1
|
Liampas A, Tseriotis VS, Mavridis T, Vavougios GD, Zis P, Hadjigeorgiou GM, Bargiotas P, Pourzitaki C, Artemiadis A. Effects of natalizumab on oligoclonal bands in the cerebrospinal fluid of patients with multiple sclerosis: a systematic review and meta-analysis. Neurol Sci 2024:10.1007/s10072-024-07930-w. [PMID: 39673046 DOI: 10.1007/s10072-024-07930-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/04/2024] [Indexed: 12/15/2024]
Abstract
INTRODUCTION Oligoclonal bands (OCBs) in cerebrospinal fluid (CSF) are utilized for diagnosing multiple sclerosis (MS), as they are found in 95% of patients. Additionally, OCBs are linked to disease prognosis. The primary contributors to OCB production are long-lived plasma cells. This study aims to quantify the impact of natalizumab (NTZ) on OCB levels in the CSF of MS patients. METHODS A systematic search on MEDLINE, SCOPUS and Web of Science for English-written and peer-reviewed longitudinal studies on adults was performed. Methodological quality was assessed with the Newcastle-Ottawa Scale. Proportional meta-analysis was performed in R using a generalized linear mixed-effects model. We investigated heterogeneity with influence diagnostics, sensitivity analysis and meta-regression. RESULTS Eight eligible studies of adequate quality with a total sample of 326 relapsing-remitting MS patients were included. A summary rate of 14.07% [95% CI, 4.48%-36.36%] for complete loss of OCBs and 42.02% [95% CI, 15.23%-74.51%] for reduction in OCB number or intensity was observed, both with considerable heterogeneity. Pooled estimates dropped (11% [95% CI, 0.04%-0.29%] and 34% [95% CI, 0.11%-0.68%] respectively) after the identification of an influential study. Multivariable meta-regression identified IgG index as a factor contributing to heterogeneity (adj. p = 0.0279), regarding reduction of OCB number or intensity. DISCUSSION In conclusion, our systematic review and meta-analysis showed that NTZ can lead to reduction of intrathecal OCBs in MS patients, indicating a possible effect of NTZ on memory plasma cells, which are the main source of OCBs in MS.
Collapse
Affiliation(s)
- Andreas Liampas
- Department of Neurology, Nicosia General Hospital, Nicosia Limassol Old Road 215, P.C. 2029, Strovolos, Nicosia, Cyprus.
| | - Vasilis-Spyridon Tseriotis
- "Agios Pavlos" General Hospital of Thessaloniki, 161 Ethnikis Antistaseos Leof., P.C. 55134, 12 Kalamaria, Thessaloniki, Greece
- Laboratory of Clinical Pharmacology, Aristotle University of Thessaloniki, P.C. 54124, Thessaloniki, Greece
| | - Theodoros Mavridis
- Department of Neurology, Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital, Dublin, Incorporating the National Children's Hospital (AMNCH), Dublin, D24 NR0A, Ireland
- 21St Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528, Athens, Greece
| | - George D Vavougios
- Medical School, University of Cyprus, 75 Kallipoleos Street, P.C. 1678, 10 Nicosia, Nicosia, Cyprus
| | - Panagiotis Zis
- Medical School, University of Cyprus, 75 Kallipoleos Street, P.C. 1678, 10 Nicosia, Nicosia, Cyprus
| | - Georgios M Hadjigeorgiou
- Medical School, University of Cyprus, 75 Kallipoleos Street, P.C. 1678, 10 Nicosia, Nicosia, Cyprus
| | - Panagiotis Bargiotas
- Medical School, University of Cyprus, 75 Kallipoleos Street, P.C. 1678, 10 Nicosia, Nicosia, Cyprus
| | - Chryssa Pourzitaki
- Laboratory of Clinical Pharmacology, Aristotle University of Thessaloniki, P.C. 54124, Thessaloniki, Greece
| | - Artemios Artemiadis
- Medical School, University of Cyprus, 75 Kallipoleos Street, P.C. 1678, 10 Nicosia, Nicosia, Cyprus
| |
Collapse
|
2
|
Proschinger S, Belen S, Adammek F, Schlagheck ML, Rademacher A, Schenk A, Warnke C, Bloch W, Zimmer P. Sportizumab - Multimodal progressive exercise over 10 weeks decreases Th17 frequency and CD49d expression on CD8 + T cells in relapsing-remitting multiple sclerosis: A randomized controlled trial. Brain Behav Immun 2024; 124:397-408. [PMID: 39675643 DOI: 10.1016/j.bbi.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND Multiple Sclerosis (MS) represents a neuroinflammatory autoimmune disease characterized by the predominance of circulating T cell subsets with proinflammatory characteristics and increased central nervous system (CNS)-homing potential. Substantial evidence confirms various beneficial effects of chronic exercise interventions in MS, but it is unknown how long-term multi-modal intense exercise affects MS-associated lymphocytes that are commonly targeted by medication in persons with relapsing remitting MS (pwRRMS). METHODS A total of 45 participants with defined RRMS were randomized to either the exercise (n = 22) or passive waitlist-control group (n = 23). A 10-week intervention consisting of progressive resistance and strength-endurance exercises was applied (3x/week à 60 min). Blood was drawn before (T1) and after (T2) the intervention period. Flow cytometry was used for phenotyping lymphocyte subsets. RESULTS Relative protein expression of CD49d within CD8+ T cells, quantified via mean fluorescence intensity (MFI), is significantly associated with the Expanded Disability Status Scale (p = 0.007, r = 0.440), decreased in the exercise group (p = 0.001) only, and was significantly lower in the exercise compared to the control group at T2 (p < 0.001). T helper (Th) 17 cell frequency decreased only in the exercise group (p < 0.001). CD8+CD20+ T cell frequency was significantly lower in the exercise compared to the control group at T2 (p = 0.003), without showing significant time effects. CONCLUSION The 10-week multimodal exercise intervention mainly affected circulating T cells harboring a pathophysiological phenotype in MS. The findings of a decreased frequency of pathogenic Th17 cells and the reduced CNS-homing potential of CD8+ T cells, indicated by reduced CD49d MFI, substantiate the positive effects of exercise on cellular biomarkers involved in disease activity and progression in MS. To confirm exercise-mediated beneficial effects on both disease domains, clinical endpoints (i.e., relapse rate, lesion formation, EDSS score) should be assessed together with these cellular and molecular markers in studies with a larger sample size and a duration of six to twelve months or longer.
Collapse
Affiliation(s)
- Sebastian Proschinger
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany
| | - Sergen Belen
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany; Department for Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Frederike Adammek
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany
| | - Marit Lea Schlagheck
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany
| | | | - Alexander Schenk
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany
| | - Clemens Warnke
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Wilhelm Bloch
- Department for Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Philipp Zimmer
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany.
| |
Collapse
|
3
|
Foley JF, Defer G, Ryerson LZ, Cohen JA, Arnold DL, Butzkueven H, Cutter GR, Giovannoni G, Killestein J, Wiendl H, Li K, Dsilva L, Toukam M, Ferber K, Sohn J, Engelman H, Lasky T. Pharmacokinetics and Pharmacodynamics of Natalizumab 6-Week Dosing vs Continued 4-Week Dosing for Relapsing-Remitting Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200321. [PMID: 39393045 PMCID: PMC11488827 DOI: 10.1212/nxi.0000000000200321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 08/30/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND AND OBJECTIVES Exposure to natalizumab, an efficacious treatment for relapsing-remitting multiple sclerosis (RRMS), is associated with increased risk of progressive multifocal leukoencephalopathy (PML). Compared with every-4-week (Q4W) dosing, extended-interval dosing of natalizumab is associated with decreased risk of PML. Clinical efficacy was maintained in the majority of patients switched to every-6-week (Q6W) dosing in the phase 3b NOVA clinical trial. In this article, we report pharmacokinetics (PK) and pharmacodynamics (PD) of Q6W vs Q4W dosing in NOVA. METHODS In NOVA study Part 1, participants with RRMS (aged 18-60 years) and Expanded Disability Status Scale score <5.5, who were stable on IV natalizumab Q4W dosing for ≥12 months, were randomized to continue IV Q4W dosing or switched to IV Q6W dosing of natalizumab and followed for 72 weeks. Exploratory outcomes were measurements of trough serum natalizumab concentration, α4-integrin saturation, and soluble vascular cell adhesion molecule-1 (sVCAM-1) concentration. A mixed model of repeated measures was used to estimate mean treatment differences between groups. Patient-level PK and PD data were examined in those with relapse or radiologic disease activity. RESULTS In NOVA, 486 (Q6W, n = 245; Q4W, n = 241) and 487 (Q6W, n = 246; Q4W, n = 241) participants were included in the PK and PD populations, respectively. Mean trough natalizumab concentrations ranged from 10 to 21 μg/mL (Q6W) and 33-38 μg/mL (Q4W), and mean α4-integrin saturation remained above 65.5% (Q6W) and above 77.9% (Q4W). In the Q6W group, mean sVCAM-1 levels increased 23.6% by week 24 and remained elevated throughout the study, while mean sVCAM-1 levels remained generally stable in the Q4W group. Most participants with T2 lesion activity or relapse activity, in either treatment arm, maintained trough natalizumab levels >10 μg/mL and trough α4-integrin saturation >50%. DISCUSSION Compared with Q4W dosing, Q6W dosing was associated with a 60%-70% decrease in mean trough natalizumab levels and a 9%-16% decrease in mean α4-integrin saturation. At the patient level, neither natalizumab concentration nor α4-integrin saturation was consistently predictive of lesion or relapse activity, suggesting that trough natalizumab and α4-integrin saturation measurements should be interpreted with caution in clinical practice. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov, NCT03689972; EudraCT, 2018-002145-11. Submitted 2018-09-27. First patient enrolled: 2018-12-26. https://clinicaltrials.gov/study/NCT03689972.
Collapse
Affiliation(s)
- John F Foley
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Gilles Defer
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Lana Zhovtis Ryerson
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Jeffrey A Cohen
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Douglas L Arnold
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Helmut Butzkueven
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Gary R Cutter
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Gavin Giovannoni
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Joep Killestein
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Heinz Wiendl
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Kexuan Li
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Liesel Dsilva
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Marie Toukam
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Kyle Ferber
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Jihee Sohn
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Holly Engelman
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| | - Tyler Lasky
- From the Rocky Mountain MS Clinic (J.F.F.), Salt Lake City, UT; Department of Neurology (G.D.), Centre Hospitalier Universitaire de Caen, France; Hackensack Meridian Medical Group - Neurology (L.Z.R.), Jersey Shore University Medical Center, Neptune City, NJ; Mellen MS Center (J.A.C.), Neurological Institute, Cleveland Clinic, OH; Montréal Neurological Institute (D.L.A.), McGill University; NeuroRx Research (D.L.A.), Montréal, Quebec, Canada; Department of Neuroscience (H.B.), Central Clinical School, Monash University, Melbourne, Victoria, Australia; University of Alabama at Birmingham (G.R.C.), School of Public Health; Blizard Institute (G.G.), Barts and The London School of Medicine and Dentistry; Queen Mary University of London (G.G.), United Kingdom; Department of Neurology (J.K.), Amsterdam University Medical Centers, Vrije Universiteit, Netherlands; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Biogen (K.L., L.D., M.T., K.F., J.S., T.L.), Cambridge, MA; and Ashfield MedComms (H.E.), Middletown, CT
| |
Collapse
|
4
|
Orrù V, Serra V, Marongiu M, Lai S, Lodde V, Zoledziewska M, Steri M, Loizedda A, Lobina M, Piras MG, Virdis F, Delogu G, Marini MG, Mingoia M, Floris M, Masala M, Castelli MP, Mostallino R, Frau J, Lorefice L, Farina G, Fronza M, Carmagnini D, Carta E, Pilotto S, Chessa P, Devoto M, Castiglia P, Solla P, Zarbo RI, Idda ML, Pitzalis M, Cocco E, Fiorillo E, Cucca F. Implications of disease-modifying therapies for multiple sclerosis on immune cells and response to COVID-19 vaccination. Front Immunol 2024; 15:1416464. [PMID: 39076966 PMCID: PMC11284103 DOI: 10.3389/fimmu.2024.1416464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/27/2024] [Indexed: 07/31/2024] Open
Abstract
Introduction Disease-modifying therapies (DMTs) have been shown to improve disease outcomes in multiple sclerosis (MS) patients. They may also impair the immune response to vaccines, including the SARS-CoV-2 vaccine. However, available data on both the intrinsic immune effects of DMTs and their influence on cellular response to the SARS-CoV-2 vaccine are still incomplete. Methods Here, we evaluated the immune cell effects of 3 DMTs on the response to mRNA SARS-CoV-2 vaccination by comparing MS patients treated with one specific therapy (fingolimod, dimethyl fumarate, or natalizumab) with both healthy controls and untreated patients. We profiled 23 B-cell traits, 57 T-cell traits, and 10 cytokines, both at basal level and after stimulation with a pool of SARS-CoV-2 spike peptides, in 79 MS patients, treated with DMTs or untreated, and 32 healthy controls. Measurements were made before vaccination and at three time points after immunization. Results and Discussion MS patients treated with fingolimod showed the strongest immune cell dysregulation characterized by a reduction in all measured lymphocyte cell classes; the patients also had increased immune cell activation at baseline, accompanied by reduced specific immune cell response to the SARS-CoV-2 vaccine. Also, anti-spike specific B cells progressively increased over the three time points after vaccination, even when antibodies measured from the same samples instead showed a decline. Our findings demonstrate that repeated booster vaccinations in MS patients are crucial to overcoming the immune cell impairment caused by DMTs and achieving an immune response to the SARS-CoV-2 vaccine comparable to that of healthy controls.
