1
|
Yang JY, Sun SZ, Sun YY, Zuo TQ, Li XT, Zhao LY, Wu HY, Peng W. Analysis of diroximel fumarate data for patients with relapsing forms of multiple sclerosis using related adverse events from the FDA adverse reporting system. Expert Opin Drug Saf 2025:1-8. [PMID: 40171720 DOI: 10.1080/14740338.2025.2488318] [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: 09/13/2024] [Accepted: 03/05/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Diroximel fumarate (DRF) is an oral fumarate used to treat relapsing forms of multiple sclerosis (RMS). This study comprehensively analyzed the adverse events (AEs) associated with DRF for treating RMS based on data from the FDA Adverse Event Reporting System (FAERS) database. RESEARCH DESIGN AND METHODS This study collected data on AEs associated with DRF treatment of RMS from the FAERS database between 2019 and 2024. We used reporting odds ratio (ROR), proportional reporting ratio (PRR), Bayesian Confidence Propagation Neural Network (BCPNN), and Multi-item Gamma Poisson Shrinker (MGPS) for signal detection. RESULTS This study collected 7,944,554 AE reports, of which 7,868 were associated with DRF. A total of 120 preferred terms (PTs) were included in the analysis, relating to 27 system organ classes (SOCs). This study identified several clinically significant new potential AEs, including decreased immune responsiveness (n = 26, ROR 4.45, PRR 4.45, IC 2.15, EBGM 4.43), female breast cancer (n = 50, ROR 4.07, PRR 4.07, IC 2.02, EBGM 4.05), transient blindness (n = 19, ROR 7.27, PRR 7.26, IC 2.85, EBGM 7.21) and others. CONCLUSIONS Our study identified several potentially important AEs that were not mentioned in the DRF instructions. However, further epidemiologic studies are needed to validate these findings.
Collapse
Affiliation(s)
- Jing-Ya Yang
- The First Clinical School, Shandong University of Traditional Chinese Medicine, Shandong, Jinan, China
| | - Sheng-Zhu Sun
- The First Clinical School, Shandong University of Traditional Chinese Medicine, Shandong, Jinan, China
| | - Yi-Yan Sun
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian-Qi Zuo
- The First Clinical School, Shandong University of Traditional Chinese Medicine, Shandong, Jinan, China
| | - Xiao-Tong Li
- Department of Neurology, Taian Hospital of Traditional Chinese Medicine, Taian, Shandong, China
| | - Lei-Yong Zhao
- The First School of Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Hong-Yun Wu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wei Peng
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
2
|
Dempsey J, Balshi A, Sloane J. Herpes zoster infection in a patient with relapsing-remitting multiple sclerosis treated with diroximel fumarate. BMJ Case Rep 2025; 18:e264081. [PMID: 40180342 DOI: 10.1136/bcr-2024-264081] [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: 04/05/2025] Open
Abstract
We present a case of a woman in her 50s with relapsing-remitting multiple sclerosis, treated with diroximel fumarate, who developed an itching, burning vesicular rash following a C3 dermatomal distribution on her left face and neck. She was diagnosed with herpes zoster, treated with valacyclovir and gabapentin, and made a full recovery. Ensuring patients are protected against varicella-zoster virus before starting diroximel fumarate can prevent this painful infection. Thus, we recommend vaccination against the varicella-zoster virus prior to diroximel fumarate initiation be incorporated as the standard of care.
Collapse
Affiliation(s)
- John Dempsey
- SUNY Upstate Medical University Norton College of Medicine, Syracuse, New York, USA
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Alexandra Balshi
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Jacob Sloane
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
3
|
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 2025; 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] [MESH Headings] [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
|
4
|
Jeuris J, Vangheluwe R, Van Cauter S. Varicella-Zoster virus associated vasculopathy in a young male multiple sclerosis patient, treated with dimethyl fumarate. Acta Neurol Belg 2025:10.1007/s13760-025-02723-0. [PMID: 39875790 DOI: 10.1007/s13760-025-02723-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/09/2025] [Indexed: 01/30/2025]
Abstract
With the increasing use of disease modifying therapies as treatment for multiple sclerosis, knowledge of the rare but possibly severe adverse events becomes increasingly important. We present a case of Varicella-Zoster virus associated vasculopathy in a young male multiple sclerosis patient, treated with dimethyl fumarate. We aim to address this rare but potentially deadly complication of varicella-Zoster virus infection and spread awareness about the increased risk in this patient population.
Collapse
Affiliation(s)
- Jules Jeuris
- Ziekenhuis Oost Limburg, Genk, Belgium.
- KU-Leuven, Leuven, België.
| | | | | |
Collapse
|
5
|
Dalmau J, Dalakas MC, Kolson DL, Pröbstel AK, Paul F, Zamvil SS. Ten Years of Neurology® Neuroimmunology & Neuroinflammation: Decade in Review. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200363. [PMID: 39724529 DOI: 10.1212/nxi.0000000000200363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Affiliation(s)
- Josep Dalmau
- IDIBAPS-CaixaResearch Institute, University Hospital Clínic of Barcelona, Barcelona, Spain
- University of Pennsylvania, Philadelphia
| | - Marinos C Dalakas
- University of Athens Medical School, Greece
- Jefferson University, Philadelphia, PA
| | | | - Anne-Katrin Pröbstel
- Departments of Neurology, University Hospital of Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Switzerland
| | | | - Scott S Zamvil
- Department of Neurology, University of California, San Francisco
| |
Collapse
|
6
|
Balshi A, Dempsey J, Sloane JA. Atypical disseminated herpes zoster infections in patients with demyelinating disease treated with dimethyl fumarate. J Neurovirol 2024; 30:556-558. [PMID: 39433711 DOI: 10.1007/s13365-024-01230-x] [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: 08/23/2024] [Revised: 09/11/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024]
Abstract
We report two patients who developed atypical, disseminated herpes zoster infections while on dimethyl fumarate (DMF) treatment, one with varicella zoster virus (VZV) encephalitis and another with herpes zoster oticus resulting in lasting motor and sensory deficits. We recommend vaccination against VZV prior to DMF initiation be incorporated as standard of care, as ensuring patients are protected against VZV before starting DMF can prevent such severe outcomes.
Collapse
Affiliation(s)
- Alexandra Balshi
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Kirstein 215, Boston, MA, 02215, USA
| | - John Dempsey
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Kirstein 215, Boston, MA, 02215, USA
| | - Jacob A Sloane
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Kirstein 215, Boston, MA, 02215, USA.
| |
Collapse
|
7
|
Shalaby NM, Rushdi R, Alroughany R, Ahmed S, Merghany N, Shehata H, Al-Hashel J, Nada M, Gad A, Hassan A, Kishk N, Hamdy S, Abdelnaseer M, Hegazy M, Ahmed S, Abdel-Aal AR, El Shebawy H. Impact of Fingolimod Discontinuation Strategy on Recurrence of Disease Activity in Individuals With Multiple Sclerosis. Int J MS Care 2024; 26:329-340. [PMID: 39588274 PMCID: PMC11588075 DOI: 10.7224/1537-2073.2023-050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
BACKGROUND For individuals with multiple sclerosis (MS), treatment interruption can result in relapse/recurrence of the disease activity. Currently, there are no consensus guidelines about whether an abrupt stop with a short washout period or gradual tapering is better for fingolimod (Gilenya) cessation. We investigated the impact of the fingolimod discontinuation strategy on the recurrence of disease activity and the rebound occurrence of symptoms during washout. METHODS This was a retrospective, observational, multicenter study of individuals with MS in Egypt and Kuwait. The charts of patients on fingolimod therapy were screened to collect data on the impact of drug cessation strategies on disease activity and relapse occurrence. Disease relapse after cessation was defined as a relapse that occurred in the previous 12 months despite using a first-line treatment option or 2 relapses in the previous 12 months. RESULTS In a cohort of 100 patients, 58 had an abrupt cessation and 42 had a gradual tapering. Compared with abrupt cessation, gradual tapering was associated with a significantly lower rate of disease relapse (4.8% vs 81%, respectively; P = .001). Abrupt cessation also resulted in increased MRI findings of new lesions (24.1%; P = .29), enhancing lesions (32.8%; P = .5), and enlarging lesions (6.9%; P = .59); however, none of the MRI findings were significant. Other risk factors showed no significant association with disease relapse after fingolimod cessation. CONCLUSIONS Gradual fingolimod tapering is highly recommended to decrease the risk of rebound and severe disease reactivation. A prolonged washout should be avoided for lymphocyte recovery.
Collapse
Affiliation(s)
- Nevin M. Shalaby
- From the Department of Neurology, Cairo University, Cairo, Egypt
| | - Rufaidah Rushdi
- the Department of Pediatrics, Cairo University, Cairo, Egypt
| | - Raed Alroughany
- the Department of Neurology, Al-Amiri Hospital, Kuwait City, Kuwait
| | - Samar Ahmed
- the Department of Neurology, Minia University, Minia, Egypt
- the Department of Neurology, Ibn Sina Hospital, Kuwait City, Kuwait
| | - Nahla Merghany
- From the Department of Neurology, Cairo University, Cairo, Egypt
| | - Hatem Shehata
- From the Department of Neurology, Cairo University, Cairo, Egypt
| | - Jasem Al-Hashel
- the Department of Neurology, Ibn Sina Hospital, Kuwait City, Kuwait
| | - Mona Nada
- From the Department of Neurology, Cairo University, Cairo, Egypt
| | - Adel Gad
- From the Department of Neurology, Cairo University, Cairo, Egypt
| | - Amr Hassan
- From the Department of Neurology, Cairo University, Cairo, Egypt
| | - Nirmeen Kishk
- From the Department of Neurology, Cairo University, Cairo, Egypt
| | - Sherif Hamdy
- From the Department of Neurology, Cairo University, Cairo, Egypt
| | | | - Mohamed Hegazy
- From the Department of Neurology, Cairo University, Cairo, Egypt
| | - Sandra Ahmed
- From the Department of Neurology, Cairo University, Cairo, Egypt
| | | | - Haidy El Shebawy
- From the Department of Neurology, Cairo University, Cairo, Egypt
| |
Collapse
|
8
|
Dempsey JP, Wu L, Balshi A, Jun C, Baber U, Sloane JA. Worsening of lymphopenia in patients with multiple sclerosis when switched from dimethyl fumarate to diroximel fumarate. Mult Scler Relat Disord 2024; 89:105737. [PMID: 39029343 DOI: 10.1016/j.msard.2024.105737] [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: 12/22/2023] [Revised: 06/02/2024] [Accepted: 06/19/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Diroximel fumarate (DRF) and dimethyl fumarate (DMF) are similar disease-modifying therapies (DMTs) that reduce disease activity in patients with relapsing-remitting multiple sclerosis (MS). We expect that patients on DRF would experience a similar incidence and severity of lymphopenia, given that it is a well-documented side effect of DMF treatment. METHODS We utilized linear mixed-effects models to test for differences in white blood cell count (WBC), absolute lymphocyte count (ALC), absolute CD3+ count, absolute CD4+ count, and absolute CD8+ count over time in clinically stable patients with MS on DMF who switched to DRF. RESULTS Twenty-two patients with MS who were clinically stable on DMF switched to DRF. Linear mixed-effects models showed a decrease in ALC when switching medications (β = -225.70, p < 0.040). In addition, the models showed a decrease in absolute CD8+ counts after switches from DMF to DRF (β = -85.59, p = 0.034). CONCLUSION Patients with MS who are stable on DMF and switch to DRF may experience worsening of lymphopenia and lower absolute CD8+ counts, which may increase their risk of opportunistic infections. These findings indicate that close lymphocyte subset monitoring is clinically important when switching patients with MS from DMF to DRF.
Collapse
Affiliation(s)
- John Patrick Dempsey
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lisa Wu
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alexandra Balshi
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Claire Jun
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ursela Baber
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jacob A Sloane
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Arneth B. Current Knowledge about CD3 +CD20 + T Cells in Patients with Multiple Sclerosis. Int J Mol Sci 2024; 25:8987. [PMID: 39201672 PMCID: PMC11354236 DOI: 10.3390/ijms25168987] [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: 05/24/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/03/2024] Open
Abstract
Multiple sclerosis (MS) is a disease of the central nervous system (CNS) characterized by inflammation and autoimmune responses. This review explores the participation of T cells, particularly certain CD3+CD20+ T cells, in the clinical manifestations of MS and highlights their presence in diagnosed patients. These T cells show aberrant expression of CD20, normally considered a B-cell marker. In this review, relevant journal articles available in PubMed and CINAHL were identified by employing diverse search terms, such as MS, CD3+CD20+ T cells, the incidence and significance of CD3+CD20+ T cells in MS patients, and the impact of rituximab treatment. The search was limited to articles published in the ten-year period from 2014 to 2024. The results of this review suggest that most scholars agree on the presence of CD3+CD20+ T cells in cerebrospinal fluid. Emerging concepts relate to the fundamental role of CD20-expressing T cells in determining the target and efficacy of MS therapeutics and the presence of T cells in the cerebrospinal fluid of MS patients. The results clearly show that CD20+ T cells indicate disease chronicity and high disease activity.
