1
|
Aharoni R, Milo R, Arnon R. Glatiramer Acetate for the Treatment of Multiple Sclerosis: From First-Generation Therapy to Elucidation of Immunomodulation and Repair. Pharmacol Rev 2024; 76:1133-1158. [PMID: 39406508 DOI: 10.1124/pharmrev.124.000927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 10/18/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS), with a putative autoimmune origin and complex pathogenesis. Modification of the natural history of MS by reducing relapses and slowing disability accumulation was first attained in the 1990 s with the development of the first-generation disease-modifying therapies. Glatiramer acetate (GA), a copolymer of L-alanine, L-lysine, L-glutamic acid, and L-tyrosine, was discovered due to its ability to suppress the animal model of MS, experimental autoimmune encephalomyelitis. Extensive clinical trials and long-term assessments established the efficacy and the safety of GA. Furthermore, studies of the therapeutic processes induced by GA in animal models and in MS patients indicate that GA affects various levels of the innate and the adaptive immune response, generating deviation from proinflammatory to anti-inflammatory pathways. This includes competition for binding to antigen presenting cells; driving dendritic cells, monocytes, and B-cells toward anti-inflammatory responses; and stimulating T-helper 2 and T-regulatory cells. The immune cells stimulated by GA reach the CNS and secrete in situ anti-inflammatory cytokines alleviating the pathological processes. Furthermore, cumulative findings reveal that in addition to its immunomodulatory effect, GA promotes neuroprotective repair processes such as neurotrophic factors secretion, remyelination, and neurogenesis. This review aims to provide an overview of MS pathology diagnosis and treatment as well as the diverse mechanism of action of GA. SIGNIFICANCE STATEMENT: Understanding the complex MS immune pathogenesis provided multiple targets for therapeutic intervention, resulting in a plethora of agents, with various mechanisms of action, efficacy, and safety profiles. However, promoting repair beyond the body's limited spontaneous extent is still a major challenge. GA, one of the first approved disease-modifying therapies, induces diverse immunomodulatory effects. Furthermore, GA treatment results in elevated neurotrophic factors secretion, remyelination and neurogenesis, supporting the notion that immunomodulatory treatment can support in situ a growth-promoting and repair environment.
Collapse
Affiliation(s)
- Rina Aharoni
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel (Ri.A., Ru.A.); and Department of Neurology, Barzilai Medical Center, Ashkelon, Israel, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel (R.M.)
| | - Ron Milo
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel (Ri.A., Ru.A.); and Department of Neurology, Barzilai Medical Center, Ashkelon, Israel, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel (R.M.)
| | - Ruth Arnon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel (Ri.A., Ru.A.); and Department of Neurology, Barzilai Medical Center, Ashkelon, Israel, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel (R.M.)
| |
Collapse
|
2
|
Aviel G, Elkahal J, Umansky KB, Bueno-Levy H, Petrover Z, Kotlovski Y, Lendengolts D, Kain D, Shalit T, Zhang L, Miyara S, Kramer MP, Merbl Y, Kozlovski S, Alon R, Aharoni R, Arnon R, Mishali D, Katz U, Nachman D, Asleh R, Amir O, Tzahor E, Sarig R. Repurposing of glatiramer acetate to treat cardiac ischemia in rodent models. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1049-1066. [PMID: 39215106 DOI: 10.1038/s44161-024-00524-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 07/10/2024] [Indexed: 09/04/2024]
Abstract
Myocardial injury may ultimately lead to adverse ventricular remodeling and development of heart failure (HF), which is a major cause of morbidity and mortality worldwide. Given the slow pace and substantial costs of developing new therapeutics, drug repurposing is an attractive alternative. Studies of many organs, including the heart, highlight the importance of the immune system in modulating injury and repair outcomes. Glatiramer acetate (GA) is an immunomodulatory drug prescribed for patients with multiple sclerosis. Here, we report that short-term GA treatment improves cardiac function and reduces scar area in a mouse model of acute myocardial infarction and a rat model of ischemic HF. We provide mechanistic evidence indicating that, in addition to its immunomodulatory functions, GA exerts beneficial pleiotropic effects, including cardiomyocyte protection and enhanced angiogenesis. Overall, these findings highlight the potential repurposing of GA as a future therapy for a myriad of heart diseases.
Collapse
Affiliation(s)
- Gal Aviel
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jacob Elkahal
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Kfir Baruch Umansky
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hanna Bueno-Levy
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Zachary Petrover
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yulia Kotlovski
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Daria Lendengolts
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - David Kain
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Shalit
- Bioinformatics Unit, G-INCPM, Weizmann Institute of Science, Rehovot, Israel
| | - Lingling Zhang
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Shoval Miyara
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Matthias P Kramer
- The Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yifat Merbl
- The Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Stav Kozlovski
- The Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ronen Alon
- The Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Rina Aharoni
- The Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ruth Arnon
- The Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - David Mishali
- Pediatric Heart Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Uriel Katz
- Pediatric Heart Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dean Nachman
- Heart Institute, Hadassah Medical Center and Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Rabea Asleh
- Heart Institute, Hadassah Medical Center and Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Offer Amir
- Heart Institute, Hadassah Medical Center and Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Eldad Tzahor
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Rachel Sarig
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
3
|
Nouh RA, Kamal A, Oyewole O, Abbas WA, Abib B, Omar A, Mansour ST, Abdelnaser A. Unveiling the Potential of Cannabinoids in Multiple Sclerosis and the Dawn of Nano-Cannabinoid Medicine. Pharmaceutics 2024; 16:241. [PMID: 38399295 PMCID: PMC10891830 DOI: 10.3390/pharmaceutics16020241] [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: 10/09/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 02/25/2024] Open
Abstract
Multiple sclerosis is the predominant autoimmune disorder affecting the central nervous system in adolescents and adults. Specific treatments are categorized as disease-modifying, whereas others are symptomatic treatments to alleviate painful symptoms. Currently, no singular conventional therapy is universally effective for all patients across all stages of the illness. Nevertheless, cannabinoids exhibit significant promise in their capacity for neuroprotection, anti-inflammation, and immunosuppression. This review will examine the traditional treatment for multiple sclerosis, the increasing interest in using cannabis as a treatment method, its role in protecting the nervous system and regulating the immune system, commercially available therapeutic cannabinoids, and the emerging use of cannabis in nanomedicine. In conclusion, cannabinoids exhibit potential as a disease-modifying treatment rather than merely symptomatic relief. However, further research is necessary to unveil their role and establish the safety and advancements in nano-cannabinoid medicine, offering the potential for reduced toxicity and fewer adverse effects, thereby maximizing the benefits of cannabinoids.
Collapse
Affiliation(s)
- Roua A. Nouh
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo, P.O. Box 74, New Cairo 11835, Egypt; (R.A.N.); (O.O.); (W.A.A.); (A.O.)
| | - Ahmed Kamal
- Biochemistry Department, Faculty of Science, Suez University, P.O. Box 43221, Suez 43533, Egypt;
| | - Oluwaseyi Oyewole
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo, P.O. Box 74, New Cairo 11835, Egypt; (R.A.N.); (O.O.); (W.A.A.); (A.O.)
| | - Walaa A. Abbas
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo, P.O. Box 74, New Cairo 11835, Egypt; (R.A.N.); (O.O.); (W.A.A.); (A.O.)
| | - Bishoy Abib
- Department of Chemistry, School of Sciences and Engineering, The American University in Cairo, P.O. Box 74, New Cairo 11835, Egypt; (B.A.); (S.T.M.)
| | - Abdelrouf Omar
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo, P.O. Box 74, New Cairo 11835, Egypt; (R.A.N.); (O.O.); (W.A.A.); (A.O.)
| | - Somaia T. Mansour
- Department of Chemistry, School of Sciences and Engineering, The American University in Cairo, P.O. Box 74, New Cairo 11835, Egypt; (B.A.); (S.T.M.)
| | - Anwar Abdelnaser
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box 74, New Cairo 11835, Egypt
| |
Collapse
|
4
|
Gouttefangeas C, Klein R, Maia A. The good and the bad of T cell cross-reactivity: challenges and opportunities for novel therapeutics in autoimmunity and cancer. Front Immunol 2023; 14:1212546. [PMID: 37409132 PMCID: PMC10319254 DOI: 10.3389/fimmu.2023.1212546] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/24/2023] [Indexed: 07/07/2023] Open
Abstract
T cells are main actors of the immune system with an essential role in protection against pathogens and cancer. The molecular key event involved in this absolutely central task is the interaction of membrane-bound specific T cell receptors with peptide-MHC complexes which initiates T cell priming, activation and recall, and thus controls a range of downstream functions. While textbooks teach us that the repertoire of mature T cells is highly diverse, it is clear that this diversity cannot possibly cover all potential foreign peptides that might be encountered during life. TCR cross-reactivity, i.e. the ability of a single TCR to recognise different peptides, offers the best solution to this biological challenge. Reports have shown that indeed, TCR cross-reactivity is surprisingly high. Hence, the T cell dilemma is the following: be as specific as possible to target foreign danger and spare self, while being able to react to a large spectrum of body-threatening situations. This has major consequences for both autoimmune diseases and cancer, and significant implications for the development of T cell-based therapies. In this review, we will present essential experimental evidence of T cell cross-reactivity, implications for two opposite immune conditions, i.e. autoimmunity vs cancer, and how this can be differently exploited for immunotherapy approaches. Finally, we will discuss the tools available for predicting cross-reactivity and how improvements in this field might boost translational approaches.