Collapse
Affiliation(s)
- Valeria Orrù
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Valentina Serra
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Michele Marongiu
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Sandra Lai
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Valeria Lodde
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Magdalena Zoledziewska
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - Maristella Steri
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - Annalisa Loizedda
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - Monia Lobina
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Maria Grazia Piras
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Francesca Virdis
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - Giuseppe Delogu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | - Maura Mingoia
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - Matteo Floris
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Marco Masala
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - M. Paola Castelli
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Rafaela Mostallino
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Jessica Frau
- Regional Multiple Sclerosis Center, Azienda Sanitaria Locale (ASL) Cagliari, Cagliari, Italy
| | - Lorena Lorefice
- Regional Multiple Sclerosis Center, Azienda Sanitaria Locale (ASL) Cagliari, Cagliari, Italy
| | - Gabriele Farina
- Neurology Unit, Azienza Ospedaliera Universitaria (AOU) Sassari, Sassari, Italy
| | - Marzia Fronza
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Daniele Carmagnini
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Elisa Carta
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Silvy Pilotto
- Neurology Unit, Azienza Ospedaliera Universitaria (AOU) Sassari, Sassari, Italy
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Paola Chessa
- Neurology Unit, Azienza Ospedaliera Universitaria (AOU) Sassari, Sassari, Italy
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Marcella Devoto
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Paolo Castiglia
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Paolo Solla
- Neurology Unit, Azienza Ospedaliera Universitaria (AOU) Sassari, Sassari, Italy
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Roberto Ignazio Zarbo
- Neurology Unit, Azienza Ospedaliera Universitaria (AOU) Sassari, Sassari, Italy
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Maria Laura Idda
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Maristella Pitzalis
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - Eleonora Cocco
- Regional Multiple Sclerosis Center, Azienda Sanitaria Locale (ASL) Cagliari, Cagliari, Italy
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Edoardo Fiorillo
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Francesco Cucca
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
5
|
Page LJ, Pay IF, Castellana ET, Heussen R, Hoyt T, Foley J, Messmer BT. Intact natalizumab pharmacokinetics is impacted by endogenous IgG4 concentration. Mult Scler Relat Disord 2024; 87:105667. [PMID: 38759421 DOI: 10.1016/j.msard.2024.105667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/02/2024] [Accepted: 05/05/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Natalizumab (NAT) pharmacokinetics and pharmacodynamics are complicated by arm exchange with endogenous IgG4, resulting in a mixture of a more potent intact, bivalent form and a less potent, functionally monovalent form. Total NAT and endogenous IgG4 concentrations vary considerably across patients. This study assessed the concentration of intact NAT, and how it relates to total NAT and endogenous IgG4 levels in blood and saliva. METHODS Paired serum and saliva samples from a small cohort of relapsing-remitting multiple sclerosis patients were measured for levels of intact NAT, total NAT, IgG and IgG4. RESULTS Intact NAT concentration was dependent on both total NAT and endogenous IgG4 levels. Low endogenous IgG4 led to a higher ratio of intact NAT to total NAT, while the opposite was observed in subjects with high endogenous IgG4. Serum and saliva measurements show good concordance. CONCLUSIONS Intact NAT concentration is influenced by both NAT pharmacokinetics and endogenous IgG4 levels. Patients with low IgG4 levels can have high concentrations of intact NAT even with lower levels of total NAT, which may explain cases of NAT-associated progressive multifocal leukoencephalopathy (PML) in such patients. Monitoring both forms of NAT could better guide dosing, maximizing drug efficacy and safety.
Collapse
Affiliation(s)
- Lesley J Page
- Abreos Biosciences, 3550 General Atomics Court(,) G02/rm140, San Diego, CA, USA.
| | - Iona F Pay
- Abreos Biosciences, 3550 General Atomics Court(,) G02/rm140, San Diego, CA, USA
| | - Edward T Castellana
- Abreos Biosciences, 3550 General Atomics Court(,) G02/rm140, San Diego, CA, USA
| | - Raphaela Heussen
- Abreos Biosciences, 3550 General Atomics Court(,) G02/rm140, San Diego, CA, USA
| | - Tamara Hoyt
- Rocky Mountain MS Clinic, 370 E 9th Ave, Salt Lake City, Utah 84103
| | - John Foley
- Rocky Mountain MS Clinic, 370 E 9th Ave, Salt Lake City, Utah 84103
| | - Bradley T Messmer
- Abreos Biosciences, 3550 General Atomics Court(,) G02/rm140, San Diego, CA, USA
| |
Collapse
|
6
|
Abdelrahman A, Alvarez E. Advances in Multiple Sclerosis Neurotherapeutics, Neuroprotection, and Risk Mitigation Strategies. Neurol Clin 2024; 42:115-135. [PMID: 37980110 DOI: 10.1016/j.ncl.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2023]
Abstract
The treatment of patients with relapsing multiple sclerosis (MS) has advanced tremendously over the past few decades. More efficacious therapies have been approved, which can significantly reduce the inflammatory process of relapsing MS. Neuroprotection by controlling this pathophysiology is important given our current limitations to control progressive MS and induce neurorepair. Here, the authors discuss the current landscape of neurotherapeutics for relapsing MS focusing on newer disease-modifying treatments and their use. Risk mitigation of these medications can greatly improve their safety and improve their benefit-risk balance. The authors discuss treatment strategies for risk mitigation including treatment discontinuation and de-escalation.
Collapse
Affiliation(s)
- Ahmad Abdelrahman
- Department of Neurology, Rocky Mountain MS Center at the University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Enrique Alvarez
- Department of Neurology, Rocky Mountain MS Center at the University of Colorado Anschutz Medical Center, University of Colorado, Aurora, CO, USA.
| |
Collapse
|
7
|
Frisch ES, Häusler D, Weber MS. Natalizumab Promotes Activation of Peripheral Monocytes in Patients With Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:10/4/e200114. [PMID: 37072216 PMCID: PMC10112857 DOI: 10.1212/nxi.0000000000200114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/16/2023] [Indexed: 04/20/2023]
Abstract
OBJECTIVES Natalizumab (NTZ), a monoclonal antibody against very late antigen-4 (VLA-4), is one of the most efficient therapies to prevent acute relapses in multiple sclerosis (MS). VLA-4 is the key adhesion molecule for peripheral immune cells, especially lymphocytes to enter the CNS. While its blockade thus virtually abrogates CNS infiltration of these cells, long-term exposure to natalizumab may also affect immune cell function. METHODS In this study, we report that in patients with MS, NTZ treatment is associated with an enhanced activation status of peripheral monocytes. RESULTS Expression of 2 independent activation markers, CD69 and CD150, was significantly higher on blood monocytes from NTZ-treated patients when compared with those from matched untreated patients with MS, while other properties such as cytokine production remained unchanged. DISCUSSION These findings consolidate the concept that peripheral immune cells remain fully competent on NTZ treatment, an excellent asset rare among MS treatments. However, they also suggest that NTZ may exert nondesirable effects on the progressive aspect of MS, where myeloid cells and their chronic activation are attributed a prominent pathophysiologic role.
Collapse
Affiliation(s)
- Esther S Frisch
- From the Department of Neuropathology (E.S.F., D.H., M.S.W.), University Medical Center; Department of Neurology (E.S.F., M.S.W.), University Medical Center; and Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP (D.H., M.S.W.), Göttingen, Germany
| | - Darius Häusler
- From the Department of Neuropathology (E.S.F., D.H., M.S.W.), University Medical Center; Department of Neurology (E.S.F., M.S.W.), University Medical Center; and Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP (D.H., M.S.W.), Göttingen, Germany
| | - Martin S Weber
- From the Department of Neuropathology (E.S.F., D.H., M.S.W.), University Medical Center; Department of Neurology (E.S.F., M.S.W.), University Medical Center; and Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP (D.H., M.S.W.), Göttingen, Germany.
| |
Collapse
|
8
|
Oliveira DS, Castro AR, Ruano L. Recurrent skin infections associated with natalizumab treatment. Neurol Sci 2023; 44:1093-1095. [PMID: 36331655 DOI: 10.1007/s10072-022-06481-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Daniela Santos Oliveira
- Serviço de Neurologia, Centro Hospitalar de Entre o Douro e Vouga, Rua Dr. Cândido Pinho, No. 5, 4520-220, Santa Maria da Feira, Portugal.
| | - Ana Rita Castro
- Serviço de Neurologia, Centro Hospitalar de Entre o Douro e Vouga, Rua Dr. Cândido Pinho, No. 5, 4520-220, Santa Maria da Feira, Portugal
| | - Luís Ruano
- Serviço de Neurologia, Centro Hospitalar de Entre o Douro e Vouga, Rua Dr. Cândido Pinho, No. 5, 4520-220, Santa Maria da Feira, Portugal
- Instituto de Saúde Pública, Universidade do Porto, Rua das Taipas, No. 135, 4050-600, Porto, Portugal
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Rua das Taipas, No. 135, 4050-600, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| |
Collapse
|
9
|
Barzegar M, Houshi S, Sadeghi E, Hashemi MS, Pishgahi G, Bagherieh S, Afshari-Safavi A, Mirmosayyeb O, Shaygannejad V, Zabeti A. Association of Disease-Modifying Therapies with COVID-19 Susceptibility and Severity in Patients with Multiple Sclerosis: A Systematic Review and Network Meta-Analysis. Mult Scler Int 2022; 2022:9388813. [PMID: 36187599 PMCID: PMC9519336 DOI: 10.1155/2022/9388813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/03/2022] [Indexed: 12/24/2022] Open
Abstract
Background We conducted this study to assess the effect of disease-modifying therapies (DMTs) on coronavirus disease (COVID-19) susceptibility and severity in people with multiple sclerosis (MS). Methods Available studies from PubMed, Scopus, EMBASE, Web of Science, and gray literature, including reference lists and conference abstracts, were searched from December 1, 2019, to July 26, 2021. We included cross-sectional, case-control, and cohort studies assessing the association of DMTs with risk of contracting COVID-19 or its outcomes in MS patients on univariate or multivariate regression analyses. We conducted a network meta-analysis (NMA) to compare the risk of COVID-19 and developing severe infection across DMTs. Results Out of the initial 3893 records and 1883 conference abstracts, a total of 10 studies were included. Pairwise comparisons showed that none of the DMTs meaningfully affect the risk of acquiring infection. There was significant total heterogeneity and inconsistency across this NMA. In comparison with no DMT, dimethyl fumarate (0.62 (0.42, 0.93)), fingolimod (0.55 (0.32, 0.94)), natalizumab (0.50 (0.31, 0.81)), and interferon (0.42 (0.22, 0.79)) were associated with a decreased risk of severe COVID-19; but, rituximab was observed to increase the risk (1.94 (1.20, 3.12)). Compared to rituximab or ocrelizumab, all DMTs were associated with a decreased risk. Pairwise comparisons showed no differences across other DMTs. Interferon and rituximab were associated with the lowest and highest risks of severe COVID-19. Conclusion Our study showed an increased risk of severe COVID-19 in patients on rituximab and ocrelizumab. No association with COVID-19 severity across other DMTs was observed.
Collapse
Affiliation(s)
- Mahdi Barzegar
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shakiba Houshi
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Erfan Sadeghi
- Department of Biostatistics and Epidemiology, Faculty of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mozhgan Sadat Hashemi
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasem Pishgahi
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Bagherieh
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Afshari-Safavi
- Department of Biostatistics and Epidemiology, Faculty of Health, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Omid Mirmosayyeb
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vahid Shaygannejad
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Aram Zabeti
- Department of Neurology and Rehabilitation Medicine, Waddell center in Multiple Sclerosis, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
10
|
Häusler D, Akgün K, Stork L, Lassmann H, Ziemssen T, Brück W, Metz I. CNS inflammation after natalizumab therapy for multiple sclerosis: A retrospective histopathological and CSF cohort study. Brain Pathol 2021; 31:e12969. [PMID: 33955606 PMCID: PMC8549024 DOI: 10.1111/bpa.12969] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/29/2021] [Accepted: 04/12/2021] [Indexed: 11/30/2022] Open
Abstract
Natalizumab, a recombinant humanized monoclonal antibody directed against the α4 subunit of the integrins α4ß1 and α4ß7, has been approved for the treatment of active relapsing-remitting MS. Although natalizumab is a highly beneficial drug that effectively reduces the risk of sustained disability progression and the rate of clinical relapses, some patients do not respond to it, and some are at higher risk of developing progressive multifocal leukoencephalopathy (PML). The histopathological effects after natalizumab therapy are still unknown. We, therefore, performed a detailed histological characterization of the CNS inflammatory cell infiltrate of 24 brain specimens from natalizumab treated patients, consisting of 20 biopsies and 4 autopsies and 21 MS controls. To complement the analysis, immune cells in blood and cerebrospinal fluid (CSF) of 30 natalizumab-treated patients and 42 MS controls were quantified by flow cytometry. Inflammatory infiltrates within lesions were mainly composed of T cells and macrophages, some B cells, plasma cells, and dendritic cells. There was no significant difference in the numbers of T cells or macrophages and microglial cells in lesions of natalizumab-treated patients as compared to controls. A shift towards cytotoxic T cells of a memory phenotype was observed in the CSF. Plasma cells were significantly increased in active demyelinating lesions of natalizumab-treated patients, but no correlation to clinical disability was observed. Dendritic cells within lesions were found to be reduced with longer ongoing therapy duration. Our findings suggest that natalizumab does not completely prevent immune cells from entering the CNS and is associated with an accumulation of plasma cells, the pathogenic and clinical significance of which is not known. As B cells are considered to serve as a reservoir of the JC virus, the observed plasma cell accumulation and reduction in dendritic cells in the CNS of natalizumab-treated patients may potentially play a role in PML development.
