1
|
Mohammed EMA. Understanding Multiple Sclerosis Pathophysiology and Current Disease-Modifying Therapies: A Review of Unaddressed Aspects. FRONT BIOSCI-LANDMRK 2024; 29:386. [PMID: 39614433 DOI: 10.31083/j.fbl2911386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 12/01/2024]
Abstract
Multiple sclerosis (MS) is a complex autoimmune disorder of the central nervous system (CNS) with an unknown etiology and pathophysiology that is not completely understood. Although great strides have been made in developing disease-modifying therapies (DMTs) that have significantly improved the quality of life for MS patients, these treatments do not entirely prevent disease progression or relapse. Identifying the unaddressed pathophysiological aspects of MS and developing targeted therapies to fill in these gaps are essential in providing long-term relief for patients. Recent research has uncovered some aspects of MS that remain outside the scope of available DMTs, and as such, yield only limited benefits. Despite most MS pathophysiology being targeted by DMTs, many patients still experience disease progression or relapse, indicating that a more detailed understanding is necessary. Thus, this literature review seeks to explore the known aspects of MS pathophysiology, identify the gaps in present DMTs, and explain why current treatments cannot entirely arrest MS progression.
Collapse
Affiliation(s)
- Eiman M A Mohammed
- Kuwait Cancer Control Centre, Department of Medical Laboratory, Molecular Genetics Laboratory, Ministry of Health, 13001 Shuwaikh, Kuwait
| |
Collapse
|
2
|
In Silico Drug Repurposing in Multiple Sclerosis Using scRNA-Seq Data. Int J Mol Sci 2023; 24:ijms24020985. [PMID: 36674506 PMCID: PMC9864606 DOI: 10.3390/ijms24020985] [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: 12/07/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system still lacking a cure. Treatment typically focuses on slowing the progression and managing MS symptoms. Single-cell transcriptomics allows the investigation of the immune system-the key player in MS onset and development-in great detail increasing our understanding of MS mechanisms and stimulating the discovery of the targets for potential therapies. Still, de novo drug development takes decades; however, this can be reduced by drug repositioning. A promising approach is to select potential drugs based on activated or inhibited genes and pathways. In this study, we explored the public single-cell RNA data from an experiment with six patients on single-cell RNA peripheral blood mononuclear cells (PBMC) and cerebrospinal fluid cells (CSF) of patients with MS and idiopathic intracranial hypertension. We demonstrate that AIM2 inflammasome, SMAD2/3 signaling, and complement activation pathways are activated in MS in different CSF and PBMC immune cells. Using genes from top-activated pathways, we detected several promising small molecules to reverse MS immune cells' transcriptomic signatures, including AG14361, FGIN-1-27, CA-074, ARP 101, Flunisolide, and JAK3 Inhibitor VI. Among these molecules, we also detected an FDA-approved MS drug Mitoxantrone, supporting the reliability of our approach.