Collapse
Affiliation(s)
- Cécile Gouttefangeas
- Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany
| | - Reinhild Klein
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Ana Maia
- Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| |
Collapse
|
5
|
AbdelRazek MA, Tummala S, Khalid F, Tauhid S, Jalkh Y, Khalil S, Hurwitz S, Zurawski J, Bakshi R. Exploring the effect of glatiramer acetate on cerebral gray matter atrophy in multiple sclerosis. J Neurol Sci 2023; 444:120501. [PMID: 36481574 DOI: 10.1016/j.jns.2022.120501] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND PURPOSE Cerebral gray matter (GM) atrophy is a proposed measure of neuroprotection in multiple sclerosis (MS). Glatiramer acetate (GA) limits clinical relapses, MRI lesions, and whole brain atrophy in relapsing-remitting MS (RRMS). The effect of GA on GM atrophy remains unclear. We assessed GM atrophy in patients with RRMS starting GA therapy in comparison to a cohort of patients with clinically benign RRMS (BMS). DESIGN/METHODS We studied 14 patients at GA start [age (mean ± SD) 44.2 ± 7.0 years, disease duration (DD) 7.2 ± 6.4 years, Expanded Disability Status Scale score (EDSS) (median,IQR) 1.0,2.0] and 6 patients with BMS [age 43.0 ± 6.1 years, DD 18.1 ± 8.4 years, EDSS 0.5,1.0]. Brain MRI was obtained at baseline and one year later (both groups) and two years later in all patients in the GA group except one who was lost to follow-up. Semi-automated algorithms assessed cerebral T2 hyperintense lesion volume (T2LV), white matter fraction (WMF), GM fraction (GMF), and brain parenchymal fraction (BPF). The exact Wilcoxon-Mann-Whitney test compared the groups. The Wilcoxon signed rank test assessed longitudinal changes within groups. RESULTS During the first year, MRI changes did not differ significantly between groups (p > 0.15). Within the BMS group, WMF and BPF decreased during the first year (p = 0.03). Within the GA group, there was no significant change in MRI measures during each annual period (p > 0.05). Over two years, the GA group had a significant increase in T2LV and decrease in WMF (p < 0.05), while GMF and BPF remained stable (p > 0.05). MRI changes in brain volumes (GMF or WMF) in the first year in the GA group were not significantly different from those in the BMS group (p > 0.5). CONCLUSIONS In this pilot study with a small sample size, patients with RRMS started on GA did not show significant GM or whole brain atrophy over 2 years, resembling MS patients with a clinically benign disease course.
Collapse
Affiliation(s)
| | - Subhash Tummala
- Departments of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Fariha Khalid
- Departments of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shahamat Tauhid
- Departments of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Youmna Jalkh
- Departments of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Samar Khalil
- Departments of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shelley Hurwitz
- Departments of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jonathan Zurawski
- Departments of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rohit Bakshi
- Departments of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Departments of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Liu C, Zhu J, Mi Y, Jin T. Impact of disease-modifying therapy on dendritic cells and exploring their immunotherapeutic potential in multiple sclerosis. J Neuroinflammation 2022; 19:298. [PMID: 36510261 PMCID: PMC9743681 DOI: 10.1186/s12974-022-02663-z] [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: 06/28/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022] Open
Abstract
Dendritic cells (DCs) are the most potent professional antigen-presenting cells (APCs), which play a pivotal role in inducing either inflammatory or tolerogenic response based on their subtypes and environmental signals. Emerging evidence indicates that DCs are critical for initiation and progression of autoimmune diseases, including multiple sclerosis (MS). Current disease-modifying therapies (DMT) for MS can significantly affect DCs' functions. However, the study on the impact of DMT on DCs is rare, unlike T and B lymphocytes that are the most commonly discussed targets of these therapies. Induction of tolerogenic DCs (tolDCs) with powerful therapeutic potential has been well-established to combat autoimmune responses in laboratory models and early clinical trials. In contrast to in vitro tolDC induction, in vivo elicitation by specifically targeting multiple cell-surface receptors has shown greater promise with more advantages. Here, we summarize the role of DCs in governing immune tolerance and in the process of initiating and perpetuating MS as well as the effects of current DMT drugs on DCs. We then highlight the most promising cell-surface receptors expressed on DCs currently being explored as the viable pharmacological targets through antigen delivery to generate tolDCs in vivo.
Collapse
Affiliation(s)
- Caiyun Liu
- grid.430605.40000 0004 1758 4110Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- grid.430605.40000 0004 1758 4110Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China ,grid.24381.3c0000 0000 9241 5705Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrcs, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Yan Mi
- grid.430605.40000 0004 1758 4110Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Tao Jin
- grid.430605.40000 0004 1758 4110Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
7
|
Kunkl M, Amormino C, Tedeschi V, Fiorillo MT, Tuosto L. Astrocytes and Inflammatory T Helper Cells: A Dangerous Liaison in Multiple Sclerosis. Front Immunol 2022; 13:824411. [PMID: 35211120 PMCID: PMC8860818 DOI: 10.3389/fimmu.2022.824411] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/13/2022] [Indexed: 11/15/2022] Open
Abstract
Multiple Sclerosis (MS) is a neurodegenerative autoimmune disorder of the central nervous system (CNS) characterized by the recruitment of self-reactive T lymphocytes, mainly inflammatory T helper (Th) cell subsets. Once recruited within the CNS, inflammatory Th cells produce several inflammatory cytokines and chemokines that activate resident glial cells, thus contributing to the breakdown of blood-brain barrier (BBB), demyelination and axonal loss. Astrocytes are recognized as key players of MS immunopathology, which respond to Th cell-defining cytokines by acquiring a reactive phenotype that amplify neuroinflammation into the CNS and contribute to MS progression. In this review, we summarize current knowledge of the astrocytic changes and behaviour in both MS and experimental autoimmune encephalomyelitis (EAE), and the contribution of pathogenic Th1, Th17 and Th1-like Th17 cell subsets, and CD8+ T cells to the morphological and functional modifications occurring in astrocytes and their pathological outcomes.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Carola Amormino
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Valentina Tedeschi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Maria Teresa Fiorillo
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| |
Collapse
|
8
|
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: 14] [Impact Index Per Article: 4.7] [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
|
9
|
Influence of immunomodulatory drugs on the gut microbiota. Transl Res 2021; 233:144-161. [PMID: 33515779 PMCID: PMC8184576 DOI: 10.1016/j.trsl.2021.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/10/2020] [Accepted: 01/26/2021] [Indexed: 12/17/2022]
Abstract
Immunomodulatory medications are a mainstay of treatment for autoimmune diseases and malignancies. In addition to their direct effects on immune cells, these medications also impact the gut microbiota. Drug-induced shifts in commensal microbes can lead to indirect but important changes in the immune response. We performed a comprehensive literature search focusing on immunotherapy/microbe interactions. Immunotherapies were categorized into 5 subtypes based on their mechanisms of action: cell trafficking inhibitors, immune checkpoint inhibitors, immunomodulators, antiproliferative drugs, and inflammatory cytokine inhibitors. Although no consistent relationships were observed between types of immunotherapy and microbiota, most immunotherapies were associated with shifts in specific colonizing bacterial taxa. The relationships between colonizing microbes and drug efficacy were not well-studied for autoimmune diseases. In contrast, the efficacy of immune checkpoint inhibitors for cancer was tied to the baseline composition of the gut microbiota. There was a paucity of high-quality data; existing data were generated using heterogeneous sampling and analytic techniques, and most studies involved small numbers of participants. Further work is needed to elucidate the extent and clinical significance of immunotherapy effects on the human microbiome.
Collapse
|
10
|
Neuhaus O, Köhler W, Then Bergh F, Kristoferitsch W, Faiss J, Rosenkranz T, Reske D, Patejdl R, Hartung HP, Zettl UK. Glatiramer Acetate Treatment in Multiple Sclerosis-Associated Fatigue-Beneficial Effects on Self-Assessment Scales But Not on Molecular Markers. Biomolecules 2021; 11:biom11030393. [PMID: 33800033 PMCID: PMC8002075 DOI: 10.3390/biom11030393] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 02/28/2021] [Accepted: 03/03/2021] [Indexed: 01/14/2023] Open
Abstract
Although fatigue is a common symptom in multiple sclerosis (MS), its pathomechanisms are incompletely understood. Glatiramer acetate (GA), an immunomodulatory agent approved for treatment of relapsing-remitting MS (RRMS), possesses unique mechanisms of action and has been shown to exhibit beneficial effects on MS fatigue. The objective of this study was to correlate clinical, neuropsychological, and immunological parameters in RRMS patients with fatigue before and during treatment with GA. In a prospective, open-label, multicenter trial, 30 patients with RRMS and fatigue were treated with GA for 12 months. Inclusion criterion was the presence of fatigue as one of the most frequent and disabling symptoms. Before and during treatment, fatigue was assessed using the Fatigue Severity Scale (FSS), the MS-FSS, and the Modified Fatigue Impact Scale (MFIS). In addition, fatigue and quality of life were assessed using the Visual Analog Scales (VAS). Laboratory assessments included screening of 188 parameters using real-time PCR microarrays followed by further analysis of several cytokines, chemokines, and neurotrophic factors. Fatigue self-assessments were completed in 25 patients. After 12 months of treatment with GA, 13 of these patients improved in all three scales (with the most prominent effects on the MFIS), whereas 5 patients had deteriorated. The remaining 7 patients exhibited inconsistent effects within the three scales. Fatigue and overall quality of life had improved, as assessed via VAS. Laboratory assessments revealed heterogeneous mRNA levels of cytokines, chemokines, and neurotrophic factors. In conclusion, we were not able to correlate clinical and molecular effects of GA in patients with RRMS and fatigue.
Collapse
Affiliation(s)
- Oliver Neuhaus
- Department of Neurology, Klinikum der Heinrich Heine Universität, 40225 Düsseldorf, Germany;
- Department of Neurology, SRH Kliniken Landkreis Sigmaringen GmbH, 72488 Sigmaringen, Germany
- Correspondence: ; Tel.: +49-7571-1002483
| | - Wolfgang Köhler
- Department of Neurology, Fachkrankenhaus Hubertusburg, 04779 Wermsdorf, Germany;
- Department of Neurology, Universität Leipzig, 04103 Leipzig, Germany;
| | | | - Wolfgang Kristoferitsch
- Department of Neurology, SMZ-Ost-Donauspital, 1220 Vienna, Austria;
- Karl Landsteiner Institute for Neuroimmunological and Neurodegenerative Disorders, 1220 Vienna, Austria
| | - Jürgen Faiss
- Department of Neurology, Asklepios Fachklinikum Teupitz, 15755 Teupitz, Germany;
- German Stroke Society, 10117 Berlin, Germany
| | - Thorsten Rosenkranz
- Department of Neurology, Asklepios Klinik St. Georg, 20099 Hamburg, Germany;
| | - Dirk Reske
- Department of Neurology, Klinikum der Universität zu Köln, 50937 Cologne, Germany;
- Department of Psychiatry, LVR-Klinik Köln, 51109 Cologne, Germany
| | - Robert Patejdl
- Department of Physiology, Universitätsmedizin Rostock, 18057 Rostock, Germany;
| | - Hans-Peter Hartung
- Department of Neurology, Klinikum der Heinrich Heine Universität, 40225 Düsseldorf, Germany;
| | - Uwe K. Zettl
- Department of Neurology, Universitätsmedizin Rostock, 18057 Rostock, Germany;
| |
Collapse
|
11
|
Song JY, Griffin JD, Larson NR, Christopher MA, Middaugh CR, Berkland CJ. Synthetic Cationic Autoantigen Mimics Glatiramer Acetate Persistence at the Site of Injection and Is Efficacious Against Experimental Autoimmune Encephalomyelitis. Front Immunol 2021; 11:603029. [PMID: 33537031 PMCID: PMC7848024 DOI: 10.3389/fimmu.2020.603029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022] Open
Abstract
A synthetic peptide, K-PLP, consisting of 11-unit poly-lysine (K11) linked via polyethylene glycol (PEG) to proteolipid protein epitope (PLP) was synthesized, characterized, and evaluated for efficacy in ameliorating experimental autoimmune encephalomyelitis (EAE) induced by PLP. K-PLP was designed to mimic the cationic nature of the relapsing-remitting multiple sclerosis treatment, glatiramer acetate (GA). With a pI of ~10, GA is able to form visible aggregates at the site of injection via electrostatic interactions with the anionic extracellular matrix. Aggregation further facilitates the retention of GA at the site of injection and draining lymph nodes, which may contribute to its mechanism of action. K-PLP with a pI of ~11, was found to form visible aggregates in the presence of glycosaminoglycans and persist at the injection site and draining lymph nodes in vivo, similar to GA. Additionally, EAE mice treated with K-PLP showed significant inhibition of clinical symptoms compared to free poly-lysine and to PLP, which are the components of K-PLP. The ability of the poly-lysine motif to retain PLP at the injection site, which increased the local exposure of PLP to immune cells may be an important factor affecting drug efficacy.
