclerosis. AJNR Am J Neuroradiol 2016; 37:1030-7. [PMID: 26965463 DOI: 10.3174/ajnr.a4690] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/12/2015] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND PURPOSE Natalizumab treatment strongly affects relapsing-remitting multiple sclerosis, possibly by restraining white matter damage. This study investigated changes in white matter diffusivity in patients with relapsing-remitting multiple sclerosis during their first year of natalizumab treatment by using diffusion tensor imaging. MATERIALS AND METHODS The study included patients with relapsing-remitting multiple sclerosis initiating natalizumab at baseline (n = 22), patients with relapsing-remitting multiple sclerosis continuing interferon-β or glatiramer acetate (n = 17), and healthy controls (n = 12). Diffusion tensor imaging parameters were analyzed at baseline and month 12. We measured the extent and severity of white matter damage with diffusion tensor imaging parameters such as fractional anisotropy, comparing the patient groups with healthy controls at both time points. RESULTS The extent and severity of white matter damage were reduced significantly in the natalizumab group with time (fractional anisotropy-based extent, 56.8% to 47.2%; severity, z = -0.67 to -0.59; P = .02); this reduction was not observed in the interferon-β/glatiramer acetate group (extent, 41.4% to 39.1%, and severity, z = -0.64 to -0.67; P = .94). Cognitive performance did not change with time in the patient groups but did correlate with the severity of damage (r = 0.53, P = < .001). CONCLUSIONS In patients with relapsing-remitting multiple sclerosis starting natalizumab treatment, the extent and severity of white matter damage were reduced significantly in the first year of treatment. These findings may aid in explaining the large observed clinical effect of natalizumab in relapsing-remitting multiple sclerosis.
Collapse
Affiliation(s)
- O T Wiebenga
- From the Departments of Radiology and Nuclear Medicine (O.T.W., G.J.A.N., M.D.S., F.B.) Anatomy and Neurosciences (O.T.W., M.M.S., H.E.H., G.J.A.N., J.J.G.G.)
| | - M M Schoonheim
- Anatomy and Neurosciences (O.T.W., M.M.S., H.E.H., G.J.A.N., J.J.G.G.)
| | - H E Hulst
- Anatomy and Neurosciences (O.T.W., M.M.S., H.E.H., G.J.A.N., J.J.G.G.)
| | - G J A Nagtegaal
- From the Departments of Radiology and Nuclear Medicine (O.T.W., G.J.A.N., M.D.S., F.B.) Anatomy and Neurosciences (O.T.W., M.M.S., H.E.H., G.J.A.N., J.J.G.G.)
| | | | - M D Steenwijk
- From the Departments of Radiology and Nuclear Medicine (O.T.W., G.J.A.N., M.D.S., F.B.)
| | | | - P J W Pouwels
- Physics and Medical Technology (P.J.W.P.), Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - F Barkhof
- From the Departments of Radiology and Nuclear Medicine (O.T.W., G.J.A.N., M.D.S., F.B.)
| | - J J G Geurts
- Anatomy and Neurosciences (O.T.W., M.M.S., H.E.H., G.J.A.N., J.J.G.G.)
| |
Collapse
|
107
|
Ley K, Rivera-Nieves J, Sandborn WJ, Shattil S. Integrin-based therapeutics: biological basis, clinical use and new drugs. Nat Rev Drug Discov 2016; 15:173-83. [PMID: 26822833 PMCID: PMC4890615 DOI: 10.1038/nrd.2015.10] [Citation(s) in RCA: 303] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Integrins are activatable molecules that are involved in adhesion and signalling. Of the 24 known human integrins, 3 are currently targeted therapeutically by monoclonal antibodies, peptides or small molecules: drugs targeting the platelet αIIbβ3 integrin are used to prevent thrombotic complications after percutaneous coronary interventions, and compounds targeting the lymphocyte α4β1 and α4β7 integrins have indications in multiple sclerosis and inflammatory bowel disease. New antibodies and small molecules targeting β7 integrins (α4β7 and αEβ7 integrins) and their ligands are in clinical development for the treatment of inflammatory bowel diseases. Integrin-based therapeutics have shown clinically significant benefits in many patients, leading to continued medical interest in the further development of novel integrin inhibitors. Of note, almost all integrin antagonists in use or in late-stage clinical trials target either the ligand-binding site or the ligand itself.
Collapse
Affiliation(s)
- Klaus Ley
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle Drive, La Jolla, Califoria 92037, USA, and the Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093 USA
| | - Jesus Rivera-Nieves
- La Jolla Institute for Allergy and the Immunology, 9420 Athena Circle Drive, La Jolla, Califoria 92037, USA, and the Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093 USA
| | - William J Sandborn
- Immunology and the Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093 USA
| | - Sanford Shattil
- Division of Haematology-Oncology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093 USA
| |
Collapse
|
108
|
Barbin L, Rousseau C, Jousset N, Casey R, Debouverie M, Vukusic S, De Sèze J, Brassat D, Wiertlewski S, Brochet B, Pelletier J, Vermersch P, Edan G, Lebrun-Frenay C, Clavelou P, Thouvenot E, Camdessanché JP, Tourbah A, Stankoff B, Al Khedr A, Cabre P, Papeix C, Berger E, Heinzlef O, Debroucker T, Moreau T, Gout O, Bourre B, Créange A, Labauge P, Magy L, Defer G, Foucher Y, Laplaud DA. Comparative efficacy of fingolimod vs natalizumab: A French multicenter observational study. Neurology 2016; 86:771-8. [PMID: 26826205 PMCID: PMC4763805 DOI: 10.1212/wnl.0000000000002395] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 10/29/2015] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To compare natalizumab and fingolimod on both clinical and MRI outcomes in patients with relapsing-remitting multiple sclerosis (RRMS) from 27 multiple sclerosis centers participating in the French follow-up cohort Observatoire of Multiple Sclerosis. METHODS Patients with RRMS included in the study were aged from 18 to 65 years with an Expanded Disability Status Scale score of 0-5.5 and an available brain MRI performed within the year before treatment initiation. The data were collected for 326 patients treated with natalizumab and 303 with fingolimod. The statistical analysis was performed using 2 different methods: logistic regression and propensity scores (inverse probability treatment weighting). RESULTS The confounder-adjusted proportion of patients with at least one relapse within the first and second year of treatment was lower in natalizumab-treated patients compared to the fingolimod group (21.1% vs 30.4% at first year, p = 0.0092; and 30.9% vs 41.7% at second year, p = 0.0059) and supported the trend observed in nonadjusted analysis (21.2% vs 27.1% at 1 year, p = 0.0775). Such statistically significant associations were also observed for gadolinium (Gd)-enhancing lesions and new T2 lesions at both 1 year (Gd-enhancing lesions: 9.3% vs 29.8%, p < 0.0001; new T2 lesions: 10.6% vs 29.6%, p < 0.0001) and 2 years (Gd-enhancing lesions: 9.1% vs 22.1%, p = 0.0025; new T2 lesions: 16.9% vs 34.1%, p = 0.0010) post treatment initiation. CONCLUSION Taken together, these results suggest the superiority of natalizumab over fingolimod to prevent relapses and new T2 and Gd-enhancing lesions at 1 and 2 years. CLASSIFICATION OF EVIDENCE This study provides Class IV evidence that for patients with RRMS, natalizumab decreases the proportion of patients with at least one relapse within the first year of treatment compared to fingolimod.
Collapse
Affiliation(s)
| | - Chloe Rousseau
- Authors' affiliations are listed at the end of the article
| | | | - Romain Casey
- Authors' affiliations are listed at the end of the article
| | | | - Sandra Vukusic
- Authors' affiliations are listed at the end of the article
| | - Jerome De Sèze
- Authors' affiliations are listed at the end of the article
| | - David Brassat
- Authors' affiliations are listed at the end of the article
| | | | - Bruno Brochet
- Authors' affiliations are listed at the end of the article
| | - Jean Pelletier
- Authors' affiliations are listed at the end of the article
| | | | - Gilles Edan
- Authors' affiliations are listed at the end of the article
| | | | | | - Eric Thouvenot
- Authors' affiliations are listed at the end of the article
| | | | - Ayman Tourbah
- Authors' affiliations are listed at the end of the article
| | - Bruno Stankoff
- Authors' affiliations are listed at the end of the article
| | | | - Philippe Cabre
- Authors' affiliations are listed at the end of the article
| | | | - Eric Berger
- Authors' affiliations are listed at the end of the article
| | | | | | | | - Olivier Gout
- Authors' affiliations are listed at the end of the article
| | | | - Alain Créange
- Authors' affiliations are listed at the end of the article
| | - Pierre Labauge
- Authors' affiliations are listed at the end of the article
| | - Laurent Magy
- Authors' affiliations are listed at the end of the article
| | - Gilles Defer
- Authors' affiliations are listed at the end of the article
| | - Yohann Foucher
- Authors' affiliations are listed at the end of the article
| | | | | |
Collapse
|
109
|
Sellebjerg F, Cadavid D, Steiner D, Villar LM, Reynolds R, Mikol D. Exploring potential mechanisms of action of natalizumab in secondary progressive multiple sclerosis. Ther Adv Neurol Disord 2016; 9:31-43. [PMID: 26788129 DOI: 10.1177/1756285615615257] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Multiple sclerosis (MS) is a common and chronic central nervous system (CNS) demyelinating disease and a leading cause of permanent disability. Patients most often present with a relapsing-remitting disease course, typically progressing over time to a phase of relentless advancement in secondary progressive MS (SPMS), for which approved disease-modifying therapies are limited. In this review, we summarize the pathophysiological mechanisms involved in the development of SPMS and the rationale and clinical potential for natalizumab, which is currently approved for the treatment of relapsing forms of MS, to exert beneficial effects in reducing disease progression unrelated to relapses in SPMS. In both forms of MS, active brain-tissue injury is associated with inflammation; but in SPMS, the inflammatory response occurs at least partly behind the blood-brain barrier and is followed by a cascade of events, including persistent microglial activation that may lead to chronic demyelination and neurodegeneration associated with irreversible disability. In patients with relapsing forms of MS, natalizumab therapy is known to significantly reduce intrathecal inflammatory responses which results in reductions in brain lesions and brain atrophy as well as beneficial effects on clinical measures, such as reduced frequency and severity of relapse and reduced accumulation of disability. Natalizumab treatment also reduces levels of cerebrospinal fluid chemokines and other biomarkers of intrathecal inflammation, axonal damage and demyelination, and has demonstrated the ability to reduce innate immune activation and intrathecal immunoglobulin synthesis in patients with MS. The efficacy of natalizumab therapy in SPMS is currently being investigated in a randomized, double-blind, placebo-controlled trial.