Collapse
Affiliation(s)
- Darius Häusler
- Institute of NeuropathologyUniversity Medical CenterGöttingenGermany
| | - Katja Akgün
- Department of NeurologyCenter of Clinical NeuroscienceCarl Gustav Carus University ClinicUniversity Hospital of DresdenDresdenGermany
| | - Lidia Stork
- Institute of NeuropathologyUniversity Medical CenterGöttingenGermany
| | - Hans Lassmann
- Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Tjalf Ziemssen
- Department of NeurologyCenter of Clinical NeuroscienceCarl Gustav Carus University ClinicUniversity Hospital of DresdenDresdenGermany
| | - Wolfgang Brück
- Institute of NeuropathologyUniversity Medical CenterGöttingenGermany
| | - Imke Metz
- Institute of NeuropathologyUniversity Medical CenterGöttingenGermany
| |
Collapse
|
11
|
Schlüter M, Oswald E, Winklmeier S, Meinl I, Havla J, Eichhorn P, Meinl E, Kümpfel T. Effects of Natalizumab Therapy on Intrathecal Immunoglobulin G Production Indicate Targeting of Plasmablasts. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/5/e1030. [PMID: 34210800 PMCID: PMC8265584 DOI: 10.1212/nxi.0000000000001030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/16/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVES To evaluate the long-term effects of natalizumab (NTZ) on different features of intrathecal immunoglobulin (Ig) synthesis in patients with multiple sclerosis (MS) and to quantify the expression of α4-integrin in stages of B-cell maturation. METHODS We combined a cross-sectional (49 NTZ-treated MS patients, mean treatment duration 5.1 years, and 47 untreated MS patients) and a longitudinal study (33 patients with MS before and during NTZ, mean treatment duration: 4.8 years), analyzing paired serum and CSF samples for IgG, IgA, and IgM levels, reactivity against selected viruses (measles virus, rubella virus, and varicella zoster virus [MRZ] reaction), and oligoclonal bands (OCBs). Banding patterns before and after therapy were directly compared by isoelectric focusing in 1 patient. In addition, we determined the expression of α4-integrin by FACS analysis on blood-derived B-cell subsets (plasmablasts, memory B cells, and naive B cells) of healthy controls. RESULTS In serum, NTZ decreased IgM and IgG, but not IgA, levels. IgM hypogammaglobulinemia occurred in 28% of NTZ-treated patients. In CSF, NTZ treatment resulted in a strong reduction of intrathecally produced IgG and, to a lesser extent, IgA, whereas IgM indices [(Ig CSF/Serum)/(Albumin CSF/Serum)] remained largely unchanged. Reduction of the IgG index correlated with NTZ treatment duration, as did serum IgM and IgA levels. MRZ reaction was unchanged and OCB persisted. Direct comparison of OCB pattern before and after NTZ revealed the persistence of individual bands. α4-Integrin expression was highest on plasmablasts (CD19+CD38+CD27+). CONCLUSION Our data indicate that NTZ reduces short-lived plasmablasts in the CNS compartment but has little effect on locally persisting long-lived plasma cells.
Collapse
Affiliation(s)
- Miriam Schlüter
- From the Institute of Clinical Neuroimmunology (M.S., E.O., S.W., I.M., J.H., E.M., T.K.), Biomedical Center and LMU Klinikum; and Institute of Laboratory Medicine (P.E.), LMU Klinikum, Munich, Germany
| | - Eva Oswald
- From the Institute of Clinical Neuroimmunology (M.S., E.O., S.W., I.M., J.H., E.M., T.K.), Biomedical Center and LMU Klinikum; and Institute of Laboratory Medicine (P.E.), LMU Klinikum, Munich, Germany
| | - Stephan Winklmeier
- From the Institute of Clinical Neuroimmunology (M.S., E.O., S.W., I.M., J.H., E.M., T.K.), Biomedical Center and LMU Klinikum; and Institute of Laboratory Medicine (P.E.), LMU Klinikum, Munich, Germany
| | - Ingrid Meinl
- From the Institute of Clinical Neuroimmunology (M.S., E.O., S.W., I.M., J.H., E.M., T.K.), Biomedical Center and LMU Klinikum; and Institute of Laboratory Medicine (P.E.), LMU Klinikum, Munich, Germany
| | - Joachim Havla
- From the Institute of Clinical Neuroimmunology (M.S., E.O., S.W., I.M., J.H., E.M., T.K.), Biomedical Center and LMU Klinikum; and Institute of Laboratory Medicine (P.E.), LMU Klinikum, Munich, Germany
| | - Peter Eichhorn
- From the Institute of Clinical Neuroimmunology (M.S., E.O., S.W., I.M., J.H., E.M., T.K.), Biomedical Center and LMU Klinikum; and Institute of Laboratory Medicine (P.E.), LMU Klinikum, Munich, Germany
| | - Edgar Meinl
- From the Institute of Clinical Neuroimmunology (M.S., E.O., S.W., I.M., J.H., E.M., T.K.), Biomedical Center and LMU Klinikum; and Institute of Laboratory Medicine (P.E.), LMU Klinikum, Munich, Germany
| | - Tania Kümpfel
- From the Institute of Clinical Neuroimmunology (M.S., E.O., S.W., I.M., J.H., E.M., T.K.), Biomedical Center and LMU Klinikum; and Institute of Laboratory Medicine (P.E.), LMU Klinikum, Munich, Germany.
| |
Collapse
|
12
|
Natalizumab differentially affects plasmablasts and B cells in multiple sclerosis. Mult Scler Relat Disord 2021; 52:102987. [PMID: 33984651 DOI: 10.1016/j.msard.2021.102987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/28/2021] [Accepted: 04/26/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Natalizumab treatment increases the frequencies of B cells in blood but reduces IgG in blood and CSF. Plasmablasts are important in the production of IgG, and the development of plasmablasts is CD49d dependent. OBJECTIVE We hypothesized that natalizumab treatment affects the development of plasmablasts. METHODS We retrospectively analyzed frequencies and absolute counts of B cell subsets by flow cytometry from a longitudinal cohort of 9 progressive multiple sclerosis (MS) patients treated with natalizumab for 60 weeks, and a cross-sectional relapsing-remitting MS (RRMS) cohort with 17 untreated and 37 treated with natalizumab (17 stable and 20 unstable patients with relapse activity). Additionally, CD49d expression on B cell subsets was examined in 10 healthy controls, and blood and cerebrospinal fluid (CSF) frequencies of B cell subsets were quantified in untreated and natalizumab treated RRMS patients. RESULTS In progressive MS, levels of IgG decreased in plasma (p<0.001) from baseline to 60 weeks follow-up. In the progressive MS and RRMS cohorts we observed that natalizumab treatment significantly increased the frequency of B cells (p=0.004; p<0.0001) and several B cell subsets, most pronounced for memory B cell subsets (p=0.0001; p<0.0001), while there was a decrease in plasmablast frequency (p=0.008; p=0.008). In both progressive MS and RRMS the absolute cell counts of B cells increased (p=0.004; p<0.001), which was explained by a significant increase in all subsets, except for plasmablasts. Furthermore, we found decreased memory B cell counts in unstable compared to stable natalizumab-treated patients (p=0.02). The expression of CD49d was higher on plasmablasts compared to other B cell subsets (p<0.0001). In CSF, plasmablasts could not be detected in patients treated with natalizumab, in contrast to an increased frequency in untreated RRMS patients. CONCLUSION We confirm previous studies showing that natalizumab increases circulating number of B cells, particularly memory cells, concomitant with a decrease in plasma IgG concentrations. Moreover, we demonstrate in two separate cohorts that natalizumab treatment markedly decreases frequencies of plasmablasts while the absolute number is stable. Additionally, plasmablasts have high expression of CD49d, and plasmablasts could not be detected in the CSF of natalizumab-treated patients. Finally, memory B cells were found to be reduced in unstable natalizumab-treated patients, which could possibly indicate increased recruitment to the CNS.
Collapse
|
13
|
Di Pilato M, Palomino-Segura M, Mejías-Pérez E, Gómez CE, Rubio-Ponce A, D'Antuono R, Pizzagalli DU, Pérez P, Kfuri-Rubens R, Benguría A, Dopazo A, Ballesteros I, Sorzano COS, Hidalgo A, Esteban M, Gonzalez SF. Neutrophil subtypes shape HIV-specific CD8 T-cell responses after vaccinia virus infection. NPJ Vaccines 2021; 6:52. [PMID: 33846352 PMCID: PMC8041892 DOI: 10.1038/s41541-021-00314-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 03/09/2021] [Indexed: 12/20/2022] Open
Abstract
Neutrophils are innate immune cells involved in the elimination of pathogens and can also induce adaptive immune responses. Nα and Nβ neutrophils have been described with distinct in vitro capacity to generate antigen-specific CD8 T-cell responses. However, how these cell types exert their role in vivo and how manipulation of Nβ/Nα ratio influences vaccine-mediated immune responses are not known. In this study, we find that these neutrophil subtypes show distinct migratory and motility patterns and different ability to interact with CD8 T cells in the spleen following vaccinia virus (VACV) infection. Moreover, after analysis of adhesion, inflammatory, and migration markers, we observe that Nβ neutrophils overexpress the α4β1 integrin compared to Nα. Finally, by inhibiting α4β1 integrin, we increase the Nβ/Nα ratio and enhance CD8 T-cell responses to HIV VACV-delivered antigens. These findings provide significant advancements in the comprehension of neutrophil-based control of adaptive immune system and their relevance in vaccine design.
Collapse
Affiliation(s)
- Mauro Di Pilato
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland. .,Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología-CSIC, Madrid, Spain. .,Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA. .,Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Miguel Palomino-Segura
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Ernesto Mejías-Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología-CSIC, Madrid, Spain.,Max von Pettenkofer-Institute, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Carmen E Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología-CSIC, Madrid, Spain
| | - Andrea Rubio-Ponce
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.,Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Rocco D'Antuono
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Crick Advanced Light Microscopy Science and Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Diego Ulisse Pizzagalli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Institute of Computational Science, Università della Svizzera Italiana, Lugano, Switzerland
| | - Patricia Pérez
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología-CSIC, Madrid, Spain
| | - Raphael Kfuri-Rubens
- Center of Integrated Protein Science Munich and Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Alberto Benguría
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Ana Dopazo
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Iván Ballesteros
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Carlos Oscar S Sorzano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología-CSIC, Madrid, Spain
| | - Andrés Hidalgo
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología-CSIC, Madrid, Spain.
| | - Santiago F Gonzalez
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.
| |
Collapse
|
14
|
Eliseeva DD, Vasiliev AV, Abramova AA, Kochergin IA, Zakharova MN. [Monoclonal antibody therapies for rapidly progressive and highly active multiple sclerosis in the era of the COVID-19 pandemic]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:31-36. [PMID: 34387443 DOI: 10.17116/jnevro202112107231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
As the COVID-19 pandemic continues, reducing the risk of infection for immunocompromised patients remains an important issue. Patients with aggressive multiple sclerosis (MS) require immunosuppressive therapy in order to control the overactive autoimmune response. Preliminary international and national trials demonstrate that older age, higher disability status and progressive MS are generally associated with a more severe clinical course of COVID-19. However, uncertainty remains about the effect of disease-modifying therapies on the COVID-19 clinical presentation. In this article, we pay special attention to monoclonal antibodies used for immune reconstitution therapy, which results in significant changes to the T-cell and/or B-cell repertoire. Based on the published data from registries in different countries, we attempted to estimate the benefits and risks of these therapies in a complicated epidemiological setting.
Collapse
Affiliation(s)
| | - A V Vasiliev
- «Neuroclinic» (Yusupov Hospital), Moscow, Russia
| | | | | | | |
Collapse
|
15
|
Bhise V, Dhib-Jalbut S. Potential Risks and Benefits of Multiple Sclerosis Immune Therapies in the COVID-19 Era: Clinical and Immunological Perspectives. Neurotherapeutics 2021; 18:244-251. [PMID: 33533012 PMCID: PMC7853164 DOI: 10.1007/s13311-021-01008-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2021] [Indexed: 02/07/2023] Open
Abstract
Coronavirus SARS-CoV2 has emerged as one of the greatest infectious disease health challenges in a century. Patients with multiple sclerosis (MS) have a particular vulnerability to infections through their use of immunosuppressive disease-modifying therapies (DMTs). Specific DMTs pose particular risk based on their mechanisms of action (MOA). As a result, patients require individualized approaches to starting new treatments and continuation of therapy. Additionally, vaccinations must be considered carefully, and individuals on long-term B cell-depleting therapies may have diminished immune responses to vaccination, based on preserved T cells and diminished but present antibody titers to influenza vaccines. We review the immunology behind these treatments and their impact on COVID-19, as well as the current recommendations for best practices for use of DMTs in patients with MS.