Collapse
Affiliation(s)
- Jimmy Y Song
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - J Daniel Griffin
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States.,Department of Bioengineering, University of Kansas, Lawrence, KS, United States
| | - Nicholas R Larson
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Matthew A Christopher
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - C Russell Middaugh
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Cory J Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States.,Department of Bioengineering, University of Kansas, Lawrence, KS, United States.,Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
12
|
Melnikov M, Sharanova S, Sviridova A, Rogovskii V, Murugina N, Nikolaeva A, Dagil Y, Murugin V, Ospelnikova T, Boyko A, Pashenkov M. The influence of glatiramer acetate on Th17-immune response in multiple sclerosis. PLoS One 2020; 15:e0240305. [PMID: 33126239 PMCID: PMC7599084 DOI: 10.1371/journal.pone.0240305] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
Glatiramer acetate (GA) is approved for the treatment of multiple sclerosis (MS). However, the mechanism of action of GA in MS is still unclear. In particular, it is not known whether GA can modulate the pro-inflammatory Th17-type immune response in MS. We investigated the effects of original GA (Copaxone®, Teva, Israel) and generic GA (Timexone®, Biocad, Russia) on Th17- and Th1-type cytokine production in vitro in 25 patients with relapsing-remitting MS and 25 healthy subjects. Both original and generic GA at concentrations 50–200 μg/ml dose-dependently inhibited interleukin-17 and interferon-γ production by anti-CD3/anti-CD28-activated peripheral blood mononuclear cells from MS patients and healthy subjects. This effect of GA was reproduced using purified CD4+ T cells, suggesting that GA can directly modulate the functions of Th17 and Th1 cells. At high concentrations (100–200 μg/ml), GA also suppressed the production of Th17-differentiation cytokines (interleukin-1β and interleukin-6) by lipopolysaccharide (LPS)-activated dendritic cells (DCs). These GA/LPS-treated DCs induced lower interleukin-17 and interferon-γ production by autologous CD4+ T cells compared to LPS-treated DCs. These data suggest that GA can inhibit Th17-immune response and that this inhibitory effect is preferentially exercised by direct influence of GA on T cells. We also demonstrate a comparable ability of original and generic GA to modulate pro-inflammatory cytokine production.
Collapse
Affiliation(s)
- Mikhail Melnikov
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Moscow, Russia
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
- Department of Neuroimmunology, Federal Center of Brain Research and Neurotechnology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
- * E-mail:
| | - Svetlana Sharanova
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Anastasiya Sviridova
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of Neuroimmunology, Federal Center of Brain Research and Neurotechnology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Vladimir Rogovskii
- Department of Neuroimmunology, Federal Center of Brain Research and Neurotechnology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
- Department of Molecular Pharmacology and Radiobiology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Nina Murugina
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Anna Nikolaeva
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Yulia Dagil
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Vladimir Murugin
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Tatiana Ospelnikova
- Laboratory of Interferons, I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia
| | - Alexey Boyko
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of Neuroimmunology, Federal Center of Brain Research and Neurotechnology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Mikhail Pashenkov
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| |
Collapse
|
13
|
Moser T, Akgün K, Proschmann U, Sellner J, Ziemssen T. The role of TH17 cells in multiple sclerosis: Therapeutic implications. Autoimmun Rev 2020; 19:102647. [PMID: 32801039 DOI: 10.1016/j.autrev.2020.102647] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 03/08/2020] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) where immunopathology is thought to be mediated by myelin-reactive CD4+ T helper (TH) cells. The TH cells most commonly implicated in the pathogenesis of the disease are of TH1 and TH17 lineage, which are defined by the production of interferon-γ and interleukin-17, respectively. Moreover, there is emerging evidence for the involvement of TH17.1 cells, which share the hallmarks of TH1 and TH17 subsets. In this review, we summarise current knowledge about the potential role of TH17 subsets in the initiation and progression of the disease and put a focus on their response to approved immunomodulatory MS drugs. In this regard, TH17 cells are abundant in peripheral blood, cerebrospinal fluid and brain lesions of MS patients, and their counts and inflammatory mediators are further increased during relapses. Fingolimod and alemtuzumab induce a paramount decrease in central memory T cells, which harbour the majority of peripheral TH17 cells, while the efficacy of natalizumab, dimethyl fumarate and importantly hematopoietic stem cell therapy correlates with TH17.1 cell inhibition. Interestingly, also CD20 antibodies target highly inflammatory TH cells and hamper TH17 differentiation by IL-6 reductions. Moreover, recovery rates of TH cells best correlate with long-term efficacy after therapeutical immunodepletion. We conclude that central memory TH17.1 cells play a pivotal role in MS pathogenesis and they represent a major target of MS therapeutics.
Collapse
Affiliation(s)
- Tobias Moser
- Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, Dresden University of Technical, Fetscherstrasse 74, 01307 Dresden, Germany; Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Ignaz-Harrer-Straße 79, 5020 Salzburg, Austria
| | - Katja Akgün
- Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, Dresden University of Technical, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Undine Proschmann
- Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, Dresden University of Technical, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Johann Sellner
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Ignaz-Harrer-Straße 79, 5020 Salzburg, Austria; Department of Neurology, Landesklinikum Mistelbach-Gänserndorf, Liechtensteinstrasse 67, 3120 Mistelbach, Austria; Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, 81675 München, Germany
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, Dresden University of Technical, Fetscherstrasse 74, 01307 Dresden, Germany.
| |
Collapse
|
14
|
Aharoni R, Eilam R, Schottlender N, Radomir L, Leistner-Segal S, Feferman T, Hirsch D, Sela M, Arnon R. Glatiramer acetate increases T- and B -regulatory cells and decreases granulocyte-macrophage colony-stimulating factor (GM-CSF) in an animal model of multiple sclerosis. J Neuroimmunol 2020; 345:577281. [DOI: 10.1016/j.jneuroim.2020.577281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 01/21/2023]
|
15
|
Gharibi T, Babaloo Z, Hosseini A, Marofi F, Ebrahimi-Kalan A, Jahandideh S, Baradaran B. The role of B cells in the immunopathogenesis of multiple sclerosis. Immunology 2020; 160:325-335. [PMID: 32249925 DOI: 10.1111/imm.13198] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/01/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
There is ongoing debate on how B cells contribute to the pathogenesis of multiple sclerosis (MS). The success of B-cell targeting therapies in MS highlighted the role of B cells, particularly the antibody-independent functions of these cells such as antigen presentation to T cells and modulation of the function of T cells and myeloid cells by secreting pathogenic and/or protective cytokines in the central nervous system. Here, we discuss the role of different antibody-dependent and antibody-independent functions of B cells in MS disease activity and progression proposing new therapeutic strategies for the optimization of B-cell targeting treatments.
Collapse
Affiliation(s)
- Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zohreh Babaloo
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezoo Hosseini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Ebrahimi-Kalan
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Jahandideh
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Häusler D, Hajiyeva Z, Traub JW, Zamvil SS, Lalive PH, Brück W, Weber MS. Glatiramer acetate immune modulates B-cell antigen presentation in treatment of MS. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 7:e698. [PMID: 32184341 PMCID: PMC7136047 DOI: 10.1212/nxi.0000000000000698] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/31/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE We examined the effect of glatiramer acetate (GA) on B-cell maturation, differentiation, and antigen presentation in MS and experimental autoimmune encephalomyelitis (EAE). METHODS A cross-sectional study of blood samples from 20 GA-treated and 18 untreated patients with MS was performed by flow cytometry; 6 GA-treated patients with MS were analyzed longitudinally. GA-mediated effects on B-cell antigen-presenting function were investigated in EAE, or, alternatively, B cells were treated with GA in vitro using vehicle as a control. RESULTS In MS, GA diminished transitional B-cell and plasmablast frequency, downregulated CD69, CD25, and CD95 expression, and decreased TNF-α production, whereas IL-10 secretion and MHC Class II expression were increased. In EAE, we observed an equivalent dampening of proinflammatory B-cell properties and an enhanced expression of MHC Class II. When used as antigen-presenting cells for activation of naive T cells, GA-treated B cells promoted development of regulatory T cells, whereas proinflammatory T-cell differentiation was diminished. CONCLUSIONS GA immune modulates B-cell function in EAE and MS and efficiently interferes with pathogenic B cell-T cell interaction.