Collapse
Affiliation(s)
- Finn Sellebjerg
- Danish Multiple Sclerosis Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Luisa Maria Villar
- Department of Immunology, Ramón y Cajal University Hospital, Institute Ramón y Cajal for Biomedical Research, Madrid, Spain
| | - Richard Reynolds
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| | | |
Collapse
|
110
|
Arpín EC, Sobrino TG, Vivero CD, del Campo Amigo Jorrín M, Regal AR, González JP, Bouzas ML. Changes in brain atrophy indices in patients with relapsing-remitting multiple sclerosis treated with natalizumab. Neurodegener Dis Manag 2016; 6:5-12. [PMID: 26782312 DOI: 10.2217/nmt.15.53] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM To evaluate the effect of natalizumab on progression of brain atrophy in multiple sclerosis (MS) patients and to search for a clinical or radiological marker of progression of brain atrophy. PATIENTS AND METHODS We retrospectively recorded demographic and clinical data, as well as the corpus callosum index (CCI) using MRI, in MS patients treated with natalizumab for 1-4 years. RESULTS In the study population (n = 29), baseline mean CCI was 0.37 ± 0.04 and final CCI 0.36 ± 0.04. 17 patients did not develop brain atrophy during follow-up. There was no statistically significant relationship between progression of atrophy and clinical and radiological parameters. CONCLUSION Natalizumab may have a neuroprotective effect.
Collapse
Affiliation(s)
- Eva Costa Arpín
- Department of Neurology, Hospital Clínico Universitario, Santiago de Compostela, Spain
| | - Tania García Sobrino
- Department of Neurology, Hospital Clínico Universitario, Santiago de Compostela, Spain
| | | | | | - Ana Rodríguez Regal
- Department of Neurology, Complejo Hospitalario de Pontevedra, Pontevedra, Spain
| | - José Prieto González
- Department of Neurology, Hospital Clínico Universitario, Santiago de Compostela, Spain
| | - Manuela Lema Bouzas
- Department of Neurology, Hospital Clínico Universitario, Santiago de Compostela, Spain
| |
Collapse
|
111
|
Ziemssen T, Derfuss T, de Stefano N, Giovannoni G, Palavra F, Tomic D, Vollmer T, Schippling S. Optimizing treatment success in multiple sclerosis. J Neurol 2015; 263:1053-65. [PMID: 26705122 PMCID: PMC4893374 DOI: 10.1007/s00415-015-7986-y] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 11/25/2015] [Indexed: 01/01/2023]
Abstract
Despite important advances in the treatment of multiple sclerosis (MS) over recent years, the introduction of several disease-modifying therapies (DMTs), the burden of progressive disability and premature mortality associated with the condition remains substantial. This burden, together with the high healthcare and societal costs associated with MS, creates a compelling case for early treatment optimization with highly efficacious therapies. Often, patients receive several first-line therapies, while more recent and in part more effective treatments are still being introduced only after these have failed. However, with the availability of highly efficacious therapies, a novel treatment strategy has emerged, where the aim is to achieve no evidence of disease activity (NEDA). Achieving NEDA necessitates regular monitoring of relapses, disability and functionality. However, there is only a poor correlation between conventional magnetic resonance imaging measures like T2 hyperintense lesion burden and the level of clinical disability. Hence, MRI-based measures of brain atrophy have emerged in recent years potentially reflecting the magnitude of MS-related neuroaxonal damage. Currently available DMTs differ markedly in their effects on brain atrophy: some, such as fingolimod, have been shown to significantly slow brain volume loss, compared to placebo, whereas others have shown either no, inconsistent, or delayed effects. In addition to regular monitoring, treatment optimization also requires early intervention with efficacious therapies, because accumulating evidence shows that effective intervention during a limited period early in the course of MS is critical for maintaining neurological function and preventing subsequent disability. Together, the advent of new MS therapies and evolving management strategies offer exciting new opportunities to optimize treatment outcomes.
Collapse
Affiliation(s)
- Tjalf Ziemssen
- MS Center Dresden, Center of Clinical Neuroscience, Neurological Clinic, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstrasse 74, 01307, Dresden, Germany.
| | - Tobias Derfuss
- MS Center Dresden, Center of Clinical Neuroscience, Neurological Clinic, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Nicola de Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Gavin Giovannoni
- Queen Mary University London, Barts and The London School of Medicine and Dentistry, London, UK
| | - Filipe Palavra
- Neurology-Neuroimmunology Department, Multiple Sclerosis Centre of Catalonia (Cemcat), Vall d'Hebron University Hospital, Barcelona, Spain
| | | | - Tim Vollmer
- University of Colorado Health Sciences Center, Aurora, CO, USA
| | - Sven Schippling
- Department of Neurology, Neuroimmunology and Multiple Sclerosis Research, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
112
|
Abstract
AbstractMultiple sclerosis is a chronic demyelinating disease characterized by focal and diffuse inflammation of the central nervous system resulting in significant physical and cognitive disabilities. Disease-modifying therapies targeting the dysfunctional immune response are most effective in the first few years after disease onset, indicating that there is a limited time window for therapy to influence the disease course. No evidence of disease activity is emerging as a new standard for treatment response and may be associated with improved long-term disability outcomes. An aggressive management strategy, including earlier use of more potent immunomodulatory agents and close monitoring of the clinical and radiologic response to treatment, is recommended to minimize early brain volume loss and slow the progression of physical and cognitive impairments in patients with relapsing-remitting multiple sclerosis.
Collapse
|
113
|
Lycke J. Monoclonal antibody therapies for the treatment of relapsing-remitting multiple sclerosis: differentiating mechanisms and clinical outcomes. Ther Adv Neurol Disord 2015; 8:274-93. [PMID: 26600872 PMCID: PMC4643868 DOI: 10.1177/1756285615605429] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Monoclonal antibody (mAb) therapies for relapsing-remitting multiple sclerosis (MS) target immune cells or other molecules involved in pathogenic pathways with extraordinary specificity. Natalizumab and alemtuzumab are the only two currently approved mAbs for the treatment of MS, having demonstrated significant reduction in clinical and magnetic resonance imaging disease activity and disability in clinical studies. Ocrelizumab and daclizumab are in the late stages of phase III trials, and several other mAbs are in the early stages of clinical evaluation. mAbs have distinct structural characteristics (e.g. chimeric, humanized, fully human) and unique targets (e.g. blocking interactions, induction of signal transduction by receptor binding, complement-dependent cytotoxicity, antibody-dependent cell-mediated cytotoxicity) conferring different mechanisms of action in MS. Because of these differences, mAbs for MS do not constitute a single treatment class; each must be considered individually when selecting appropriate therapy. Furthermore, in reviewing the data from clinical studies of mAbs, attention should be drawn to use of different comparators (e.g. placebo or interferon β-1a) and study designs. Each mAb treatment has a unique administration schedule. In the decision to select the appropriate treatment for each individual MS patient, careful review of the benefits relative to risks of mAbs is balanced against the risk of development of MS-associated disability.
Collapse
Affiliation(s)
- Jan Lycke
- Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| |
Collapse
|
114
|
Vision in a phase 3 trial of natalizumab for multiple sclerosis: relation to disability and quality of life. J Neuroophthalmol 2015; 35:6-11. [PMID: 25370598 PMCID: PMC4337583 DOI: 10.1097/wno.0000000000000173] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Supplemental Digital Content is Available in the Text. Background: Low-contrast visual acuity (LCVA), a sensitive measure of visual function in multiple sclerosis (MS), demonstrated treatment effects as a secondary outcome measure in the Phase 3 trial of natalizumab, AFFIRM. In these posttrial analyses, we studied the relation of visual function to quality of life (QOL), magnetic resonance imaging (MRI) measures, and Expanded Disability Status Scale (EDSS) scores. Methods: At baseline and at 52 and 104 weeks in AFFIRM, patients underwent binocular testing of LCVA (1.25% and 2.5% contrast) and high-contrast visual acuity (HCVA). Vision-specific QOL was assessed by the Impact of Visual Impairment Scale (IVIS), whereas the SF-36 Health Survey and Visual Analog Scale were administered as generic QOL measures and the EDSS as a measure of neurologic impairment. Results: Among QOL measures, IVIS scores showed the most significant correlations with visual dysfunction at all time points in the trial (r= −0.25 to −0.45, P < 0.0001 for LCVA and HCVA). Higher MRI T1- and T2-lesion volumes were also associated with worse vision scores at all time points (P < 0.0001). Clinically meaningful worsening (progression) of LCVA was noted in substantial proportions of patients in AFFIRM and was prevalent even among those without EDSS progression over 2 years (21.9% with LCVA progression at 2.5% contrast; 26.2% at 1.25% contrast). HCVA worsened in only 3.7% of patients without EDSS progression. Conclusions: Loss of visual function, particularly as measured by LCVA, was common in AFFIRM, occurring in >20% of patients. Both LCVA and HCVA scores reflect vision-specific aspects of QOL, but LCVA provides information about disability progression not entirely captured by the EDSS. Vision represents a key dimension of outcome assessment for MS and adds valuable information on disability and QOL that can be useful to clinicians.