Collapse
|
3
|
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: 14] [Impact Index Per Article: 4.7] [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
|
4
|
El-Emam MA, El Achy S, Abdallah DM, El-Abhar HS, Gowayed MA. Does physical exercise improve or deteriorate treatment of multiple sclerosis with mitoxantrone? Experimental autoimmune encephalomyelitis study in rats. BMC Neurosci 2022; 23:11. [PMID: 35247984 PMCID: PMC8897955 DOI: 10.1186/s12868-022-00692-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 02/02/2022] [Indexed: 12/11/2022] Open
Abstract
Background Mitoxantrone has proved efficacy in treatment of multiple sclerosis (MS). The fact that physical exercise could slow down the progression of disease and improve performance is still a debatable issue, hence; we aimed at studying whether combining mitoxantrone with exercise is of value in the management of MS. Methods Thirty-six male rats were divided into sedentary and exercised groups. During a 14-day habituation period rats were subjected to exercise training on a rotarod (30 min/day) before Experimental Autoimmune Encephalomyelitis (EAE) induction and thereafter for 17 consecutive days. On day 13 after induction, EAE groups (exercised &sedentary) were divided into untreated and mitoxantrone treated ones. Disease development was evaluated by motor performance and EAE score. Cerebrospinal fluid (CSF) was used for biochemical analysis. Brain stem and cerebellum were examined histopathological and immunohistochemically. Results Exercise training alone did not add a significant value to the studied parameters, except for reducing Foxp3 immunoreactivity in EAE group and caspase-3 in the mitoxantrone treated group. Unexpectedly, exercise worsened the mitoxantrone effect on EAE score, Bcl2 and Bax. Mitoxantrone alone decreased EAE/demyelination/inflammation scores, Foxp3 immunoreactivity, and interleukin-6, while increased the re-myelination marker BDNF without any change in tumor necrosis factor-α. It clearly interrupted the apoptotic pathway in brain stem, but worsened EAE mediated changes of the anti-apoptotic Bcl2 and pro-apoptotic marker Bax in the CSF. Conclusions The neuroprotective effect of mitoxantrone was related with remyelination, immunosuppressive and anti-inflammatory potentials. Exercise training did not show added value to mitoxantrone, in contrast, it disrupts the apoptotic pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12868-022-00692-1.
Collapse
|
5
|
DiSano KD, Gilli F, Pachner AR. Memory B Cells in Multiple Sclerosis: Emerging Players in Disease Pathogenesis. Front Immunol 2021; 12:676686. [PMID: 34168647 PMCID: PMC8217754 DOI: 10.3389/fimmu.2021.676686] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/11/2021] [Indexed: 11/25/2022] Open
Abstract
Multiple Sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. Once thought to be primarily driven by T cells, B cells are emerging as central players in MS immunopathogenesis. Interest in multiple B cell phenotypes in MS expanded following the efficacy of B cell-depleting agents targeting CD20 in relapsing-remitting MS and inflammatory primary progressive MS patients. Interestingly, these therapies primarily target non-antibody secreting cells. Emerging studies seek to explore B cell functions beyond antibody-mediated roles, including cytokine production, antigen presentation, and ectopic follicle-like aggregate formation. Importantly, memory B cells (Bmem) are rising as a key B cell phenotype to investigate in MS due to their antigen-experience, increased lifespan, and rapid response to stimulation. Bmem display diverse effector functions including cytokine production, antigen presentation, and serving as antigen-experienced precursors to antibody-secreting cells. In this review, we explore the cellular and molecular processes involved in Bmem development, Bmem phenotypes, and effector functions. We then examine how these concepts may be applied to the potential role(s) of Bmem in MS pathogenesis. We investigate Bmem both within the periphery and inside the CNS compartment, focusing on Bmem phenotypes and proposed functions in MS and its animal models. Finally, we review how current immunomodulatory therapies, including B cell-directed therapies and other immunomodulatory therapies, modify Bmem and how this knowledge may be harnessed to direct therapeutic strategies in MS.
Collapse
Affiliation(s)
- Krista D. DiSano
- Department of Neurology, Geisel School of Medicine & Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | | | | |
Collapse
|
6
|
B Cells and Antibodies as Targets of Therapeutic Intervention in Neuromyelitis Optica Spectrum Disorders. Pharmaceuticals (Basel) 2021; 14:ph14010037. [PMID: 33419217 PMCID: PMC7825598 DOI: 10.3390/ph14010037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/02/2021] [Accepted: 01/03/2021] [Indexed: 12/11/2022] Open
Abstract
The first description of neuromyelitis optica by Eugène Devic and Fernand Gault dates back to the 19th century, but only the discovery of aquaporin-4 autoantibodies in a major subset of affected patients in 2004 led to a fundamentally revised disease concept: Neuromyelits optica spectrum disorders (NMOSD) are now considered autoantibody-mediated autoimmune diseases, bringing the pivotal pathogenetic role of B cells and plasma cells into focus. Not long ago, there was no approved medication for this deleterious disease and off-label therapies were the only treatment options for affected patients. Within the last years, there has been a tremendous development of novel therapies with diverse treatment strategies: immunosuppression, B cell depletion, complement factor antagonism and interleukin-6 receptor blockage were shown to be effective and promising therapeutic interventions. This has led to the long-expected official approval of eculizumab in 2019 and inebilizumab in 2020. In this article, we review current pathogenetic concepts in NMOSD with a focus on the role of B cells and autoantibodies as major contributors to the propagation of these diseases. Lastly, by highlighting promising experimental and future treatment options, we aim to round up the current state of knowledge on the therapeutic arsenal in NMOSD.