Collapse
Affiliation(s)
- Darius Häusler
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland
| | - Zivar Hajiyeva
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland
| | - Jan W Traub
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland
| | - Scott S Zamvil
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland
| | - Patrice H Lalive
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland
| | - Wolfgang Brück
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland
| | - Martin S Weber
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland.
| |
Collapse
|
17
|
Kunkl M, Frascolla S, Amormino C, Volpe E, Tuosto L. T Helper Cells: The Modulators of Inflammation in Multiple Sclerosis. Cells 2020; 9:cells9020482. [PMID: 32093011 PMCID: PMC7072830 DOI: 10.3390/cells9020482] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative disease characterized by the progressive loss of axonal myelin in several areas of the central nervous system (CNS) that is responsible for clinical symptoms such as muscle spasms, optic neuritis, and paralysis. The progress made in more than one decade of research in animal models of MS for clarifying the pathophysiology of MS disease validated the concept that MS is an autoimmune inflammatory disorder caused by the recruitment in the CNS of self-reactive lymphocytes, mainly CD4+ T cells. Indeed, high levels of T helper (Th) cells and related cytokines and chemokines have been found in CNS lesions and in cerebrospinal fluid (CSF) of MS patients, thus contributing to the breakdown of the blood-brain barrier (BBB), the activation of resident astrocytes and microglia, and finally the outcome of neuroinflammation. To date, several types of Th cells have been discovered and designated according to the secreted lineage-defining cytokines. Interestingly, Th1, Th17, Th1-like Th17, Th9, and Th22 have been associated with MS. In this review, we discuss the role and interplay of different Th cell subpopulations and their lineage-defining cytokines in modulating the inflammatory responses in MS and the approved as well as the novel therapeutic approaches targeting T lymphocytes in the treatment of the disease.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Simone Frascolla
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Carola Amormino
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Elisabetta Volpe
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| |
Collapse
|
18
|
Rommer PS, Milo R, Han MH, Satyanarayan S, Sellner J, Hauer L, Illes Z, Warnke C, Laurent S, Weber MS, Zhang Y, Stuve O. Immunological Aspects of Approved MS Therapeutics. Front Immunol 2019; 10:1564. [PMID: 31354720 PMCID: PMC6637731 DOI: 10.3389/fimmu.2019.01564] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 06/24/2019] [Indexed: 12/21/2022] Open
Abstract
Multiple sclerosis (MS) is the most common neurological immune-mediated disease leading to disability in young adults. The outcome of the disease is unpredictable, and over time, neurological disabilities accumulate. Interferon beta-1b was the first drug to be approved in the 1990s for relapsing-remitting MS to modulate the course of the disease. Over the past two decades, the treatment landscape has changed tremendously. Currently, more than a dozen drugs representing 1 substances with different mechanisms of action have been approved (interferon beta preparations, glatiramer acetate, fingolimod, siponimod, mitoxantrone, teriflunomide, dimethyl fumarate, cladribine, alemtuzumab, ocrelizumab, and natalizumab). Ocrelizumab was the first medication to be approved for primary progressive MS. The objective of this review is to present the modes of action of these drugs and their effects on the immunopathogenesis of MS. Each agent's clinical development and potential side effects are discussed.
Collapse
Affiliation(s)
- Paulus S. Rommer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Ron Milo
- Department of Neurology, Barzilai University Medical Center, Ashkelon, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - May H. Han
- Neuroimmunology Division, Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Sammita Satyanarayan
- Neuroimmunology Division, Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Johann Sellner
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria
- Department of Neurology, Klinikum Rechts der Isar, Technische Universität, Munich, Germany
| | - Larissa Hauer
- Department of Psychiatry, Psychotherapy, and Psychosomatics, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Clemens Warnke
- Department of Neurology, Medical Faculty, University of Köln, Cologne, Germany
| | - Sarah Laurent
- Department of Neurology, Medical Faculty, University of Köln, Cologne, Germany
| | - Martin S. Weber
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
- Department of Neurology, University Medical Center, Göttingen, Germany
| | - Yinan Zhang
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Olaf Stuve
- Department of Neurology, Klinikum Rechts der Isar, Technische Universität, Munich, Germany
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, VA North Texas Health Care System, Medical Service Dallas, VA Medical Center, Dallas, TX, United States
| |
Collapse
|
19
|
A differential sex-specific pattern of IgG2 and IgG4 subclasses of anti-drug antibodies (ADAs) induced by glatiramer acetate in relapsing-remitting multiple sclerosis patients. Mult Scler Relat Disord 2019; 34:92-99. [PMID: 31272071 DOI: 10.1016/j.msard.2019.06.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 11/21/2022]
Abstract
BACKGROUND Glatiramer acetate (GA) is a drug for Multiple Sclerosis (MS) treatment. However, its administration induces anti-drug antibodies (ADA). This research evaluated the sex differences in humoral response against GA in RR-MS patients METHODS: We analyzed 69 RR-MS patients, 43 treated with GA and 26 treated with IFN-β. In all cases, the serum concentration of IgG antibodies was determined by UPLC, whereas the levels of IgG subclasses (1-4) of anti-GA antibodies and the concentration of IL-6 were detected by Multiplex and IL-10, and IFN-γ were detected by ELISA. RESULTS The total concentration of IgG antibodies in patients did not differ between treatments, whereas the IgG levels of ADA were higher in male and female patients treated with GA (P ≤ 0.0001). The subclasses of IgG anti-GA antibodies were as follows: IgG4>>IgG3>IgG1>IgG2. Statistical analysis showed differences in the IgG2 (P ≤ 0.01) and IgG4 (P ≤ 0.0001) subclasses by sex in RR-MS patients. Levels of IgG1 subclass in male patients correlated positively with the circulatory levels of IL-6 (rs = 0.587, P ≤ 0.04) and IFN-γ (rs = 0.721, P ≤ 0.001), while IgG2 subclass levels in female patients correlated with serum levels of IFN-γ (rs = 0.628, P ≤ 0.0006). Statistical analysis did not detect correlations between the levels of IgG (1-4) subclasses of anti-GA antibodies and the evaluated clinical parameters. CONCLUSION This study showed differences in the levels of IgG2 and IgG4 subclasses of ADA between male and female RR-MS patients. Further studies are necessary to take advantage of the clinical potential of this finding.
Collapse
|
20
|
Boziki M, Lagoudaki R, Melo P, Kanidou F, Bakirtzis C, Nikolaidis I, Grigoriadou E, Afrantou T, Tatsi T, Matsi S, Grigoriadis N. Induction of apoptosis in CD4(+) T-cells is linked with optimal treatment response in patients with relapsing-remitting multiple sclerosis treated with Glatiramer acetate. J Neurol Sci 2019; 401:43-50. [DOI: 10.1016/j.jns.2019.03.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/11/2019] [Accepted: 03/28/2019] [Indexed: 11/29/2022]
|
21
|
Shahi SK, Freedman SN, Murra AC, Zarei K, Sompallae R, Gibson-Corley KN, Karandikar NJ, Murray JA, Mangalam AK. Prevotella histicola, A Human Gut Commensal, Is as Potent as COPAXONE® in an Animal Model of Multiple Sclerosis. Front Immunol 2019; 10:462. [PMID: 30984162 PMCID: PMC6448018 DOI: 10.3389/fimmu.2019.00462] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 02/20/2019] [Indexed: 12/31/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the central nervous system. We and others have shown that there is enrichment or depletion of some gut bacteria in MS patients compared to healthy controls (HC), suggesting an important role of the gut bacteria in disease pathogenesis. Thus, specific gut bacteria that are lower in abundance in MS patients could be used as a potential treatment option for this disease. In particular, we and others have shown that MS patients have a lower abundance of Prevotella compared to HC, whereas the abundance of Prevotella is increased in patients that receive disease-modifying therapies such as Copaxone® (Glatiramer acetate-GA). This inverse correlation between the severity of MS disease and the abundance of Prevotella suggests its potential for use as a therapeutic option to treat MS. Notably we have previously identified a specific strain, Prevotella histicola (P. histicola), that suppresses disease in the animal model of MS, experimental autoimmune encephalomyelitis (EAE) compared with sham treatment. In the present study we analyzed whether the disease suppressing effects of P. histicola synergize with those of the disease-modifying drug Copaxone® to more effectively suppress disease compared to either treatment alone. Treatment with P. histicola was as effective in suppressing disease as treatment with Copaxone®, whereas the combination of P. histicola plus Copaxone® was not more effective than either individual treatment. P. histicola-treated mice had an increased frequency and number of CD4+FoxP3+ regulatory T cells in periphery as well as gut and a decreased frequency of pro-inflammatory IFN-γ and IL17-producing CD4 T cells in the CNS, suggesting P. histicola suppresses disease by boosting anti-inflammatory immune responses and inhibiting pro-inflammatory immune responses. In conclusion, our study indicates that the human gut commensal P. histicola can suppress disease as efficiently as Copaxone® and may provide an alternative treatment option for MS patients.
Collapse
Affiliation(s)
- Shailesh K Shahi
- Department of Pathology, University of Iowa, Iowa City, IA, United States
| | - Samantha N Freedman
- Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Alexandra C Murra
- Department of Pathology, University of Iowa, Iowa City, IA, United States
| | - Kasra Zarei
- Medical Scientist Training Program, University of Iowa, Iowa City, IA, United States
| | | | | | - Nitin J Karandikar
- Department of Pathology, University of Iowa, Iowa City, IA, United States.,Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States.,Graduate Program in Molecular Cell Biology, University of Iowa, Iowa City, IA, United States
| | - Joseph A Murray
- Department of Immunology, Mayo Clinic, Rochester, MN, United States.,Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Ashutosh K Mangalam
- Department of Pathology, University of Iowa, Iowa City, IA, United States.,Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States.,Medical Scientist Training Program, University of Iowa, Iowa City, IA, United States.,Graduate Program in Molecular Cell Biology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
22
|
Amrouche K, Pers JO, Jamin C. Glatiramer Acetate Stimulates Regulatory B Cell Functions. THE JOURNAL OF IMMUNOLOGY 2019; 202:1970-1980. [DOI: 10.4049/jimmunol.1801235] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/07/2019] [Indexed: 01/14/2023]
|
23
|
Prod'homme T, Zamvil SS. The Evolving Mechanisms of Action of Glatiramer Acetate. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a029249. [PMID: 29440323 DOI: 10.1101/cshperspect.a029249] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Glatiramer acetate (GA) is a synthetic amino acid copolymer that is approved for treatment of relapsing remitting multiple sclerosis (RRMS) and clinically isolated syndrome (CIS). GA reduces multiple sclerosis (MS) disease activity and has shown comparable efficacy with high-dose interferon-β. The mechanism of action (MOA) of GA has long been an enigma. Originally, it was recognized that GA treatment promoted expansion of GA-reactive T-helper 2 and regulatory T cells, and induced the release of neurotrophic factors. However, GA treatment influences both innate and adaptive immune compartments, and it is now recognized that antigen-presenting cells (APCs) are the initial cellular targets for GA. The anti-inflammatory (M2) APCs induced following treatment with GA are responsible for the induction of anti-inflammatory T cells that contribute to its therapeutic benefit. Here, we review studies that have shaped our current understanding of the MOA of GA.