Collapse
Affiliation(s)
- Vikram Bhise
- Departments of Pediatrics, Rutgers Robert Wood Johnson Medical School, 89 French Street, Suite 2200, New Brunswick, NJ, 08901, USA.
| | - Suhayl Dhib-Jalbut
- Department of Neurology, Rutgers Robert Wood Johnson Medical School, 125 Paterson Street, Suite 65000, New Brunswick, NJ, 08901, USA
| |
Collapse
|
16
|
Zrzavy T, Wimmer I, Rommer PS, Berger T. Immunology of COVID-19 and disease-modifying therapies: The good, the bad and the unknown. Eur J Neurol 2020; 28:3503-3516. [PMID: 33090599 PMCID: PMC7675490 DOI: 10.1111/ene.14578] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/04/2020] [Indexed: 01/08/2023]
Abstract
Objective The outbreak of the SARS‐CoV‐2 pandemic, caused by a previously unknown infectious agent, posed unprecedented challenges to healthcare systems and unmasked their vulnerability and limitations worldwide. Patients with long‐term immunomodulatory/suppressive therapies, as well as their physicians, were and are concerned about balancing the risk of infection and effects of disease‐modifying therapy. Over the last few months, knowledge regarding SARS‐CoV‐2 has been growing tremendously, and the first experiences of infections in patients with multiple sclerosis (MS) have been reported. Methods This review summarizes the currently still limited knowledge about SARS‐CoV‐2 immunology and the commonly agreed modes of action of approved drugs in immune‐mediated diseases of the central nervous system (MS and neuromyelitis optica spectrum disorder). Specifically, we discuss whether immunosuppressive/immunomodulatory drugs may increase the risk of SARS‐CoV‐2 infection and, conversely, may decrease the severity of a COVID‐19 disease course. Results At present, it can be recommended in general that none of those therapies with a definite indication needs to be stopped per se. A possibly increased risk of infection for most medications is accompanied by the possibility to reduce the severity of COVID‐19. Conclusions Despite the knowledge gain over the last few months, current evidence remains limited, and, thus, further clinical vigilance and systematic documentation is essential.
Collapse
Affiliation(s)
- Tobias Zrzavy
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Isabella Wimmer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Paulus S Rommer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
17
|
Khoy K, Mariotte D, Defer G, Petit G, Toutirais O, Le Mauff B. Natalizumab in Multiple Sclerosis Treatment: From Biological Effects to Immune Monitoring. Front Immunol 2020; 11:549842. [PMID: 33072089 PMCID: PMC7541830 DOI: 10.3389/fimmu.2020.549842] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis is a chronic demyelinating disease of the central nervous system (CNS) with an autoimmune component. Among the recent disease-modifying treatments available, Natalizumab, a monoclonal antibody directed against the alpha chain of the VLA-4 integrin (CD49d), is a potent inhibitor of cell migration toward the tissues including CNS. It potently reduces relapses and active brain lesions in the relapsing remitting form of the disease. However, it has also been associated with a severe infectious complication, the progressive multifocal leukoencephalitis (PML). Using the standard protocol with an injection every 4 weeks it has been shown by a close monitoring of the drug that trough levels soon reach a plateau with an almost saturation of the target cell receptor as well as a down modulation of this receptor. In this review, mechanisms of action involved in therapeutic efficacy as well as in PML risk will be discussed. Furthermore the interest of a biological monitoring that may be helpful to rapidly adapt treatment is presented. Indeed, development of anti-NAT antibodies, although sometimes unapparent, can be detected indirectly by normalization of CD49d expression on circulating mononuclear cells and might require to switch to another drug. On the other hand a stable modulation of CD49d expression might be useful to follow the circulating NAT levels and apply an extended interval dose scheme that could contribute to limiting the risk of PML.
Collapse
Affiliation(s)
- Kathy Khoy
- Laboratory of Immunology, Department of Biology, CHU Caen Normandie, Caen, France
| | - Delphine Mariotte
- Laboratory of Immunology, Department of Biology, CHU Caen Normandie, Caen, France
| | - Gilles Defer
- Department of Neurology, MS Expert Centre, CHU Caen Normandie, Caen, France.,UMR-S1237, Physiopathology and Imaging of Neurological Disorders, INSERM, Caen, France.,Normandie Université, UNICAEN, Caen, France
| | - Gautier Petit
- Laboratory of Immunology, Department of Biology, CHU Caen Normandie, Caen, France
| | - Olivier Toutirais
- Laboratory of Immunology, Department of Biology, CHU Caen Normandie, Caen, France.,UMR-S1237, Physiopathology and Imaging of Neurological Disorders, INSERM, Caen, France.,Normandie Université, UNICAEN, Caen, France
| | - Brigitte Le Mauff
- Laboratory of Immunology, Department of Biology, CHU Caen Normandie, Caen, France.,UMR-S1237, Physiopathology and Imaging of Neurological Disorders, INSERM, Caen, France.,Normandie Université, UNICAEN, Caen, France
| |
Collapse
|
18
|
Korsukewitz C, Reddel SW, Bar-Or A, Wiendl H. Neurological immunotherapy in the era of COVID-19 - looking for consensus in the literature. Nat Rev Neurol 2020; 16:493-505. [PMID: 32641860 PMCID: PMC7341707 DOI: 10.1038/s41582-020-0385-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2020] [Indexed: 01/08/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is concerning for patients with neuroimmunological diseases who are receiving immunotherapy. Uncertainty remains about whether immunotherapies increase the risk of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or increase the risk of severe disease and death upon infection. National and international societies have developed guidelines and statements, but consensus does not exist in several areas. In this Review, we attempt to clarify where consensus exists and where uncertainty remains to inform management approaches based on the first principles of neuroimmunology. We identified key questions that have been addressed in the literature and collated the recommendations to generate a consensus calculation in a Delphi-like approach to summarize the information. We summarize the international recommendations, discuss them in light of the first available data from patients with COVID-19 receiving immunotherapy and provide an overview of management approaches in the COVID-19 era. We stress the principles of medicine in general and neuroimmunology in particular because, although the risk of viral infection has become more relevant, most of the considerations apply to the general management of neurological immunotherapy. We also give special consideration to immunosuppressive treatment and cell-depleting therapies that might increase susceptibility to SARS-CoV-2 infection but reduce the risk of severe COVID-19.
Collapse
Affiliation(s)
- Catharina Korsukewitz
- Department of Neurology with Institute of Translational Neurology, University of Muenster, Muenster, Germany
| | - Stephen W Reddel
- Department of Neurology, Concord Hospital and The Brain and Mind Centre, University of Sydney, Sydney, Australia
| | - Amit Bar-Or
- Center for Neuroinflammation and Neurotherapeutics and the Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University of Muenster, Muenster, Germany.
| |
Collapse
|
19
|
Risiken und Chancen von Immuntherapien in Zeiten der Coronavirus-2019-Pandemie. DGNEUROLOGIE 2020. [PMCID: PMC7284681 DOI: 10.1007/s42451-020-00205-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Immuntherapien stellen die essenzielle Grundlage der Behandlung von neuroinflammatorischen Erkrankungen dar. In Zeiten der Coronavirus-2019 (COVID-19)-Pandemie ergibt sich im klinischen Alltag jedoch zunehmend die Frage, ob eine Immuntherapie bei neurologischen Patienten aufgrund des potenziellen Infektionsrisikos eingeleitet, intensiviert, pausiert oder gar beendet werden sollte. Unsicherheit besteht v. a. deshalb, weil verschiedene nationale und internationale Fachgesellschaften diesbezüglich unterschiedliche Empfehlungen veröffentlichten. In diesem Artikel soll ein Überblick über die Wirkmechanismen von Immuntherapien und den daraus abzuleitenden Infektionsrisiken in Bezug auf COVID-19 (durch den Coronavirus verursachte Erkrankung) gegeben werden. Potenzielle Chancen und vorteilhafte Effekte einzelner Substrate in der Akuttherapie von COVID-19 werden diskutiert.
Collapse
|
20
|
Marques PT, Kay CSK, Basílio FMA, Pinheiro RL, Werneck LC, Lorenzoni PJ, Scola RH. Localized sporotrichosis during natalizumab treatment in Multiple Sclerosis. Mult Scler Relat Disord 2020; 41:102029. [DOI: 10.1016/j.msard.2020.102029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/23/2020] [Accepted: 02/25/2020] [Indexed: 10/24/2022]
|
21
|
Pawlitzki M, Zettl UK, Ruck T, Rolfes L, Hartung HP, Meuth SG. Merits and culprits of immunotherapies for neurological diseases in times of COVID-19. EBioMedicine 2020; 56:102822. [PMID: 32535547 PMCID: PMC7286830 DOI: 10.1016/j.ebiom.2020.102822] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022] Open
Abstract
Immunosuppression and immunomodulation are valuable therapeutic approaches for managing neuroimmunological diseases. In times of the Coronavirus disease 2019 (COVID-19) pandemic, clinicians must deal with the question of whether immunotherapy should currently be initiated or discontinued in neurological patients. Uncertainty exists especially because different national medical associations publish different recommendations on the extent to which immunotherapies must be continued, monitored, or possibly switched during the current pandemic. Based on the most recently available data both about the novel coronavirus and the approved immunotherapies for neurological diseases, we provide an updated overview that includes current treatment strategies and the associated COVID-19 risk, but also the potential of immunotherapies to treat COVID-19.
Collapse
Affiliation(s)
- Marc Pawlitzki
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany.
| | - Uwe K Zettl
- Department of Neurology, Neuroimmunological Section, University of Rostock, Rostock, Germany
| | - Tobias Ruck
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Leoni Rolfes
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany.
| |
Collapse
|
22
|
Petersen E, Ammitzbøll C, Søndergaard H, Oturai A, Sørensen P, Nilsson A, Börnsen L, von Essen M, Sellebjerg F. Expression of melanoma cell adhesion molecule-1 (MCAM-1) in natalizumab-treated multiple sclerosis. J Neuroimmunol 2019; 337:577085. [DOI: 10.1016/j.jneuroim.2019.577085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 10/06/2019] [Accepted: 10/07/2019] [Indexed: 12/21/2022]
|
23
|
Traub JW, Pellkofer HL, Grondey K, Seeger I, Rowold C, Brück W, Husseini L, Häusser-Kinzel S, Weber MS. Natalizumab promotes activation and pro-inflammatory differentiation of peripheral B cells in multiple sclerosis patients. J Neuroinflammation 2019; 16:228. [PMID: 31733652 PMCID: PMC6858649 DOI: 10.1186/s12974-019-1593-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/23/2019] [Indexed: 12/29/2022] Open
Abstract
Background In the past, multiple sclerosis (MS) medications have been primarily designed to modulate T cell properties. Based on the emerging concept that B cells are equally important for the propagation of MS, we compared the effect of four commonly used, primarily T cell-targeting MS medications on B cells. Methods Using flow cytometry, we analyzed peripheral blood mononuclear cells (PBMC) of untreated (n = 19) and dimethyl fumarate (DMF; n = 21)-, fingolimod (FTY; n = 17)-, glatiramer acetate (GA; n = 18)-, and natalizumab (NAT; n = 20)-treated MS patients, focusing on B cell maturation, differentiation, and cytokine production. Results While GA exerted minor effects on the investigated B cell properties, DMF and FTY robustly inhibited pro-inflammatory B cell function. In contrast, NAT treatment enhanced B cell differentiation, activation, and pro-inflammatory cytokine production when compared to both intraindividual samples collected before NAT treatment initiation as well as untreated MS controls. Our mechanistic in vitro studies confirm this observation. Conclusion Our data indicate that common MS medications have differential, in part opposing effects on B cells. The observed activation of peripheral B cells upon NAT treatment may be instructive to interpret its unfavorable effect in certain B cell-mediated inflammatory conditions and to elucidate the immunological basis of MS relapses after NAT withdrawal. Trial registration Protocols were approved by the ethical review committee of the University Medical Center Göttingen (#3/4/14).