Collapse
|
7
|
Differential Effects of MS Therapeutics on B Cells-Implications for Their Use and Failure in AQP4-Positive NMOSD Patients. Int J Mol Sci 2020; 21:ijms21145021. [PMID: 32708663 PMCID: PMC7404039 DOI: 10.3390/ijms21145021] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 12/25/2022] Open
Abstract
B cells are considered major contributors to multiple sclerosis (MS) pathophysiology. While lately approved disease-modifying drugs like ocrelizumab deplete B cells directly, most MS medications were not primarily designed to target B cells. Here, we review the current understanding how approved MS medications affect peripheral B lymphocytes in humans. These highly contrasting effects are of substantial importance when considering these drugs as therapy for neuromyelitis optica spectrum disorders (NMOSD), a frequent differential diagnosis to MS, which is considered being a primarily B cell- and antibody-driven diseases. Data indicates that MS medications, which deplete B cells or induce an anti-inflammatory phenotype of the remaining ones, were effective and safe in aquaporin-4 antibody positive NMOSD. In contrast, drugs such as natalizumab and interferon-β, which lead to activation and accumulation of B cells in the peripheral blood, lack efficacy or even induce catastrophic disease activity in NMOSD. Hence, we conclude that the differential effect of MS drugs on B cells is one potential parameter determining the therapeutic efficacy or failure in antibody-dependent diseases like seropositive NMOSD.
Collapse
|
8
|
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: 114] [Impact Index Per Article: 19.0] [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
|
9
|
Napier J, Rose L, Adeoye O, Hooker E, Walsh KB. Modulating acute neuroinflammation in intracerebral hemorrhage: the potential promise of currently approved medications for multiple sclerosis. Immunopharmacol Immunotoxicol 2019; 41:7-15. [PMID: 30702002 DOI: 10.1080/08923973.2019.1566361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The secondary inflammatory injury following intracerebral hemorrhage (ICH) results in increased morbidity and mortality. White blood cells have been implicated as critical mediators of this inflammatory injury. Currently, no medications have been clinically proven to ameliorate or beneficially modulate inflammation, or to improve outcomes by any mechanism, following ICH. However, other neuroinflammatory conditions, such as multiple sclerosis, have approved pharmacologic therapies that modulate the inflammatory response and minimize the damage caused by inflammatory cells. Thus, there is substantial interest in existing therapies for neuroinflammation and their potential applicability to other acute neurological diseases such as ICH. In this review, we examined the mechanism of action of twelve currently approved medications for multiple sclerosis: alemtuzumab, daclizumab, dimethyl fumarate, fingolimod, glatiramer acetate, interferon beta-1a, interferon beta-1b, mitoxantrone, natalizumab, ocrelizumab, rituximab, teriflunomide. We analyzed the existing literature pertaining to the effects of these medications on various leukocytes and also with emphasis on mechanisms of action during the acute period following initiation of therapy. As a result, we provide a valuable summary of the current body of knowledge regarding these therapies and evidence that supports or refutes their likely promise for treating neuroinflammation following ICH.