Collapse
Affiliation(s)
- Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, Feulgenstr. 12, 35392 Giessen, Germany;
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Philipps University Marburg, Baldinger Str., 35043 Marburg, Germany
| |
Collapse
|
10
|
Kaye AD, Lacey J, Le V, Fazal A, Boggio NA, Askins DH, Anderson L, Robinson CL, Paladini A, Mosieri CN, Kaye AM, Ahmadzadeh S, Shekoohi S, Varrassi G. The Evolving Role of Monomethyl Fumarate Treatment as Pharmacotherapy for Relapsing-Remitting Multiple Sclerosis. Cureus 2024; 16:e57714. [PMID: 38711693 PMCID: PMC11070887 DOI: 10.7759/cureus.57714] [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: 02/27/2024] [Accepted: 04/05/2024] [Indexed: 05/08/2024] Open
Abstract
Multiple sclerosis is the most common autoimmune disease affecting the central nervous system (CNS) worldwide. Multiple sclerosis involves inflammatory demyelination of nerve fibers in the CNS, often presenting with recurrent episodes of focal sensory or motor deficits associated with the region of the CNS affected. The prevalence of this disease has increased rapidly over the last decade. Despite the approval of many new pharmaceutical therapies in the past 20 years, there remains a growing need for alternative therapies to manage the course of this disease. Treatments are separated into two main categories: management of acute flare versus long-term prevention of flares via disease-modifying therapy. Primary drug therapies for acute flare include corticosteroids to limit inflammation and symptomatic management, depending on symptoms. Several different drugs have been recently approved for use in modifying the course of the disease, including a group of medications known as fumarates (e.g., dimethyl fumarate, diroximel fumarate, monomethyl fumarate) that have been shown to be efficacious and relatively safe. In the present investigation, we review available evidence focused on monomethyl fumarate, also known as Bafiertam®, along with bioequivalent fumarates for the long-term treatment of relapsing-remitting multiple sclerosis.
Collapse
Affiliation(s)
- Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - John Lacey
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Viet Le
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Ahmed Fazal
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | | | - Dorothy H Askins
- Department of Anesthesiology, Tulane University, New Orleans, USA
| | - Lillian Anderson
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Christopher L Robinson
- Department of Anesthesiology, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Antonella Paladini
- Department of Life, Health, and Environmental Sciences (MESVA), University of L'Aquila, L'Aquila, ITA
| | - Chizoba N Mosieri
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Adam M Kaye
- Department of Pharmacy Practice, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, USA
| | - Shahab Ahmadzadeh
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | | |
Collapse
|
11
|
Wang AA, Luessi F, Neziraj T, Pössnecker E, Zuo M, Engel S, Hanuscheck N, Florescu A, Bugbee E, Ma XI, Rana F, Lee D, Ward LA, Kuhle J, Himbert J, Schraad M, van Puijenbroek E, Klein C, Urich E, Ramaglia V, Pröbstel AK, Zipp F, Gommerman JL. B cell depletion with anti-CD20 promotes neuroprotection in a BAFF-dependent manner in mice and humans. Sci Transl Med 2024; 16:eadi0295. [PMID: 38446903 DOI: 10.1126/scitranslmed.adi0295] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 02/13/2024] [Indexed: 03/08/2024]
Abstract
Anti-CD20 therapy to deplete B cells is highly efficacious in preventing new white matter lesions in patients with relapsing-remitting multiple sclerosis (RRMS), but its protective capacity against gray matter injury and axonal damage is unclear. In a passive experimental autoimmune encephalomyelitis (EAE) model whereby TH17 cells promote brain leptomeningeal immune cell aggregates, we found that anti-CD20 treatment effectively spared myelin content and prevented myeloid cell activation, oxidative damage, and mitochondrial stress in the subpial gray matter. Anti-CD20 treatment increased B cell survival factor (BAFF) in the serum, cerebrospinal fluid, and leptomeninges of mice with EAE. Although anti-CD20 prevented gray matter demyelination, axonal loss, and neuronal atrophy, co-treatment with anti-BAFF abrogated these benefits. Consistent with the murine studies, we observed that elevated BAFF concentrations after anti-CD20 treatment in patients with RRMS were associated with better clinical outcomes. Moreover, BAFF promoted survival of human neurons in vitro. Together, our data demonstrate that BAFF exerts beneficial functions in MS and EAE in the context of anti-CD20 treatment.
Collapse
Affiliation(s)
- Angela A Wang
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Felix Luessi
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Tradite Neziraj
- Department of Neurology, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
| | - Elisabeth Pössnecker
- Department of Neurology, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
| | - Michelle Zuo
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Sinah Engel
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Nicholas Hanuscheck
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Alexandra Florescu
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Eryn Bugbee
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Xianjie I Ma
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Fatima Rana
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Dennis Lee
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Lesley A Ward
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Jens Kuhle
- Department of Neurology, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
| | - Johannes Himbert
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Muriel Schraad
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | | | - Christian Klein
- Roche Innovation Center Zurich, Roche Glycart AG, 8952 Schlieren, Switzerland
| | - Eduard Urich
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4052 Basel, Switzerland
| | - Valeria Ramaglia
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Anne-Katrin Pröbstel
- Department of Neurology, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | | |
Collapse
|
12
|
Yang Y, Wang Y, Wang Z, Yan H, Gong Y, Hu Y, Jiang Y, Wen S, Xu F, Wang B, Humphries F, Chen Y, Wang X, Yang S. ECSIT facilitates memory CD8 + T cell development by mediating fumarate synthesis during viral infection and tumorigenesis. Nat Cell Biol 2024; 26:450-463. [PMID: 38326554 DOI: 10.1038/s41556-024-01351-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 01/07/2024] [Indexed: 02/09/2024]
Abstract
Memory CD8+ T cells play a crucial role in infection and cancer and mount rapid responses to repeat antigen exposure. Although memory cell transcriptional programmes have been previously identified, the regulatory mechanisms that control the formation of CD8+ T cells have not been resolved. Here we report ECSIT as an essential mediator of memory CD8+ T cell differentiation. Ablation of ECSIT in T cells resulted in loss of fumarate synthesis and abrogated TCF-1 expression via demethylation of the TCF-1 promoter by the histone demethylase KDM5, thereby impairing memory CD8+ T cell development in a cell-intrinsic manner. In addition, ECSIT expression correlated positively with stem-like memory progenitor exhausted CD8+ T cells and the survival of patients with cancer. Our study demonstrates that ECSIT-mediated fumarate synthesis stimulates TCF-1 activity and memory CD8+ T cell development during viral infection and tumorigenesis and highlights the utility of therapeutic fumarate analogues and PD-L1 inhibition for tumour immunotherapy.
Collapse
Affiliation(s)
- Yongbing Yang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, National Vaccine Innovation Platform, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
- Department of Medical Laboratory, Affiliated Children's Hospital of Jiangnan University, Wuxi, China
| | - Yanan Wang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, National Vaccine Innovation Platform, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Zhongcheng Wang
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Huanyu Yan
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Yi Gong
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, National Vaccine Innovation Platform, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yingchao Hu
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, National Vaccine Innovation Platform, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yuying Jiang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, National Vaccine Innovation Platform, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Shuang Wen
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, National Vaccine Innovation Platform, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Feifei Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Bingwei Wang
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Fiachra Humphries
- Division of Innate Immunity, Department of Medicine, UMass Chan Medical School, Worcester, MA, USA.
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, Nanjing, China.
| | - Xi Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.
| | - Shuo Yang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, National Vaccine Innovation Platform, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
13
|
Tieck MP, Vasilenko N, Ruschil C, Kowarik MC. Peripheral memory B cells in multiple sclerosis vs. double negative B cells in neuromyelitis optica spectrum disorder: disease driving B cell subsets during CNS inflammation. Front Cell Neurosci 2024; 18:1337339. [PMID: 38385147 PMCID: PMC10879280 DOI: 10.3389/fncel.2024.1337339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024] Open
Abstract
B cells are fundamental players in the pathophysiology of autoimmune diseases of the central nervous system, such as multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD). A deeper understanding of disease-specific B cell functions has led to the differentiation of both diseases and the development of different treatment strategies. While NMOSD is strongly associated with pathogenic anti-AQP4 IgG antibodies and proinflammatory cytokine pathways, no valid autoantibodies have been identified in MS yet, apart from certain antigen targets that require further evaluation. Although both diseases can be effectively treated with B cell depleting therapies, there are distinct differences in the peripheral B cell subsets that influence CNS inflammation. An increased peripheral blood double negative B cells (DN B cells) and plasmablast populations has been demonstrated in NMOSD, but not consistently in MS patients. Furthermore, DN B cells are also elevated in rheumatic diseases and other autoimmune entities such as myasthenia gravis and Guillain-Barré syndrome, providing indirect evidence for a possible involvement of DN B cells in other autoantibody-mediated diseases. In MS, the peripheral memory B cell pool is affected by many treatments, providing indirect evidence for the involvement of memory B cells in MS pathophysiology. Moreover, it must be considered that an important effector function of B cells in MS may be the presentation of antigens to peripheral immune cells, including T cells, since B cells have been shown to be able to recirculate in the periphery after encountering CNS antigens. In conclusion, there are clear differences in the composition of B cell populations in MS and NMOSD and treatment strategies differ, with the exception of broad B cell depletion. This review provides a detailed overview of the role of different B cell subsets in MS and NMOSD and their implications for treatment options. Specifically targeting DN B cells and plasmablasts in NMOSD as opposed to memory B cells in MS may result in more precise B cell therapies for both diseases.
Collapse
Affiliation(s)
| | | | | | - M. C. Kowarik
- Department of Neurology and Stroke, Center for Neurology, and Hertie-Institute for Clinical Brain Research Eberhard-Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
14
|
Hajizadeh M, Jabbari A, Spotin A, Hejazian SS, Mikaeili Galeh T, Hassannia H, Sahlolbei M, Pagheh AS, Ahmadpour E. Modulatory Effects of Hydatid Cyst Fluid on a Mouse Model of Experimental Autoimmune Encephalomyelitis. Vet Sci 2024; 11:34. [PMID: 38250940 PMCID: PMC10819194 DOI: 10.3390/vetsci11010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
The reduced burden of helminth parasites in industrialized countries is probably one of the reasons for the increased prevalence of autoimmune disorders such as multiple sclerosis (MS). The current study aimed to evaluate the potential preventive effects of hydatid cyst fluid (HCF) on the disease severity in an EAE mouse model of MS. EAE-induced mice were treated with HCF before and after EAE induction. An RT-PCR-based evaluation of IFN-γ, IL-1β, TNF, T-bet, IL-4, GATA3, IL-17, RoRγ, TGF-β, and FOXP3 expression levels in splenocytes and an ELISA-based analysis of IFN-γ and IL-4 levels in cell culture supernatant of splenocytes were performed. Histopathological examinations of mice during the study were also conducted. The expression levels of T-bet, IL-4, GATA3, TGF-β, and FOXP3 in EAE + HCF mice were significantly higher compared to EAE + PBS mice. In the EAE + HCF group, the expression levels of IFN-γ, IL-1β, and TNF were significantly lower than in the EAE + PBS group. The histopathological results showed significantly reduced inflammation and demyelination in EAE + HCF mice compared to EAE + PBS mice. Our study provides proof-of-concept in the EAE mouse model of MS that helminth-derived products such as HCF have a potential prophylactic effect on MS development and present a novel potential therapeutic strategy.
Collapse
Affiliation(s)
- Maryam Hajizadeh
- Infectious and Tropical Disease Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
| | - Aynaz Jabbari
- Infectious and Tropical Disease Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
| | - Adel Spotin
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran (S.S.H.)
| | - Seyyed Sina Hejazian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran (S.S.H.)
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
| | - Tahereh Mikaeili Galeh
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy 53464-58167, Iran
| | - Hadi Hassannia
- Immunogenetic Research Center, Faculty of Medicine and Amol Faculty of Paramedical Sciences, Mazandaran, University of Medical Sciences, Sari 48175-866, Iran
| | - Maryam Sahlolbei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran (S.S.H.)
| | - Abdol Sattar Pagheh
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand 14619-65381, Iran
| | - Ehsan Ahmadpour
- Infectious and Tropical Disease Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran (S.S.H.)
| |
Collapse
|
15
|
Vakrakou AG, Brinia ME, Alexaki A, Koumasopoulos E, Stathopoulos P, Evangelopoulos ME, Stefanis L, Stadelmann-Nessler C, Kilidireas C. Multiple faces of multiple sclerosis in the era of highly efficient treatment modalities: Lymphopenia and switching treatment options challenges daily practice. Int Immunopharmacol 2023; 125:111192. [PMID: 37951198 DOI: 10.1016/j.intimp.2023.111192] [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: 09/03/2023] [Revised: 10/25/2023] [Accepted: 11/03/2023] [Indexed: 11/13/2023]
Abstract
The expanded treatment landscape in relapsing-remitting multiple sclerosis (MS) has resulted in highly effective treatment options and complexity in managing disease- or drug-related events during disease progression. Proper decision-making requires thorough knowledge of the immunobiology of MS itself and an understanding of the main principles behind the mechanisms that lead to secondary autoimmunity affecting organs other than the central nervous system as well as opportunistic infections. The immune system is highly adapted to both environmental and disease-modifying agents. Immune reconstitution following cell depletion or cell entrapment therapies eliminates pathogenic aspects of the disease but can also lead to distorted immune responses with harmful effects. Atypical relapses occur with second-line treatments or after their discontinuation and require appropriate clinical decisions. Lymphopenia is a result of the mechanism of action of many drugs used to treat MS. However, persistent lymphopenia and cell-specific lymphopenia could result in disease exacerbation, secondary autoimmunity, or the emergence of opportunistic infections. Clinicians treating patients with MS should be aware of the multiple faces of MS under novel, efficient treatment modalities and understand the intricate brain-immune cell interactions in the context of an altered immune system. MS relapses and disease progression still occur despite the current treatment modalities and are mediated either by failure to control effector mechanisms inherent to MS pathophysiology or by new drug-related mechanisms. The multiple faces of MS due to the highly adapted immune system of patients impose the need for appropriate switching therapies that safeguard disease remission and further clinical improvement.