Collapse
|
115
|
Vitaliti G, Matin N, Tabatabaie O, Di Traglia M, Pavone P, Lubrano R, Falsaperla R. Natalizumab in multiple sclerosis: discontinuation, progressive multifocal leukoencephalopathy and possible use in children. Expert Rev Neurother 2015; 15:1321-1341. [PMID: 26513633 DOI: 10.1586/14737175.2015.1102061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In the early 1990s, attention was drawn to the migration of immune cells into the central nervous system via the blood-brain barrier. The literature showed that lymphocytes binding to the endothelium were successfully inhibited by an antibody against α4β1 integrin. These biological findings resulted in the development of a humanized antibody to α4 integrin - natalizumab (NTZ) - to treat multiple sclerosis (MS). Here, we provide a systematic review and meta-analysis on the efficacy and safety of natalizumab, trying to answer the question whether its use may be recommended both in adult and in pediatric age groups as standard MS treatment. Our results highlight the improvement of clinical and radiological findings in treated patients (p < 0.005), confirming NTZ efficacy. Nevertheless, if NTZ is shown to be efficient, further studies should be performed to evaluate its safety and to target the MS profile that could benefit from this treatment.
Collapse
Affiliation(s)
- Giovanna Vitaliti
- a General Paediatrics Operative Unit , Policlinico-Vittorio-Emanuele University Hospital, University of Catania , Catania , Italy
| | - Nassim Matin
- b Tehran University of Medical Sciences , Tehran , Iran
| | | | - Mario Di Traglia
- c Department of Statistics , La Sapienza University of Rome , Rome , Italy
| | - Piero Pavone
- a General Paediatrics Operative Unit , Policlinico-Vittorio-Emanuele University Hospital, University of Catania , Catania , Italy
| | - Riccardo Lubrano
- d Paediatric Department, Paediatric Nephrology Operative Unit , Sapienza University of Rome , Rome , Italy
| | - Raffaele Falsaperla
- a General Paediatrics Operative Unit , Policlinico-Vittorio-Emanuele University Hospital, University of Catania , Catania , Italy
| |
Collapse
|
116
|
Design and development of an ethnically-diverse imaging informatics-based eFolder system for multiple sclerosis patients. Comput Med Imaging Graph 2015; 46 Pt 2:257-68. [PMID: 26564667 DOI: 10.1016/j.compmedimag.2015.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 09/06/2015] [Accepted: 09/29/2015] [Indexed: 11/24/2022]
Abstract
PURPOSE MRI has been used to identify multiple sclerosis (MS) lesions in brain and spinal cord visually. Integrating patient information into an electronic patient record system has become key for modern patient care in medicine in recent years. Clinically, it is also necessary to track patients' progress in longitudinal studies, in order to provide comprehensive understanding of disease progression and response to treatment. As the amount of required data increases, there exists a need for an efficient systematic solution to store and analyze MS patient data, disease profiles, and disease tracking for both clinical and research purposes. METHOD An imaging informatics based system, called MS eFolder, has been developed as an integrated patient record system for data storage and analysis of MS patients. The eFolder system, with a DICOM-based database, includes a module for lesion contouring by radiologists, a MS lesion quantification tool to quantify MS lesion volume in 3D, brain parenchyma fraction analysis, and provide quantitative analysis and tracking of volume changes in longitudinal studies. Patient data, including MR images, have been collected retrospectively at University of Southern California Medical Center (USC) and Los Angeles County Hospital (LAC). The MS eFolder utilizes web-based components, such as browser-based graphical user interface (GUI) and web-based database. The eFolder database stores patient clinical data (demographics, MS disease history, family history, etc.), MR imaging-related data found in DICOM headers, and lesion quantification results. Lesion quantification results are derived from radiologists' contours on brain MRI studies and quantified into 3-dimensional volumes and locations. Quantified results of white matter lesions are integrated into a structured report based on DICOM-SR protocol and templates. The user interface displays patient clinical information, original MR images, and viewing structured reports of quantified results. The GUI also includes a data mining tool to handle unique search queries for MS. System workflow and dataflow steps has been designed based on the IHE post-processing workflow profile, including workflow process tracking, MS lesion contouring and quantification of MR images at a post-processing workstation, and storage of quantitative results as DICOM-SR in DICOM-based storage system. The web-based GUI is designed to display zero-footprint DICOM web-accessible data objects (WADO) and the SR objects. SUMMARY The MS eFolder system has been designed and developed as an integrated data storage and mining solution in both clinical and research environments, while providing unique features, such as quantitative lesion analysis and disease tracking over a longitudinal study. A comprehensive image and clinical data integrated database provided by MS eFolder provides a platform for treatment assessment, outcomes analysis and decision-support. The proposed system serves as a platform for future quantitative analysis derived automatically from CAD algorithms that can also be integrated within the system for individual disease tracking and future MS-related research. Ultimately the eFolder provides a decision-support infrastructure that can eventually be used as add-on value to the overall electronic medical record.
Collapse
|
117
|
Zare-Shahabadi A, Rashidian A, Sahraian MA, Rezaei N. Potential role of immunoglobulin replacement therapy on MRI measures in multiple sclerosis: a systematic review. Expert Rev Clin Immunol 2015; 11:1291-5. [PMID: 26488634 DOI: 10.1586/1744666x.2015.1102059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Multiple sclerosis is a chronic immune-mediated disease of the nervous system. In the early disease course, axonal loss and neurodegeneration occurs that could possibly lead to irreversible neurological impairments. Preventing brain atrophy may have important clinical implications affecting treatment decisions in the future. In recent years, research efforts have directed towards finding agents to modify the disease and reduce brain volume loss. Intravenous immunoglobulin (IVIg) may have some potential roles in this regard.
Collapse
Affiliation(s)
- Ameneh Zare-Shahabadi
- a Molecular Immunology Research Center, Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,b MS Research Center, Neuroscience Institute , Tehran University of Medical Sciences , Tehran , Iran.,c Psychiatry and Psychology Research Center , Tehran University of Medical Sciences , Tehran , Iran
| | - Arash Rashidian
- d Department of Health Management and Economics, School of Public Health , Tehran University of Medical Sciences , Tehran , Iran.,e Knowledge Utilization Research Center , Tehran University of Medical Sciences , Tehran , Iran
| | - Mohammad Ali Sahraian
- b MS Research Center, Neuroscience Institute , Tehran University of Medical Sciences , Tehran , Iran
| | - Nima Rezaei
- a Molecular Immunology Research Center, Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,f Universal Scientific Education and Research Network (USERN) , Tehran , Iran.,g Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center , Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
118
|
Neuroimaging of Natalizumab Complications in Multiple Sclerosis: PML and Other Associated Entities. Mult Scler Int 2015; 2015:809252. [PMID: 26483978 PMCID: PMC4592919 DOI: 10.1155/2015/809252] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/14/2015] [Accepted: 08/31/2015] [Indexed: 12/19/2022] Open
Abstract
Natalizumab (Tysabri) is a monoclonal antibody (α4 integrin antagonist) approved for treatment of multiple sclerosis, both for patients who fail therapy with other disease modifying agents and for patients with aggressive disease. Natalizumab is highly effective, resulting in significant decreases in rates of both relapse and disability accumulation, as well as marked decrease in MRI evidence of disease activity. As such, utilization of natalizumab is increasing, and the presentation of its associated complications is increasing accordingly. This review focuses on the clinical and neuroimaging features of the major complications associated with natalizumab therapy, focusing on the rare but devastating progressive multifocal leukoencephalopathy (PML). Associated entities including PML associated immune reconstitution inflammatory syndrome (PML-IRIS) and the emerging phenomenon of rebound of MS disease activity after natalizumab discontinuation are also discussed. Early recognition of neuroimaging features associated with these processes is critical in order to facilitate prompt diagnosis, treatment, and/or modification of therapies to improve patient outcomes.
Collapse
|
119
|
Ziemssen T, De Stefano N, Sormani MP, Van Wijmeersch B, Wiendl H, Kieseier BC. Optimizing therapy early in multiple sclerosis: An evidence-based view. Mult Scler Relat Disord 2015; 4:460-469. [DOI: 10.1016/j.msard.2015.07.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 06/01/2015] [Accepted: 07/15/2015] [Indexed: 01/26/2023]
|
120
|
Subei AM, Ontaneda D. Risk Mitigation Strategies for Adverse Reactions Associated with the Disease-Modifying Drugs in Multiple Sclerosis. CNS Drugs 2015; 29:759-71. [PMID: 26407624 PMCID: PMC4621807 DOI: 10.1007/s40263-015-0277-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Over the past several years, the number of disease-modifying therapies (DMTs) for the treatment of multiple sclerosis (MS) has doubled in number. The 13 approved agents have shown a wide range of efficacy and safety in their clinical trials and post-marketing experience. While the availability of the newer agents allows for a wider selection of therapy for clinicians and patients, there is a need for careful understanding of the benefits and risks of each agent. Several factors such as the medication efficacy, side-effect profile, patient's preference, and co-morbidities need to be considered. An individualized treatment approach is thus imperative. In this review, risk stratification and mitigation strategies of the various disease-modifying agents are discussed.