Collapse
Affiliation(s)
| | - Scott S Zamvil
- Department of Neurology and Program in Immunology, University of California, San Francisco, San Francisco, California 94158
| |
Collapse
|
24
|
Effect of glatiramer acetate on cerebral grey matter pathology in patients with relapsing-remitting multiple sclerosis. Mult Scler Relat Disord 2019; 27:305-311. [DOI: 10.1016/j.msard.2018.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/08/2018] [Accepted: 11/11/2018] [Indexed: 01/17/2023]
|
25
|
Song JY, Larson NR, Thati S, Torres-Vazquez I, Martinez-Rivera N, Subelzu NJ, Leon MA, Rosa-Molinar E, Schöneich C, Forrest ML, Middaugh CR, Berkland CJ. Glatiramer acetate persists at the injection site and draining lymph nodes via electrostatically-induced aggregation. J Control Release 2018; 293:36-47. [PMID: 30414463 DOI: 10.1016/j.jconrel.2018.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/30/2018] [Accepted: 11/04/2018] [Indexed: 01/01/2023]
Abstract
Glatiramer acetate (GA) is widely prescribed for the treatment of relapsing-remitting multiple sclerosis, however, the mechanism of action is still not fully understood. We investigated the structural properties of GA and examined alterations to the drug upon injection into the subcutaneous space. First, a variety of biophysical characterization techniques were employed to characterize GA in solution. GA was found to exist as alpha helices in solution with a hydrodynamic radius of ~3 nm in size. To simulate GA behavior at the site of injection, GA was injected into a solution of 1.5 MDa hyaluronic acid (HA). Visible aggregates were observed immediately upon injection and subsequent testing indicated aggregation was driven by electrostatic interactions between the positively-charged GA and negatively-charged HA. In vivo testing confirmed GA formed spherical particles in the nano- to micrometer size range, suggesting this mechanism contributes to persistence at the injection site and in draining lymph nodes. The aggregates were found to associate with glycosaminoglycans, suggesting an electrostatic mechanism of induced aggregation like the simulated injection. These novel observations may help explain the complex immunomodulatory mechanisms of GA and adverse injection site reactions seen in patients.
Collapse
Affiliation(s)
- Jimmy Y Song
- Department of Pharmaceutical Chemistry, University of Kansas, USA
| | | | - Sharadvi Thati
- Department of Pharmaceutical Chemistry, University of Kansas, USA
| | - Irma Torres-Vazquez
- Microscopy and Analytical Imaging Laboratory, University of Kansas, USA; Department of Pharmacology and Toxicology, University of Kansas, USA
| | - Noraida Martinez-Rivera
- Microscopy and Analytical Imaging Laboratory, University of Kansas, USA; Department of Pharmacology and Toxicology, University of Kansas, USA
| | | | | | - Eduardo Rosa-Molinar
- Department of Bioengineering, University of Kansas, USA; Microscopy and Analytical Imaging Laboratory, University of Kansas, USA; Department of Pharmacology and Toxicology, University of Kansas, USA
| | | | - M Laird Forrest
- Department of Pharmaceutical Chemistry, University of Kansas, USA
| | | | - Cory J Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, USA; Department of Chemistry, University of Kansas, USA; Department of Bioengineering, University of Kansas, USA; Department of Chemical and Petroleum Engineering, University of Kansas, USA.
| |
Collapse
|
26
|
van der Touw W, Kang K, Luan Y, Ma G, Mai S, Qin L, Bian G, Zhang R, Mungamuri SK, Hu HM, Zhang CC, Aaronson SA, Feldmann M, Yang WC, Chen SH, Pan PY. Glatiramer Acetate Enhances Myeloid-Derived Suppressor Cell Function via Recognition of Paired Ig-like Receptor B. THE JOURNAL OF IMMUNOLOGY 2018; 201:1727-1734. [PMID: 30068593 DOI: 10.4049/jimmunol.1701450] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 06/25/2018] [Indexed: 01/26/2023]
Abstract
Glatiramer acetate (GA; Copaxone) is a copolymer therapeutic that is approved by the Food and Drug Administration for the relapsing-remitting form of multiple sclerosis. Despite an unclear mechanism of action, studies have shown that GA promotes protective Th2 immunity and stimulates release of cytokines that suppress autoimmunity. In this study, we demonstrate that GA interacts with murine paired Ig-like receptor B (PIR-B) on myeloid-derived suppressor cells and suppresses the STAT1/NF-κB pathways while promoting IL-10/TGF-β cytokine release. In inflammatory bowel disease models, GA enhanced myeloid-derived suppressor cell-dependent CD4+ regulatory T cell generation while reducing proinflammatory cytokine secretion. Human monocyte-derived macrophages responded to GA by reducing TNF-α production and promoting CD163 expression typical of alternative maturation despite the presence of GM-CSF. Furthermore, GA competitively interacts with leukocyte Ig-like receptors B (LILRBs), the human orthologs of PIR-B. Because GA limited proinflammatory activation of myeloid cells, therapeutics that target LILRBs represent novel treatment modalities for autoimmune indications.
Collapse
Affiliation(s)
- William van der Touw
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Kyeongah Kang
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX 77030
| | - Yi Luan
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ge Ma
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Sunny Mai
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX 77030
| | - Lihui Qin
- Department of Pathology, Weill Cornell Medical College, New York, NY 10065
| | - Guanglin Bian
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ruihua Zhang
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Sathish Kumar Mungamuri
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Hong-Ming Hu
- Laboratory of Cancer Immunobiology, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR 97213
| | - Cheng Cheng Zhang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Stuart A Aaronson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Marc Feldmann
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopedics, Rheumatology, and Musculoskeletal Science, University of Oxford, Oxford OX3 7FZ, United Kingdom
| | - Wen-Chin Yang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Shu-Hsia Chen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; .,Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX 77030.,Center for Infectious Diseases and Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan; and.,Cancer Center, Houston Methodist Research Institute, Houston, TX 77030
| | - Ping-Ying Pan
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; .,Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX 77030.,Cancer Center, Houston Methodist Research Institute, Houston, TX 77030
| |
Collapse
|
27
|
First-line therapy in relapsing remitting multiple sclerosis. Rev Neurol (Paris) 2018; 174:419-428. [DOI: 10.1016/j.neurol.2018.03.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/11/2018] [Accepted: 03/12/2018] [Indexed: 11/21/2022]
|
28
|
D Alessandro J, Garofalo K, Zhao G, Honan C, Duffner J, Capila I, Fier I, Kaundinya G, Kantor D, Ganguly T. Demonstration of Biological and Immunological Equivalence of a Generic Glatiramer Acetate. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2018; 16:714-723. [PMID: 28240190 PMCID: PMC5684786 DOI: 10.2174/1871527316666170223162747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/24/2017] [Accepted: 02/02/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND In April 2015, the US Food and Drug Administration approved the first generic glatiramer acetate, Glatopa® (M356), as fully substitutable for Copaxone® 20 mg/mL for relapsing forms of multiple sclerosis (MS). This approval was accomplished through an Abbreviated New Drug Application that demonstrated equivalence to Copaxone. METHOD This article will provide an overview of the methods used to establish the biological and immunological equivalence of the two glatiramer acetate products, including methods evaluating antigenpresenting cell (APC) biology, T-cell biology, and other immunomodulatory effects. RESULTS In vitro and in vivo experiments from multiple redundant orthogonal assays within four biological processes (aggregate biology, APC biology, T-cell biology, and B-cell biology) modulated by glatiramer acetate in MS established the biological and immunological equivalence of Glatopa and Copaxone and are described. The following were observed when comparing Glatopa and Copaxone in these experiments: equivalent delays in symptom onset and reductions in "disease" intensity in experimental autoimmune encephalomyelitis; equivalent dose-dependent increases in Glatopa- and Copaxone- induced monokine-induced interferon-gamma release from THP-1 cells; a shift to a T helper 2 phenotype resulting in the secretion of interleukin (IL)-4 and downregulation of IL-17 release; no differences in immunogenicity and the presence of equivalent "immunofingerprints" between both versions of glatiramer acetate; and no stimulation of histamine release with either glatiramer acetate in basophilic leukemia 2H3 cell lines. CONCLUSION In summary, this comprehensive approach across different biological and immunological pathways modulated by glatiramer acetate consistently supported the biological and immunological equivalence of Glatopa and Copaxone.
Collapse
Affiliation(s)
| | - Kevin Garofalo
- Research Department, Momenta Pharmaceuticals, Inc., Cambridge, MA. United States
| | - Ganlin Zhao
- Division of Bioequivalence I, Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD. United States
| | - Christopher Honan
- Research Department, Momenta Pharmaceuticals, Inc., Cambridge, MA. United States
| | - Jay Duffner
- Research Department, Momenta Pharmaceuticals, Inc., Cambridge, MA. United States
| | - Ishan Capila
- Research Department, Momenta Pharmaceuticals, Inc., Cambridge, MA. United States
| | - Ian Fier
- Research Department, Momenta Pharmaceuticals, Inc., Cambridge, MA. United States
| | - Ganesh Kaundinya
- Research Department, Momenta Pharmaceuticals, Inc., Cambridge, MA. United States
| | - Daniel Kantor
- Division of Neurology, Florida Atlantic University, Boca Raton, FL. United States
| | - Tanmoy Ganguly
- Momenta Pharmaceuticals, Inc., 675 West Kendall Street, Cambridge, MA 02142. United States
| |
Collapse
|
29
|
Abstract
Growing evidence indicates that B cells play a key role in the pathogenesis of multiple sclerosis (MS). B cells occupy distinct central nervous system (CNS) compartments in MS, including the cerebrospinal fluid and white matter lesions. Also, it is now known that, in addition to entering the CNS, B cells can circulate into the periphery via a functional lymphatic system. Data suggest that the role of B cells in MS mainly involves their in situ activation in demyelinating lesions, leading to altered pro- and anti-inflammatory cytokine secretion, and a highly effective antigen-presenting cell function, resulting in activation of memory or naïve T cells. Clinically, B cell-depleting agents show significant efficacy in MS. In addition, many disease-modifying therapies (DMTs) traditionally understood to target T cells are now known to influence B cell number and function. One of the earliest DMTs to be developed, glatiramer acetate (GA), has been shown to reduce the total frequency of B cells, plasmablasts, and memory B cells. It also appears to promote a shift toward reduced inflammation by increasing anti-inflammatory cytokine release and/or reducing pro-inflammatory cytokine release by B cells. In the authors' opinion, this may be mediated by cross-reactivity of B cell receptors for GA with antigen (possibly myelin basic protein) expressed in the MS lesion. More research is required to further characterize the role of B cells and their bidirectional trafficking in the pathogenesis of MS. This may uncover novel targets for MS treatments and facilitate the development of B cell biomarkers of drug response.