Collapse
Affiliation(s)
- Aigli G Vakrakou
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece; Department of Neuropathology, University of Göttingen Medical Center, Göttingen, Germany.
| | - Maria-Evgenia Brinia
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia Alexaki
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Koumasopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panos Stathopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Eleftheria Evangelopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Leonidas Stefanis
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Constantinos Kilidireas
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece; Department of Neurology, Henry Dunant Hospital Center, Athens, Greece
| |
Collapse
|
16
|
Hojjati S, Ernerudh J, Vrethem M, Mellergård J, Raffetseder J. Dimethyl fumarate treatment in relapsing remitting MS changes the inflammatory CSF protein profile by a prominent decrease in T-helper 1 immunity. Mult Scler Relat Disord 2023; 80:105126. [PMID: 37952502 DOI: 10.1016/j.msard.2023.105126] [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: 09/26/2023] [Accepted: 11/04/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Dimethyl fumarate (DMF) is a common treatment for multiple sclerosis (MS), but its mechanisms of action are not fully understood. Targeted proteomics offers insights into effects of DMF and biomarkers for treatment responses. OBJECTIVES To assess influence of DMF on inflammation- and neuro-associated proteins in plasma and cerebrospinal fluid (CSF) in MS and to reveal biomarkers for predicting treatment responses. METHODS Using the high-sensitivity and high-specificity method of proximity extension assay (PEA), we measured 182 inflammation- and neuro-associated proteins in paired plasma (n = 28) and CSF (n = 12) samples before and after one year of DMF treatment. Disease activity was evaluated through clinical examination and MRI. Statistical tests, network analysis, and regression models were used. RESULTS Several proteins including T-helper 1 (Th1)-associated proteins (CXCL10, CXCL11, granzyme A, IL-12p70, lymphotoxin-alpha) were consistently decreased in CSF, while IL-7 was increased after one year of treatment. The changes in plasma protein levels did not follow the same pattern as in CSF. Logistic regression models identified potential biomarker candidates (including plexins and neurotrophins) for prediction of treatment response. CONCLUSIONS DMF treatment induced prominent changes in CSF proteins, consistently reducing Th1-associated pro-inflammatory proteins. Neurodegeneration-related CSF proteins were able to predict treatment response. Protein biomarkers hold promise for personalized medicine.
Collapse
Affiliation(s)
- Sara Hojjati
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Jan Ernerudh
- Department of Clinical Immunology and Transfusion Medicine in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Magnus Vrethem
- Department of Neurology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johan Mellergård
- Department of Neurology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| | - Johanna Raffetseder
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
17
|
AlShammari RZ, AlOqayli FA, Alnafeesy SK, Al Thubaiti I. Reactivation of Herpes Zoster in a Young Patient With Multiple Sclerosis Under Dimethyl Fumarate Treatment and Normal Lymphocyte Subsets Count: A Case Report. Cureus 2023; 15:e51412. [PMID: 38292998 PMCID: PMC10827281 DOI: 10.7759/cureus.51412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/31/2023] [Indexed: 02/01/2024] Open
Abstract
Herpes zoster (HZ) infection results from the reactivation of the varicella-zoster virus (VZV), which remains dormant in the dorsal root ganglia after an initial chickenpox infection. Although HZ appears more common in people with multiple sclerosis (MS) than expected in the general population, few studies have investigated this association, particularly with a normal absolute lymphocyte count (ALC). Additionally, no reported cases have discussed the clinical presentation of such patients. This report describes the case of a 26-year-old female with a known history of relapsing-remitting MS on dimethyl fumarate (DMF) treatment. She presented with a history of painful erythematous blisters, diagnosed as acute HZ infection with a normal ALC. This case provides evidence that warrants further research and attention to the management of patients with MS receiving DMF, particularly regarding infectious risks. It highlights the importance of pharmacovigilance and the potential benefits of VZV and HZ immunization in DMF recipients.
Collapse
Affiliation(s)
| | - Fatimah A AlOqayli
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, SAU
| | - Saleh K Alnafeesy
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, SAU
| | | |
Collapse
|
18
|
Balshi A, Saart E, Pandeya S, Dempsey J, Baber U, Sloane JA. High CD4+:CD8+ ratios with herpes zoster infections in patients with multiple sclerosis on dimethyl fumarate. Mult Scler 2023; 29:1465-1470. [PMID: 37572049 DOI: 10.1177/13524585231189641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/14/2023]
Abstract
BACKGROUND Dimethyl fumarate (DMF) depletes CD8+ and CD4+ T cells, and cases of herpes zoster (HZ) in patients with multiple sclerosis (MS) on DMF have been documented. OBJECTIVES To evaluate lymphocyte subsets in patients with MS who developed HZ on DMF (Tecfidera) compared to matched controls who did not develop HZ. METHODS We used linear mixed-effects models to test for differences in white blood cell count, lymphocyte percentage, absolute lymphocyte count, CD3+ percentage, absolute CD3+ count, CD4+ percentage, absolute CD4+ count, CD8+ percentage, absolute CD8+ count, and CD4+:CD8+ ratio over time in HZ and non-HZ groups. RESULTS Eighteen patients developed HZ while on DMF. The linear mixed-effects model for CD4+:CD8+ ratio showed a significant difference between the HZ and non-HZ groups (p = 0.033). CD4+:CD8+ ratio decreased over time in the HZ group and increased over time in the non-HZ group. CONCLUSION Patients with MS who develop HZ while on DMF have high CD4+:CD8+ ratios, suggesting an imbalance of CD4+ and CD8+ cells that may put a patient at risk for developing HZ while on DMF. This result emphasizes the need for lymphocyte subset monitoring (including CD4+:CD8+ ratios) on DMF, as well as vaccination prior to DMF initiation.
Collapse
Affiliation(s)
- Alexandra Balshi
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Emma Saart
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Sarbesh Pandeya
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - John Dempsey
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ursela Baber
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jacob A Sloane
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
19
|
Brune-Ingebretsen S, Høgestøl EA, de Rosbo NK, Berg-Hansen P, Brunborg C, Blennow K, Zetterberg H, Paul F, Uccelli A, Villoslada P, Harbo HF, Berge T. Immune cell subpopulations and serum neurofilament light chain are associated with increased risk of disease worsening in multiple sclerosis. J Neuroimmunol 2023; 382:578175. [PMID: 37573634 DOI: 10.1016/j.jneuroim.2023.578175] [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: 03/13/2023] [Revised: 05/18/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023]
Abstract
Changes is lymphocyte subpopulations in peripheral blood have been proposed as biomarkers for evaluation of disease activity in multiple sclerosis (MS). Serum neurofilament light chain (sNfL) is a biomarker reflecting neuro-axonal injury in MS that could be used to monitor disease activity, response to drugs and to prognosticate disease course. Here we show a moderate correlation between sNfL and lymphocyte cell subpopulations, and our data furthermore suggest that sNfL and specific immune cell subpopulations together could predict future disease worsening in MS.
Collapse
Affiliation(s)
- Synne Brune-Ingebretsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway.
| | - Einar A Høgestøl
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Nicole Kerlero de Rosbo
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy; TomaLab, Institute of Nanotechnology, National Research Council (CNR), Rome, Italy
| | - Pål Berg-Hansen
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Cathrine Brunborg
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom; UK Dementia Research Institute at UCL, London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitaetsmedizin Berlin, Berlin, Germany; NeuroCure Clinical Research Center, Charité-Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Antonio Uccelli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy; Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Pablo Villoslada
- Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain
| | - Hanne F Harbo
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Tone Berge
- Department of Research, Innovation and Education, Oslo University Hospital, Oslo, Norway; Department of Mechanical, Electronic and Chemical Engineering, Oslo Metropolitan University, Oslo, Norway
| |
Collapse
|
20
|
Zhu C, Kalincik T, Horakova D, Zhou Z, Buzzard K, Skibina O, Alroughani R, Izquierdo G, Eichau S, Kuhle J, Patti F, Grand’Maison F, Hodgkinson S, Grammond P, Lechner-Scott J, Butler E, Prat A, Girard M, Duquette P, Macdonell RAL, Weinstock-Guttman B, Ozakbas S, Slee M, Sa MJ, Van Pesch V, Barnett M, Van Wijmeersch B, Gerlach O, Prevost J, Terzi M, Boz C, Laureys G, Van Hijfte L, Kermode AG, Garber J, Yamout B, Khoury SJ, Merlo D, Monif M, Jokubaitis V, van der Walt A, Butzkueven H. Comparison Between Dimethyl Fumarate, Fingolimod, and Ocrelizumab After Natalizumab Cessation. JAMA Neurol 2023; 80:739-748. [PMID: 37273217 PMCID: PMC10242509 DOI: 10.1001/jamaneurol.2023.1542] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/27/2023] [Indexed: 06/06/2023]
Abstract
Importance Natalizumab cessation is associated with a risk of rebound disease activity. It is important to identify the optimal switch disease-modifying therapy strategy after natalizumab to limit the risk of severe relapses. Objectives To compare the effectiveness and persistence of dimethyl fumarate, fingolimod, and ocrelizumab among patients with relapsing-remitting multiple sclerosis (RRMS) who discontinued natalizumab. Design, Setting, and Participants In this observational cohort study, patient data were collected from the MSBase registry between June 15, 2010, and July 6, 2021. The median follow-up was 2.7 years. This was a multicenter study that included patients with RRMS who had used natalizumab for 6 months or longer and then were switched to dimethyl fumarate, fingolimod, or ocrelizumab within 3 months after natalizumab discontinuation. Patients without baseline data were excluded from the analysis. Data were analyzed from May 24, 2022, to January 9, 2023. Exposures Dimethyl fumarate, fingolimod, and ocrelizumab. Main Outcomes and Measures Primary outcomes were annualized relapse rate (ARR) and time to first relapse. Secondary outcomes were confirmed disability accumulation, disability improvement, and subsequent treatment discontinuation, with the comparisons for the first 2 limited to fingolimod and ocrelizumab due to the small number of patients taking dimethyl fumarate. The associations were analyzed after balancing covariates using an inverse probability of treatment weighting method. Results Among 66 840 patients with RRMS, 1744 had used natalizumab for 6 months or longer and were switched to dimethyl fumarate, fingolimod, or ocrelizumab within 3 months of natalizumab discontinuation. After excluding 358 patients without baseline data, a total of 1386 patients (mean [SD] age, 41.3 [10.6] years; 990 female [71%]) switched to dimethyl fumarate (138 [9.9%]), fingolimod (823 [59.4%]), or ocrelizumab (425 [30.7%]) after natalizumab. The ARR for each medication was as follows: ocrelizumab, 0.06 (95% CI, 0.04-0.08); fingolimod, 0.26 (95% CI, 0.12-0.48); and dimethyl fumarate, 0.27 (95% CI, 0.12-0.56). The ARR ratio of fingolimod to ocrelizumab was 4.33 (95% CI, 3.12-6.01) and of dimethyl fumarate to ocrelizumab was 4.50 (95% CI, 2.89-7.03). Compared with ocrelizumab, the hazard ratio (HR) of time to first relapse was 4.02 (95% CI, 2.83-5.70) for fingolimod and 3.70 (95% CI, 2.35-5.84) for dimethyl fumarate. The HR of treatment discontinuation was 2.57 (95% CI, 1.74-3.80) for fingolimod and 4.26 (95% CI, 2.65-6.84) for dimethyl fumarate. Fingolimod use was associated with a 49% higher risk for disability accumulation compared with ocrelizumab. There was no significant difference in disability improvement rates between fingolimod and ocrelizumab. Conclusion and Relevance Study results show that among patients with RRMS who switched from natalizumab to dimethyl fumarate, fingolimod, or ocrelizumab, ocrelizumab use was associated with the lowest ARR and discontinuation rates, and the longest time to first relapse.