Collapse
Affiliation(s)
- Adnan M Subei
- Mellen Center for MS Treatment and Research, Cleveland Clinic Foundation, 9500 Euclid Avenue/U10, Cleveland, OH, 44195, USA.
| | - Daniel Ontaneda
- Mellen Center for MS Treatment and Research, Cleveland Clinic Foundation, 9500 Euclid Avenue/U10, Cleveland, OH, 44195, USA.
| |
Collapse
|
121
|
Tanaka M. [Natalizumab treatment in multiple sclerosis]. Rinsho Shinkeigaku 2015; 55:537-43. [PMID: 26156254 DOI: 10.5692/clinicalneurol.cn-000690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Multiple sclerosis is a neurodegenerative and inflammatory immune disorder in the central nervous system (CNS). Most patients show relapse-remitting clinical course. Some disease modifying medications for preventing the relapses including natalizumab (NTZ) are approved in Japan. Natalizumab (NTZ), a monoclonal antibody against α4 integrin present on the surface of lymphocytes, inhibits the binding of lymphocytes to vascular cell adhesion molecule-1 (VCAM-1) of endothelial cells and blocks the penetration of encephalitogenic T cells into the CNS. This review provides an update on the efficacy of an inhibition of relapses, adverse effects including progressive multifocal leukoencephalopathy, treatment after NTZ discontinuation, and body weight based treatment.
Collapse
Affiliation(s)
- Masami Tanaka
- Multiple Sclerosis Center, National Hospital Organization, Utano National Hospital
| |
Collapse
|
122
|
Vidal-Jordana A, Sastre-Garriga J, Rovira A, Montalban X. Treating relapsing-remitting multiple sclerosis: therapy effects on brain atrophy. J Neurol 2015; 262:2617-26. [PMID: 26041617 DOI: 10.1007/s00415-015-7798-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 05/22/2015] [Accepted: 05/25/2015] [Indexed: 01/06/2023]
Abstract
Multiple sclerosis (MS) is an immune-mediated disease of the central nervous system with a complex and heterogeneous pathology that may ultimately lead to neurodegeneration and brain atrophy. Brain volume loss in MS is known to occur early in the disease course and to be clinically relevant, as it has been related to disability progression. Nowadays, brain volume loss is relatively easy to measure with different automated, reproducible and accurate software tools. Therefore, most of (if not all) the newest clinical trials have incorporated brain volume outcomes as a measure of treatment effect. With this review, we aimed to update and summarize all existing data regarding brain volume and RRMS treatment in clinical trials as well as in open-label observational studies of drugs with positive results in its primary outcome in at least one phase III trial as of March 2014.
Collapse
Affiliation(s)
- Angela Vidal-Jordana
- Department of Neurology-Neuroimmunology and Multiple Sclerosis Centre of Catalonia (Cemcat), Edifici Cemcat, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, P. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Jaume Sastre-Garriga
- Department of Neurology-Neuroimmunology and Multiple Sclerosis Centre of Catalonia (Cemcat), Edifici Cemcat, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, P. Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| | - Alex Rovira
- Magnetic Resonance Unit, Radiology Department, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Montalban
- Department of Neurology-Neuroimmunology and Multiple Sclerosis Centre of Catalonia (Cemcat), Edifici Cemcat, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, P. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| |
Collapse
|
123
|
Nakamura K, Brown RA, Narayanan S, Collins DL, Arnold DL. Diurnal fluctuations in brain volume: Statistical analyses of MRI from large populations. Neuroimage 2015; 118:126-32. [PMID: 26049148 DOI: 10.1016/j.neuroimage.2015.05.077] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 05/10/2015] [Accepted: 05/26/2015] [Indexed: 01/18/2023] Open
Abstract
We investigated fluctuations in brain volume throughout the day using statistical modeling of magnetic resonance imaging (MRI) from large populations. We applied fully automated image analysis software to measure the brain parenchymal fraction (BPF), defined as the ratio of the brain parenchymal volume and intracranial volume, thus accounting for variations in head size. The MRI data came from serial scans of multiple sclerosis (MS) patients in clinical trials (n=755, 3269 scans) and from subjects participating in the Alzheimer's Disease Neuroimaging Initiative (ADNI, n=834, 6114 scans). The percent change in BPF was modeled with a linear mixed effect (LME) model, and the model was applied separately to the MS and ADNI datasets. The LME model for the MS datasets included random subject effects (intercept and slope over time) and fixed effects for the time-of-day, time from the baseline scan, and trial, which accounted for trial-related effects (for example, different inclusion criteria and imaging protocol). The model for ADNI additionally included the demographics (baseline age, sex, subject type [normal, mild cognitive impairment, or Alzheimer's disease], and interaction between subject type and time from baseline). There was a statistically significant effect of time-of-day on the BPF change in MS clinical trial datasets (-0.180 per day, that is, 0.180% of intracranial volume, p=0.019) as well as the ADNI dataset (-0.438 per day, that is, 0.438% of intracranial volume, p<0.0001), showing that the brain volume is greater in the morning. Linearly correcting the BPF values with the time-of-day reduced the required sample size to detect a 25% treatment effect (80% power and 0.05 significance level) on change in brain volume from 2 time-points over a period of 1year by 2.6%. Our results have significant implications for future brain volumetric studies, suggesting that there is a potential acquisition time bias that should be randomized or statistically controlled to account for the day-to-day brain volume fluctuations.
Collapse
Affiliation(s)
- Kunio Nakamura
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada; Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, ND20, Cleveland, OH 44195, USA.
| | - Robert A Brown
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Sridar Narayanan
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada; NeuroRx Research, 3575 Park Avenue, Suite #5322, Montreal, Quebec H2X 4B3, Canada
| | - D Louis Collins
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Douglas L Arnold
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada; NeuroRx Research, 3575 Park Avenue, Suite #5322, Montreal, Quebec H2X 4B3, Canada
| | | |
Collapse
|
124
|
|
125
|
Giovannoni G, Turner B, Gnanapavan S, Offiah C, Schmierer K, Marta M. Is it time to target no evident disease activity (NEDA) in multiple sclerosis? Mult Scler Relat Disord 2015. [PMID: 26195051 DOI: 10.1016/j.msard.2015.04.006] [Citation(s) in RCA: 281] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The management of multiple sclerosis is becoming increasingly complex with the emergence of new and more effective disease-modifying therapies (DMT). We propose a new treatment paradigm that individualises treatment based on a choice between two interchangeable therapeutic strategies of maintenance-escalation or induction therapy. We propose treating- to-target of no evident disease activity (NEDA) as defined using clinical and MRI criteria. This algorithm requires active monitoring with a rebaselining MRI, at a point in time after the specific DMT concerned has had sufficient time to work, and at least annual MRI studies to monitor for subclinical relapses. Disease activity on the maintenance-escalation therapy arm of the algorithm indicates a sub-optimal treatment response and should trigger a discussion about switching, or escalating, therapy or the consideration of switching to the induction therapy arm of the algorithm. In comparison, disease activity on an induction therapy arm would be an indication for retreatment or a switch to the maintenance-escalation therapy arm. We envisage the definition of NEDA evolving with time as new technological innovations are adopted into clinical practice, for example the normalisation of whole, or regional, brain atrophy rates and cerebrospinal fluid neurofilament levels.
Collapse
Affiliation(s)
- Gavin Giovannoni
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, 4 Newark Street, London E1 2AT, UK; Department of Neurology, Royal London Hospital, Barts Health NHS Trust, London, UK.
| | - Benjamin Turner
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, 4 Newark Street, London E1 2AT, UK; Department of Neurology, Royal London Hospital, Barts Health NHS Trust, London, UK.
| | - Sharmilee Gnanapavan
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, 4 Newark Street, London E1 2AT, UK; Department of Neurology, Royal London Hospital, Barts Health NHS Trust, London, UK.
| | - Curtis Offiah
- Department of Neuroradiology, Royal London Hospital, Barts Health NHS Trust, London, UK.
| | - Klaus Schmierer
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, 4 Newark Street, London E1 2AT, UK; Department of Neurology, Royal London Hospital, Barts Health NHS Trust, London, UK.
| | - Monica Marta
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, 4 Newark Street, London E1 2AT, UK; Department of Neurology, Royal London Hospital, Barts Health NHS Trust, London, UK.
| |
Collapse
|
126
|
Freedman MS, Abdoli M. Evaluating response to disease-modifying therapy in relapsing multiple sclerosis. Expert Rev Neurother 2015; 15:407-23. [DOI: 10.1586/14737175.2015.1023711] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
127
|
Mattoscio M, Nicholas R, Sormani MP, Malik O, Lee JS, Waldman AD, Dazzi F, Muraro PA. Hematopoietic mobilization: Potential biomarker of response to natalizumab in multiple sclerosis. Neurology 2015; 84:1473-82. [PMID: 25762712 DOI: 10.1212/wnl.0000000000001454] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/22/2014] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To ascertain the mobilization from the bone marrow and the functional relevance of the increased number of circulating hematopoietic stem and progenitor cells (HSPC) induced by the anti-α-4 integrin antibody natalizumab in patients with multiple sclerosis (MS). METHODS We evaluated CD45(low)CD34+ HSPC frequency by flow cytometry in blood from 45 natalizumab-treated patients (12 of whom were prospectively followed during the first year of treatment as part of a pilot cohort and 16 prospectively followed for validation), 10 untreated patients with MS, and 24 healthy donors. In the natalizumab-treated group, we also assessed sorted HSPC cell cycle status, T- and B-lymphocyte subpopulation frequencies (n = 29), and HSPC differentiation potential (n = 10). RESULTS Natalizumab-induced circulating HSPC were predominantly quiescent, suggesting recent mobilization from the bone marrow, and were capable of differentiating ex vivo. Circulating HSPC numbers were significantly increased during natalizumab, but heterogeneously, allowing the stratification of mobilizer and nonmobilizer subgroups. Nonmobilizer status was associated with persistence of disease activity during treatment. The frequency of B cells and CD103+CD8+ regulatory T cells persistently increased, more significantly in mobilizer patients, who also showed a specific naive/memory B-cell profile. CONCLUSIONS The data suggest that natalizumab-induced circulating HSPC increase is the result of true mobilization from the bone marrow and has clinical and immunologic relevance. HSPC mobilization, associated with clinical remission and increased proportion of circulating B and regulatory T cells, may contribute to the treatment's mode of action; thus, HSPC blood counts could represent an early biomarker of responsiveness to natalizumab.