Collapse
Affiliation(s)
- Jan W Traub
- Institute of Neuropathology, University Medical Center, Robert-Koch-Straße 40, 37099 Göttingen, Germany.,Department of Neurology, University Medical Center, Robert-Koch-Straße 40, 37099 Göttingen, Germany
| | - Hannah L Pellkofer
- Department of Neurology, University Medical Center, Robert-Koch-Straße 40, 37099 Göttingen, Germany.,Institute of Clinical Neuroimmunology, Ludwig Maximilian University, Großhaderner Straße 9, 82152 Munich, Germany
| | - Katja Grondey
- Institute of Neuropathology, University Medical Center, Robert-Koch-Straße 40, 37099 Göttingen, Germany
| | - Ira Seeger
- Institute of Neuropathology, University Medical Center, Robert-Koch-Straße 40, 37099 Göttingen, Germany
| | - Christoph Rowold
- Institute of Neuropathology, University Medical Center, Robert-Koch-Straße 40, 37099 Göttingen, Germany.,Department of Neurology, University Medical Center, Robert-Koch-Straße 40, 37099 Göttingen, Germany
| | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center, Robert-Koch-Straße 40, 37099 Göttingen, Germany
| | - Leila Husseini
- Department of Neurology, University Medical Center, Robert-Koch-Straße 40, 37099 Göttingen, Germany
| | - Silke Häusser-Kinzel
- Institute of Neuropathology, University Medical Center, Robert-Koch-Straße 40, 37099 Göttingen, Germany
| | - Martin S Weber
- Institute of Neuropathology, University Medical Center, Robert-Koch-Straße 40, 37099 Göttingen, Germany. .,Department of Neurology, University Medical Center, Robert-Koch-Straße 40, 37099 Göttingen, Germany.
| |
Collapse
|
24
|
Largey F, Jelcic I, Sospedra M, Heesen C, Martin R, Jelcic I. Effects of natalizumab therapy on intrathecal antiviral antibody responses in MS. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2019; 6:6/6/e621. [PMID: 31554671 PMCID: PMC6807967 DOI: 10.1212/nxi.0000000000000621] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 07/31/2019] [Indexed: 12/25/2022]
Abstract
Objective To investigate the effects of natalizumab (NAT) treatment on intrathecally produced antiviral antibodies in MS. Methods We performed a longitudinal, observational study analyzing both serum and CSF samples collected before and during NAT treatment for antibodies against measles, rubella, mumps, influenza, entero, herpes, and polyoma viruses, including JC polyomavirus (JCV) and its nearest homologue BK polyomavirus (BKV), and bacterial control antigens by ELISA to determine the antigen-specific CSF antibody index (CAI). CAI ≥1.5 indicated intrathecal synthesis of antigen-specific antibodies. Oligoclonal bands (OCBs) by isoelectric focusing and total IgG, IgM, and IgA by immunonephelometry were analyzed additionally. Results Intrathecal synthesis of JCV- and BKV-specific IgG was detected in 20% of patients with MS at baseline and was lost significantly more frequently during NAT treatment compared with other intrathecal antiviral and antibacterial antibody reactivities. Peripheral JCV- and BKV-specific antibody responses persisted, and no cross-reactivity between JCV- and BKV-specific CSF antibodies was found. Intrathecal production of antibodies against measles, rubella, and zoster antigens (MRZ reaction) was most prevalent and persisted (73.3% before vs 66.7% after 1 year of NAT therapy). CSF OCBs also persisted (93.3% vs 80.0%), but total CSF IgG and IgM levels declined significantly. Conclusions These data indicate that JCV-specific antibodies are produced intrathecally in a minority of patients with MS, and NAT treatment affects the intrathecal humoral immune response against JCV relatively specifically compared with other neurotropic viruses. Further studies are needed to determine whether this effect translates to higher risk of progressive multifocal leukoencephalopathy development.
Collapse
Affiliation(s)
- Fabienne Largey
- From the Neuroimmunology and Multiple Sclerosis Research Section (F.L., Ivan Jelcic, M.S., R.M., Ilijas Jelcic), Department of Neurology, University Hospital of Zurich, Switzerland; and Institute for Neuroimmunology and Multiple Sclerosis (inims) (C.H.), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ivan Jelcic
- From the Neuroimmunology and Multiple Sclerosis Research Section (F.L., Ivan Jelcic, M.S., R.M., Ilijas Jelcic), Department of Neurology, University Hospital of Zurich, Switzerland; and Institute for Neuroimmunology and Multiple Sclerosis (inims) (C.H.), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mireia Sospedra
- From the Neuroimmunology and Multiple Sclerosis Research Section (F.L., Ivan Jelcic, M.S., R.M., Ilijas Jelcic), Department of Neurology, University Hospital of Zurich, Switzerland; and Institute for Neuroimmunology and Multiple Sclerosis (inims) (C.H.), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Heesen
- From the Neuroimmunology and Multiple Sclerosis Research Section (F.L., Ivan Jelcic, M.S., R.M., Ilijas Jelcic), Department of Neurology, University Hospital of Zurich, Switzerland; and Institute for Neuroimmunology and Multiple Sclerosis (inims) (C.H.), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roland Martin
- From the Neuroimmunology and Multiple Sclerosis Research Section (F.L., Ivan Jelcic, M.S., R.M., Ilijas Jelcic), Department of Neurology, University Hospital of Zurich, Switzerland; and Institute for Neuroimmunology and Multiple Sclerosis (inims) (C.H.), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ilijas Jelcic
- From the Neuroimmunology and Multiple Sclerosis Research Section (F.L., Ivan Jelcic, M.S., R.M., Ilijas Jelcic), Department of Neurology, University Hospital of Zurich, Switzerland; and Institute for Neuroimmunology and Multiple Sclerosis (inims) (C.H.), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
25
|
Chisari CG, Toscano S, D’Amico E, Lo Fermo S, Zanghì A, Arena S, Zappia M, Patti F. An update on the safety of treating relapsing-remitting multiple sclerosis. Expert Opin Drug Saf 2019; 18:925-948. [DOI: 10.1080/14740338.2019.1658741] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Clara G. Chisari
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Simona Toscano
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Emanuele D’Amico
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Salvatore Lo Fermo
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Aurora Zanghì
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Sebastiano Arena
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Mario Zappia
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Francesco Patti
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| |
Collapse
|
26
|
Evaluation of natalizumab pharmacokinetics and pharmacodynamics with standard and extended interval dosing. Mult Scler Relat Disord 2019; 31:65-71. [DOI: 10.1016/j.msard.2019.03.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/18/2019] [Accepted: 03/23/2019] [Indexed: 12/22/2022]
|
27
|
Yshii L, Pignolet B, Mauré E, Pierau M, Brunner-Weinzierl M, Hartley O, Bauer J, Liblau R. IFN-γ is a therapeutic target in paraneoplastic cerebellar degeneration. JCI Insight 2019; 4:127001. [PMID: 30944244 DOI: 10.1172/jci.insight.127001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/21/2019] [Indexed: 12/18/2022] Open
Abstract
Paraneoplastic neurological disorders result from an autoimmune response against neural self-antigens that are ectopically expressed in neoplastic cells. In paraneoplastic disorders associated to autoantibodies against intracellular proteins, such as paraneoplastic cerebellar degeneration (PCD), current data point to a major role of cell-mediated immunity. In an animal model, in which a neo-self-antigen was expressed in both Purkinje neurons and implanted breast tumor cells, immune checkpoint blockade led to complete tumor control at the expense of cerebellum infiltration by T cells and Purkinje neuron loss, thereby mimicking PCD. Here, we identify 2 potential therapeutic targets expressed by cerebellum-infiltrating T cells in this model, namely α4 integrin and IFN-γ. Mice with PCD were treated with anti-α4 integrin antibodies or neutralizing anti-IFN-γ antibodies at the onset of neurological signs. Although blocking α4 integrin had little or no impact on disease development, treatment using the anti-IFN-γ antibody led to almost complete protection from PCD. These findings strongly suggest that the production of IFN-γ by cerebellum-invading T cells plays a major role in Purkinje neuron death. Our successful preclinical use of neutralizing anti-IFN-γ antibody for the treatment of PCD offers a potentially new therapeutic opportunity for cancer patients at the onset of paraneoplastic neurological disorders.
Collapse
Affiliation(s)
- Lidia Yshii
- Centre de Physiopathologie Toulouse-Purpan (CPTP), Université de Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Béatrice Pignolet
- Centre de Physiopathologie Toulouse-Purpan (CPTP), Université de Toulouse, CNRS, Inserm, UPS, Toulouse, France.,Department of Clinical Neurosciences, Toulouse University Hospital, Toulouse, France
| | - Emilie Mauré
- Centre de Physiopathologie Toulouse-Purpan (CPTP), Université de Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Mandy Pierau
- Department of Experimental Pediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Monika Brunner-Weinzierl
- Department of Experimental Pediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Oliver Hartley
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jan Bauer
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Roland Liblau
- Centre de Physiopathologie Toulouse-Purpan (CPTP), Université de Toulouse, CNRS, Inserm, UPS, Toulouse, France
| |
Collapse
|
28
|
Li R, Bar-Or A. The Multiple Roles of B Cells in Multiple Sclerosis and Their Implications in Multiple Sclerosis Therapies. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a029108. [PMID: 29661809 DOI: 10.1101/cshperspect.a029108] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Increasing evidence has suggested that both antibody-dependent and antibody-independent functions of B cells are involved in multiple sclerosis (MS). The contrasting results of distinct B-cell targeting therapies in MS patients underscores the importance of elucidating these multiple B-cell functions. In this review, we discuss the generation of autoreactive B cells, migration of B cells into the central nervous system (CNS), and how different functions of B cells may contribute to MS disease activity and potentially mitigation in both the periphery and CNS compartments. In addition, we propose several future therapeutic strategies that may better target/shape B-cell responses for long-term treatment of MS.
Collapse
Affiliation(s)
- Rui Li
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Amit Bar-Or
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
29
|
Regulation of cell migration by α4 and α9 integrins. Biochem J 2019; 476:705-718. [PMID: 30819933 DOI: 10.1042/bcj20180415] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/09/2019] [Accepted: 02/12/2019] [Indexed: 12/15/2022]
Abstract
Integrins are heterodimeric transmembrane receptors that play an essential role in enabling cells to sense and bind to extracellular ligands. Activation and clustering of integrins leads to the formation of focal adhesions at the plasma membrane that subsequently initiate signalling pathways to control a broad range of functional endpoints including cell migration, proliferation and survival. The α4 and α9 integrins form a small sub-family of receptors that share some specific ligands and binding partners. Although relatively poorly studied compared with other integrin family members, emerging evidence suggests that despite restricted cell and tissue expression profiles, these integrins play a key role in the regulation of signalling pathways controlling cytoskeletal remodelling and migration in both adherent and non-adherent cell types. This review summarises the known shared and specific roles for α4 and α9 integrins and highlights the importance of these receptors in controlling cell migration within both homeostatic and disease settings.
Collapse
|
30
|
Khachanova NV. Highly active multiple sclerosis: options for monoclonal antibody therapy. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:49-57. [DOI: 10.17116/jnevro20191191049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
31
|
Kaufmann M, Haase R, Proschmann U, Ziemssen T, Akgün K. Real-World Lab Data in Natalizumab Treated Multiple Sclerosis Patients Up to 6 Years Long-Term Follow Up. Front Neurol 2018; 9:1071. [PMID: 30581413 PMCID: PMC6292961 DOI: 10.3389/fneur.2018.01071] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/23/2018] [Indexed: 12/12/2022] Open
Abstract
Natalizumab inhibits the transmigration of immune cells across the blood-brain barrier thus inhibiting inflammation in the central nervous system. Generally, this blockade at the blood-brain barrier has significant influence on the circulating lymphocytes. Up to date, only short-term data on peripheral blood parameters are available which are mostly from controlled clinical trials and not from real-world experience. Real-world lab data of 120 patients diagnosed with highly active disease course of relapsing-remitting multiple sclerosis (RRMS) were analyzed during natalizumab treatment. Patient sampling was performed by consecutive recruitment in the Multiple Sclerosis Center Dresden. Lab testing was performed before and at every third infusion up to 72 months follow-up. After first natalizumab infusion, absolute numbers of all major lymphocyte populations including CD4+ T-cells, CD8+ T-cells, CD19+ B-cells, and NK-cells significantly increased and remained stable during the whole observation period of 72 months. Upon lymphocyte subsets, CD19+ B-cells presented a disproportionate increase up to levels higher than normal level in most of the treated patients. Neutralizing antibodies to natalizumab abrogated the described changes. Intra-individual variation of lymphocytes and its subsets remained in a narrow range for the whole treatment period. CD4/CD8 ratio did not change compared to baseline measurement up to 6 years of natalizumab treatment. Monocytes, eosinophils, and basophils, but not neutrophils persistently increased during natalizumab treatment. Hematological parameters including erythrocyte, platelet count, hemoglobin, and hematocrit remained unchanged compared to baseline. Interestingly, immature precursor cells including erythroblasts were detectable in 36,8% of the treated patients during natalizumab therapy, but not in the pretreatment period. Asymptomatic elevations of liver enzymes were rare, mostly only transient and lower than 3x upper normal limit. Kidney function parameters remained stable within physiological ranges in most patients. CRP levels >20 mg/dl were recognized only in 10 patients during natalizumab therapy and were mostly linked to respiratory tract infections. In our present analysis, we report persistent, but stable increases of peripheral immune cell subtypes in natalizumab treated patients. Additional serological analyses confirm excellent tolerability and safety even 6 years after natalizumab initiation in post-marketing experience.