Collapse
Affiliation(s)
- Chao Zhu
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Tomas Kalincik
- Clinical Outcomes Research Unit (CORe), Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
- Department of Neurology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Dana Horakova
- Charles University in Prague and General University Hospital, Prague, Czech Republic
| | - Zhen Zhou
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Katherine Buzzard
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Box Hill Hospital, Melbourne, Victoria, Australia
| | - Olga Skibina
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Box Hill Hospital, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | | | | | - Sara Eichau
- Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Jens Kuhle
- University Hospital and University of Basel, Basel, Switzerland
| | - Francesco Patti
- Multiple Sclerosis Center, University of Catania, Catania, Italy
| | | | | | | | | | - Ernest Butler
- Monash Medical Centre, Melbourne, Victoria, Australia
| | - Alexandre Prat
- CHUM MS Center and Université de Montréal, Montréal, Québec, Canada
| | - Marc Girard
- CHUM MS Center and Université de Montréal, Montréal, Québec, Canada
| | - Pierre Duquette
- CHUM MS Center and Université de Montréal, Montréal, Québec, Canada
| | | | | | | | - Mark Slee
- Flinders University, Adelaide, South Australia, Australia
| | - Maria Jose Sa
- Centro Hospitalar Universitario de São João, Porto, Portugal
| | | | | | - Bart Van Wijmeersch
- Rehabilitation and MS-Centre Overpelt and Hasselt University, Hasselt, Belgium
| | - Oliver Gerlach
- Zuyderland Medical Center, Sittard-Geleen, the Netherlands
| | | | | | - Cavit Boz
- KTU Medical Faculty Farabi Hospital, Trabzon, Turkey
| | | | | | - Allan G. Kermode
- University of Western Australia, Nedlands, Western Australia, Australia
| | - Justin Garber
- Westmead Hospital, Sydney, New South Wales, Australia
| | - Bassem Yamout
- American University of Beirut Medical Center, Beirut, Lebanon
| | - Samia J. Khoury
- American University of Beirut Medical Center, Beirut, Lebanon
| | - Daniel Merlo
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Mastura Monif
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Vilija Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Anneke van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Pant A, Dasgupta D, Tripathi A, Pyaram K. Beyond Antioxidation: Keap1-Nrf2 in the Development and Effector Functions of Adaptive Immune Cells. Immunohorizons 2023; 7:288-298. [PMID: 37099275 PMCID: PMC10579846 DOI: 10.4049/immunohorizons.2200061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/05/2023] [Indexed: 04/27/2023] Open
Abstract
Ubiquitously expressed in mammalian cells, the Kelch-like ECH-associated protein 1 (Keap1)-NF erythroid 2-related factor 2 (Nrf2) complex forms the evolutionarily conserved antioxidation system to tackle oxidative stress caused by reactive oxygen species. Reactive oxygen species, generated as byproducts of cellular metabolism, were identified as essential second messengers for T cell signaling, activation, and effector responses. Apart from its traditional role as an antioxidant, a growing body of evidence indicates that Nrf2, tightly regulated by Keap1, modulates immune responses and regulates cellular metabolism. Newer functions of Keap1 and Nrf2 in immune cell activation and function, as well as their role in inflammatory diseases such as sepsis, inflammatory bowel disease, and multiple sclerosis, are emerging. In this review, we highlight recent findings about the influence of Keap1 and Nrf2 in the development and effector functions of adaptive immune cells, that is, T cells and B cells, and discuss the knowledge gaps in our understanding. We also summarize the research potential and targetability of Nrf2 for treating immune pathologies.
Collapse
Affiliation(s)
- Anil Pant
- Department of Veterinary Pathobiology, School of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX
| | - Debolina Dasgupta
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS
| | - Aprajita Tripathi
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS
| | - Kalyani Pyaram
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
22
|
Baeva ME, Metz LM, Greenfield J, Camara-Lemarroy CR. Simple Parameters from Complete Blood Count Predict Lymphopenia, Adverse Effects and Efficacy in People with MS treated with Dimethyl Fumarate. Mult Scler Relat Disord 2023; 74:104699. [PMID: 37031552 DOI: 10.1016/j.msard.2023.104699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/28/2023] [Accepted: 04/02/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Dimethyl fumarate (DMF) is a first-line oral therapy for relapsing-remitting multiple sclerosis (RRMS). This retrospective study aims to determine the utility of routine complete blood counts (CBC) in predicting lymphopenia, adverse effects and efficacy in a real-world clinical setting. METHODS The Calgary Multiple Sclerosis (MS) Clinic manages over 1800 people with MS on disease-modifying therapies (DMT). Data of patients with relapsing-remitting MS (pwMS) who initiated DMF between July 1, 2013 and December 31, 2014 were included. Patients were followed for one year. DMT use is carefully monitored and pwMS need a screening CBC and have regular CBCs done at follow-up. Demographic, clinical, MRI and relapse information are collected prospectively in a clinic database. We analyzed CBCs at baseline and month 3. RESULTS We identified 139 pwMS in the study period who started DMF. Median follow-up time on-drug was 12 (0.16-12) months. In our study, 15.8% of pwMS developed lymphopenia grade 2 or higher. Baseline lymphocyte counts and older age were significant predictors of lymphopenia. Higher baseline eosinophil counts predicted flushing/gastrointestinal adverse effects, and higher baseline monocyte counts were predictive of breakthrough disease activity. Neutrophil and platelet to lymphocyte ratios, markers that have been associated with overall mortality in the general population, were increased at month 3. CONCLUSIONS Routinely obtained CBCs during the screening and monitoring of people with MS starting DMF offer clinically useful information and generate interesting hypotheses. Age and baseline lymphocyte counts are reinforced as clinically useful predictors of lymphopenia. Our novel findings that baseline eosinophil and monocyte counts could offer insights into usual adverse effects and efficacy, respectively, should be further investigated as a potentially new set of biomarkers.
Collapse
|
23
|
Kohle F, Dalakas MC, Lehmann HC. Repurposing MS immunotherapies for CIDP and other autoimmune neuropathies: unfulfilled promise or efficient strategy? Ther Adv Neurol Disord 2023; 16:17562864221137129. [PMID: 36620728 PMCID: PMC9810996 DOI: 10.1177/17562864221137129] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/19/2022] [Indexed: 01/03/2023] Open
Abstract
Despite advances in the treatment of chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) and other common autoimmune neuropathies (AN), still-many patients with these diseases do not respond satisfactorily to the available treatments. Repurposing of disease-modifying therapies (DMTs) from other autoimmune conditions, particularly multiple sclerosis (MS) and neuromyelitis optica spectrum disorders (NMOSD), is a promising strategy that may accelerate the establishment of novel treatment choices for AN. This approach appears attractive due to homologies in the pathogenesis of these diseases and the extensive post-marketing experience that has been gathered from treating MS and NMOSD patients. The idea is also strengthened by a number of studies that explored the efficacy of DMTs in animal models of AN but also in some CIDP patients. We here review the available preclinical and clinical data of approved MS therapeutics in terms of their applicability to AN, especially CIDP. Promising therapeutic approaches appear to be B cell-directed and complement-targeting strategies, such as anti-CD20/anti-CD19 agents, Bruton's tyrosine kinase inhibitors and anti-C5 agents, as they exert their effects in the periphery. This is a major advantage because, in contrast to MS, their action in the periphery is sufficient to exert significant immunomodulation.
Collapse
Affiliation(s)
- Felix Kohle
- Department of Neurology, Faculty of Medicine,
University of Cologne and University Hospital Cologne, Cologne,
Germany
| | - Marinos C. Dalakas
- Department of Neurology, Thomas Jefferson
University, Philadelphia, PA, USA
- Neuroimmunology Unit, National and Kapodistrian
University of Athens Medical School, Athens, Greece
| | - Helmar C. Lehmann
- Department of Neurology, Faculty of Medicine,
University of Cologne and University Hospital Cologne, Kerpener Strasse, 62,
50937 Cologne, Germany
| |
Collapse
|
24
|
Mey GM, Mahajan KR, DeSilva TM. Neurodegeneration in multiple sclerosis. WIREs Mech Dis 2023; 15:e1583. [PMID: 35948371 PMCID: PMC9839517 DOI: 10.1002/wsbm.1583] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/28/2022] [Accepted: 07/11/2022] [Indexed: 01/31/2023]
Abstract
Axonal loss in multiple sclerosis (MS) is a key component of disease progression and permanent neurologic disability. MS is a heterogeneous demyelinating and neurodegenerative disease of the central nervous system (CNS) with varying presentation, disease courses, and prognosis. Immunomodulatory therapies reduce the frequency and severity of inflammatory demyelinating events that are a hallmark of MS, but there is minimal therapy to treat progressive disease and there is no cure. Data from patients with MS, post-mortem histological analysis, and animal models of demyelinating disease have elucidated patterns of MS pathogenesis and underlying mechanisms of neurodegeneration. MRI and molecular biomarkers have been proposed to identify predictors of neurodegeneration and risk factors for disease progression. Early signs of axonal dysfunction have come to light including impaired mitochondrial trafficking, structural axonal changes, and synaptic alterations. With sustained inflammation as well as impaired remyelination, axons succumb to degeneration contributing to CNS atrophy and worsening of disease. These studies highlight the role of chronic demyelination in the CNS in perpetuating axonal loss, and the difficulty in promoting remyelination and repair amidst persistent inflammatory insult. Regenerative and neuroprotective strategies are essential to overcome this barrier, with early intervention being critical to rescue axonal integrity and function. The clinical and basic research studies discussed in this review have set the stage for identifying key propagators of neurodegeneration in MS, leading the way for neuroprotective therapeutic development. This article is categorized under: Immune System Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Gabrielle M. Mey
- Department of NeurosciencesLerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve UniversityClevelandOhioUSA
| | - Kedar R. Mahajan
- Department of NeurosciencesLerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve UniversityClevelandOhioUSA
- Mellen Center for MS Treatment and ResearchNeurological Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Tara M. DeSilva
- Department of NeurosciencesLerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
25
|
High-dimensional immune profiling identifies a biomarker to monitor dimethyl fumarate response in multiple sclerosis. Proc Natl Acad Sci U S A 2022; 119:e2205042119. [PMID: 35881799 PMCID: PMC9351505 DOI: 10.1073/pnas.2205042119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Dimethyl fumarate (DMF) is an immunomodulatory treatment for multiple sclerosis (MS). Despite its wide clinical use, the mechanisms underlying clinical response are not understood. This study aimed to reveal immune markers of therapeutic response to DMF treatment in MS. For this purpose, we prospectively collected peripheral blood mononuclear cells (PBMCs) from a highly characterized cohort of 44 individuals with MS before and at 12 and 48 wk of DMF treatment. Single cells were profiled using high-dimensional mass cytometry. To capture the heterogeneity of different immune subsets, we adopted a bioinformatic multipanel approach that allowed cell population-cluster assignment of more than 50 different parameters, including lineage and activation markers as well as chemokine receptors and cytokines. Data were further analyzed in a semiunbiased fashion implementing a supervised representation learning approach to capture subtle longitudinal immune changes characteristic for therapy response. With this approach, we identified a population of memory T helper cells expressing high levels of neuroinflammatory cytokines (granulocyte-macrophage colony-stimulating factor [GM-CSF], interferon γ [IFNγ]) as well as CXCR3, whose abundance correlated with treatment response. Using spectral flow cytometry, we confirmed these findings in a second cohort of patients. Serum neurofilament light-chain levels confirmed the correlation of this immune cell signature with axonal damage. The identified cell population is expanded in peripheral blood under natalizumab treatment, substantiating a specific role in treatment response. We propose that depletion of GM-CSF-, IFNγ-, and CXCR3-expressing T helper cells is the main mechanism of action of DMF and allows monitoring of treatment response.
Collapse
|
26
|
Dorcet G, Migné H, Biotti D, Bost C, Lerebours F, Ciron J, Treiner E. Early B cells repopulation in multiple sclerosis patients treated with rituximab is not predictive of a risk of relapse or clinical progression. J Neurol 2022; 269:5443-5453. [PMID: 35652942 PMCID: PMC9159933 DOI: 10.1007/s00415-022-11197-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND It is currently unknown whether early B cell reconstitution (EBR) in MS patients under rituximab is associated with a risk of relapse or progression. OBJECTIVES Analyzing EBR in rituximab-treated patients and its putative association with clinical findings. METHODS Prospective lymphocytes immunophenotyping was performed in a monocentric cohort of MS patients treated by rituximab for 2 years. EBR was defined when B cells concentration was > 5 cells/mm3. B cell subsets were retrospectively associated with clinical data. Clinical and radiological monitoring included relapses, EDSS (Expanded Disability Status Scale), SDMT (Symbol Digit Modalities Test), and MRI. RESULTS 182 patients were analyzed (61 remitting-relapsing and 121 progressive-active). 38.5% experienced EBR at least once, but very few (7/182) showed systematic reconstitution. Most patients remained stable upon treatment, regardless of the occurrence of EBR. Dynamics of B cell reconstitution featured increased naïve/transitional B cells, and decreased memory subsets. Homeostasis of the B cell compartment differed at baseline between patients experiencing or not EBR upon treatment. In patients with EBR, reciprocal dynamics of transitional and pro-inflammatory double-negative B cell subsets was associated with better response to rituximab treatment. CONCLUSION EBR is common in rituximab-treated MS patients and is not associated with clinical disease activity. EBR in the peripheral blood may reflect regulatory immunological phenomena in subgroup of patients.