Collapse
Affiliation(s)
- Jarred Napier
- a College of Medicine , University of Cincinnati , Cincinnati , OH , USA
| | - Lucas Rose
- a College of Medicine , University of Cincinnati , Cincinnati , OH , USA
| | - Opeolu Adeoye
- b Department of Emergency Medicine , University of Cincinnati , Cincinnati , OH , USA.,c Gardner Neuroscience Institute , University of Cincinnati , Cincinnati , OH , USA
| | - Edmond Hooker
- b Department of Emergency Medicine , University of Cincinnati , Cincinnati , OH , USA
| | - Kyle B Walsh
- b Department of Emergency Medicine , University of Cincinnati , Cincinnati , OH , USA.,c Gardner Neuroscience Institute , University of Cincinnati , Cincinnati , OH , USA
| |
Collapse
|
10
|
General Principles of Immunotherapy in Neurological Diseases. CONTEMPORARY CLINICAL NEUROSCIENCE 2019. [DOI: 10.1007/978-3-030-19515-1_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
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: 2.6] [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
|
12
|
Procaccini C, De Rosa V, Pucino V, Formisano L, Matarese G. Animal models of Multiple Sclerosis. Eur J Pharmacol 2015; 759:182-91. [PMID: 25823807 PMCID: PMC7094661 DOI: 10.1016/j.ejphar.2015.03.042] [Citation(s) in RCA: 240] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 01/30/2015] [Accepted: 03/12/2015] [Indexed: 12/26/2022]
Abstract
Multiple Sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) which involves a complex interaction between immune system and neural cells. Animal modeling has been critical for addressing MS pathogenesis. The three most characterized animal models of MS are (1) the experimental autoimmune/allergic encephalomyelitis (EAE); (2) the virally-induced chronic demyelinating disease, known as Theiler׳s murine encephalomyelitis virus (TMEV) infection and (3) the toxin-induced demyelination. All these models, in a complementary way, have allowed to reach a good knowledge of the pathogenesis of MS. Specifically, EAE is the model which better reflects the autoimmune pathogenesis of MS and is extremely useful to study potential experimental treatments. Furthermore, both TMEV and toxin-induced demyelination models are suitable for characterizing the role of the axonal injury/repair and the remyelination process in MS. In conclusion, animal models, despite their limitations, remain the most useful instrument for implementing the study of MS.
Collapse
MESH Headings
- Animals
- Cardiovirus Infections/pathology
- Cardiovirus Infections/virology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Humans
- Mice
- Mice, Transgenic
- Multiple Sclerosis/etiology
- Multiple Sclerosis/genetics
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Theilovirus/pathogenicity
Collapse
Affiliation(s)
- Claudio Procaccini
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR) c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italy
| | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR) c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italy; Unità di NeuroImmunologia, IRCCS Fondazione Santa Lucia, 00143 Roma, Italy
| | - Valentina Pucino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli Federico II, 80131 Napoli, Italy
| | - Luigi Formisano
- Divisione di Farmacologia, Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, 82100 Benevento, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, Baronissi Campus, 84081 Baronissi, Salerno, Italy; IRCCS Multimedica, 20138 Milano, Italy.
| |
Collapse
|
13
|
Role of the immunogenic and tolerogenic subsets of dendritic cells in multiple sclerosis. Mediators Inflamm 2015; 2015:513295. [PMID: 25705093 PMCID: PMC4325219 DOI: 10.1155/2015/513295] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 01/01/2015] [Accepted: 01/01/2015] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated disorder in the central nervous system (CNS) characterized by inflammation and demyelination as well as axonal and neuronal degeneration. So far effective therapies to reverse the disease are still lacking; most therapeutic drugs can only ameliorate the symptoms or reduce the frequency of relapse. Dendritic cells (DCs) are professional antigen presenting cells (APCs) that are key players in both mediating immune responses and inducing immune tolerance. Increasing evidence indicates that DCs contribute to the pathogenesis of MS and might provide an avenue for therapeutic intervention. Here, we summarize the immunogenic and tolerogenic roles of DCs in MS and review medicinal drugs that may affect functions of DCs and have been applied in clinic for MS treatment. We also describe potential therapeutic molecules that can target DCs by inducing anti-inflammatory cytokines and inhibiting proinflammatory cytokines in MS.