Collapse
Affiliation(s)
- Miriam Mattoscio
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Richard Nicholas
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Maria P Sormani
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Omar Malik
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Jean S Lee
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Adam D Waldman
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Francesco Dazzi
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy
| | - Paolo A Muraro
- From the Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories (M.M., R.N., O.M., P.A.M.), and the Department of Medicine, Division of Experimental Medicine, Centre for Haematology (F.D.), Imperial College London, UK; the Departments of Neurosciences (R.N., O.M.) and Imaging (J.S.L., A.D.W.), Imperial College Healthcare NHS Trust, London, UK; and the Biostatistics Unit, Department of Health Sciences (M.P.S.), University of Genoa, Italy.
| |
Collapse
|
128
|
Wiebenga OT, Klauser AM, Schoonheim MM, Nagtegaal GJA, Steenwijk MD, van Rossum JA, Polman CH, Barkhof F, Pouwels PJW, Geurts JJG. Enhanced axonal metabolism during early natalizumab treatment in relapsing-remitting multiple sclerosis. AJNR Am J Neuroradiol 2015; 36:1116-23. [PMID: 25742985 DOI: 10.3174/ajnr.a4252] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 12/19/2014] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND PURPOSE The considerable clinical effect of natalizumab in patients with relapsing-remitting multiple sclerosis might be explained by its possible beneficial effect on axonal functioning. In this longitudinal study, the effect of natalizumab on absolute concentrations of total N-acetylaspartate, a marker for neuronal integrity, and other brain metabolites is investigated in patients with relapsing-remitting multiple sclerosis by using MR spectroscopic imaging. MATERIALS AND METHODS In this explorative observational study, 25 patients with relapsing-remitting multiple sclerosis initiating natalizumab treatment were included and scanned every 6 months for 18 months. Additionally 18 matched patients with relapsing-remitting multiple sclerosis continuing treatment with interferon-β or glatiramer acetate were included along with 12 healthy controls. Imaging included short TE 2D-MR spectroscopic imaging with absolute metabolite quantification of total N-acetylaspartate, creatine and phosphocreatine, choline-containing compounds, myo-inositol, and glutamate. Concentrations were determined for lesional white matter, normal-appearing white matter, and gray matter. RESULTS At baseline in both patient groups, lower concentrations of total N-acetylaspartate and creatine and phosphocreatine were found in lesional white matter compared with normal-appearing white matter and additionally lower glutamate in lesional white matter of patients receiving natalizumab. In those patients, a significant yearly metabolite increase was found for lesional white matter total N-acetylaspartate (7%, P < .001), creatine and phosphocreatine (6%, P = .042), and glutamate (10%, P = .028), while lesion volumes did not change. In patients receiving interferon-β/glatiramer acetate, no significant change was measured in lesional white matter for any metabolite, while whole-brain normalized lesion volumes increased. CONCLUSIONS Patients treated with natalizumab showed an increase in total N-acetylaspartate, creatine and phosphocreatine, and glutamate in lesional white matter. These increasing metabolite concentrations might be a sign of enhanced axonal metabolism.
Collapse
Affiliation(s)
- O T Wiebenga
- From the Departments of Radiology and Nuclear Medicine (O.T.W., G.J.A.N., M.D.S., F.B.) Anatomy and Neurosciences (O.T.W., A.M.K., M.M.S., G.J.A.N., J.J.G.G.)
| | - A M Klauser
- Anatomy and Neurosciences (O.T.W., A.M.K., M.M.S., G.J.A.N., J.J.G.G.)
| | - M M Schoonheim
- Anatomy and Neurosciences (O.T.W., A.M.K., M.M.S., G.J.A.N., J.J.G.G.)
| | - G J A Nagtegaal
- From the Departments of Radiology and Nuclear Medicine (O.T.W., G.J.A.N., M.D.S., F.B.) Anatomy and Neurosciences (O.T.W., A.M.K., M.M.S., G.J.A.N., J.J.G.G.)
| | - M D Steenwijk
- From the Departments of Radiology and Nuclear Medicine (O.T.W., G.J.A.N., M.D.S., F.B.)
| | | | | | - F Barkhof
- From the Departments of Radiology and Nuclear Medicine (O.T.W., G.J.A.N., M.D.S., F.B.)
| | - P J W Pouwels
- Physics and Medical Technology (P.J.W.P.), Neuroscience Campus Amsterdam and VU University Medical Center, Amsterdam, the Netherlands
| | - J J G Geurts
- Anatomy and Neurosciences (O.T.W., A.M.K., M.M.S., G.J.A.N., J.J.G.G.)
| |
Collapse
|
129
|
Zhen C, Feng X, Li Z, Wang Y, Li B, Li L, Quan M, Wang G, Guo L. Suppression of murine experimental autoimmune encephalomyelitis development by 1,25-dihydroxyvitamin D3 with autophagy modulation. J Neuroimmunol 2015; 280:1-7. [DOI: 10.1016/j.jneuroim.2015.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 01/28/2015] [Indexed: 12/21/2022]
|
130
|
Prosperini L, De Angelis F, De Angelis R, Fanelli F, Pozzilli C. Sustained disability improvement is associated with T1 lesion volume shrinkage in natalizumab-treated patients with multiple sclerosis. J Neurol Neurosurg Psychiatry 2015; 86:236-8. [PMID: 24790213 DOI: 10.1136/jnnp-2014-307786] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Luca Prosperini
- Multiple Sclerosis Centre, S. Andrea Hospital, Rome, Italy Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - Floriana De Angelis
- Multiple Sclerosis Centre, S. Andrea Hospital, Rome, Italy Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - Rosanna De Angelis
- Multiple Sclerosis Centre, S. Andrea Hospital, Rome, Italy Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - Fulvia Fanelli
- Multiple Sclerosis Centre, S. Andrea Hospital, Rome, Italy Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - Carlo Pozzilli
- Multiple Sclerosis Centre, S. Andrea Hospital, Rome, Italy Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| |
Collapse
|
131
|
Abstract
PURPOSE OF REVIEW We summarize MRI measures currently available to assess treatment efficacy and safety in multiple sclerosis (MS) clinical trials and discuss novel metrics that could enter the clinical arena in the near future. RECENT FINDINGS In relapsing remitting MS, MRI measures of disease activity (new T2 and gadolinium-enhancing lesions) provide a good surrogacy of treatment effect on relapse rate and disability progression; however, their value in progressive MS remains elusive. For the progressive disease forms, these measures need to be combined with quantities assessing the extent of irreversible tissue loss, which have already been introduced in some clinical trials (e.g., evolution of active lesions into permanent black holes and brain atrophy). Novel measures (e.g., quantification of gray matter and spinal cord atrophy) have demonstrated a great value in explaining patients' clinical outcome, but still need to be fully validated. Despite showing promise, evaluations of cortical lesions, of microscopic tissue abnormalities, and of functional cortical reorganization are still some way off for monitoring of treatment effects. SUMMARY Trial outcomes in MS should include measures of inflammation and neurodegeneration, which should be combined according to the disease clinical phenotype, phase of the study, and the supposed mechanism of action of the drug tested.
Collapse
|
132
|
Calabrese M, Gajofatto A, Benedetti MD. Therapeutic strategies for relapsing-remitting multiple sclerosis: a special focus on reduction of grey matter damage as measured by brain atrophy. Expert Rev Neurother 2014; 14:1417-28. [PMID: 25391525 DOI: 10.1586/14737175.2014.979794] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In the past two decades, several pathological and radiological findings convincingly demonstrated that damage of the cortical and deep grey matter is a key issue in multiple sclerosis with a significant impact on physical and cognitive disability. Moreover, it has become increasingly evident that the effect of available therapies on the inflammatory white matter damage is not a guarantee of a meaningful effect on the neurodegenerative process mainly affecting the grey matter. Despite the efficacy of all approved disease-modifying drugs should be measured considering such a relevant aspect of the disease, data from clinical trials are few, scattered and heterogeneous. The aim of this review is to summarize the evidence so far acquired on the effect of reducing grey matter damage produced by current and emerging disease-modifying therapies for multiple sclerosis.
Collapse
Affiliation(s)
- Massimiliano Calabrese
- Neurology section, Department of Neurological and Movement Sciences, Policlinico di Borgo Roma, Azienda Ospedaliera Universitaria Integrata Verona, University of Verona, Piazzale Ludovico Antonio Scuro, 37134, Verona, Italy
| | | | | |
Collapse
|
133
|
Sastre-Garriga J, Tur C, Pareto D, Vidal-Jordana A, Auger C, Río J, Huerga E, Tintoré M, Rovira A, Montalban X. Brain atrophy in natalizumab-treated patients: A 3-year follow-up. Mult Scler 2014; 21:749-56. [DOI: 10.1177/1352458514556300] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 09/26/2014] [Indexed: 11/16/2022]
Abstract
Background: A pseudoatrophy effect has been held responsible for the lack of net impact of natalizumab on brain volume outcomes in 2-year trials, but no data are available beyond 24 months. Objective: We aimed to investigate brain volume dynamics in natalizumab-treated patients in up to 3 years after therapy initiation with clinical correlations. Methods: Patients on natalizumab for at least 3 years were clinically assessed 3-monthly. Magnetic resonance imaging scans were performed at baseline and yearly. Brain volume changes were obtained with SIENA. Multivariate models were used to investigate the association between baseline inflammation and changes in brain volume and disability. Results: Sixty-two patients with multiple sclerosis were analysed. Mean age and disease duration were 34.7 (SD: 8.3) and 10.4 (SD: 6.6) years. Presence of gadolinium enhancement at baseline was not associated with Expanded Disability Status Scale changes ( p=0.468), but was associated with larger brain volume decreases ( p=0.005) in the first ( p=0.024) and second year ( p=0.019) but not in the third year ( p=0.863). Brain volume changes at 12 and 36 months were marginally associated with disability status at month 12 ( p=0.094) and 36 ( p=0.084), respectively. Conclusions: Baseline inflammation affects brain volume measures up to 24 months after natalizumab initiation. A marginal association of brain volume changes with disability is present.