Collapse
Affiliation(s)
- Guillaume Dorcet
- Department of Neurology, CRC-SEP, University Hospital of Toulouse, Toulouse, France.,INSERM U1291-CNRS 5051, INFINITy, Toulouse, France
| | - Hugo Migné
- Immunology Laboratory, Biology Department, University Hospital of Toulouse, Toulouse, France
| | - Damien Biotti
- Department of Neurology, CRC-SEP, University Hospital of Toulouse, Toulouse, France.,INSERM U1291-CNRS 5051, INFINITy, Toulouse, France
| | - Chloé Bost
- INSERM U1291-CNRS 5051, INFINITy, Toulouse, France.,Immunology Laboratory, Biology Department, University Hospital of Toulouse, Toulouse, France
| | - Fleur Lerebours
- Department of Neurology, CRC-SEP, University Hospital of Toulouse, Toulouse, France
| | - Jonathan Ciron
- Department of Neurology, CRC-SEP, University Hospital of Toulouse, Toulouse, France.,INSERM U1291-CNRS 5051, INFINITy, Toulouse, France
| | - Emmanuel Treiner
- INSERM U1291-CNRS 5051, INFINITy, Toulouse, France. .,Immunology Laboratory, Biology Department, University Hospital of Toulouse, Toulouse, France.
| |
Collapse
|
27
|
Borrelli S, Mathias A, Goff GL, Pasquier RD, Théaudin M, Pot C. Delayed and recurrent dimethyl fumarate induced-lymphopenia in patients with Multiple sclerosis. Mult Scler Relat Disord 2022; 63:103887. [DOI: 10.1016/j.msard.2022.103887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/11/2022] [Accepted: 05/13/2022] [Indexed: 11/24/2022]
|
28
|
Apóstolos SLP, Boaventura M, Mendes NT, Teixeira LS, Campana IG. How to choose initial treatment in multiple sclerosis patients: a case-based approach. ARQUIVOS DE NEURO-PSIQUIATRIA 2022; 80:159-172. [PMID: 35976318 PMCID: PMC9491420 DOI: 10.1590/0004-282x-anp-2022-s128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Immunotherapy dramatically changed the natural history of multiple sclerosis (MS), which was classically associated with severe disability. Treatment strategies advocate that early control of disease activity is crucial to avoid progressive disability, and the use of high efficacy drugs may be beneficial, but safety is a concern. Choosing the disease-modifying therapy is challenging in clinical practice and should be further discussed. OBJECTIVE To discuss the state of art of selecting the initial therapy for relapsing MS patients. METHODS We used a case-based approach followed by clinical discussion, exploring therapeutic options in different MS settings. RESULTS We presented clinical cases profile compatible with the use of MS therapies, classified into moderate and high efficacy. In the moderate efficacy group, we discussed interferons, glatiramer acetate, teriflunomide and dimethyl fumarate, while in the high efficacy group we discussed fingolimod, cladribine, natalizumab, ocrelizumab, alemtuzumab and ofatumumab. CONCLUSION Advances in MS treatment are remarkable. Strong evidence supports the use of early high efficacy therapy. However, biomarkers, clinical and radiologic prognostic factors, as well as patients' individual issues, should be valued and considered for a personalized treatment decision.
Collapse
Affiliation(s)
- Samira Luisa Pereira Apóstolos
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo SP, Brazil
| | - Mateus Boaventura
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo SP, Brazil
| | - Natalia Trombini Mendes
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo SP, Brazil
| | - Larissa Silva Teixeira
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo SP, Brazil
| | - Igor Gusmão Campana
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo SP, Brazil
| |
Collapse
|
29
|
Diebold M, Galli E, Kopf A, Sanderson N, Callegari I, Ingelfinger F, Núñez NG, Benkert P, Kappos L, Kuhle J, Becher B, Claassen M, Derfuss T. Immunological predictors of dimethyl fumarate-induced lymphopenia. Ann Neurol 2022; 91:676-681. [PMID: 35170072 PMCID: PMC9314128 DOI: 10.1002/ana.26328] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 11/16/2022]
Abstract
Treatment with dimethyl fumarate (DMF) leads to lymphopenia and infectious complications in a subset of patients with multiple sclerosis (MS). Here, we aimed to reveal immune markers of DMF‐associated lymphopenia. This prospective observational study longitudinally assessed 31 individuals with MS by single‐cell mass cytometry before and after 12 and 48 weeks of DMF therapy. Employing a neural network‐based representation learning approach, we identified a CCR4‐expressing T helper cell population negatively associated with relevant lymphopenia. CCR4‐expressing T helper cells represent a candidate prognostic biomarker for the development of relevant lymphopenia in patients undergoing DMF treatment. ANN NEUROL 2022;91:676–681
Collapse
Affiliation(s)
- Martin Diebold
- MS Center, Neurologic Clinic and Policlinic, Department of Biomedicine and Research Center for Clinical Neuroimmunology and Neuroscience Basel(RC2NB), University Hospital Basel, University of Basel, Switzerland.,Institute of Neuropathology, Neurozentrum, University Hospital Freiburg, Breisacher Straße 64, 79106, Freiburg, Germany
| | - Edoardo Galli
- MS Center, Neurologic Clinic and Policlinic, Department of Biomedicine and Research Center for Clinical Neuroimmunology and Neuroscience Basel(RC2NB), University Hospital Basel, University of Basel, Switzerland.,Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Andreas Kopf
- Institute of Molecular Systems Biology, ETH Zurich, Switzerland.,Swiss Institute of Bioinformatics (SIB), Zurich, 8093, Switzerland.,Life Science Graduate School Zurich, PhD Program Systems Biology, Winterthurerstr. 190, Zurich, 8057, Switzerland
| | - Nicholas Sanderson
- MS Center, Neurologic Clinic and Policlinic, Department of Biomedicine and Research Center for Clinical Neuroimmunology and Neuroscience Basel(RC2NB), University Hospital Basel, University of Basel, Switzerland
| | - Ilaria Callegari
- MS Center, Neurologic Clinic and Policlinic, Department of Biomedicine and Research Center for Clinical Neuroimmunology and Neuroscience Basel(RC2NB), University Hospital Basel, University of Basel, Switzerland
| | - Florian Ingelfinger
- Institute of Experimental Immunology, University of Zurich, Switzerland.,Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | | | - Pascal Benkert
- MS Center, Neurologic Clinic and Policlinic, Department of Biomedicine and Research Center for Clinical Neuroimmunology and Neuroscience Basel(RC2NB), University Hospital Basel, University of Basel, Switzerland.,Clinical Trial Unit, University Hospital Basel, University of Basel, Switzerland
| | - Ludwig Kappos
- MS Center, Neurologic Clinic and Policlinic, Department of Biomedicine and Research Center for Clinical Neuroimmunology and Neuroscience Basel(RC2NB), University Hospital Basel, University of Basel, Switzerland
| | - Jens Kuhle
- MS Center, Neurologic Clinic and Policlinic, Department of Biomedicine and Research Center for Clinical Neuroimmunology and Neuroscience Basel(RC2NB), University Hospital Basel, University of Basel, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Manfred Claassen
- Division of Clinical Bioinformatics, Department of Internal Medicine I, University of Tübingen, Tübingen, Germany
| | - Tobias Derfuss
- MS Center, Neurologic Clinic and Policlinic, Department of Biomedicine and Research Center for Clinical Neuroimmunology and Neuroscience Basel(RC2NB), University Hospital Basel, University of Basel, Switzerland
| |
Collapse
|
30
|
Wrona D, Majkutewicz I, Świątek G, Dunacka J, Grembecka B, Glac W. Dimethyl Fumarate as the Peripheral Blood Inflammatory Mediators Inhibitor in Prevention of Streptozotocin-Induced Neuroinflammation in Aged Rats. J Inflamm Res 2022; 15:33-52. [PMID: 35027835 PMCID: PMC8749052 DOI: 10.2147/jir.s342280] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/23/2021] [Indexed: 12/17/2022] Open
Abstract
Purpose Intracerebroventricular-(ICV)-streptozotocin-(STZ)-induced neuroinflammation is a model of Alzheimer’s disease (AD) compatible with the inflammation hypothesis of ageing (“inflammaging” state). Previously, we observed age-dependent (young vs aged) dimethyl fumarate (DMF)-induced anti-inflammatory and neuroprotective effects in the brain along with improvement in cognitive functions in rats with the ICV-STZ-induced model of AD. To evaluate whether DMF reduces neuroinflammation based on the peripheral inflammatory response inhibition, we determined peripheral inflammatory mediators in young and aged rats with the ICV-STZ-induced AD pathology following DMF therapy. Materials and Methods Young (4-month-old) and aged (22-month-old) rats were fed with 0.4% DMF rat chow for 21 consecutive days after ICV-STZ (3 mg/ventricle) injections. After behavioral testing, blood and spleens were collected to determine the numbers of leukocytes (WBC), lymphocytes and their subpopulations, haematological parameters, the concanavalin (Con)-A-induced production and plasma concentration of interferon (IFN)-γ, interleukin (IL)-6, IL-10 and corticosterone (COR). Results Age-dependent anti-inflammatory effect of the DMF treatment in rats with ICV-STZ injections manifested as decreased peripheral WBC and lymphocyte numbers, including TCD3+CD4+CD8−, TCD3+CD4−CD8+, B (CD45RA+) and NK (161a+), in aged rats. Furthermore, DMF lowered the blood and spleen lymphocyte production of pro-inflammatory IFN-γ and IL-6 in young and aged rats, whereas it enhanced the plasma level of anti-inflammatory IL-10 and lymphocyte’s ability to produce it in aged rats only. In parallel to changes in peripheral WBC numbers in the model of AD, DMF decreased the red blood cell number, haemoglobin concentration, haematocrit and mean platelet volume in aged, but not young, rats. In contrast to controls, DMF did not influence the COR response in STZ groups. Conclusion Besides preventing neuroinflammation, DMF acts on the pro-/anti-inflammatory balance in the periphery and causes an anti-inflammatory shift in T lymphocytes which could contribute to DMF’s therapeutic effects in the ICV-STZ-induced model of AD, in particular, in aged rats.
Collapse
Affiliation(s)
- Danuta Wrona
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Gdansk, 80-308, Poland
| | - Irena Majkutewicz
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Gdansk, 80-308, Poland
| | - Grzegorz Świątek
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Gdansk, 80-308, Poland
| | - Joanna Dunacka
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Gdansk, 80-308, Poland
| | - Beata Grembecka
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Gdansk, 80-308, Poland
| | - Wojciech Glac
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Gdansk, 80-308, Poland
| |
Collapse
|
31
|
Reynolds A, Gaughan M, Holden D, Redenbaugh V, Dunne J, Redmond J, Conlon N. The effects of dimethyl fumarate and fingolimod on T-cell lymphocyte proliferation in patients with multiple sclerosis. Ir J Med Sci 2022; 191:2759-2762. [PMID: 35028898 DOI: 10.1007/s11845-021-02913-8] [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: 03/14/2021] [Accepted: 12/25/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The disease-modifying therapies (DMT), dimethyl fumarate (DMF) and fingolimod (FTY) improve the outcomes in multiple sclerosis (MS) by reducing relapses and numbers and volume of lesions. They mediate their effects through reduction of immune reactivation, which may potentially lead to lymphopaenia and increased risk of infections. Previous studies have examined the effects of these therapies on lymphocyte subsets; however, the in vivo effects on circulating lymphocyte proliferation require further elucidation. The aim of this study was to determine the effects of DMF and FTY on T-cell proliferation in patients with MS. METHOD We examined T-cell lymphocyte proliferation and lymphocyte subsets in ten patients (five on DMF, five on FTY) before starting DMT and again 4 to 11 months after being maintained on DMT. RESULTS In the FTY-treated group, the mean percentage proliferation was significantly lower using both assays (PHA assay mean percentage change - 51.2 ± 25.97, p < 0.05; anti-CD3/CD28 assay mean percentage change - 39.74 ± 27.85, p < 0.05). There was no statistical difference in T-cell lymphocyte proliferation in the DMF-treated group for either assay (PHA, p = 0.316; anti-CD3/CD28, p = 0.373). CONCLUSIONS This pilot study suggests that the T-lymphocytes of patients on FTY have an abnormal proliferation response as well as being reduced in the circulation.
Collapse
Affiliation(s)
- Audrey Reynolds
- Department of Neurology, St James's Hospital, Dublin 8, Ireland.
| | - Maria Gaughan
- Department of Neurology, St James's Hospital, Dublin 8, Ireland
| | - Dean Holden
- Department of Immunology, St James's Hospital, Dublin 8, Ireland
| | | | - Jean Dunne
- Department of Immunology, St James's Hospital, Dublin 8, Ireland
| | - Janice Redmond
- Department of Neurology, St James's Hospital, Dublin 8, Ireland
| | - Niall Conlon
- Department of Immunology, St James's Hospital, Dublin 8, Ireland
| |
Collapse
|
32
|
Lu CH, Huang YF, Chu IM. Design of Oral Sustained-Release Pellets by Modeling and Simulation Approach to Improve Compliance for Repurposing Sobrerol. Pharmaceutics 2022; 14:pharmaceutics14010167. [PMID: 35057064 PMCID: PMC8777650 DOI: 10.3390/pharmaceutics14010167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/20/2021] [Accepted: 01/07/2022] [Indexed: 12/10/2022] Open
Abstract
Sobrerol, an oral mucolytic agent, in a recent study showed promise for treating multiple sclerosis. A human equivalent dose of 486 mg of sobrerol administered thrice daily (i.e., 1459 mg of daily dose) demonstrated the highest therapeutic efficacy for repurposing use, which also points out the poor compliance of administration. In this study, oral sustained-release pellets of sobrerol were successfully developed with evaluated manufacturing conditions and drug release kinetics. For design of the target drug product, we used a modeling and simulation approach to establish a predictive model of oral pharmacokinetic profile, by exploring the characteristics and correlations corresponding to the pharmacokinetics and pharmacodynamics of sobrerol, such as absorption lag time (0.18 h), time-scaling in vitro–in vivo correlation (tin-vitro = 0.494 tin-vivo − 0.0904), gastrointestinal transit time (8 h), minimum effective concentration (1.61 μg/mL), and duration of action (12.8 h). Results showed that the frequency of administration and the daily dose remarkably reduced by 33.3% (i.e., from thrice to twice daily) and 22.8%, respectively, which indicates that this prototype approach can be adopted for rapidly developing a modified-release dosage form of sobrerol, with improvement of compliance of administration and therapeutic efficacy.