Collapse
|
14
|
Tila D, Yazdani-Arazi SN, Ghanbarzadeh S, Arami S, Pourmoazzen Z. pH-sensitive, polymer modified, plasma stable niosomes: promising carriers for anti-cancer drugs. EXCLI JOURNAL 2015; 14:21-32. [PMID: 26417350 PMCID: PMC4553888 DOI: 10.17179/excli2013-609] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 01/07/2014] [Indexed: 12/14/2022]
Abstract
The aim of this study was the design and evaluation of a novel plasma stable, pH-sensitive niosomal formulation of Mitoxantrone by a modified ethanol injection method. Cholesterol hemisuccinate was added instead of cholesterol in order to produce pH-sensitivity property and using PEG-Poly (monomethyl itaconate)-CholC6 (PEG-PMMI-CholC6) copolymer introduced simultaneously pH-sensitivity and plasma stability properties in prepared niosomes. The pH-sensitivity and cytotoxicity of Mitoxantrone niosomes were evaluated in vitro in phosphate buffer with different pHs as well as using human ovarian cancer cell line (OVCAR-3), human breast cancer cell line (MCF-7) and human umbilical vein endothelial cells (HUVEC). Results showed that both cholesterol derivatives bearing formulations had pH-sensitive property and were found to release their contents under mild acidic conditions rapidly. In addition, the PEG-PMMI-CholC6-based niosomes could reserve the pH-sensitivity after incubation in plasma. Both Mitoxantrone-loaded pH-sensitive niosomes showed higher cytotoxicity than the conventional niosomes on OVCAR-3 and MCF-7 cell lines. However, both pH-sensitive niosomes exhibited lower cytotoxic effect on HUVEC cell line. Plasma stable, pH-sensitive niosomes could improve the cytotoxic effect and reduce the side effects of anti-tumor drugs.
Collapse
Affiliation(s)
- Dena Tila
- Tabriz University of Medical Sciences, Tabriz, Iran, Research Center for Pharmaceutical Nanotechnology
| | | | - Saeed Ghanbarzadeh
- Tabriz University of Medical Sciences, Tabriz, Iran, Research Center for Pharmaceutical Nanotechnology ; Tabriz University of Medical Sciences, Tabriz, Iran, Department of Pharmaceutics, Faculty of Pharmacy ; Tabriz University of Medical Sciences, Tabriz, Iran, Student Research Committee, Faculty of Pharmacy
| | - Sanam Arami
- Tabriz University of Medical Sciences, Tabriz, Iran, Research Center for Pharmaceutical Nanotechnology ; Tabriz University of Medical Sciences, Tabriz, Iran, Student Research Committee, Faculty of Pharmacy
| | - Zhaleh Pourmoazzen
- Chemistry Department, Science Faculty, Azarbaijan Shahid Madani University,Tabriz, Iran
| |
Collapse
|
15
|
Grey (née Cotte) S, Salmen (née Stroet) A, von Ahsen N, Starck M, Winkelmann A, Zettl UK, Comabella M, Montalban X, Zipp F, Fleischer V, Kruse N, Gold R, Chan A. Lack of efficacy of mitoxantrone in primary progressive Multiple Sclerosis irrespective of pharmacogenetic factors: A multi-center, retrospective analysis. J Neuroimmunol 2015; 278:277-9. [DOI: 10.1016/j.jneuroim.2014.11.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 11/17/2014] [Accepted: 11/20/2014] [Indexed: 11/15/2022]
|
16
|
Forbes NE, Krishnan R, Diallo JS. Pharmacological modulation of anti-tumor immunity induced by oncolytic viruses. Front Oncol 2014; 4:191. [PMID: 25101247 PMCID: PMC4108035 DOI: 10.3389/fonc.2014.00191] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/07/2014] [Indexed: 01/05/2023] Open
Abstract
Oncolytic viruses (OVs) not only kill cancer cells by direct lysis but also generate a significant anti-tumor immune response that allows for prolonged cancer control and in some cases cures. How to best stimulate this effect is a subject of intense investigation in the OV field. While pharmacological manipulation of the cellular innate anti-viral immune response has been shown by several groups to improve viral oncolysis and spread, it is increasingly clear that pharmacological agents can also impact the anti-tumor immune response generated by OVs and related tumor vaccination strategies. This review covers recent progress in using pharmacological agents to improve the activity of OVs and their ability to generate robust anti-tumor immune responses.