Collapse
Affiliation(s)
- J Sastre-Garriga
- Servei de Neurologia / Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - C Tur
- Servei de Neurologia / Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - D Pareto
- Unitat de Ressonància Magnètica (Servei de Radiologia), Hospital universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - A Vidal-Jordana
- Servei de Neurologia / Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - C Auger
- Unitat de Ressonància Magnètica (Servei de Radiologia), Hospital universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - J Río
- Servei de Neurologia / Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - E Huerga
- Unitat de Ressonància Magnètica (Servei de Radiologia), Hospital universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M Tintoré
- Servei de Neurologia / Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - A Rovira
- Unitat de Ressonància Magnètica (Servei de Radiologia), Hospital universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - X Montalban
- Servei de Neurologia / Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
134
|
Therapeutic approaches to disease modifying therapy for multiple sclerosis in adults: An Australian and New Zealand perspective Part 2 New and emerging therapies and their efficacy. J Clin Neurosci 2014; 21:1847-56. [DOI: 10.1016/j.jocn.2014.01.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/28/2014] [Indexed: 12/16/2022]
|
135
|
Natalizumab reduces relapse clinical severity and improves relapse recovery in MS. Mult Scler Relat Disord 2014; 3:705-11. [DOI: 10.1016/j.msard.2014.08.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 08/27/2014] [Accepted: 08/31/2014] [Indexed: 11/22/2022]
|
136
|
Abstract
The increasing availability of new agents to treat multiple sclerosis poses new challenges for clinicians who seek therapies that are both safe and effective for their patients. The introduction of additional effective therapies has been accompanied by the recognition of serious side effects. The clinician now must weigh both the benefits and risks of therapies to help patients decide which treatment best fits each patient׳s risk/benefit profile. An optimal selection of therapies relies on a complete understanding of the risks of therapies and the factors that may help evaluate and mitigate those risks. An individualized treatment approach that incorporates patient and disease factors is needed for each patient. In this review we present risk stratification and mitigation strategies of disease modifying agents for multiple sclerosis.
Collapse
Affiliation(s)
- Daniel Ontaneda
- Mellen Center, Department of Neurology, Neurological Institute, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Samuel Cohn
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Robert J. Fox
- Mellen Center, Department of Neurology, Neurological Institute, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
137
|
Nakamura K, Brown RA, Araujo D, Narayanan S, Arnold DL. Correlation between brain volume change and T2 relaxation time induced by dehydration and rehydration: implications for monitoring atrophy in clinical studies. Neuroimage Clin 2014; 6:166-70. [PMID: 25379428 PMCID: PMC4215533 DOI: 10.1016/j.nicl.2014.08.014] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 07/25/2014] [Accepted: 08/19/2014] [Indexed: 11/25/2022]
Abstract
Brain volume change measured from magnetic resonance imaging (MRI) provides a widely used and useful in vivo measure of irreversible tissue loss. These measurements, however, can be influenced by reversible factors such as shifts in brain water content. Given the strong effect of water on T2 relaxation, we investigated whether an estimate of T2 relaxation time would correlate with brain volume changes induced by physiologically manipulating hydration status. We used a clinically feasible estimate of T2 ("pseudo-T2") computed from a dual turbo spin-echo MRI sequence and correlated pseudo-T2 changes to percent brain volume changes in 12 healthy subjects after dehydration overnight (16-hour thirsting) and rehydration (drinking 1.5 L of water). We found that the brain volume significantly increased between the dehydrated and rehydrated states (mean brain volume change = 0.36%, p = 0.0001) but did not change significantly during the dehydration interval (mean brain volume change = 0.04%, p = 0.57). The changes in brain volume and pseudo-T2 significantly correlated with each other, with marginal and conditional correlations (R (2)) of 0.44 and 0.65, respectively. Our results show that pseudo-T2 may be used in conjunction with the measures of brain volume to distinguish reversible water fluctuations and irreversible brain tissue loss (atrophy) and to investigate disease mechanisms related to neuro-inflammation, e.g., in multiple sclerosis, where edema-related water fluctuations may occur with disease activity and anti-inflammatory treatment.
Collapse
Affiliation(s)
- Kunio Nakamura
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Robert A. Brown
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - David Araujo
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Sridar Narayanan
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Douglas L. Arnold
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
138
|
Salhofer-Polanyi S, Baumgartner A, Kraus J, Maida E, Schmied M, Leutmezer F. What to expect after natalizumab cessation in a real-life setting. Acta Neurol Scand 2014; 130:97-102. [PMID: 24720783 DOI: 10.1111/ane.12250] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND To minimize the risk of progressive multifocal leucoencephalopathy (PML), treatment with natalizumab is often stopped after 2 years, but evidence upon rebound of disease activity is limited and controversial. OBJECTIVE To evaluate effects of natalizumab discontinuation on clinical disease activity within twelve months after cessation. METHODS We retrospectively analyzed data of 201 patients with MS who discontinued natalizumab between 2007 and 2012. Mean change scores of annualized relapse rate (ARR) and expanded disability status scale (EDSS) were calculated for detection of rebound disease activity after twelve months. RESULTS Natalizumab exposure did not exceed 2 years in 50.2% of patients, and the most common reasons for discontinuation were a long treatment period and concern of PML (56%). A total of 11.9% experienced a rebound phenomenon within twelve months. Mean ARR prenatalizumab was lower (P = 0.001, 95% CI -1.0-0.000) and treatment response to natalizumab poorer (P < 0.001, 95% CI 0.4-1.3) in patients with rebound compared to those without, but rebound was not associated with brief exposure to natalizumab (P = 0.159, 95% CI -9.3-1.5). 86.1% of patients switched to another therapy. Patients without rebound were found more often in the group starting an alternative treatment early (P = 0.013). CONCLUSION Our data suggest that rebound of MS disease activity affects a subgroup of patients (11.9%), especially those with low disease activity before natalizumab therapy and a longer treatment gap after its withdrawal.
Collapse
Affiliation(s)
| | - A. Baumgartner
- Department of Neurology; Medical University of Vienna; Vienna Austria
| | - J. Kraus
- Paracelsus Medical University; Salzburg Austria
| | | | - M. Schmied
- Department of Neurology; Medical University of Vienna; Vienna Austria
| | - F. Leutmezer
- Department of Neurology; Medical University of Vienna; Vienna Austria
| | | |
Collapse
|
139
|
Lavery AM, Verhey LH, Waldman AT. Outcome measures in relapsing-remitting multiple sclerosis: capturing disability and disease progression in clinical trials. Mult Scler Int 2014; 2014:262350. [PMID: 24883205 PMCID: PMC4026972 DOI: 10.1155/2014/262350] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/25/2014] [Indexed: 11/24/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disease that manifests as acute relapses and progressive disability. As a primary endpoint for clinical trials in MS, disability is difficult to both characterize and measure. Furthermore, the recovery from relapses and the rate of disability vary considerably among patients. Given these challenges, investigators have developed and studied the performance of various outcome measures and surrogate endpoints in MS clinical trials. This review defines the outcome measures and surrogate endpoints used to date in MS clinical trials and presents challenges in the design of both adult and pediatric trials.
Collapse
Affiliation(s)
- Amy M. Lavery
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Leonard H. Verhey
- The Pediatric Demyelinating Disease Program, Program in Neuroscience & Mental Health, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada M5G 1X8
| | - Amy T. Waldman
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Departments of Neurology and Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
140
|
Shimizu F, Omoto M, Sano Y, Mastui N, Miyashiro A, Tasaki A, Maeda T, Koga M, Kaji R, Kanda T. Sera from patients with multifocal motor neuropathy disrupt the blood-nerve barrier. J Neurol Neurosurg Psychiatry 2014; 85:526-37. [PMID: 23926278 DOI: 10.1136/jnnp-2013-305405] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE In multifocal motor neuropathy (MMN), the destruction of the blood-nerve barrier (BNB) has been considered to be the key step in the disease process. The purpose of the present study was to ascertain whether sera from patients with MMN can open the BNB, and which component of patient sera is the most important for this disruption. METHODS We evaluated the effects of sera from patients with MMN, patients with amyotrophic lateral sclerosis, and control subjects on the expression of tight junction proteins and vascular cell adhesion molecule-1 (VCAM-1), and on the transendothelial electrical resistance (TEER) in human peripheral nerve microvascular endothelial cells (PnMECs). RESULTS The sera from patients with MMN decreased the claudin-5 protein expression and the TEER in PnMECs. However, this effect was reversed after application of an anti-vascular endothelial growth factor (anti-VEGF) neutralising antibody. The VEGF secreted by PnMECs was significantly increased after exposure to the sera from patients with MMN. The sera from patients with MMN also increased the VCAM-1 protein expression by upregulating the nuclear factor kappa-B (NF-κB) signalling. The immunoglobulin G purified from MMN sera decreased the expression of claudin-5 and increased the VCAM-1 expression in PnMECs. CONCLUSIONS The sera from MMN patients may disrupt the BNB function via the autocrine secretion of VEGF in PnMECs, or the exposure to autoantibodies against PnMECs that are contained in the MMN sera. Autoantibodies against PnMECs in MMN sera may activate the BNB by upregulating the VCAM-1 expression, thereby allowing for the entry of a large number of circulating inflammatory cells into the peripheral nervous system.