Collapse
Affiliation(s)
- Chu-Hsun Lu
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu 30011, Taiwan;
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
- Correspondence: (C.-H.L.); (I.-M.C.)
| | - Yu-Feng Huang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu 30011, Taiwan;
| | - I-Ming Chu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
- Correspondence: (C.-H.L.); (I.-M.C.)
| |
Collapse
|
33
|
Ahmadi A, Fallah Vastani Z, Abounoori M, Azizi M, Labani‐Motlagh A, Mami S, Mami S. The role of NK and NKT cells in the pathogenesis and improvement of multiple sclerosis following disease-modifying therapies. Health Sci Rep 2022; 5:e489. [PMID: 35229046 PMCID: PMC8865072 DOI: 10.1002/hsr2.489] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 11/09/2021] [Accepted: 12/07/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an autoimmune inflammatory disease of the central nervous system (CNS) that T cells become autoreactive by recognizing CNS antigens. Both innate and adaptive immune systems are involved in the pathogenesis of MS. In recent years, the impact of innate immune cells on MS pathogenesis has received more attention. CD56bright NK cells, as an immunoregulatory subset of NK cells, can increase the production of cytokines that modulate adaptive immune responses, whereas CD56dim NK cells are more active in cytolysis functions. These two main subsets of NK cells may have different effects on the onset or progression of MS. Invariant NKT (iNKT) cells are other immune cells involved in the control of autoimmune diseases; however, variant NKT (vNKT) cells, despite limited information, could play a role in MS remission via an immunoregulatory pathway. AIM We aimed to evaluate the influence of MS therapeutic agents on NK and NKT cells and NK cell subtypes. MATERIALS AND METHODS The possible mechanism of each MS therapeutic agent has been presented here, focusing on the effects of different disease-modifying therapies on the number of NK and NKT subtypes. RESULTS Expansion of CD56bright NK cells, reduction in the CD56dim cells, and enhancement in NKT cells are the more important innate immune cells alterations following the disease-modifying therapies. CONCLUSION Expansion of CD56bright NK cells or reduction in the CD56dim cells has been associated with a successful response to different treatments in MS. iNKT and vNKT cells could have beneficial effects on MS improving. It seems that they are enhanced due to some of MS drugs, leading to disease improvement. However, a reduction in the number of NKT cells could be due to the adverse effects of some of MS drugs on the bone marrow.
Collapse
Affiliation(s)
- Alireza Ahmadi
- Student Research Committee, Department of Laboratory Sciences, Faculty of Allied Medical SciencesIlam University of Medical SciencesIlamIran
| | - Zahra Fallah Vastani
- Student Research Committee, Department of Laboratory Sciences, Faculty of Allied Medical SciencesIlam University of Medical SciencesIlamIran
| | - Mahdi Abounoori
- Student Research Committee, School of MedicineMazandaran University of Medical SciencesSariIran
| | - Mahdieh Azizi
- Department of Immunology, School of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Alireza Labani‐Motlagh
- Department of Pulmonary ImmunologyThe University of Texas Health Science Center at TylerTexasUSA
| | - Sajad Mami
- Department of laboratory and clinical science, faculty of veterinary medicineIlam UniversityIlamIran
| | - Sanaz Mami
- Department of Immunology, School of MedicineIlam University of Medical SciencesIlamIran
| |
Collapse
|
34
|
Talbot J, Højsgaard Chow H, Holm Hansen R, von Essen MR, Sellebjerg F. Immunological effects of dimethyl fumarate treatment in blood and CSF of patients with primary progressive MS. J Neuroimmunol 2021; 361:577756. [PMID: 34739914 DOI: 10.1016/j.jneuroim.2021.577756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/10/2021] [Accepted: 10/17/2021] [Indexed: 01/22/2023]
Abstract
Dimethyl fumarate is an efficient therapy used widely in patients with relapsing-remitting multiple sclerosis (RRMS). However, lacking effect of treatment has recently been reported in patients with primary progressive MS (PPMS) (Højsgaard Chow et al., 2021). In order to further analyze the immunological treatment response we investigated the systemic and intrathecal immunological effects of dimethyl fumarate (DMF) treatment in 50 patients with PPMS who participated in a 48-week randomized controlled trial with dimethyl fumarate vs placebo. We found substantial systemic immunomodulatory effects of DMF treatment comparable with those observed in patients with RRMS. However, intrathecal effects were limited and restricted to CD4+ T cells presumably resulting in higher concentrations of intrathecal IL-7.
Collapse
Affiliation(s)
- J Talbot
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark.
| | - H Højsgaard Chow
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - R Holm Hansen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - M Rode von Essen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - F Sellebjerg
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
35
|
Zhai X, Liu K, Fang H, Zhang Q, Gao X, Liu F, Zhou S, Wang X, Niu Y, Hong Y, Lin SH, Liu WH, Xiao C, Li Q, Xiao N. Mitochondrial C1qbp promotes differentiation of effector CD8 + T cells via metabolic-epigenetic reprogramming. SCIENCE ADVANCES 2021; 7:eabk0490. [PMID: 34860557 PMCID: PMC8641941 DOI: 10.1126/sciadv.abk0490] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/15/2021] [Indexed: 05/27/2023]
Abstract
Early-activated CD8+ T cells increase both aerobic glycolysis and mitochondrial oxidative phosphorylation (OXPHOS). However, whether and how the augmentation of OXPHOS regulates differentiation of effector CD8+ T cell remains unclear. Here, we found that C1qbp was intrinsically required for such differentiation in antiviral and antitumor immune responses. Activated C1qbp-deficient CD8+ T cells failed to increase mitochondrial respiratory capacities, resulting in diminished acetyl–coenzyme A as well as elevated fumarate and 2-hydroxyglutarate. Consequently, hypoacetylation of H3K27 and hypermethylation of H3K27 and CpG sites were associated with transcriptional down-regulation of effector signature genes. The effector differentiation of C1qbp-sufficient or C1qbp-deficient CD8+ T cells was reversed by fumarate or a combination of histone deacetylase inhibitor and acetate. Therefore, these findings identify C1qbp as a pivotal positive regulator in the differentiation of effector CD8+ T cells and highlight a metabolic-epigenetic axis in this process.
Collapse
Affiliation(s)
- Xingyuan Zhai
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Kai Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Hongkun Fang
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Quan Zhang
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Xianjun Gao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Fang Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Shangshang Zhou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xinming Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yujia Niu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yazhen Hong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Shu-Hai Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wen-Hsien Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Changchun Xiao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Qiyuan Li
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Nengming Xiao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| |
Collapse
|
36
|
Liebmann M, Korn L, Janoschka C, Albrecht S, Lauks S, Herrmann AM, Schulte-Mecklenbeck A, Schwab N, Schneider-Hohendorf T, Eveslage M, Wildemann B, Luessi F, Schmidt S, Diebold M, Bittner S, Gross CC, Kovac S, Zipp F, Derfuss T, Kuhlmann T, König S, Meuth SG, Wiendl H, Klotz L. Dimethyl fumarate treatment restrains the antioxidative capacity of T cells to control autoimmunity. Brain 2021; 144:3126-3141. [PMID: 34849598 PMCID: PMC8634070 DOI: 10.1093/brain/awab307] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/11/2021] [Accepted: 07/25/2021] [Indexed: 02/02/2023] Open
Abstract
Dimethyl fumarate, an approved treatment for relapsing-remitting multiple sclerosis, exerts pleiotropic effects on immune cells as well as CNS resident cells. Here, we show that dimethyl fumarate exerts a profound alteration of the metabolic profile of human CD4+ as well as CD8+ T cells and restricts their antioxidative capacities by decreasing intracellular levels of the reactive oxygen species scavenger glutathione. This causes an increase in mitochondrial reactive oxygen species levels accompanied by an enhanced mitochondrial stress response, ultimately leading to impaired mitochondrial function. Enhanced mitochondrial reactive oxygen species levels not only result in enhanced T-cell apoptosis in vitro as well as in dimethyl fumarate-treated patients, but are key for the well-known immunomodulatory effects of dimethyl fumarate both in vitro and in an animal model of multiple sclerosis, i.e. experimental autoimmune encephalomyelitis. Indeed, dimethyl fumarate immune-modulatory effects on T cells were completely abrogated by pharmacological interference of mitochondrial reactive oxygen species production. These data shed new light on dimethyl fumarate as bona fide immune-metabolic drug that targets the intracellular stress response in activated T cells, thereby restricting mitochondrial function and energetic capacity, providing novel insight into the role of oxidative stress in modulating cellular immune responses and T cell-mediated autoimmunity.
Collapse
Affiliation(s)
- Marie Liebmann
- Department of Neurology with Institute of Translational Neurology, University Hospital of Münster, Münster 48149, Germany
| | - Lisanne Korn
- Department of Neurology with Institute of Translational Neurology, University Hospital of Münster, Münster 48149, Germany
| | - Claudia Janoschka
- Department of Neurology with Institute of Translational Neurology, University Hospital of Münster, Münster 48149, Germany
| | - Stefanie Albrecht
- Institute of Neuropathology, University Hospital Münster, Münster 48149, Germany
| | - Sarah Lauks
- Department of Neurology with Institute of Translational Neurology, University Hospital of Münster, Münster 48149, Germany
| | - Alexander M Herrmann
- Department of Neurology, University Hospital Düsseldorf, Düsseldorf 40225, Germany
| | - Andreas Schulte-Mecklenbeck
- Department of Neurology with Institute of Translational Neurology, University Hospital of Münster, Münster 48149, Germany
| | - Nicholas Schwab
- Department of Neurology with Institute of Translational Neurology, University Hospital of Münster, Münster 48149, Germany
| | - Tilman Schneider-Hohendorf
- Department of Neurology with Institute of Translational Neurology, University Hospital of Münster, Münster 48149, Germany
| | - Maria Eveslage
- Institute of Biostatistics and Clinical Research, University of Münster, Münster 48149, Germany
| | - Brigitte Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg 69120, Germany
| | - Felix Luessi
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany
| | | | - Martin Diebold
- Laboratory of Clinical Neuroimmunology, Neurologic Clinic and Policlinic, Departments of Biomedicine and Clinical Research, University Hospital Basel, and University of Basel, Basel 4031, Switzerland
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany
| | - Catharina C Gross
- Department of Neurology with Institute of Translational Neurology, University Hospital of Münster, Münster 48149, Germany
| | - Stjepana Kovac
- Department of Neurology with Institute of Translational Neurology, University Hospital of Münster, Münster 48149, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany
| | - Tobias Derfuss
- Laboratory of Clinical Neuroimmunology, Neurologic Clinic and Policlinic, Departments of Biomedicine and Clinical Research, University Hospital Basel, and University of Basel, Basel 4031, Switzerland
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Münster 48149, Germany
| | - Simone König
- Core Unit Proteomics, Interdisciplinary Clinical Research Center, University of Münster, Münster 48149, Germany
| | - Sven G Meuth
- Department of Neurology, University Hospital Düsseldorf, Düsseldorf 40225, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital of Münster, Münster 48149, Germany
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital of Münster, Münster 48149, Germany
| |
Collapse
|
37
|
Lymphocyte Counts and Multiple Sclerosis Therapeutics: Between Mechanisms of Action and Treatment-Limiting Side Effects. Cells 2021; 10:cells10113177. [PMID: 34831400 PMCID: PMC8625745 DOI: 10.3390/cells10113177] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 01/18/2023] Open
Abstract
Although the detailed pathogenesis of multiple sclerosis (MS) is not completely understood, a broad range of disease-modifying therapies (DMTs) are available. A common side effect of nearly every MS therapeutic agent is lymphopenia, which can be both beneficial and, in some cases, treatment-limiting. A sound knowledge of the underlying mechanism of action of the selected agent is required in order to understand treatment-associated changes in white blood cell counts, as well as monitoring consequences. This review is a comprehensive summary of the currently available DMTs with regard to their effects on lymphocyte count. In the first part, we describe important general information about the role of lymphocytes in the course of MS and the essentials of lymphopenic states. In the second part, we introduce the different DMTs according to their underlying mechanism of action, summarizing recommendations for lymphocyte monitoring and definitions of lymphocyte thresholds for different therapeutic regimens.
Collapse
|
38
|
Barešić M, Reihl Crnogaj M, Zadro I, Anić B. Demyelinating disease (multiple sclerosis) in a patient with psoriatic arthritis treated with adalimumab: a case-based review. Rheumatol Int 2021; 41:2233-2239. [PMID: 34557936 DOI: 10.1007/s00296-021-04995-0] [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: 05/31/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
Over the past two decades, tumor necrosis factor-α (TNF-α) inhibitors became one of the most important drugs in the treatment of patients with psoriatic arthritis. Unfortunately, some of the patients exhibit unwanted side effects of the treatment. We describe a patient with psoriasis, psoriatic arthritis and uveitis who was treated with adalimumab and after 4 months of the treatment developed clinical and neuroradiological signs of demyelinating disease of the central nervous system. She experienced no signs and symptoms of neurological disease prior to adalimumab administration. After a detailed neurological work-up she was diagnosed with relapsing-remitting type of multiple sclerosis and treated with oral and pulse glucocorticoids and later with dimethyl fumarate. Adalimumab was discontinued. The question remains was the demyelination induced by the TNF-α blockade or was it unmasked by the introduction of the cytokine blocking agent. In patients suffering from inflammatory arthritis, treating disease to target as well as a close follow-up and knowledge of potential side effects of treatment remains crucial in good clinical practice.