Collapse
Affiliation(s)
- Maxi Kaufmann
- MS Center Dresden, Center of Clinical Neuroscience, Carl Gustav Carus University Hospital, University of Technology Dresden, Dresden, Germany
| | - Rocco Haase
- MS Center Dresden, Center of Clinical Neuroscience, Carl Gustav Carus University Hospital, University of Technology Dresden, Dresden, Germany
| | - Undine Proschmann
- MS Center Dresden, Center of Clinical Neuroscience, Carl Gustav Carus University Hospital, University of Technology Dresden, Dresden, Germany
| | - Tjalf Ziemssen
- MS Center Dresden, Center of Clinical Neuroscience, Carl Gustav Carus University Hospital, University of Technology Dresden, Dresden, Germany
| | - Katja Akgün
- MS Center Dresden, Center of Clinical Neuroscience, Carl Gustav Carus University Hospital, University of Technology Dresden, Dresden, Germany
| |
Collapse
|
32
|
Malhotra S, Villar LM, Costa C, Midaglia L, Cubedo M, Medina S, Fissolo N, Río J, Castilló J, Álvarez-Cermeño JC, Sánchez A, Montalban X, Comabella M. Circulating EZH2-positive T cells are decreased in multiple sclerosis patients. J Neuroinflammation 2018; 15:296. [PMID: 30367633 PMCID: PMC6202809 DOI: 10.1186/s12974-018-1336-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/16/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recent studies in experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis (MS), suggest an involvement of the histone methyltransferase enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) in important processes such as cell adhesion and migration. METHODS Here, we aimed to expand these initial observations by investigating the role of EZH2 in MS. mRNA expression levels for EZH2 were measured by real-time PCR in peripheral blood mononuclear cells (PBMC) from 121 MS patients (62 untreated and 59 receiving treatment) and 24 healthy controls. RESULTS EZH2 expression levels were decreased in PBMC from untreated patients compared to that from controls, and treatment significantly upregulated EZH2 expression. Expression of miR-124 was increased in MS patients compared to controls. Blood immunophenotyping revealed EZH2 expression mostly restricted to CD4+ and CD8+ T cells, and circulating EZH2+ CD4+ and CD8+ T cells were decreased in untreated MS patients compared to controls. CD8+ T cells expressing EZH2 exhibited a predominant central memory phenotype, whereas EZH2+ CD4+ T cells were of effector memory nature, and both T cell subsets produced TNF-α. EZH2+ T cells were enriched in the cerebrospinal fluid compartment compared to blood and were found in chronic active lesions from MS patients. EZH2 inhibition and microarray analysis in PBMC was associated with significant downregulation of key T cell adhesion molecules. CONCLUSION These findings suggest a role of EZH2 in the migration of T cells in MS patients. The observation of TNF-α expression by CD4+ and CD8+ T cells expressing EZH2 warrants additional studies to explore more in depth the pathogenic potential of EZH2+-positive cells in MS.
Collapse
Affiliation(s)
- Sunny Malhotra
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Luisa M Villar
- Departments of Neurology and Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacion Sanitaria, Madrid, Spain
| | - Carme Costa
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Luciana Midaglia
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta Cubedo
- Departament d'Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Silvia Medina
- Departments of Neurology and Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacion Sanitaria, Madrid, Spain
| | - Nicolás Fissolo
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jordi Río
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joaquín Castilló
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - José C Álvarez-Cermeño
- Departments of Neurology and Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacion Sanitaria, Madrid, Spain
| | - Alex Sánchez
- Unitat d'Estadística i Bioinformàtica, Institut de Recerca, HUVH, Barcelona, Spain.,Genetics, Microbiology and Statistics Department, Universitat de Barcelona, Barcelona, Spain
| | - Xavier Montalban
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
33
|
Negron A, Robinson RR, Stüve O, Forsthuber TG. The role of B cells in multiple sclerosis: Current and future therapies. Cell Immunol 2018; 339:10-23. [PMID: 31130183 DOI: 10.1016/j.cellimm.2018.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 02/07/2023]
Abstract
While it was long held that T cells were the primary mediators of multiple sclerosis (MS) pathogenesis, the beneficial effects observed in response to treatment with Rituximab (RTX), a monoclonal antibody (mAb) targeting CD20, shed light on a key contributor to MS that had been previously underappreciated: B cells. This has been reaffirmed by results from clinical trials testing the efficacy of subsequently developed B cell-depleting mAbs targeting CD20 as well as studies revisiting the effects of previous disease-modifying therapies (DMTs) on B cell subsets thought to modulate disease severity. In this review, we summarize current knowledge regarding the complex roles of B cells in MS pathogenesis and current and potential future B cell-directed therapies.
Collapse
Affiliation(s)
- Austin Negron
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | - Rachel R Robinson
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA
| | | |
Collapse
|
34
|
Li R, Patterson KR, Bar-Or A. Reassessing B cell contributions in multiple sclerosis. Nat Immunol 2018; 19:696-707. [PMID: 29925992 DOI: 10.1038/s41590-018-0135-x] [Citation(s) in RCA: 271] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023]
Abstract
There is growing recognition that B cell contributions to normal immune responses extend well beyond their potential to become antibody-producing cells, including roles at the innate-adaptive interface and their potential to modulate the responses of other immune cells such as T cells and myeloid cells. These B cell functions can have both pathogenic and protective effects in the context of central nervous system (CNS) inflammation. Here, we review recent advances in the field of multiple sclerosis (MS), which has traditionally been viewed as primarily a T cell-mediated disease, and we consider antibody-dependent and, particularly, emerging antibody-independent functions of B cells that may be relevant in both the peripheral and CNS disease compartments.
Collapse
Affiliation(s)
- Rui Li
- Center for Neuroinflammation and Experimental Therapeutics (CNET) and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristina R Patterson
- Center for Neuroinflammation and Experimental Therapeutics (CNET) and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics (CNET) and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Affiliation(s)
- Jorge Correale
- Department of Neurology, Raúl Carrea Institute for Neurological Research (FLENI), Buenos Aires, Argentina
| |
Collapse
|
36
|
Stewart-Hutchinson PJ, Szasz TP, Jaeger ER, Onken MD, Cooper JA, Morley SC. Technical Advance: New in vitro method for assaying the migration of primary B cells using an endothelial monolayer as substrate. J Leukoc Biol 2017. [PMID: 28637896 DOI: 10.1189/jlb.1ta0117-008r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Migration of B cells supports their development and recruitment into functional niches. Therefore, defining factors that control B cell migration will lead to a better understanding of adaptive immunity. In vitro cell migration assays with B cells have been limited by poor adhesion of cells to glass coated with adhesion molecules. We have developed a technique using monolayers of endothelial cells as the substrate for B cell migration and used this technique to establish a robust in vitro assay for B cell migration. We use TNF-α to up-regulate surface expression of the adhesion molecule VCAM-1 on endothelial cells. The ligand VLA-4 is expressed on B cells, allowing them to interact with the endothelial monolayer and migrate on its surface. We tested our new method by examining the role of L-plastin (LPL), an F-actin-bundling protein, in B cell migration. LPL-deficient (LPL-/-) B cells displayed decreased speed and increased arrest coefficient compared with wild-type (WT) B cells, following chemokine stimulation. However, the confinement ratios for WT and LPL-/- B cells were similar. Thus, we demonstrate how the use of endothelial monolayers as a substrate will support future interrogation of molecular pathways essential to B cell migration.
Collapse
Affiliation(s)
- Phillip J Stewart-Hutchinson
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Taylor P Szasz
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Emily R Jaeger
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael D Onken
- Departments of Biochemistry and Molecular Biophysics and Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA; and
| | - John A Cooper
- Departments of Biochemistry and Molecular Biophysics and Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA; and
| | - Sharon Celeste Morley
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA; .,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
37
|
Sabol RA, Noxon V, Sartor O, Berger JR, Qureshi Z, Raisch DW, Norris LB, Yarnold PR, Georgantopoulos P, Hrushesky WJ, Bobolts L, Ray P, Lebby A, Kane RC, Bennett CL. Melanoma complicating treatment with natalizumab for multiple sclerosis: A report from the Southern Network on Adverse Reactions (SONAR). Cancer Med 2017. [PMID: 28635055 PMCID: PMC5504343 DOI: 10.1002/cam4.1098] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
A 43-year-old female with multiple sclerosis developed urethral melanoma. The only potential risk factor was treatment with natalizumab, a humanized monoclonal antibody against α4 integrins. To investigate the risk-exposure relationship, we reviewed this case, all other published cases, and cases of natalizumab-associated melanoma reported to regulatory agencies. Data sources included the Food and Drug Administration's (FDA) Adverse Event Reporting System (FAERS) (2004-2014), a FDA Advisory Committee Meeting Report, and peer-reviewed publications. In the United States, the manufacturer maintains an FDA-mandated Tysabri Safety Surveillance Program (part of the Tysabri Outcomes Unified Commitment to Health (TOUCH)) of natalizumab-treated patients. We statistically compared reporting completeness for natalizumab-associated melanoma cases in FAERs for which information was obtained entirely from the TOUCH program versus cases where FAERS information was supplemented by TOUCH program information. FAERS included 137 natalizumab-associated melanoma reports in patients with multiple sclerosis. Median age at melanoma diagnosis was 45 years (range: 21-74 years). Changes in preexisting nevi occurred in 16%, history of cutaneous nevi occurred in 22%, diagnosis within 2 years of beginning natalizumab occurred in 34%, and 74% had primary surgical treatment. Among seven natalizumab-treated MS patients who developed biopsy-confirmed melanoma on treatment and reported in the literature, median age at diagnosis was 41 years (range: 38-48 years); and the melanoma diagnosis occurred following a median of 12 natalizumab doses (range: 1-77 doses). A history of mole or nevi was noted in four patients and a history of prior melanoma was noted in one patient. Completeness scores for reports were significantly lower for FAERS cases reported from the TOUCH program versus FAERS cases supplemented by TOUCH information (median score of 2 vs. 4 items out of 8-possible items, P < 0.0007). Clinicians should monitor existing nevi and maintain suspicion for melanoma developing in natalizumab-treated patients. The TOUCH Safety Surveillance Program, currently focused on progressive multifocal leukoencephalopathy, should be expanded to include information on other serious complications including malignancies, particularly if they are immunologic in nature.
Collapse
Affiliation(s)
- Rachel A Sabol
- Tulane University School of Medicine, New Orleans, Louisiana
| | - Virginia Noxon
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - Oliver Sartor
- Tulane University School of Medicine, New Orleans, Louisiana
| | - Joseph R Berger
- Department of Neurology, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Zaina Qureshi
- The Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
| | - Dennis W Raisch
- University of New Mexico, College of Pharmacy, Albuquerque, New Mexico
| | - LeAnn B Norris
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - Paul R Yarnold
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - Peter Georgantopoulos
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - William J Hrushesky
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | | | - Paul Ray
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - Akida Lebby
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - Robert C Kane
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - Charles L Bennett
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina.,The Medical University of South Carolina Hollings Cancer Center, Charleston, South Carolina.,William Jennings Bryan Dorn Veterans Administration Medical Center, Columbia, South Carolina
| |
Collapse
|
38
|
Characterization of naïve, memory and effector T cells in progressive multiple sclerosis. J Neuroimmunol 2017; 310:17-25. [PMID: 28778440 DOI: 10.1016/j.jneuroim.2017.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/02/2017] [Accepted: 06/02/2017] [Indexed: 01/09/2023]
Abstract
We characterized naïve, central memory (CM), effector memory (EM) and terminally differentiated effector memory (TEMRA) CD4+ and CD8+ T cells and their expression of CD49d and CD26 in peripheral blood in patients with multiple sclerosis (MS) and healthy controls. CD26+ CD28+ CD4+ TEMRA T cells were increased in all subtypes of MS, and CD26+ CD28+ CD8+ TEMRA T cells were increased in relapsing-remitting and secondary progressive MS. Conversely, in progressive MS, CD49d+ CM T cells were decreased and natalizumab increased the circulating number of all six subsets but reduced the frequency of most subsets expressing CD49d and CD26.
Collapse
|
39
|
Petersen ER, Søndergaard HB, Oturai AB, Jensen P, Sorensen PS, Sellebjerg F, Börnsen L. Soluble serum VCAM-1, whole blood mRNA expression and treatment response in natalizumab-treated multiple sclerosis. Mult Scler Relat Disord 2016; 10:66-72. [PMID: 27919501 DOI: 10.1016/j.msard.2016.09.001] [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] [Received: 04/07/2016] [Revised: 07/12/2016] [Accepted: 09/05/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Natalizumab reduces disease activity in multiple sclerosis (MS). Natalizumab binds to the very late antigen-4 and inhibits vascular cell adhesion molecule-1 (VCAM-1)-mediated transmigration of immune cells across the blood-brain-barrier. This is associated with decreased serum concentrations of soluble (s)VCAM-1 and an altered composition of immune cell-subsets in the blood. OBJECTIVE We aimed to examine if sVCAM-1 serum concentrations and whole blood mRNA expression levels of immune activation biomarkers is associated with disease activity in natalizumab-treated MS-patients. METHODS sVCAM-1 serum concentrations and whole blood mRNA expression were measured in blood samples from untreated RRMS-patients and from two independent groups of natalizumab-treated patients. RESULTS sVCAM-1 serum concentrations and whole blood expression of HLX1 and IL1B mRNA were lower, whereas expression of EBI3 mRNA was higher in natalizumab-treated MS-patients. Five genes were differentially expressed in clinically unstable natalizumab-treated MS-patients in the discovery but not in the validation group. CONCLUSION Decreased serum concentrations of sVCAM-1 and altered whole blood mRNA expression levels of a panel of immunomarkers, associated with natalizumab-treatment, are not sensitive markers of MS disease activity. However, decreased expression of pro-inflammatory HLX1 and IL1B and increased expression of immunoregulatory EBI3 may indicate a less pathogenic immune activation status in natalizumab-treated MS.