Collapse
Affiliation(s)
- Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, , Ube, Yamaguchi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Dehydration affects spinal cord cross-sectional area measurement on MRI in healthy subjects. Spinal Cord 2014; 52:616-20. [PMID: 24777158 DOI: 10.1038/sc.2014.66] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 03/27/2014] [Accepted: 04/01/2014] [Indexed: 11/08/2022]
Abstract
STUDY DESIGN This was a prospective cohort observational study. OBJECTIVE To determine the effect of dehydration and rehydration on spinal cord cross-sectional area (CSA) measurement on magnetic resonance imaging (MRI). SETTING MRI Research Centre, University of British Columbia, Canada. METHODS Ten healthy subjects (aged 21-32 years) were scanned on a 3T MRI scanner at four time points: (1) baseline, (2) rescan after 1 h, (3) the next day after fasting for a minimum of 14 h and (4) after rehydration with 1.5 l of water over the course of 1 h. Two independent, established semi-automatic CSA measurement techniques (one based on two-dimensional (2D) edge detection, the other on three-dimensional (3D) surface fitting) were applied to a 3D T1-weighted scan of each subject at each time point, with the operator blinded to scan order. The percentage change in CSA from baseline to each subsequent time point was calculated. One-tailed paired t-tests were used to assess the significance of the changes from baseline. RESULTS A decrease in CSA following dehydration was detected by both measurement methods, with a mean change of -0.654% (s.d.=0.778, P<0.05) and -0.650% (s.d.=1.071, P<0.05) for the first and second methods, respectively. CONCLUSION Dehydration can confound CSA measurements on MRI. The magnitude of the effect is significant relative to short-term pathological changes that have been observed in diseases such as multiple sclerosis.
Collapse
|
142
|
Waubant E, Maghzi AH, Revirajan N, Spain R, Julian L, Mowry EM, Marcus J, Liu S, Jin C, Green A, McCulloch CE, Pelletier D. A randomized controlled phase II trial of riluzole in early multiple sclerosis. Ann Clin Transl Neurol 2014; 1:340-7. [PMID: 25356404 PMCID: PMC4184685 DOI: 10.1002/acn3.60] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 03/03/2014] [Accepted: 03/14/2014] [Indexed: 12/22/2022] Open
Abstract
Objectives We evaluated the effect of riluzole versus placebo added to weekly IM interferon beta-1a in early multiple sclerosis (MS). Methods This is a randomized (1:1), double-blind, placebo-controlled trial of riluzole 50 mg twice daily in subjects with MS onset less than 1 year prior. Trial participation was up to 3 years. The primary endpoint was change in percent brain volume change. Secondary endpoints included changes in normalized gray and normal-appearing white matter volumes, retinal nerve fiber layer thickness (RNFL), MS Functional Composite and Symbol Digit Modalities Test scores. Mixed model regression analysis was used to compare the changes over time between groups. Results Forty-three subjects were randomized to study drug (22 riluzole, 21 placebo). Baseline characteristics were overall similar between groups except for older age (P = 0.042), higher normalized cerebrospinal fluid volume (P = 0.050), lower normalized gray matter volume (P = 0.14), and thinner RNFL (P = 0.043) in the riluzole group. In the primary analysis, percent brain volume change in the placebo group decreased at a rate of 0.49% per year whereas the riluzole group decreased by 0.86% per year (0.37% more per year; 95% CI −0.78, 0.024; P = 0.065). Although age did not influence the rate of brain volume decline, the difference between groups was attenuated after adjustment for baseline normalized gray matter and lesion volume (0.26% more per year in riluzole group; 95% CI −0.057, 0.014; P = 0.22). Analyses of secondary outcomes showed no differences between groups. Interpretation This trial provides class 1 evidence that riluzole treatment does not meaningfully reduce brain atrophy progression in early MS.
Collapse
Affiliation(s)
- Emmanuelle Waubant
- MS Center, Neurology Department, University of California San Francisco San Francisco, California ; Department of Pediatrics, University of California San Francisco San Francisco, California
| | - Amir-Hadi Maghzi
- MS Center, Neurology Department, University of California San Francisco San Francisco, California
| | - Nisha Revirajan
- MS Center, Neurology Department, University of California San Francisco San Francisco, California
| | - Rebecca Spain
- Portland VA Medical Center & Oregon Health & Science University Portland, Oregon
| | - Laura Julian
- MS Center, Neurology Department, University of California San Francisco San Francisco, California
| | - Ellen M Mowry
- Department of Neurology, Hopkins MS Center Baltimore, Maryland
| | - Jacqueline Marcus
- MS Center, Neurology Department, University of California San Francisco San Francisco, California
| | - Shuang Liu
- Department of Neurology, Yale University School of Medicine New Haven, Connecticut
| | - Chengshi Jin
- Department of Epidemiology and Biostatistics, University of California San Francisco San Francisco, California
| | - Ari Green
- MS Center, Neurology Department, University of California San Francisco San Francisco, California ; Department of Ophthalmology, University of California San Francisco San Francisco, California
| | - Charles E McCulloch
- Department of Epidemiology and Biostatistics, University of California San Francisco San Francisco, California
| | - Daniel Pelletier
- Department of Neurology, Yale University School of Medicine New Haven, Connecticut
| |
Collapse
|
143
|
Abstract
Natalizumab (Tysabri®) is a humanized monoclonal antibody against the α4 chain of integrins and was the first targeted therapy to be approved for the treatment of relapsing-remitting multiple sclerosis (RRMS). Natalizumab acts as a selective adhesion molecule antagonist, which binds very late antigen (VLA)-4 and inhibits the translocation of activated VLA-4-expressing leukocytes across the blood-brain barrier into the CNS. In a pivotal phase III clinical trial, natalizumab 300 mg intravenously every 4 weeks for 2 years in adults with RRMS significantly reduced the annualized relapse rate and the risk of sustained progression of disability compared with placebo, as well as significantly increasing the proportion of relapse-free patients at 1 and 2 years. Natalizumab also significantly reduced the number of T2-hyperintense, gadolinium-enhancing and T1-hypointense lesions on magnetic resonance imaging, and significantly reduced the volume of T2-hyperintense and T1-hypointense lesions compared with placebo. Natalizumab recipients generally experienced improved health-related quality of life at 1-2 years. Natalizumab was generally well tolerated in pivotal trials. The only adverse events that were more frequent with natalizumab monotherapy than with placebo were fatigue and allergic reactions. The main safety and tolerability issue with natalizumab is the incidence of progressive multifocal leukoencephalopathy (PML). As long as the risk of PML is managed effectively, natalizumab is a valuable therapeutic option for adults with highly active relapsing forms of multiple sclerosis.
Collapse
|
144
|
Tasaki A, Shimizu F, Sano Y, Fujisawa M, Takahashi T, Haruki H, Abe M, Koga M, Kanda T. Autocrine MMP-2/9 secretion increases the BBB permeability in neuromyelitis optica. J Neurol Neurosurg Psychiatry 2014; 85:419-30. [PMID: 24259591 DOI: 10.1136/jnnp-2013-305907] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Pathological breakdown of the blood-brain barrier (BBB) is thought to constitute the beginning of the disease process in neuromyelitis optica (NMO). In the current study, we investigated possible molecular mechanisms responsible for the breakdown of BBB using NMO sera. METHODS We analysed the effects of sera obtained from anti-aquaporin 4 (AQP4) antibody-positive NMO spectrum disorder (NMOSD) patients, multiple sclerosis (MS) patients and control subjects on the production of claudin-5, matrix-metalloproteinases (MMPs)-2/9, and vascular cell adhesion protein-1 (VCAM-1) in human brain microvascular endothelial cells (BMECs). We also examined whether immunoglobulin G (IgG) purified from NMOSD sera influences the claudin-5 or VCAM-1 protein expression. RESULTS The disturbance of BBB properties in BMECs following exposure to NMOSD sera was restored after adding the MMP inhibitor, GM6001. The secretion of MMP-2/9 by BMECs significantly increased after applying the NMOSD sera. The sera from NMOSD patients also increased both the MMP-2/9 secretion and the VCAM-1 protein level by BMECs. The IgG purified from NMOSD sera did not influence the BBB properties or the amount of MMP-2/9 proteins, although it did increase the amount of VCAM-1 proteins in BMECs. Reduction in anti-AQP4 antibody titre was not correlated with a reduction in VCAM-1 expression. CONCLUSIONS The autocrine secretion of MMP-2/9 by BMECs induced by humoral factors, other than IgG, in sera obtained from NMOSD patients potentially increases BBB permeability. IgG obtained from NMOSD sera, apart from anti-AQP4 antibodies, affect the BBB by upregulating VCAM, thereby facilitating the entry of inflammatory cells into the central nervous system.
Collapse
Affiliation(s)
- Ayako Tasaki
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, , Ube, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Shimizu F, Tasaki A, Sano Y, Ju M, Nishihara H, Oishi M, Koga M, Kawai M, Kanda T. Sera from remitting and secondary progressive multiple sclerosis patients disrupt the blood-brain barrier. PLoS One 2014; 9:e92872. [PMID: 24686948 PMCID: PMC3970956 DOI: 10.1371/journal.pone.0092872] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 02/26/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Pathological destruction of blood-brain barrier (BBB) has been thought to be the initial key event in the process of developing multiple sclerosis (MS). The purpose of the present study was to clarify the possible molecular mechanisms responsible for the malfunction of BBB by sera from relapse-remitting MS (RRMS) and secondary progressive MS (SPMS) patients. METHODS We evaluated the effects of sera from the patients in the relapse phase of RRMS (RRMS-R), stable phase of RRMS (RRMS-S) and SPMS on the expression of tight junction proteins and vascular cell adhesion protein-1 (VCAM-1), and on the transendothelial electrical resistance (TEER) in human brain microvascular endothelial cells (BMECs). RESULTS Sera from the RRMS-R or SPMS patients decreased the claudin-5 protein expression and the TEER in BMECs. In RRMS-R, this effect was restored after adding an MMP inhibitor, and the MMP-2/9 secretion by BMECs was significantly increased after the application of patients' sera. In SPMS, the immunoglobulin G (IgG) purified from patients' sera also decreased the claudin-5 protein expression and the TEER in BMECs. The sera and purified IgG from all MS patients increased the VCAM-1 protein expression in BMECs. CONCLUSIONS The up-regulation of autocrine MMP-2/9 by BMECs after exposure to sera from RRMS-R patients or the autoantibodies against BMECs from SPMS patients can compromise the BBB. Both RRMS-S and SPMS sera increased the VCAM-1 expression in the BBB, thus indicating that targeting the VCAM-1 in the BBB could represent a possible therapeutic strategy for even the stable phase of MS and SPMS.