Collapse
Affiliation(s)
- Marko Barešić
- Division of Clinical Immunology and Rheumatology, Department of Internal Medicine, School of Medicine, University Hospital Center Zagreb, University of Zagreb, Zagreb, Croatia.
| | - Mirna Reihl Crnogaj
- Division of Physical Medicine, Rehabilitation and Rheumatology, National Memorial Hospital Vukovar, Vukovar, Croatia
| | - Ivana Zadro
- Department of Neurology, School of Medicine, University Hospital Center Zagreb, University of Zagreb, Zagreb, Croatia
| | - Branimir Anić
- Division of Clinical Immunology and Rheumatology, Department of Internal Medicine, School of Medicine, University Hospital Center Zagreb, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
39
|
Baeva ME, Baev PB, Nelson J, Kazimirchik A, Vorobeychik G. A retrospective analysis of changes in lymphocyte levels in patients with multiple sclerosis during and after Tecfidera® treatment. Mult Scler J Exp Transl Clin 2021; 7:20552173211029674. [PMID: 34345437 PMCID: PMC8283074 DOI: 10.1177/20552173211029674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/14/2021] [Indexed: 11/16/2022] Open
Abstract
Background There are currently no best practice recommendations for lymphocyte subset monitoring for patients with multiple sclerosis (pwMS) on disease-modifying therapies including Tecfidera® (dimethyl fumarate, DMF). However, there have been several cases of pwMS on DMF without severe lymphopenia who had high CD4:CD8 T cell ratios and went on to develop progressive multifocal leukoencephalopathy. Objective Our objective was to characterize the changes in immune profile during and after DMF treatment in pwMS. Methods A retrospective analysis of longitudinal data from 299 pwMS who have been treated with DMF at the Fraser Health Multiple Sclerosis Clinic in British Columbia, Canada. The blood test results were taken from January 1, 2013 to April 1, 2020. Results Our results suggest that CD8+ T cells had the highest proportional decrease compared to other lymphocyte subset populations and overall lymphocyte count in response to DMF treatment. CD56+ Natural Killer cells were similarly decreased in response to DMF treatment. CD4:CD8 T cell ratio was the measurement that had the highest rate of change in response to DMF initiation and discontinuation. Conclusion CD8+ T cell count and CD4:CD8 T cell ratio may be a more sensitive measurement of the immune landscape of patients with MS on DMF.
Collapse
Affiliation(s)
- Maria-Elizabeth Baeva
- Fraser Health Multiple Sclerosis Clinic, Burnaby Hospital, Burnaby Hospital, Burnaby, British Columbia, Canada
| | - Philip Boris Baev
- Fraser Health Multiple Sclerosis Clinic, Burnaby Hospital, Burnaby Hospital, Burnaby, British Columbia, Canada
| | - Jill Nelson
- Fraser Health Multiple Sclerosis Clinic, Burnaby Hospital, Burnaby Hospital, Burnaby, British Columbia, Canada
| | - Anna Kazimirchik
- Fraser Health Multiple Sclerosis Clinic, Burnaby Hospital, Burnaby Hospital, Burnaby, British Columbia, Canada
| | - Galina Vorobeychik
- Fraser Health Multiple Sclerosis Clinic, Burnaby Hospital, Burnaby Hospital, Burnaby, British Columbia, Canada
| |
Collapse
|
40
|
Veroni C, Aloisi F. The CD8 T Cell-Epstein-Barr Virus-B Cell Trialogue: A Central Issue in Multiple Sclerosis Pathogenesis. Front Immunol 2021; 12:665718. [PMID: 34305896 PMCID: PMC8292956 DOI: 10.3389/fimmu.2021.665718] [Citation(s) in RCA: 39] [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] [Received: 02/08/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
The cause and the pathogenic mechanisms leading to multiple sclerosis (MS), a chronic inflammatory disease of the central nervous system (CNS), are still under scrutiny. During the last decade, awareness has increased that multiple genetic and environmental factors act in concert to modulate MS risk. Likewise, the landscape of cells of the adaptive immune system that are believed to play a role in MS immunopathogenesis has expanded by including not only CD4 T helper cells but also cytotoxic CD8 T cells and B cells. Once the key cellular players are identified, the main challenge is to define precisely how they act and interact to induce neuroinflammation and the neurodegenerative cascade in MS. CD8 T cells have been implicated in MS pathogenesis since the 80's when it was shown that CD8 T cells predominate in MS brain lesions. Interest in the role of CD8 T cells in MS was revived in 2000 and the years thereafter by studies showing that CNS-recruited CD8 T cells are clonally expanded and have a memory effector phenotype indicating in situ antigen-driven reactivation. The association of certain MHC class I alleles with MS genetic risk implicates CD8 T cells in disease pathogenesis. Moreover, experimental studies have highlighted the detrimental effects of CD8 T cell activation on neural cells. While the antigens responsible for T cell recruitment and activation in the CNS remain elusive, the high efficacy of B-cell depleting drugs in MS and a growing number of studies implicate B cells and Epstein-Barr virus (EBV), a B-lymphotropic herpesvirus that is strongly associated with MS, in the activation of pathogenic T cells. This article reviews the results of human studies that have contributed to elucidate the role of CD8 T cells in MS immunopathogenesis, and discusses them in light of current understanding of autoreactivity, B-cell and EBV involvement in MS, and mechanism of action of different MS treatments. Based on the available evidences, an immunopathological model of MS is proposed that entails a persistent EBV infection of CNS-infiltrating B cells as the target of a dysregulated cytotoxic CD8 T cell response causing CNS tissue damage.
Collapse
Affiliation(s)
| | - Francesca Aloisi
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
41
|
Anagnostouli MC, Velonakis G, Dalakas MC. Aggressive Herpes Zoster in Young Patients With Multiple Sclerosis Under Dimethyl Fumarate: Significance of CD8 + and Natural Killer Cells. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/4/e1017. [PMID: 34049996 PMCID: PMC8168045 DOI: 10.1212/nxi.0000000000001017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 03/22/2021] [Indexed: 11/15/2022]
Affiliation(s)
- Maria C Anagnostouli
- From the Multiple Sclerosis and Demyelinating Diseases Unit and Immunogenetics Laboratory (M.C.A.), 1st Department of Neurology, Medical School, National and Kapodistrian University of Athens, NKUA, Aeginition University Hospital, Athens, Greece; Research Unit of Radiology (G.V.), 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, Greece; Neuroimmunology Unit (M.C.D.), Department of Pathophysiology, National and Kapodistrian University of Athens, Greece; and Department of Neurology (M.C.D.), Thomas Jefferson University, Philadelphia, PA.
| | - Georgios Velonakis
- From the Multiple Sclerosis and Demyelinating Diseases Unit and Immunogenetics Laboratory (M.C.A.), 1st Department of Neurology, Medical School, National and Kapodistrian University of Athens, NKUA, Aeginition University Hospital, Athens, Greece; Research Unit of Radiology (G.V.), 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, Greece; Neuroimmunology Unit (M.C.D.), Department of Pathophysiology, National and Kapodistrian University of Athens, Greece; and Department of Neurology (M.C.D.), Thomas Jefferson University, Philadelphia, PA
| | - Marinos C Dalakas
- From the Multiple Sclerosis and Demyelinating Diseases Unit and Immunogenetics Laboratory (M.C.A.), 1st Department of Neurology, Medical School, National and Kapodistrian University of Athens, NKUA, Aeginition University Hospital, Athens, Greece; Research Unit of Radiology (G.V.), 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, Greece; Neuroimmunology Unit (M.C.D.), Department of Pathophysiology, National and Kapodistrian University of Athens, Greece; and Department of Neurology (M.C.D.), Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
42
|
Lee CH, Jiang B, Nakhaei-Nejad M, Barilla D, Blevins G, Giuliani F. Cross-sectional analysis of peripheral blood mononuclear cells in lymphopenic and non-lymphopenic relapsing-remitting multiple sclerosis patients treated with dimethyl fumarate. Mult Scler Relat Disord 2021; 52:103003. [PMID: 34118574 DOI: 10.1016/j.msard.2021.103003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/24/2021] [Accepted: 04/29/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Relapsing-remitting multiple sclerosis (RRMS) is an autoimmune disorder of the central nervous system. Dimethyl Fumarate is a disease-modifying medication used to treat RRMS patients that can induce lymphopenia. We aimed to immunophenotype peripheral blood mononuclear cells (PBMC) in RRMS patients cross-sectionally and examine the characteristics and modifications of lymphopenia over time. METHODS Characterization of PBMC was done by multiparametric flow cytometry. Patients had been on treatment for up to 4 years and were grouped into lymphopenic (DMF-L) and non-lymphopenic (DMF-N) patients. RESULTS Lymphopenia affected the cell population changes over time, with other patient characteristics (gender, age, and previous treatment status) also having significant effects. In both lymphopenic and non-lymphopenic patients, PBMC percentages were reduced over time. While overall T and B cells frequencies were not affected, males, older patients and untreated patients had significant changes in B cell subpopulations over time. CD4+ to CD8+T cell ratio increased significantly in lymphopenic patients over time. CD4-CD8-T cell population was similarly reduced in both lymphopenic and non-lymphopenic patients, over time. While the monocyte and NK overall populations were not changed, non-classical monocyte subpopulation decreased over time in lymphopenic patients. We also found CD56-CD16+ and CD56-CD16- NK cells frequencies changed over time in lymphopenic patients. Immune populations showed correlations with clinical outcomes measured by EDSS and relapse rate. Analysis of the overall immunophenotype showed that, while groups divided by other patient characteristics showed differences, the lymphopenia status overrode these differences, resulting in similar immunophenotype within DMF-L. CONCLUSIONS Our data provide evidence that under the same therapy, lymphopenia affects how the immunophenotype changes over time and can override the differences associated with other patient characteristics and possibly mask other significant changes in the immune profile of patients.
Collapse
Affiliation(s)
- Chieh-Hsin Lee
- Division of Neurology, Department of Medicine, University of Alberta, Alberta Canada T6G 2M8, Edmonton, Canada
| | - Bei Jiang
- Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, Canada
| | - Maryam Nakhaei-Nejad
- Division of Neurology, Department of Medicine, University of Alberta, Alberta Canada T6G 2M8, Edmonton, Canada
| | - David Barilla
- Division of Neurology, Department of Medicine, University of Alberta, Alberta Canada T6G 2M8, Edmonton, Canada
| | - Gregg Blevins
- Division of Neurology, Department of Medicine, University of Alberta, Alberta Canada T6G 2M8, Edmonton, Canada
| | - Fabrizio Giuliani
- Division of Neurology, Department of Medicine, University of Alberta, Alberta Canada T6G 2M8, Edmonton, Canada.
| |
Collapse
|
43
|
Morrison PJ, Suhrkamp I, Gerdes S, Mrowietz U. Oral dimethyl fumarate induces changes within the peripheral neutrophil compartment of patients with psoriasis that are linked with skin improvement. Br J Dermatol 2021; 185:605-615. [PMID: 33657656 DOI: 10.1111/bjd.19899] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Dimethyl fumarate (DMF) is a treatment for moderate-to-severe psoriasis and multiple sclerosis. DMF therapy typically improves skin inflammation within the first 3 months of treatment. DMF is a prodrug that generates the hydroxycarboxylic acid receptor 2 (HCA2) agonist, monomethyl fumarate (MMF). Despite widespread clinical use, DMF's mechanism of action is not fully understood. OBJECTIVES We wished to characterize the changes induced by DMF in peripheral neutrophils within the first 3 months of treatment to better understand its early antipsoriatic effects. METHODS Flow cytometry was used to assess T-cell and neutrophil frequencies, apoptosis and activation phenotype. In vitro culture of neutrophils with DMF and MMF was used to evaluate apoptosis and HCA2 internalization. Serum levels of neutrophil degranulation products were measured by enzyme-linked immunosorbent assay. RESULTS Patients with psoriasis had significantly higher leucocyte counts at baseline compared with controls, with a large population of pro-inflammatory CD62Llo CD11bbright neutrophils. Analysis revealed that DMF treatment reduced the frequency of CD62Llo CD11bbright neutrophils and serum levels of neutrophil activation markers. This reduction was not linked to increased apoptosis. CONCLUSIONS Our results reveal a novel in vivo effect of DMF therapy on pro-inflammatory neutrophils that likely contributes to this treatment's antipsoriatic efficacy.