Collapse
Affiliation(s)
- E R Petersen
- Danish Multiple Sclerosis Center, Department of neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| | - H B Søndergaard
- Danish Multiple Sclerosis Center, Department of neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - A B Oturai
- Danish Multiple Sclerosis Center, Department of neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Peh Jensen
- Danish Multiple Sclerosis Center, Department of neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - P S Sorensen
- Danish Multiple Sclerosis Center, Department of neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - F Sellebjerg
- Danish Multiple Sclerosis Center, Department of neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - L Börnsen
- Danish Multiple Sclerosis Center, Department of neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
McGinley MP, Moss BP, Cohen JA. Safety of monoclonal antibodies for the treatment of multiple sclerosis. Expert Opin Drug Saf 2016; 16:89-100. [DOI: 10.1080/14740338.2017.1250881] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Marisa P. McGinley
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brandon P. Moss
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jeffrey A. Cohen
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
41
|
Saraste M, Penttilä TL, Airas L. Natalizumab treatment leads to an increase in circulating CXCR3-expressing B cells. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2016; 3:e292. [PMID: 27800533 PMCID: PMC5079379 DOI: 10.1212/nxi.0000000000000292] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/12/2016] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To study the effects of natalizumab treatment on subgroups of circulating peripheral blood B cell populations. METHODS We studied the proportions and absolute numbers of CD19+CD20+, CD10+, and CD5+ B cell populations, and determined very late activation antigen-4 and chemokine receptor CXCR3, CCR5, and CCR6 expression on B cells in the peripheral blood of 14 natalizumab-treated patients with relapsing-remitting multiple sclerosis. Five blood samples per patient were obtained longitudinally before and during the first year of treatment. Blood samples were analyzed by 6-color flow cytometry. RESULTS Proportions of B cells and CD10+ pre-B cells were significantly increased, and very late activation antigen-4 expression on the B cell surface was significantly decreased already after 1 week of natalizumab treatment. Natalizumab-induced sustained increase in the proportion and absolute number of CXCR3-expressing B cells was statistically significant after 1 month of treatment. There were no changes in the proportions of CCR5- or CCR6-expressing B cells. CONCLUSIONS The rapid and persistent increase in circulating CXCR3-expressing B cells in response to natalizumab treatment possibly reflects the relevance of this chemokine receptor in controlling migration of B cells into the CNS in humans in vivo.
Collapse
Affiliation(s)
- Maija Saraste
- Department of Neurology (M.S), University of Turku; TYKSLAB (T.-L.P.), Laboratory of Clinical Haematology, Turku University Hospital; and Division of Clinical Neurosciences (L.A.), Turku University Hospital, and University of Turku, Finland
| | - Tarja-Leena Penttilä
- Department of Neurology (M.S), University of Turku; TYKSLAB (T.-L.P.), Laboratory of Clinical Haematology, Turku University Hospital; and Division of Clinical Neurosciences (L.A.), Turku University Hospital, and University of Turku, Finland
| | - Laura Airas
- Department of Neurology (M.S), University of Turku; TYKSLAB (T.-L.P.), Laboratory of Clinical Haematology, Turku University Hospital; and Division of Clinical Neurosciences (L.A.), Turku University Hospital, and University of Turku, Finland
| |
Collapse
|
42
|
Leukocyte adhesion molecule dynamics after Natalizumab withdrawal in Multiple Sclerosis. Clin Immunol 2016; 171:18-24. [DOI: 10.1016/j.clim.2016.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/30/2016] [Accepted: 08/01/2016] [Indexed: 11/20/2022]
|
43
|
Iannetta M, Zingaropoli MA, Bellizzi A, Morreale M, Pontecorvo S, D’Abramo A, Oliva A, Anzivino E, Lo Menzo S, D’Agostino C, Mastroianni CM, Millefiorini E, Pietropaolo V, Francia A, Vullo V, Ciardi MR. Natalizumab Affects T-Cell Phenotype in Multiple Sclerosis: Implications for JCV Reactivation. PLoS One 2016; 11:e0160277. [PMID: 27486658 PMCID: PMC4972347 DOI: 10.1371/journal.pone.0160277] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 07/15/2016] [Indexed: 01/12/2023] Open
Abstract
The anti-CD49d monoclonal antibody natalizumab is currently an effective therapy against the relapsing-remitting form of multiple sclerosis (RRMS). Natalizumab therapeutic efficacy is limited by the reactivation of the John Cunningham polyomavirus (JCV) and development of progressive multifocal leukoencephalopathy (PML). To correlate natalizumab-induced phenotypic modifications of peripheral blood T-lymphocytes with JCV reactivation, JCV-specific antibodies (serum), JCV-DNA (blood and urine), CD49d expression and relative abundance of peripheral blood T-lymphocyte subsets were longitudinally assessed in 26 natalizumab-treated RRMS patients. Statistical analyses were performed using GraphPad Prism and R. Natalizumab treatment reduced CD49d expression on memory and effector subsets of peripheral blood T-lymphocytes. Moreover, accumulation of peripheral blood CD8+ memory and effector cells was observed after 12 and 24 months of treatment. CD4+ and CD8+ T-lymphocyte immune-activation was increased after 24 months of treatment. Higher percentages of CD8+ effectors were observed in subjects with detectable JCV-DNA. Natalizumab reduces CD49d expression on CD8+ T-lymphocyte memory and effector subsets, limiting their migration to the central nervous system and determining their accumulation in peripheral blood. Impairment of central nervous system immune surveillance and reactivation of latent JCV, can explain the increased risk of PML development in natalizumab-treated RRMS subjects.
Collapse
MESH Headings
- Adult
- Antibodies, Viral/blood
- DNA, Viral/analysis
- DNA, Viral/blood
- Female
- Humans
- JC Virus/drug effects
- JC Virus/physiology
- Leukoencephalopathy, Progressive Multifocal/complications
- Leukoencephalopathy, Progressive Multifocal/immunology
- Leukoencephalopathy, Progressive Multifocal/virology
- Male
- Multiple Sclerosis, Relapsing-Remitting/complications
- Multiple Sclerosis, Relapsing-Remitting/immunology
- Multiple Sclerosis, Relapsing-Remitting/therapy
- Multiple Sclerosis, Relapsing-Remitting/virology
- Natalizumab/adverse effects
- Natalizumab/pharmacology
- Phenotype
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Treatment Outcome
- Virus Activation/drug effects
Collapse
Affiliation(s)
- Marco Iannetta
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
- Inserm, U1016, Institut Cochin, Paris, France
- * E-mail:
| | | | - Anna Bellizzi
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Manuela Morreale
- Department of Medical and Surgical Sciences and Biotechnology, Neurovascular Diagnosis Unit, Section of Neurology, Sapienza University, Rome, Italy
- Department of Neurology and Psychiatry, Multiple Sclerosis Center, Sapienza University, Rome, Italy
| | - Simona Pontecorvo
- Department of Neurology and Psychiatry, Multiple Sclerosis Center, Sapienza University, Rome, Italy
| | - Alessandra D’Abramo
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Alessandra Oliva
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Elena Anzivino
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Sara Lo Menzo
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Claudia D’Agostino
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | | | - Enrico Millefiorini
- Department of Neurology and Psychiatry, Multiple Sclerosis Center, Sapienza University, Rome, Italy
| | - Valeria Pietropaolo
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Ada Francia
- Department of Neurology and Psychiatry, Multiple Sclerosis Center, Sapienza University, Rome, Italy
| | - Vincenzo Vullo
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Maria Rosa Ciardi
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| |
Collapse
|
44
|
Zhovtis Ryerson L, Frohman TC, Foley J, Kister I, Weinstock-Guttman B, Tornatore C, Pandey K, Donnelly S, Pawate S, Bomprezzi R, Smith D, Kolb C, Qureshi S, Okuda D, Kalina J, Rimler Z, Green R, Monson N, Hoyt T, Bradshaw M, Fallon J, Chamot E, Bucello M, Beh S, Cutter G, Major E, Herbert J, Frohman EM. Extended interval dosing of natalizumab in multiple sclerosis. J Neurol Neurosurg Psychiatry 2016; 87:885-9. [PMID: 26917698 DOI: 10.1136/jnnp-2015-312940] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/06/2016] [Indexed: 11/03/2022]
Abstract
BACKGROUND Natalizumab (NTZ), a monoclonal antibody to human α4β1/β7 integrin, is an effective therapy for multiple sclerosis (MS), albeit associated with progressive multifocal leukoencephalopathy (PML). Clinicians have been extending the dose of infusions with a hypothesis of reducing PML risk. The aim of the study is to evaluate the clinical consequences of reducing NTZ frequency of infusion up to 8 weeks 5 days. METHODS A retrospective chart review in 9 MS centres was performed in order to identify patients treated with extended interval dosing (EID) regimens of NTZ. Patients were stratified into 3 groups based on EID NTZ treatment schedule in individual centres: early extended dosing (EED; n=249) every 4 weeks 3 days to 6 weeks 6 days; late extended dosing (LED; n=274) every 7 weeks to 8 weeks 5 days; variable extended dosing (n=382) alternating between EED and LED. These groups were compared with patients on standard interval dosing (SID; n=1093) every 4 weeks. RESULTS 17% of patients on SID had new T2 lesions compared with 14% in EID (p=0.02); 7% of patients had enhancing T1 lesions in SID compared with 9% in EID (p=0.08); annualised relapse rate was 0.14 in the SID group, and 0.09 in the EID group. No evidence of clinical or radiographic disease activity was observed in 62% of SID and 61% of EID patients (p=0.83). No cases of PML were observed in EID group compared with 4 cases in SID cohort. CONCLUSIONS Dosing intervals up to 8 weeks 5 days did not diminish effectiveness of NTZ therapy. Further monitoring is ongoing to evaluate if the risk of PML is reduced in patients on EID.
Collapse
Affiliation(s)
- L Zhovtis Ryerson
- Department of Neurology, Langone Medical Center, New York University, New York, New York, USA
| | - T C Frohman
- Departments of Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - J Foley
- Rocky Mountain MS Clinic, Salt Lake City, Utah, USA
| | - I Kister
- Department of Neurology, Langone Medical Center, New York University, New York, New York, USA
| | | | | | - K Pandey
- Barnabas Health MS Center, Livingston, New Jersey, USA
| | - S Donnelly
- CUNY Graduate Center, New York, New York, USA
| | - S Pawate
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - R Bomprezzi
- University of Massachusetts School of Medicine, Worcester, Massachusetts, USA
| | - D Smith
- Multiple Sclerosis Center of Connecticut, Norwich, Connecticut, USA
| | - C Kolb
- University of Buffalo, Buffalo, New York, USA
| | - S Qureshi
- Departments of Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - D Okuda
- Departments of Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - J Kalina
- Department of Neurology, Langone Medical Center, New York University, New York, New York, USA
| | - Z Rimler
- Department of Neurology, Langone Medical Center, New York University, New York, New York, USA
| | - R Green
- Barnabas Health MS Center, Livingston, New Jersey, USA
| | - N Monson
- Departments of Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - T Hoyt
- Rocky Mountain MS Clinic, Salt Lake City, Utah, USA
| | - M Bradshaw
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - J Fallon
- Department of Neurology, Langone Medical Center, New York University, New York, New York, USA
| | - E Chamot
- University of Alabama School of Public Health, Birmingham, Alabama, USA
| | - M Bucello
- University of Buffalo, Buffalo, New York, USA
| | - S Beh
- Departments of Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - G Cutter
- University of Alabama School of Public Health, Birmingham, Alabama, USA
| | - E Major
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - J Herbert
- Department of Neurology, Langone Medical Center, New York University, New York, New York, USA
| | - E M Frohman
- Departments of Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA Department of Bioengineering, University of Texas at Dallas, Dallas, Texas, USA Department of Behavioural and Brain Sciences, University of Texas at Dallas, Dallas, Texas, USA
| |
Collapse
|
45
|
Abstract
Multiple sclerosis (MS) is a complex disease that causes a great deal of disability, especially in the young adult population. There have been several immunomodulatory agents that have been approved by the Food and Drug Administration for MS, including glatiramer acetate, interferon-β 1a and -β 1b, mitoxantrone, and corticosteroids. The effectiveness of these therapies has not been optimal, and drugs, such as monoclonal antibodies, that more selectively target the pathogenetic process of MS have been sought. These agents have their own intrinsic limitations such as systemic inflammatory reactions, induction of neutralizing antiantibodies, and even life-threatening infectious processes. The agent that has been in the forefront of the discussion is natalizumab, a monoclonal antibody (mAb) against α 4 integrin, which shows much promise in suppressing MS activity. However, 3 individuals treated with natalizumab developed a life-threatening infection, progressive multifocal leukoencephalopathy. This article reviews the role of mAbs in the treatment of MS, particularly their advantages over other drugs and their limitations, which have to be overcome for these agents to be at the forefront in the treatment of MS.
Collapse
Affiliation(s)
| | - Jane W. Chan
- University of Kentucky College of Medicine, Lexington,
| |
Collapse
|
46
|
Bhise V, Dhib-Jalbut S. Further understanding of the immunopathology of multiple sclerosis: impact on future treatments. Expert Rev Clin Immunol 2016; 12:1069-89. [PMID: 27191526 DOI: 10.1080/1744666x.2016.1191351] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The understanding of the immunopathogenesis of multiple sclerosis (MS) has expanded with more research into T-cell subtypes, cytokine contributors, B-cell participation, mitochondrial dysfunction, and more. Treatment options have rapidly expanded with three relatively recent oral therapy alternatives entering the arena. AREAS COVERED In the following review, we discuss current mechanisms of immune dysregulation in MS, how they relate to current treatments, and the impact these findings will have on the future of therapy. Expert commentary: The efficacy of these medications and understanding their mechanisms of actions validates the immunopathogenic mechanisms thought to underlie MS. Further research has exposed new targets, while new promising therapies have shed light on new aspects into the pathophysiology of MS.