Collapse
|
30
|
Deciphering the Role of B Cells in Multiple Sclerosis-Towards Specific Targeting of Pathogenic Function. Int J Mol Sci 2017; 18:ijms18102048. [PMID: 28946620 PMCID: PMC5666730 DOI: 10.3390/ijms18102048] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 12/25/2022] Open
Abstract
B cells, plasma cells and antibodies may play a key role in the pathogenesis of multiple sclerosis (MS). This notion is supported by various immunological changes observed in MS patients, such as activation and pro-inflammatory differentiation of peripheral blood B cells, the persistence of clonally expanded plasma cells producing immunoglobulins in the cerebrospinal fluid, as well as the composition of inflammatory central nervous system lesions frequently containing co-localizing antibody depositions and activated complement. In recent years, the perception of a respective pathophysiological B cell involvement was vividly promoted by the empirical success of anti-CD20-mediated B cell depletion in clinical trials; based on these findings, the first monoclonal anti-CD20 antibody—ocrelizumab—is currently in the process of being approved for treatment of MS. In this review, we summarize the current knowledge on the role of B cells, plasma cells and antibodies in MS and elucidate how approved and future treatments, first and foremost anti-CD20 antibodies, therapeutically modify these B cell components. We will furthermore describe regulatory functions of B cells in MS and discuss how the evolving knowledge of these therapeutically desirable B cell properties can be harnessed to improve future safety and efficacy of B cell-directed therapy in MS.
Collapse
|
31
|
Grebenciucova E, Pruitt A. Infections in Patients Receiving Multiple Sclerosis Disease-Modifying Therapies. Curr Neurol Neurosci Rep 2017; 17:88. [PMID: 28940162 DOI: 10.1007/s11910-017-0800-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW This paper will systemically review the risk of infections associated with current disease-modifying treatments and will discuss pre-treatment testing recommendations, infection monitoring strategies, and patient education. RECENT FINDINGS Aside from glatiramer acetate and interferon-beta therapies, all other multiple sclerosis treatments to various degrees impair immune surveillance and may predispose patients to the development of both community-acquired and opportunistic infections. Some of these infections are rarely seen in neurologic practice, and neurologists should be aware of how to monitor for these infections and how to educate patients about medication-specific risks. Of particular interest in this discussion is the risk of PML in association with the recently approved B cell depleting therapy, ocrelizumab, particularly when switching from natalizumab. The risk of infection in association with MS treatments has become one of the most important factors in the choice of therapy. Balance of the overall risk versus benefit should be continuously re-evaluated during treatment.
Collapse
Affiliation(s)
- Elena Grebenciucova
- Multiple Sclerosis Division, Davee Department of Neurology and Clinical Neurosciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Amy Pruitt
- Multiple Sclerosis Division, the Department of Neurology, Perelman School of Medicine, The University of Pennsylvania, 3400 Convention Avenue, Philadelphia, PA, 19104, USA
| |
Collapse
|
32
|
Abstract
Glatiramer acetate is a mixture of synthetic peptides that are cross-reactive with MBP. The antigen-based therapy induces a shift to an anti-inflammatory Th2 bias and is used in the treatment of relapsing-remitting multiple sclerosis. Like other peptide antigens, GA induces an antibody response in all patients. In contrast to biologically active agents, such as the recombinant interferon beta drugs, GA is a peptide antigen that lacks intrinsic biological activity. In vitro and in vivo data have shown that GA-reactive antibodies are not neutralizing. Antibodies do not alter the principal immunological effects of GA, including binding to MHC Class II molecules, activation and proliferation of GA-reactive T cells, and the release of anti-inflammatory Th2 cytokines. Higher antibody titres do not appear to be associated with a deterioration in clinical endpoints, such as relapse rate, EDSS progression or the occurrence of side effects in MS patients treated with GA. The presence of GA-reactive antibodies may promote remyelination and enhance the immunological and clinical effects of GA, indicating that they may be part of GA's mechanism of action. Multiple Sclerosis 2007; 13: S28—S35. http://msj.sagepub.com
Collapse
|
33
|
Spadaro M, Montarolo F, Perga S, Martire S, Brescia F, Malucchi S, Bertolotto A. Biological activity of glatiramer acetate on Treg and anti-inflammatory monocytes persists for more than 10years in responder multiple sclerosis patients. Clin Immunol 2017. [PMID: 28642148 DOI: 10.1016/j.clim.2017.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Glatiramer acetate (GA) is a widely used treatment for multiple sclerosis (MS), with incompletely defined mechanism of action. Short-term studies suggested its involvement in the modulation of anti-inflammatory cytokines and regulatory T cells (Treg), while long-term effect is still unknown. To investigate this aspect, we analyzed by flow-cytometry peripheral-blood Treg, natural killer (NK), CD4 and CD8 T-cells and anti-inflammatory CD14+CD163+ monocytes from 37 healthy donor and 90 RRMS patients divided in untreated, treated with GA for 12months and from 34 to 192months. While NK, CD4 and CD8 T-cells did not show any significant differences among groups over time, we demonstrated that GA increased the anti-inflammatory monocytes and restored the Treg level in both GA-treated groups. Both these effects are a characteristic of responder patients and are observed not just in short-term but even after as long as a decade of GA treatment.
Collapse
Affiliation(s)
- Michela Spadaro
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Clinical Neurobiology Unit, Orbassano, Turin, Italy; AOU S. Luigi Gonzaga, Neurologia 2 - CReSM (Centro Riferimento Regionale Sclerosi Multipla), Orbassano, Turin, Italy.
| | - Francesca Montarolo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Clinical Neurobiology Unit, Orbassano, Turin, Italy; AOU S. Luigi Gonzaga, Neurologia 2 - CReSM (Centro Riferimento Regionale Sclerosi Multipla), Orbassano, Turin, Italy
| | - Simona Perga
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Clinical Neurobiology Unit, Orbassano, Turin, Italy; AOU S. Luigi Gonzaga, Neurologia 2 - CReSM (Centro Riferimento Regionale Sclerosi Multipla), Orbassano, Turin, Italy
| | - Serena Martire
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Clinical Neurobiology Unit, Orbassano, Turin, Italy; AOU S. Luigi Gonzaga, Neurologia 2 - CReSM (Centro Riferimento Regionale Sclerosi Multipla), Orbassano, Turin, Italy
| | - Federica Brescia
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Clinical Neurobiology Unit, Orbassano, Turin, Italy; AOU S. Luigi Gonzaga, Neurologia 2 - CReSM (Centro Riferimento Regionale Sclerosi Multipla), Orbassano, Turin, Italy
| | - Simona Malucchi
- AOU S. Luigi Gonzaga, Neurologia 2 - CReSM (Centro Riferimento Regionale Sclerosi Multipla), Orbassano, Turin, Italy
| | - Antonio Bertolotto
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Clinical Neurobiology Unit, Orbassano, Turin, Italy; AOU S. Luigi Gonzaga, Neurologia 2 - CReSM (Centro Riferimento Regionale Sclerosi Multipla), Orbassano, Turin, Italy
| |
Collapse
|
34
|
Kelly SH, Shores LS, Votaw NL, Collier JH. Biomaterial strategies for generating therapeutic immune responses. Adv Drug Deliv Rev 2017; 114:3-18. [PMID: 28455189 PMCID: PMC5606982 DOI: 10.1016/j.addr.2017.04.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 04/19/2017] [Accepted: 04/21/2017] [Indexed: 01/04/2023]
Abstract
Biomaterials employed to raise therapeutic immune responses have become a complex and active field. Historically, vaccines have been developed primarily to fight infectious diseases, but recent years have seen the development of immunologically active biomaterials towards an expanding list of non-infectious diseases and conditions including inflammation, autoimmunity, wounds, cancer, and others. This review structures its discussion of these approaches around a progression from single-target strategies to those that engage increasingly complex and multifactorial immune responses. First, the targeting of specific individual cytokines is discussed, both in terms of delivering the cytokines or blocking agents, and in terms of active immunotherapies that raise neutralizing immune responses against such single cytokine targets. Next, non-biological complex drugs such as randomized polyamino acid copolymers are discussed in terms of their ability to raise multiple different therapeutic immune responses, particularly in the context of autoimmunity. Last, biologically derived matrices and materials are discussed in terms of their ability to raise complex immune responses in the context of tissue repair. Collectively, these examples reflect the tremendous diversity of existing approaches and the breadth of opportunities that remain for generating therapeutic immune responses using biomaterials.
Collapse
Affiliation(s)
- Sean H Kelly
- Duke University, Department of Biomedical Engineering, United States
| | - Lucas S Shores
- Duke University, Department of Biomedical Engineering, United States
| | - Nicole L Votaw
- Duke University, Department of Biomedical Engineering, United States
| | - Joel H Collier
- Duke University, Department of Biomedical Engineering, United States.
| |
Collapse
|
35
|
Grossman I, Knappertz V, Laifenfeld D, Ross C, Zeskind B, Kolitz S, Ladkani D, Hayardeny L, Loupe P, Laufer R, Hayden M. Pharmacogenomics strategies to optimize treatments for multiple sclerosis: Insights from clinical research. Prog Neurobiol 2017; 152:114-130. [DOI: 10.1016/j.pneurobio.2016.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 02/10/2016] [Accepted: 02/27/2016] [Indexed: 12/13/2022]
|
36
|
Ahn YH, Jeon SB, Chang CY, Goh EA, Kim SS, Kim HJ, Song J, Park EJ. Glatiramer acetate attenuates the activation of CD4 + T cells by modulating STAT1 and -3 signaling in glia. Sci Rep 2017; 7:40484. [PMID: 28094337 PMCID: PMC5240344 DOI: 10.1038/srep40484] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/06/2016] [Indexed: 12/21/2022] Open
Abstract
Interactions between immune effector cells of the central nervous system appear to directly or indirectly influence the progress/regression of multiple sclerosis (MS). Here, we report that glial STAT1 and -3 are distinctively phosphorylated following the interaction of activated lymphocytes and glia, and this effect is significantly inhibited by glatiramer acetate (GA), a disease-modifying drug for MS. GA also reduces the activations of STAT1 and -3 by MS-associated stimuli such as IFNγ or LPS in primary glia, but not neurons. Experiments in IFNγ- and IFNγ receptor-deficient mice revealed that GA-induced inhibitions of STAT signaling are independent of IFNγ and its receptor. Interestingly, GA induces the expression levels of suppressor of cytokine signaling-1 and -3, representative negative regulators of STAT signaling in glia. We further found that GA attenuates the LPS-triggered enhancement of IL-2, a highly produced cytokine in patients with active MS, in CD4+ T cells co-cultured with glia, but not in CD4+ T cells alone. Collectively, these results provide that activation of glial STATs is an essential event in the interaction between glia and T cells, which is a possible underlying mechanism of GA action in MS. These findings provide an insight for the development of targeted therapies against MS.