Collapse
Affiliation(s)
- P J Morrison
- Psoriasis Center, Department of Dermatology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, 24105, Germany
| | - I Suhrkamp
- Psoriasis Center, Department of Dermatology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, 24105, Germany
| | - S Gerdes
- Psoriasis Center, Department of Dermatology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, 24105, Germany
| | - U Mrowietz
- Psoriasis Center, Department of Dermatology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, 24105, Germany
| |
Collapse
|
44
|
COVID-19 in multiple sclerosis patients treated with dimethyl fumarate. J Neurol 2021; 268:3132-3134. [PMID: 33611610 PMCID: PMC7896828 DOI: 10.1007/s00415-021-10446-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/04/2021] [Indexed: 12/29/2022]
|
45
|
Increased multiple sclerosis disease activity in patients transitioned from fingolimod to dimethyl fumarate: a case series. BMC Neurol 2021; 21:48. [PMID: 33530945 PMCID: PMC7851905 DOI: 10.1186/s12883-021-02058-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 01/13/2021] [Indexed: 01/07/2023] Open
Abstract
Background Fingolimod is a S1P1 receptor modulator that prevents activated lymphocyte egress from lymphoid tissues causing lymphopenia, mainly affecting CD4+ T lymphocytes. Withdrawal from fingolimod can be followed by severe disease reactivation, and this coincides with return of autoreactive lymphocytes into circulation. The CD8+ T cytotoxic population returns prior to the regulatory CD4+ T lymphocytes leading to a state of dysregulation, which may contribute to the rebound and severity of clinical relapses. On the other hand, dimethyl fumarate (DMF) preferentially reduces CD8+ T lymphocytes, has the same efficacy as fingolimod, and therefore, was expected to be a suitable oral alternative to reduce the rebound associated with fingolimod withdrawal. Case presentation We present six patients with relapsing-remitting MS who developed an unexpected increase in disease activity after transitioning from fingolimod to DMF. All patients were clinically and radiologically stable on fingolimod for at least 1 year. The switch in therapy was due to significantly low CD4+ T lymphocyte count ≤65 cells/ul (normal range 490–1740 cells/ul), after discussing the results with the patients and the potential risk for opportunistic infections including cryptococcal infections. DMF was introduced following a washout period of 5 to 11 weeks to allow reconstitution of the immune system and for the absolute lymphocyte count to reach ≥500 cells/ul. Every patient who experienced a relapse had several enhancing lesions in the brain and/or spinal cord between 12 to 23 weeks after cessation of fingolimod and 1 to 18 weeks after starting DMF. All relapses were treated with intravenous methylprednisolone with good clinical responses. Conclusion The anticipated beneficial response of DMF treatment to mitigate rebound after fingolimod therapy cessation was not observed. Our patients experienced rebound disease despite being on treatment with DMF. Additional studies are necessary to understand which treatments are most effective to transition to after discontinuing fingolimod.
Collapse
|
46
|
Lower lymphocyte counts and older age are associated with reduced multiple sclerosis disease activity during dimethyl fumarate treatment. Mult Scler Relat Disord 2021; 49:102781. [PMID: 33524927 DOI: 10.1016/j.msard.2021.102781] [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: 09/27/2020] [Revised: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND delayed-release dimethyl fumarate (DMF) is a disease modifying therapy for relapsing-remitting multiple sclerosis (MS) with antioxidant and anti-inflammatory properties. The drug causes lymphocyte count reduction, which can lead to lymphopenia development during treatment. This is an important safety issue, due to infectious risk, mainly progressive multifocal leukoencephalopathy (PML). If the lymphocyte count influences the response to treatment is still a matter of debate, as there are contrasting contrasting data in the literature. Considering this, we aimed to identify DMF induced lymphopenia risk factors and to evaluate lymphopenia impact on MS disease activity in a real world setting. METHODS a retrospective study on 135 MS patients receiving DMF with a mean treatment duration of 32.3±15.9 months was performed. Baseline and follow-up demographic, clinical, magnetic resonance imaging (MRI) and laboratory data were collected. RESULTS 44 patients (32.6%) developed lymphopenia, with 11 (8.1%) grade 1, 23 (17.0%) grade 2 and 10 (7.4 %) grade 3. Older age and lower basal absolute lymphocyte count were found to be associated with lymphopenia development on a binary regression model (p<0.001 and p=0.009). When compared with non lymphopenic+lymphopenia grade 1 patients, those experiencing lymphopenia grade 2+3 had longer disease activity free survival (p<0.001), fewer clinical relapses (p=0.005) and lower MRI disease activity (p≤0.001). On Cox regression model, older age and lymphopenia grade 2+3 were found to be protective factors against disease activity (HR=0.966; 95% C.I.=0.942-0.992; p=0.009 for age; HR=0.137; 95% C.I.=0.043-0.439; p=0.001 for lymphopenia grade 2+3) and MRI disease activity (HR=0.968; 95% C.I.=0.941-0.997; p=0.030 for age; HR=0.142; 95% C.I.=0.034-0.591; p=0.007 for lymphopenia grade 2+3). Only lymphopenia grade 2+3 was found to be a predictor of clinical relapses (HR=0.970; 95% C.I.=0.936-1.005; p=0.095 for age; HR=0.115; 95% C.I.=0.016-0.854; p=0.034 for lymphopenia grade 2+3), with a protective effect. CONCLUSION older age and lower basal lymphocyte count were found to be associated with lymphopenia development. Lymphopenia grade 2+3 and older age could be protective against clinical and radiologic disease activity during DMF treatment.
Collapse
|
47
|
Wipke BT, Hoepner R, Strassburger-Krogias K, Thomas AM, Gianni D, Szak S, Brennan MS, Pistor M, Gold R, Chan A, Scannevin RH. Different Fumaric Acid Esters Elicit Distinct Pharmacologic Responses. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/2/e950. [PMID: 33468560 PMCID: PMC7862084 DOI: 10.1212/nxi.0000000000000950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/25/2020] [Indexed: 01/29/2023]
Abstract
Objective To test the hypothesis that dimethyl fumarate (DMF, Tecfidera) elicits different biological changes from DMF combined with monoethyl fumarate (MEF) (Fumaderm, a psoriasis therapy), we investigated DMF and MEF in rodents and cynomolgus monkeys. Possible translatability of findings was explored with lymphocyte counts from a retrospective cohort of patients with MS. Methods In rodents, we evaluated pharmacokinetic and pharmacodynamic effects induced by DMF and MEF monotherapies or in combination (DMF/MEF). Clinical implications were investigated in a retrospective, observational analysis of patients with MS treated with DMF/MEF (n = 36). Results In rodents and cynomolgus monkeys, monomethyl fumarate (MMF, the primary metabolite of DMF) exhibited higher brain penetration, whereas MEF was preferentially partitioned into the kidney. In mice, transcriptional profiling for DMF and MEF alone identified both common and distinct pharmacodynamic responses, with almost no overlap between DMF- and MEF-induced differentially expressed gene profiles in immune tissues. The nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-mediated oxidative stress response pathway was exclusively regulated by DMF, whereas apoptosis pathways were activated by MEF. DMF/MEF treatment demonstrated that DMF and MEF functionally interact to modify DMF- and MEF-specific responses in unpredictable ways. In patients with MS, DMF/MEF treatment led to early and pronounced suppression of lymphocytes, predominantly CD8+ T cells. In a multivariate regression analysis, the absolute lymphocyte count (ALC) was associated with age at therapy start, baseline ALC, and DMF/MEF dosage but not with previous immunosuppressive medication and sex. Furthermore, the ALC increased in a small cohort of patients with MS (n = 6/7) after switching from DMF/MEF to DMF monotherapy. Conclusions Fumaric acid esters exhibit different biodistribution and may elicit different biological responses; furthermore, pharmacodynamic effects of combinations differ unpredictably from monotherapy. The strong potential to induce lymphopenia in patients with MS may be a result of activation of apoptosis pathways by MEF compared with DMF.
Collapse
Affiliation(s)
- Brian T Wipke
- From Biogen, Inc (B.T.W., A.M.T., D.G., S.S., M.S.B., R.H.S.), Cambridge, MA; Department of Neurology (R.H., M.P., A.C.), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Neurology (K.S.-K., R.G.), St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Robert Hoepner
- From Biogen, Inc (B.T.W., A.M.T., D.G., S.S., M.S.B., R.H.S.), Cambridge, MA; Department of Neurology (R.H., M.P., A.C.), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Neurology (K.S.-K., R.G.), St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Katrin Strassburger-Krogias
- From Biogen, Inc (B.T.W., A.M.T., D.G., S.S., M.S.B., R.H.S.), Cambridge, MA; Department of Neurology (R.H., M.P., A.C.), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Neurology (K.S.-K., R.G.), St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Ankur M Thomas
- From Biogen, Inc (B.T.W., A.M.T., D.G., S.S., M.S.B., R.H.S.), Cambridge, MA; Department of Neurology (R.H., M.P., A.C.), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Neurology (K.S.-K., R.G.), St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Davide Gianni
- From Biogen, Inc (B.T.W., A.M.T., D.G., S.S., M.S.B., R.H.S.), Cambridge, MA; Department of Neurology (R.H., M.P., A.C.), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Neurology (K.S.-K., R.G.), St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Suzanne Szak
- From Biogen, Inc (B.T.W., A.M.T., D.G., S.S., M.S.B., R.H.S.), Cambridge, MA; Department of Neurology (R.H., M.P., A.C.), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Neurology (K.S.-K., R.G.), St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Melanie S Brennan
- From Biogen, Inc (B.T.W., A.M.T., D.G., S.S., M.S.B., R.H.S.), Cambridge, MA; Department of Neurology (R.H., M.P., A.C.), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Neurology (K.S.-K., R.G.), St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Maximilian Pistor
- From Biogen, Inc (B.T.W., A.M.T., D.G., S.S., M.S.B., R.H.S.), Cambridge, MA; Department of Neurology (R.H., M.P., A.C.), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Neurology (K.S.-K., R.G.), St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Ralf Gold
- From Biogen, Inc (B.T.W., A.M.T., D.G., S.S., M.S.B., R.H.S.), Cambridge, MA; Department of Neurology (R.H., M.P., A.C.), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Neurology (K.S.-K., R.G.), St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Andrew Chan
- From Biogen, Inc (B.T.W., A.M.T., D.G., S.S., M.S.B., R.H.S.), Cambridge, MA; Department of Neurology (R.H., M.P., A.C.), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Neurology (K.S.-K., R.G.), St. Josef Hospital, Ruhr University Bochum, Bochum, Germany.
| | - Robert H Scannevin
- From Biogen, Inc (B.T.W., A.M.T., D.G., S.S., M.S.B., R.H.S.), Cambridge, MA; Department of Neurology (R.H., M.P., A.C.), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Neurology (K.S.-K., R.G.), St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
48
|
Timpani CA, Rybalka E. Calming the (Cytokine) Storm: Dimethyl Fumarate as a Therapeutic Candidate for COVID-19. Pharmaceuticals (Basel) 2020; 14:15. [PMID: 33375288 PMCID: PMC7824470 DOI: 10.3390/ph14010015] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022] Open
Abstract
COVID-19 has rapidly spread worldwide and incidences of hospitalisation from respiratory distress are significant. While a vaccine is in the pipeline, there is urgency for therapeutic options to address the immune dysregulation, hyperinflammation and oxidative stress that can lead to death. Given the shared pathogenesis of severe cases of COVID-19 with aspects of multiple sclerosis and psoriasis, we propose dimethyl fumarate as a viable treatment option. Currently approved for multiple sclerosis and psoriasis, dimethyl fumarate is an immunomodulatory, anti-inflammatory and anti-oxidative drug that could be rapidly implemented into the clinic to calm the cytokine storm which drives severe COVID-19.
Collapse
Affiliation(s)
- Cara A. Timpani
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia;
- Australian Institute for Musculoskeletal Science, St Albans, VIC 3021, Australia
| | - Emma Rybalka
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia;
- Australian Institute for Musculoskeletal Science, St Albans, VIC 3021, Australia
| |
Collapse
|
49
|
Abstract
Background: Only progressive multifocal leukoencephalopathy (PML) is currently described in the dimethyl fumarate (DMF) prescribing information. Objectives: To describe opportunistic infections (OIs), other than PML, reported in association with DMF. Methods: The FDA Adverse Event Reporting System (FAERS) and medical literature were searched. Results: We retrieved 34 cases of serious OIs with a causal association with DMF, including 11 central nervous system (CNS) infections and 23 extra-CNS infections. Six OIs occurred with normal circulating absolute lymphocyte counts. The median latency from DMF initiation was 13 months and was variable. Conclusion: DMF is associated with the development of OIs that require invasive diagnostic and/or therapeutic procedures. Patients should be monitored for OIs when treated with DMF regardless of circulating absolute lymphocyte counts.
Collapse
|
50
|
Moise N, Friedman A. A mathematical model of the multiple sclerosis plaque. J Theor Biol 2020; 512:110532. [PMID: 33152395 DOI: 10.1016/j.jtbi.2020.110532] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/15/2022]
Abstract
Multiple sclerosis is an autoimmune disease that affects white matter in the central nervous system. It is one of the primary causes of neurological disability among young people. Its characteristic pathological lesion is called a plaque, a zone of inflammatory activity and tissue destruction that expands radially outward by destroying the myelin and oligodendrocytes of white matter. The present paper develops a mathematical model of the multiple sclerosis plaques. Although these plaques do not provide reliable information of the clinical disability in MS, they are nevertheless useful as a primary outcome measure of Phase II trials. The model consists of a system of partial differential equations in a simplified geometry of the lesion, consisting of three domains: perivascular space, demyelinated plaque, and white matter. The model describes the activity of various pro- and anti-inflammatory cells and cytokines in the plaque, and quantifies their effect on plaque growth. We show that volume growth of plaques are in qualitative agreement with reported clinical studies of several currently used drugs. We then use the model to explore treatments with combinations of such drugs, and with experimental drugs. We finally consider the benefits of early vs. delayed treatment.
Collapse
Affiliation(s)
- Nicolae Moise
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Biomedical Engineering, Ohio State University, Columbus, OH, USA
| | - Avner Friedman
- Mathematical Biosciences Institute & Department of Mathematics, Ohio State University, Columbus, OH, USA.
| |
Collapse
|