Collapse
Affiliation(s)
- Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Ayako Tasaki
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Mihua Ju
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Hideaki Nishihara
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Mariko Oishi
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Michiaki Koga
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Motoharu Kawai
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
- * E-mail:
| |
Collapse
|
146
|
Abstract
PURPOSE OF REVIEW The treatment of multiple sclerosis (MS) is evolving beyond the current parenteral immunomodulators and early oral alternatives, offering physicians considerable choice of therapies. Although all agents are tested in similarly designed clinical studies, comparison of their outcomes is not possible except in carefully controlled head-to-head comparator studies. In this review, the current, recent, and most imminent therapies are discussed and an overall summary is presented along with a discussion of how they are perceived relative to the older or other recent agents. RECENT FINDINGS The list of potentially effective agents for the treatment of MS may be exhaustive, but several have now completed their phase 3 trials and have received or imminently expect government approval. This review discusses these new agents in terms of their perceived mechanisms of action and their respective results, and attempts to position them among the currently approved and utilized agents for MS. SUMMARY Although it is not yet possible to predict which treatment is best suited to a given patient, it is nevertheless important to have a perspective of the possible agents and their efficacy and safety, and a plan regarding how to use them in order to maximize benefit and minimize harm in controlling relapsing MS.
Collapse
Affiliation(s)
- Mark S Freedman
- The Ottawa Hospital, 501 Smyth Rd, Ottawa, ON K1H 8L6, Canada.
| |
Collapse
|
147
|
Kurth F, Luders E, Sicotte NL, Gaser C, Giesser BS, Swerdloff RS, Montag MJ, Voskuhl RR, Mackenzie-Graham A. Neuroprotective effects of testosterone treatment in men with multiple sclerosis. NEUROIMAGE-CLINICAL 2014; 4:454-60. [PMID: 24634831 PMCID: PMC3952353 DOI: 10.1016/j.nicl.2014.03.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/03/2014] [Accepted: 03/04/2014] [Indexed: 11/16/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory and neurodegenerative disease of the central nervous system. While current medication reduces relapses and inflammatory activity, it has only a modest effect on long-term disability and gray matter atrophy. Here, we have characterized the potential neuroprotective effects of testosterone on cerebral gray matter in a pilot clinical trial. Ten men with relapsing-remitting MS were included in this open-label phase II trial. Subjects were observed without treatment for 6 months, followed by testosterone treatment for another 12 months. Focal gray matter loss as a marker for neurodegeneration was assessed using voxel-based morphometry. During the non-treatment phase, significant voxel-wise gray matter decreases were widespread (p≤ 0.05 corrected). However, during testosterone treatment, gray matter loss was no longer evident. In fact, a significant gray matter increase in the right frontal cortex was observed (p≤ 0.05 corrected). These observations support the potential of testosterone treatment to stall (and perhaps even reverse) neurodegeneration associated with MS. Furthermore, they warrant the investigation of testosterone's neuroprotective effects in larger, placebo controlled MS trials as well as in other neurodegenerative diseases. This is the first report of gray matter increase as the result of treatment in MS.
Collapse
Affiliation(s)
- Florian Kurth
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA ; Brain Mapping Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Eileen Luders
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA ; Brain Mapping Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Nancy L Sicotte
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA ; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Christian Gaser
- Department of Psychiatry, Jena University Hospital, Jena, Germany ; Department of Neurology, Jena University Hospital, Jena, Germany
| | - Barbara S Giesser
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ronald S Swerdloff
- Department of Medicine, Harbor-UCLA Medical Center, Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - Michael J Montag
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Rhonda R Voskuhl
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Allan Mackenzie-Graham
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA ; Brain Mapping Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
148
|
Alroughani RA, Aref HM, Bohlega SA, Dahdaleh MP, Feki I, Al Jumah MA, Al-Kawi MZ, Koussa SF, Sahraian MA, Alsharoqi IA, Yamout BI. Natalizumab treatment for multiple sclerosis: Middle East and North Africa regional recommendations for patient selection and monitoring. BMC Neurol 2014; 14:27. [PMID: 24521176 PMCID: PMC3927624 DOI: 10.1186/1471-2377-14-27] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 02/07/2014] [Indexed: 01/09/2023] Open
Abstract
Background Natalizumab, a highly specific α4-integrin antagonist, , has recently been registered across the Middle East and North Africa region. It improves clinical and magnetic resonance imaging (MRI) outcomes and reduces the rate of relapse and disability progression in relapsing-remitting multiple sclerosis (MS). Natalizumab is recommended for patients who fail first-line disease-modifying therapy or who have very active disease. Progressive multifocal leukoencephalopathy is a rare, serious adverse event associated with natalizumab. We aim to develop regional recommendations for the selection and monitoring of MS patients to be treated with natalizumab in order to guide local neurological societies. Methods After a review of available literature, a group of neurologists with expertise in the management of MS met to discuss the evidence and develop regional recommendations to guide appropriate use of natalizumab in the region. Results Disease breakthrough is defined as either clinical (relapse or disability progression) or radiological activity (new T2 lesion or gadolinium-enhancing lesions on MRI), or a combination of both. Natalizumab is recommended as an escalation therapy in patients with breakthrough disease based on its established efficacy in Phase III studies. Several factors including prior immunosuppressant therapy, anti-John Cunningham virus (JCV) antibody status and patient choice will affect the selection of natalizumab. In highly active MS, natalizumab is considered as a first-line therapy for naive patients with disabling relapses in association with MRI activity. The anti-JCV antibody test is used to assess anti-JCV antibody status and identify the risk of PML. While seronegative patients should continue treatment with natalizumab, anti-JCV antibody testing every 6 months and annual MRI scans are recommended as part of patient monitoring. In seropositive patients, the expected benefits of natalizumab treatment have to be weighed against the risks of PML. Clinical vigilance and follow-up MRI scans remain the cornerstone of monitoring. After 2 years of natalizumab therapy, monitoring should include more frequent MRI scans (every 3–4 months) for seropositive patients, and the risk-benefit ratio should be reassessed and discussed with patients. Conclusions Recommendations have been developed to guide neurologists in the Middle East and North Africa on patient selection for natalizumab treatment and monitoring.
Collapse
|
149
|
De Stefano N, Airas L, Grigoriadis N, Mattle HP, O'Riordan J, Oreja-Guevara C, Sellebjerg F, Stankoff B, Walczak A, Wiendl H, Kieseier BC. Clinical relevance of brain volume measures in multiple sclerosis. CNS Drugs 2014; 28:147-56. [PMID: 24446248 DOI: 10.1007/s40263-014-0140-z] [Citation(s) in RCA: 226] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Multiple sclerosis (MS) is a chronic disease with an inflammatory and neurodegenerative pathology. Axonal loss and neurodegeneration occurs early in the disease course and may lead to irreversible neurological impairment. Changes in brain volume, observed from the earliest stage of MS and proceeding throughout the disease course, may be an accurate measure of neurodegeneration and tissue damage. There are a number of magnetic resonance imaging-based methods for determining global or regional brain volume, including cross-sectional (e.g. brain parenchymal fraction) and longitudinal techniques (e.g. SIENA [Structural Image Evaluation using Normalization of Atrophy]). Although these methods are sensitive and reproducible, caution must be exercised when interpreting brain volume data, as numerous factors (e.g. pseudoatrophy) may have a confounding effect on measurements, especially in a disease with complex pathological substrates such as MS. Brain volume loss has been correlated with disability progression and cognitive impairment in MS, with the loss of grey matter volume more closely correlated with clinical measures than loss of white matter volume. Preventing brain volume loss may therefore have important clinical implications affecting treatment decisions, with several clinical trials now demonstrating an effect of disease-modifying treatments (DMTs) on reducing brain volume loss. In clinical practice, it may therefore be important to consider the potential impact of a therapy on reducing the rate of brain volume loss. This article reviews the measurement of brain volume in clinical trials and practice, the effect of DMTs on brain volume change across trials and the clinical relevance of brain volume loss in MS.
Collapse
Affiliation(s)
- Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Viale Bracci 2, Siena, 53100, Italy,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Singer BA. Fingolimod for the treatment of relapsing multiple sclerosis. Expert Rev Neurother 2014; 13:589-602. [PMID: 23738997 DOI: 10.1586/ern.13.52] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fingolimod 0.5 mg (Gilenya(™), Novartis Pharmaceuticals Corporation, FL, USA) is the first once-daily oral therapy approved for relapsing forms of multiple sclerosis (MS) in the USA and for rapidly evolving severe MS or highly active disease despite IFN-β treatment in Europe. An extensive clinical development program has established fingolimod as an effective therapy that reduces relapses by approximately half compared with placebo or intramuscular IFN-β1a. Over 2 years of postmarketing experience in >63,000 MS patients (with >73,000 patient-years of exposure) across the world has contributed to a well-characterized safety profile for fingolimod, and its side effects are manageable through patient monitoring. This article discusses the unique mechanisms of action of fingolimod in the immune and nervous systems, the key data underlying its efficacy and safety profile and perspectives on the role of fingolimod in current and future treatment strategies for MS.
Collapse
Affiliation(s)
- Barry A Singer
- The MS Center for Innovations in Care, Missouri Baptist Medical Center, St Louis, MO 63131, USA.
| |
Collapse
|