Collapse
Affiliation(s)
- Ye-Hyeon Ahn
- Cancer Immunology Branch, National Cancer Center, Goyang, South Korea
- Dept.of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Sae-Bom Jeon
- Cancer Immunology Branch, National Cancer Center, Goyang, South Korea
| | - Chi Young Chang
- Cancer Immunology Branch, National Cancer Center, Goyang, South Korea
| | - Eun-Ah Goh
- Dept. of System Cancer Science, Graduate School of Cancer Science and Policy, Goyang, South Korea
| | - Sang Soo Kim
- Radiation Medicine Branch, National Cancer Center, Goyang, South Korea
| | - Ho Jin Kim
- Dept. of System Cancer Science, Graduate School of Cancer Science and Policy, Goyang, South Korea
- Dept. of Neurology, National Cancer Center, Goyang, South Korea
| | - Jaewhan Song
- Dept.of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Eun Jung Park
- Cancer Immunology Branch, National Cancer Center, Goyang, South Korea
- Dept. of System Cancer Science, Graduate School of Cancer Science and Policy, Goyang, South Korea
| |
Collapse
|
37
|
Abstract
Over the last decade, evidence condensed that B cells, B cell-derived plasma cells and antibodies play a key role in the pathogenesis and progression of multiple sclerosis (MS). In many patients with MS, peripheral B cells show signs of chronic activation; within the cerebrospinal fluid clonally expanded plasma cells produce oligoclonal immunoglobulins, which remain a hallmark diagnostic finding. Confirming the clinical relevance of these immunological alterations, recent trials testing anti-CD20-mediated depletion of peripheral B cells showed an instantaneous halt in development of new central nervous system lesions and occurrence of relapses. Notwithstanding this enormous success, not all B cells or B cell subsets may contribute in a pathogenic manner, and may, in contrast, exert anti-inflammatory and, thus, therapeutically desirable properties in MS. Naïve B cells, in MS patients similar to healthy controls, are a relevant source of regulatory cytokines such as interleukin-10, which dampens the activity of other immune cells and promotes recovery from acute disease flares in experimental MS models. In this review, we describe in detail pathogenic but also regulatory properties of B and plasma cells in the context of MS and its animal model experimental autoimmune encephalomyelitis. In the second part, we review what impact current and future therapies may have on these B cell properties. Within this section, we focus on the highly encouraging data on anti-CD20 antibodies as future therapy for MS. Lastly, we discuss how B cell-directed therapy in MS could be possibly advanced even further in regard to efficacy and safety by integrating the emerging information on B cell regulation in MS into future therapeutic strategies.
Collapse
Affiliation(s)
- Silke Kinzel
- Department of Neuropathology, University Medical Center, Georg August University, Robert-Koch-Str. 40, 37099, Göttingen, Germany
| | - Martin S Weber
- Department of Neuropathology, University Medical Center, Georg August University, Robert-Koch-Str. 40, 37099, Göttingen, Germany.
- Department of Neurology, University Medical Center, 37075, Göttingen, Germany.
| |
Collapse
|
38
|
De Riccardis L, Ferramosca A, Danieli A, Trianni G, Zara V, De Robertis F, Maffia M. Metabolic response to glatiramer acetate therapy in multiple sclerosis patients. BBA CLINICAL 2016; 6:131-137. [PMID: 27785417 PMCID: PMC5079236 DOI: 10.1016/j.bbacli.2016.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 10/12/2016] [Accepted: 10/17/2016] [Indexed: 12/15/2022]
Abstract
Glatiramer acetate (GA; Copaxone) is a random copolymer of glutamic acid, lysine, alanine, and tyrosine used for the treatment of patients with multiple sclerosis (MS). Its mechanism of action has not been already fully elucidated, but it seems that GA has an immune-modulatory effect and neuro-protective properties. Lymphocyte mitochondrial dysfunction underlines the onset of several autoimmune disorders. In MS first diagnosis patients, CD4+, the main T cell subset involved in the pathogenesis of MS, undergo a metabolic reprogramming that consist in the up-regulation of glycolysis and in the down-regulation of oxidative phosphorylation. Currently, no works exist about CD4+ T cell metabolism in response to GA treatment. In order to provide novel insight into the potential use of GA in MS treatment, blood samples were collected from 20 healthy controls (HCs) and from 20 RR MS patients prior and every 6 months during the 12 months of GA administration. GA treated patients' CD4+ T cells were compared with those from HCs analysing their mitochondrial activity through polarographic and enzymatic methods in association with their antioxidant status, through the analysis of SOD, GPx and CAT activities. Altogether, our findings suggest that GA is able to reduce CD4+ T lymphocytes' dysfunctions by increasing mitochondrial activity and their response to oxidative stress. GA is able to reduce CD4 + T cell's dysfunctions in MS patients; A CD4 + T cell metabolic response in GA treated patients is proposed; Metabolic response relies on changes in mitochondrial activity and in antioxidative status.
Collapse
Key Words
- CAT, catalase
- CD4+ T cells
- CNS, central nervous system
- CS, citrate synthase
- EAE, experimental autoimmune encephalomyelitis
- GPX, glutathione peroxidase
- GR, glutathione reductase
- Glycolysis
- HK, hexokinase
- MCT, mono-carboxylate transporters
- MS, Multiple Sclerosis
- Multiple sclerosis
- OXPHOS
- OXPHOS, oxidative phosphorylation
- Oxidative stress
- PBMC, peripheral blood mononuclear cell
- PFK, phosphofructokinase
- RCR, respiratory control ratio
- ROS, reactive oxygen species
- RRMS, Relapsing-Remitting Multiple Sclerosis
- SOD, superoxide dismutase
- Th, T helper
Collapse
Affiliation(s)
- Lidia De Riccardis
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Prov.le Lecce-Monteroni, Lecce, Italy
| | - Alessandra Ferramosca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Prov.le Lecce-Monteroni, Lecce, Italy
| | - Antonio Danieli
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Prov.le Lecce-Monteroni, Lecce, Italy
| | - Giorgio Trianni
- Department of Neurology, "Vito Fazzi" Hospital, ASL-Lecce, Italy
| | - Vincenzo Zara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Prov.le Lecce-Monteroni, Lecce, Italy
| | | | - Michele Maffia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Prov.le Lecce-Monteroni, Lecce, Italy
| |
Collapse
|
39
|
Multiple Sclerosis and Obesity: Possible Roles of Adipokines. Mediators Inflamm 2016; 2016:4036232. [PMID: 27721574 PMCID: PMC5046034 DOI: 10.1155/2016/4036232] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/22/2016] [Accepted: 08/08/2016] [Indexed: 12/21/2022] Open
Abstract
Multiple Sclerosis (MS) is an autoimmune disorder of the Central Nervous System that has been associated with several environmental factors, such as diet and obesity. The possible link between MS and obesity has become more interesting in recent years since the discovery of the remarkable properties of adipose tissue. Once MS is initiated, obesity can contribute to increased disease severity by negatively influencing disease progress and treatment response, but, also, obesity in early life is highly relevant as a susceptibility factor and causally related risk for late MS development. The aim of this review was to discuss recent evidence about the link between obesity, as a chronic inflammatory state, and the pathogenesis of MS as a chronic autoimmune and inflammatory disease. First, we describe the main cells involved in MS pathogenesis, both from neural tissue and from the immune system, and including a new participant, the adipocyte, focusing on their roles in MS. Second, we concentrate on the role of several adipokines that are able to participate in the mediation of the immune response in MS and on the possible cross talk between the latter. Finally, we explore recent therapy that involves the transplantation of adipocyte precursor cells for the treatment of MS.
Collapse
|
40
|
Yokoyama K, Hattori N. Immunomodulatory effects of glatiramer acetate as they relate to stage-specific immune dysregulation in multiple sclerosis. Nihon Yakurigaku Zasshi 2016; 148:105-20. [PMID: 27478050 DOI: 10.1254/fpj.148.105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
41
|
Comi G, Amato MP, Bertolotto A, Centonze D, De Stefano N, Farina C, Gallo P, Ghezzi A, Grimaldi LM, Mancardi G, Marrosu MG, Montanari E, Patti F, Pozzilli C, Provinciali L, Salvetti M, Tedeschi G, Trojano M. The heritage of glatiramer acetate and its use in multiple sclerosis. ACTA ACUST UNITED AC 2016. [DOI: 10.1186/s40893-016-0010-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
42
|
Jansson A, Ernerudh J, Kvarnström M, Ekerfelt C, Vrethem M. Elispot assay detection of cytokine secretion in multiple sclerosis patients treated with interferon-b1a or glatiramer acetate compared with untreated patients. Mult Scler 2016; 9:440-5. [PMID: 14582766 DOI: 10.1191/1352458503ms951oa] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The mechanisms behind the beneficial effects of interferon-b1a (IFN-b1a) and glatiramer acetate (GA) in the treatment of multiple sclerosis (MS) are still uncertain. A ltered cytokine patterns have been suggested including inhibition of proinflammatory cytokines like interferon-g (IFN-g) and enhancement of anti-inflammato ry cytokines such as interleukin-4 (IL-4). Twenty-nine patients with MS (10 untreated, nine treated with IFN-b1a and 10 with GA) were investigated with elispot of peripheral blood mononuclear cells. Spontaneous and myelin induced (myelin basic protein (MBP), myelin oligodendro cyte glycoprotein (MO G)-14-39 and MO G 63-87) IFN-g, IL-4, IL-5 and IL-10 secretion was studied. We found a significant reduction of spontaneous IFN-g, IL-4 and IL-5, but no difference in IL-10 secreting cells in both groups of treated patients compared with the untreated patients. Myelin-specific responses showed a significant decrease of IFN-g and an increase of IL-5, but no change in IL-4 and IL-10 secreting cells in treated compared with untreated patients. Both treatment groups revealed similar cytokine secretion patterns except for a more pronounced decrease of both spontaneous and MO G 14-39 induced IL-4 secretion in the IFN-b1a treated group. Thus, immunological effects of IFN-b1a and G A were similar showing that disease promoting Th1 (IFN-g) cells were reduced while the potentially beneficial Th2 response (IL-4) was maintained.