Collapse
Affiliation(s)
- Vikram Bhise
- a Rutgers Biomedical and Health Sciences - Departments of Pediatrics , Robert Wood Johnson Medical School , New Brunswick , NJ , USA
| | - Suhayl Dhib-Jalbut
- b Rutgers Biomedical and Health Sciences - Departments of Neurology , Robert Wood Johnson Medical School , New Brunswick , NJ , USA
| |
Collapse
|
47
|
Interferon-β therapy specifically reduces pathogenic memory B cells in multiple sclerosis patients by inducing a FAS-mediated apoptosis. Immunol Cell Biol 2016; 94:886-894. [PMID: 27265253 DOI: 10.1038/icb.2016.55] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/06/2016] [Accepted: 05/29/2016] [Indexed: 12/27/2022]
Abstract
Growing evidences put B lymphocytes on a central stage in multiple sclerosis (MS) immunopathology. While investigating the effects of interferon-β (IFN-β) therapy, one of the most used first-line disease-modifying drugs for the treatment of relapsing-remitting MS, in circulating B-cell sub-populations, we found a specific and marked decrease of CD27+ memory B cells. Interestingly, memory B cells are considered a population with a great disease-driving relevance in MS and resulted to be also target of B-cell depleting therapies. In addition, Epstein-Barr virus (EBV), associated with MS etiopathogenesis, harbors in this cell type and an IFN-β-induced reduction of the memory B-cell compartment, in turn, resulted in a decreased expression of the EBV gene latent membrane protein 2A in treated patients. We found that in vivo IFN-β therapy specifically and highly induced apoptosis in memory B cells, in accordance with a strong increase of the apoptotic markers Annexin-V and active caspase-3, via a mechanism requiring the FAS-receptor/TACI (transmembrane activator and CAML interactor) signaling. Thus, efficacy of IFN-β therapy in MS may rely not only on its recognized anti-inflammatory activities but also on the specific depletion of memory B cells, considered to be a pathogenic cell subset, reducing their inflammatory impact in target organs.
Collapse
|
48
|
Kimura K, Nakamura M, Sato W, Okamoto T, Araki M, Lin Y, Murata M, Takahashi R, Yamamura T. Disrupted balance of T cells under natalizumab treatment in multiple sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2016; 3:e210. [PMID: 27006971 PMCID: PMC4784802 DOI: 10.1212/nxi.0000000000000210] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/11/2015] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To compare effects of natalizumab on inflammatory and regulatory T cells with regard to expression of α4-integrin (CD49d). METHODS Twenty-seven natalizumab-naive and 8 natalizumab-treated patients with multiple sclerosis (MS), 7 patients with neuromyelitis optica (NMO) or NMO spectrum disorder, and 8 healthy controls were included. The positive rate of CD49d was analyzed and compared among T helper 1 (Th1), T helper 17 (Th17), and regulatory T (Treg) cells (CD49d+Th1, CD49d+Th17, and CD49d+Treg, respectively). RESULTS Natalizumab treatment increased CD49d ratios, CD49d+Th1/CD49d+Treg, and CD49d+Th17/CD49d+Treg. This indicates larger reduction of the CD49d+ population in Treg cells than in Th1 or Th17 cells. The CD49d ratios of 2 patients who experienced exacerbation during natalizumab treatment were remarkably higher than those of the other natalizumab-treated patients. Natalizumab treatment increased the expression of TBX21, RORC, interferon (IFN)-γ, and interleukin (IL)-17A, and decreased the expression of FOXP3 in CD49d+ memory CD4 T cells. Natalizumab treatment also increased the amount of IFN-γ and IL-17A secreted by CD49d+ memory CD4 T cells. CONCLUSIONS The reduction rate of the CD49d+ population in Treg cells was larger than that in Th1 or Th17 cells. Although the large reduction in CD49d+ population is beneficial for MS, the proinflammatory state of residual CD49d+ cells might, in part, explain the presence of disease activity under natalizumab treatment.
Collapse
Affiliation(s)
- Kimitoshi Kimura
- Department of Immunology, National Institute of Neuroscience (K.K., M.N., W.S., Y.L., T.Y.), and Multiple Sclerosis Center (W.S., T.O., M.A., Y.L., T.Y.) and Department of Neurology (Y.L., M.M.), National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Tokyo; and Department of Neurology (K.K., R.T.), Kyoto University Graduate School of Medicine, Japan
| | - Masakazu Nakamura
- Department of Immunology, National Institute of Neuroscience (K.K., M.N., W.S., Y.L., T.Y.), and Multiple Sclerosis Center (W.S., T.O., M.A., Y.L., T.Y.) and Department of Neurology (Y.L., M.M.), National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Tokyo; and Department of Neurology (K.K., R.T.), Kyoto University Graduate School of Medicine, Japan
| | - Wakiro Sato
- Department of Immunology, National Institute of Neuroscience (K.K., M.N., W.S., Y.L., T.Y.), and Multiple Sclerosis Center (W.S., T.O., M.A., Y.L., T.Y.) and Department of Neurology (Y.L., M.M.), National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Tokyo; and Department of Neurology (K.K., R.T.), Kyoto University Graduate School of Medicine, Japan
| | - Tomoko Okamoto
- Department of Immunology, National Institute of Neuroscience (K.K., M.N., W.S., Y.L., T.Y.), and Multiple Sclerosis Center (W.S., T.O., M.A., Y.L., T.Y.) and Department of Neurology (Y.L., M.M.), National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Tokyo; and Department of Neurology (K.K., R.T.), Kyoto University Graduate School of Medicine, Japan
| | - Manabu Araki
- Department of Immunology, National Institute of Neuroscience (K.K., M.N., W.S., Y.L., T.Y.), and Multiple Sclerosis Center (W.S., T.O., M.A., Y.L., T.Y.) and Department of Neurology (Y.L., M.M.), National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Tokyo; and Department of Neurology (K.K., R.T.), Kyoto University Graduate School of Medicine, Japan
| | - Youwei Lin
- Department of Immunology, National Institute of Neuroscience (K.K., M.N., W.S., Y.L., T.Y.), and Multiple Sclerosis Center (W.S., T.O., M.A., Y.L., T.Y.) and Department of Neurology (Y.L., M.M.), National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Tokyo; and Department of Neurology (K.K., R.T.), Kyoto University Graduate School of Medicine, Japan
| | - Miho Murata
- Department of Immunology, National Institute of Neuroscience (K.K., M.N., W.S., Y.L., T.Y.), and Multiple Sclerosis Center (W.S., T.O., M.A., Y.L., T.Y.) and Department of Neurology (Y.L., M.M.), National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Tokyo; and Department of Neurology (K.K., R.T.), Kyoto University Graduate School of Medicine, Japan
| | - Ryosuke Takahashi
- Department of Immunology, National Institute of Neuroscience (K.K., M.N., W.S., Y.L., T.Y.), and Multiple Sclerosis Center (W.S., T.O., M.A., Y.L., T.Y.) and Department of Neurology (Y.L., M.M.), National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Tokyo; and Department of Neurology (K.K., R.T.), Kyoto University Graduate School of Medicine, Japan
| | - Takashi Yamamura
- Department of Immunology, National Institute of Neuroscience (K.K., M.N., W.S., Y.L., T.Y.), and Multiple Sclerosis Center (W.S., T.O., M.A., Y.L., T.Y.) and Department of Neurology (Y.L., M.M.), National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Tokyo; and Department of Neurology (K.K., R.T.), Kyoto University Graduate School of Medicine, Japan
| |
Collapse
|
49
|
Teniente-Serra A, Grau-López L, Mansilla MJ, Fernández-Sanmartín M, Ester Condins A, Ramo-Tello C, Martínez-Cáceres E. Multiparametric flow cytometric analysis of whole blood reveals changes in minor lymphocyte subpopulations of multiple sclerosis patients. Autoimmunity 2016; 49:219-28. [PMID: 26829210 DOI: 10.3109/08916934.2016.1138271] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The objective of this study is to characterise the functionally relevant minor lymphocyte subpopulations in whole blood of multiple sclerosis (MS) patients and their potential utility as biomarkers for treatment follow up. MATERIAL AND METHODS Peripheral blood from 40 healthy donors (HD) and 66 MS patients [23 relapsing-remitting (RRMS) without treatment, 27 RRMS undergoing treatment (16 IFN-β, 11 natalizumab), and 16 progressive forms (eight secondary progressive and eight primary progressive)] was analysed by multiparametric flow cytometry. RESULTS Untreated MS patients showed a decrease in early effector memory (CD45RA(-)CCR7(-)CD27(+)) CD4(+) and CD8(+) T cells and an increase in Th17 lymphocytes in peripheral blood compared with HD. Regarding the effect of treatment, whereas no differences in relative percentages of cellular subpopulations were observed in patients under IFN-β treatment, those under treatment with natalizumab had an increased percentage of early effector memory CD4(+) (CD45RA(-)CCR7(-)CD27(+)), central memory CD8(+) (CD45RA(-)CCR7(+)CD27(+)) T cells, recent thymic emigrants (CD4(+) CD45RA(+)CCR7(+)CD27(+)CD31(+)PTK7(+)) and transitional B cells (CD19(+)CD27(-)CD24(hi)CD38(hi)). CONCLUSIONS Multiparametric flow cytometry analysis of whole blood is a robust, reproducible, and sensitive technology to monitor the effect of MS treatments even in minor lymphocyte subpopulations that might represent useful biomarkers of treatment response.
Collapse
Affiliation(s)
- Aina Teniente-Serra
- a Immunology Division, Germans Trias i Pujol University Hospital and Research Institute (IGTP) , Campus Can Ruti , Badalona , Barcelona .,b Department of Cell Biology , Physiology and Immunology, Universitat Autònoma de Barcelona , Bellaterra , Barcelona
| | - Laia Grau-López
- c Multiple Sclerosis Unit, Department of Neurosciences. Germans Trias i Pujol University Hospital , Badalona , Barcelona , and
| | - M José Mansilla
- a Immunology Division, Germans Trias i Pujol University Hospital and Research Institute (IGTP) , Campus Can Ruti , Badalona , Barcelona .,b Department of Cell Biology , Physiology and Immunology, Universitat Autònoma de Barcelona , Bellaterra , Barcelona
| | - Marco Fernández-Sanmartín
- d Flow Cytometry Facility, Germans Trias i Pujol Research Institute (IGTP) , Campus Can Ruti , Badalona , Barcelona , and
| | | | - Cristina Ramo-Tello
- c Multiple Sclerosis Unit, Department of Neurosciences. Germans Trias i Pujol University Hospital , Badalona , Barcelona , and
| | - Eva Martínez-Cáceres
- a Immunology Division, Germans Trias i Pujol University Hospital and Research Institute (IGTP) , Campus Can Ruti , Badalona , Barcelona .,b Department of Cell Biology , Physiology and Immunology, Universitat Autònoma de Barcelona , Bellaterra , Barcelona
| |
Collapse
|
50
|
Li R, Rezk A, Healy LM, Muirhead G, Prat A, Gommerman JL, Bar-Or A. Cytokine-Defined B Cell Responses as Therapeutic Targets in Multiple Sclerosis. Front Immunol 2016; 6:626. [PMID: 26779181 PMCID: PMC4705194 DOI: 10.3389/fimmu.2015.00626] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/30/2015] [Indexed: 02/04/2023] Open
Abstract
Important antibody-independent pathogenic roles of B cells are emerging in autoimmune diseases, including multiple sclerosis (MS). The contrasting results of different treatments targeting B cells in patients (in spite of predictions of therapeutic benefits from animal models) call for a better understanding of the multiple roles that distinct human B cell responses likely play in MS. In recent years, both murine and human B cells have been identified with distinct functional properties related to their expression of particular cytokines. These have included regulatory (Breg) B cells (secreting interleukin (IL)-10 or IL-35) and pro-inflammatory B cells (secreting tumor necrosis factor α, LTα, IL-6, and granulocyte macrophage colony-stimulating factor). Better understanding of human cytokine-defined B cell responses is necessary in both health and diseases, such as MS. Investigation of their surface phenotype, distinct functions, and the mechanisms of regulation (both cell intrinsic and cell extrinsic) may help develop effective treatments that are more selective and safe. In this review, we focus on mechanisms by which cytokine-defined B cells contribute to the peripheral immune cascades that are thought to underlie MS relapses, and the impact of B cell-directed therapies on these mechanisms.
Collapse
Affiliation(s)
- Rui Li
- Neuroimmunology Unit, Montreal Neurological Institute and Hospital, McGill University , Montreal, QC , Canada
| | - Ayman Rezk
- Neuroimmunology Unit, Montreal Neurological Institute and Hospital, McGill University , Montreal, QC , Canada
| | - Luke M Healy
- Neuroimmunology Unit, Montreal Neurological Institute and Hospital, McGill University , Montreal, QC , Canada
| | - Gillian Muirhead
- Neuroimmunology Unit, Montreal Neurological Institute and Hospital, McGill University , Montreal, QC , Canada
| | - Alexandre Prat
- Neuroimmunology Unit, Department of Neuroscience, Centre de Recherche du CHUM (CRCHUM), Université de Montreal , Montreal, QC , Canada
| | | | - Amit Bar-Or
- Neuroimmunology Unit, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada; Experimental Therapeutics Program, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | | |
Collapse
|