Collapse
Affiliation(s)
- A Jansson
- Clinical Research Centre and Department of Molecular and Clinical Medicine, Division of Clinical Immunology, Linköping University, S-581 85 Linköping, Sweden
| | | | | | | | | |
Collapse
|
43
|
Luessi F, Zipp F, Witsch E. Dendritic cells as therapeutic targets in neuroinflammation. Cell Mol Life Sci 2016; 73:2425-50. [PMID: 26970979 PMCID: PMC11108452 DOI: 10.1007/s00018-016-2170-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 02/02/2016] [Accepted: 02/25/2016] [Indexed: 12/23/2022]
Abstract
Multiple sclerosis (MS) is the most common chronic inflammatory demyelinating disorder of the central nervous system characterized by infiltration of immune cells and progressive damage to myelin sheaths and neurons. There is still no cure for the disease, but drug regimens can reduce the frequency of relapses and slightly delay progression. Myeloid cells or antigen-presenting cells (APCs) such as dendritic cells (DC), macrophages, and resident microglia, are key players in both mediating immune responses and inducing immune tolerance. Mounting evidence indicates a contribution of these myeloid cells to the pathogenesis of multiple sclerosis and to the effects of treatment, the understanding of which might provide strategies for more potent novel therapeutic interventions. Here, we review recent insights into the role of APCs, with specific focus on DCs in the modulation of neuroinflammation in MS.
Collapse
Affiliation(s)
- Felix Luessi
- Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University of Mainz,Rhine Main Neuroscience Network (rmn2), Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University of Mainz,Rhine Main Neuroscience Network (rmn2), Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Esther Witsch
- Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University of Mainz,Rhine Main Neuroscience Network (rmn2), Langenbeckstrasse 1, 55131, Mainz, Germany.
| |
Collapse
|
44
|
Administration of Myelin Basic Protein Peptides Encapsulated in Mannosylated Liposomes Normalizes Level of Serum TNF-α and IL-2 and Chemoattractants CCL2 and CCL4 in Multiple Sclerosis Patients. Mediators Inflamm 2016; 2016:2847232. [PMID: 27239100 PMCID: PMC4864531 DOI: 10.1155/2016/2847232] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 04/06/2016] [Indexed: 11/17/2022] Open
Abstract
We have previously shown that immunodominant MBP peptides encapsulated in mannosylated liposomes (Xemys) effectively suppressed experimental allergic encephalomyelitis (EAE). Within the frames of the successfully completed phase I clinical trial, we investigated changes in the serum cytokine profile after Xemys administration in MS patients. We observed a statistically significant decrease of MCP-1/CCL2, MIP-1β/CCL4, IL-7, and IL-2 at the time of study completion. In contrast, the serum levels of TNF-α were remarkably elevated. Our data suggest that the administration of Xemys leads to a normalization of cytokine status in MS patients to values commonly reported for healthy subjects. These data are an important contribution for the upcoming Xemys clinical trials.
Collapse
|
45
|
Functional effects of the antigen glatiramer acetate are complex and tightly associated with its composition. J Neuroimmunol 2016; 290:84-95. [DOI: 10.1016/j.jneuroim.2015.11.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/16/2015] [Accepted: 11/23/2015] [Indexed: 11/23/2022]
|
46
|
Abstract
A rapidly changing set of drugs for treatment of multiple sclerosis (MS) leads to the necessity of searching for predictors of their efficacy. Understanding of pathogenetic processes in MS and mechanisms of action of different drugs play an important role in the search for markers of potential responders. The author analyses the presently accumulated information on the original drug copaxone (glatiramer acetate) including current concepts on the mechanism of action, long-term safety and efficacy. Data on the frequency and significance of adverse effects during treatment with glatiramer acetate as well as on the influence of the drug on pregnancy, postpartum course of MS and development of the infant who received glatiramer acetate prenatally compared to other disease-modifying drugs are presented.
Collapse
Affiliation(s)
- D S Kasatkin
- Department of Nervous Diseases with Medical Genetics and Neurosurgery 'Yaroslavl state medical University', Yaroslavl, Russia
| |
Collapse
|
47
|
D’Alessandro JS, Duffner J, Pradines J, Capila I, Garofalo K, Kaundinya G, Greenberg BM, Kantor D, Ganguly TC. Equivalent Gene Expression Profiles between Glatopa™ and Copaxone®. PLoS One 2015; 10:e0140299. [PMID: 26473741 PMCID: PMC4608686 DOI: 10.1371/journal.pone.0140299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/23/2015] [Indexed: 11/18/2022] Open
Abstract
Glatopa™ is a generic glatiramer acetate recently approved for the treatment of patients with relapsing forms of multiple sclerosis. Gene expression profiling was performed as a means to evaluate equivalence of Glatopa and Copaxone®. Microarray analysis containing 39,429 unique probes across the entire genome was performed in murine glatiramer acetate--responsive Th2-polarized T cells, a test system highly relevant to the biology of glatiramer acetate. A closely related but nonequivalent glatiramoid molecule was used as a control to establish assay sensitivity. Multiple probe-level (Student's t-test) and sample-level (principal component analysis, multidimensional scaling, and hierarchical clustering) statistical analyses were utilized to look for differences in gene expression induced by the test articles. The analyses were conducted across all genes measured, as well as across a subset of genes that were shown to be modulated by Copaxone. The following observations were made across multiple statistical analyses: the expression of numerous genes was significantly changed by treatment with Copaxone when compared against media-only control; gene expression profiles induced by Copaxone and Glatopa were not significantly different; and gene expression profiles induced by Copaxone and the nonequivalent glatiramoid were significantly different, underscoring the sensitivity of the test system and the multiple analysis methods. Comparative analysis was also performed on sets of transcripts relevant to T-cell biology and antigen presentation, among others that are known to be modulated by glatiramer acetate. No statistically significant differences were observed between Copaxone and Glatopa in the expression levels (magnitude and direction) of these glatiramer acetate-regulated genes. In conclusion, multiple methods consistently supported equivalent gene expression profiles between Copaxone and Glatopa.
Collapse
Affiliation(s)
| | - Jay Duffner
- Momenta Pharmaceuticals, Inc., Cambridge, MA, United States of America
| | - Joel Pradines
- Momenta Pharmaceuticals, Inc., Cambridge, MA, United States of America
| | - Ishan Capila
- Momenta Pharmaceuticals, Inc., Cambridge, MA, United States of America
| | - Kevin Garofalo
- Momenta Pharmaceuticals, Inc., Cambridge, MA, United States of America
| | - Ganesh Kaundinya
- Momenta Pharmaceuticals, Inc., Cambridge, MA, United States of America
| | - Benjamin M. Greenberg
- The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Daniel Kantor
- Kantor Neurology, Coconut Creek, FL, United States of America
| | - Tanmoy C. Ganguly
- Momenta Pharmaceuticals, Inc., Cambridge, MA, United States of America
- * E-mail:
| |
Collapse
|
48
|
Telesford K, Ochoa-Repáraz J, Kasper LH. Gut commensalism, cytokines, and central nervous system demyelination. J Interferon Cytokine Res 2015; 34:605-14. [PMID: 25084177 DOI: 10.1089/jir.2013.0134] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
There is increasing support for the importance of risk factors such as genetic makeup, obesity, smoking, vitamin D insufficiency, and antibiotic exposure contributing to the development of autoimmune diseases, including human multiple sclerosis (MS). Perhaps the greatest environmental risk factor associated with the development of immune-mediated conditions is the gut microbiome. Microbial and helminthic agents are active participants in shaping the immune systems of their hosts. This concept is continually reinforced by studies in the burgeoning area of commensal-mediated immunomodulation. The clinical importance of these findings for MS is suggested by both their participation in disease and, perhaps of greater clinical importance, attenuation of disease severity. Observations made in murine models of central nervous system demyelinating disease and a limited number of small studies in human MS suggest that immune homeostasis within the gut microbiome may be of paramount importance in maintaining a disease-free state. This review describes three immunological factors associated with the gut microbiome that are central to cytokine network activities in MS pathogenesis: T helper cell polarization, T regulatory cell function, and B cell activity. Comparisons are drawn between the regulatory mechanisms attributed to first-line therapies and those described in commensal-mediated amelioration of central nervous system demyelination.
Collapse
Affiliation(s)
- Kiel Telesford
- 1 Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth , Lebanon , New Hampshire
| | | | | |
Collapse
|
49
|
Affiliation(s)
- Mark D Willis
- Institute of Psychological Medicine and Clinical Neuroscience, Cardiff University, University Hospital of Wales, Heath Park, Cardiff, UK
| | - Neil P Robertson
- Institute of Psychological Medicine and Clinical Neuroscience, Cardiff University, University Hospital of Wales, Heath Park, Cardiff, UK
| |
Collapse
|
50
|
Abstract
The role of CD8+ T cells in the process of autoimmune pathology has been both understudied and controversial. Multiple sclerosis (MS) is an inflammatory, demyelinating disorder of the central nervous system (CNS) with underlying T cell-mediated immunopathology. CD8+ T cells are the predominant T cells in human MS lesions, showing oligoclonal expansion at the site of pathology. It is still unclear whether these cells represent pathogenic immune responses or disease-regulating elements. Through studies in human MS and its animal model, experimental autoimmune encephalomyelitis (EAE), we have discovered two novel CD8+ T cell populations that play an essential immunoregulatory role in disease: (1) MHC class Ia-restricted neuroantigen-specific "autoregulatory" CD8+ T cells and (2) glatiramer acetate (GA/Copaxone(®)) therapy-induced Qa-1/HLA-E-restricted GA-specific CD8+ T cells. These CD8+ Tregs suppress proliferation of pathogenic CD4+ CD25- T cells when stimulated by their cognate antigens. Similarly, CD8+ Tregs significantly suppress EAE when transferred either pre-disease induction or during peak disease. The mechanism of disease inhibition depends, at least in part, on an antigen-specific, contact-dependent process and works through modulation of CD4+ T cell responses as well as antigen-presenting cells through a combination of cytotoxicity and cytokine-mediated modulation. This review provides an overview of our understanding of CD8+ T cells in immune-mediated disease, focusing particularly on our findings regarding regulatory CD8+ T cells both in MS and in EAE. Clinical relevance of these novel CD8-regulatory populations is discussed, providing insights into a potentially intriguing, novel therapeutic strategy for these diseases.
Collapse
|