Collapse
Affiliation(s)
- Nicole E Forbes
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, ON , Canada ; Faculty of Medicine, University of Ottawa , Ottawa, ON , Canada
| | - Ramya Krishnan
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, ON , Canada ; Faculty of Medicine, University of Ottawa , Ottawa, ON , Canada
| | - Jean-Simon Diallo
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, ON , Canada ; Faculty of Medicine, University of Ottawa , Ottawa, ON , Canada
| |
Collapse
|
17
|
Cocco E, Marrosu MG. The current role of mitoxantrone in the treatment of multiple sclerosis. Expert Rev Neurother 2014; 14:607-16. [PMID: 24834466 DOI: 10.1586/14737175.2014.915742] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mitoxantrone is an immunosuppressive drug approved for aggressive relapsing and progressive multiple sclerosis. In recent years, its use has decreased due to the risk of severe adverse events and the introduction of novel therapies, such as natalizumab or fingolimod. Mitoxantrone is effective in reducing inflammatory activity by decreasing the number of relapses and MRI lesions and simultaneously decreasing the worsening of disability. Apart from its role as a second/third-line therapy, some studies suggest its use as an induction therapy. However, mitoxantrone use is limited because of its potential risk of severe adverse events, such as cardiotoxicity and the induction of therapy-related acute leukemia. Genetic markers are on evaluation to predict side effects and therapeutic efficacy, which is consistent with the direction of personalized treatment. Considering its efficacy and the potential risks, mitoxantrone use is limited to active patients after a careful, individualized evaluation of the risk/benefit balance.
Collapse
Affiliation(s)
- Eleonora Cocco
- Multiple Sclerosis Center, Department of Public Health, Clinical and molecular medicine, University of Cagliari, Cagliari, Italy
| | | |
Collapse
|
18
|
Okuda DT. Immunosuppressive treatments in multiple sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2014; 122:503-11. [DOI: 10.1016/b978-0-444-52001-2.00022-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
19
|
Rossato LG, Costa VM, Dallegrave E, Arbo M, Dinis-Oliveira RJ, Santos-Silva A, Duarte JA, de Lourdes Bastos M, Palmeira C, Remião F. Cumulative mitoxantrone-induced haematological and hepatic adverse effects in a subchronic in vivo study. Basic Clin Pharmacol Toxicol 2013; 114:254-62. [PMID: 24119282 DOI: 10.1111/bcpt.12143] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 09/10/2013] [Indexed: 01/16/2023]
Abstract
Mitoxantrone (MTX) is an antineoplastic agent that can induce hepato- and haematotoxicity. This work aimed to investigate the occurrence of cumulative early and late MTX-induced hepatic and haematological disturbances in an vivo model. A control group and two groups treated with three cycles of 2.5 mg/kg MTX at days 0, 10 and 20 were formed. One of the treated groups suffered euthanasia on day 22 (MTX22) to evaluate early MTX toxic effects, while the other suffered euthanasia on day 48 (MTX48), to allow the evaluation of MTX late effects. An early immunosuppression with a drop in the IgG levels was observed, causing a slight decrease in the plasma total protein content. The early bone marrow depression was followed by signs of recovery in MTX48. The genotoxic potential of MTX was demonstrated by the presence of several micronuclei in MTX22 leucocytes. Increases in plasma iron and cholesterol levels in the MTX22 rats were observed, while in both groups increases in the unconjugated bilirubin, C4 complement, and decreases in the triglycerides, alanine aminotransferase, alkaline phosphatase and transferrin were found in plasma samples. On MTX 48, the liver histology showed more hepatotoxic signs, the hepatic levels of reduced and oxidized glutathione were increased, and ATP hepatic levels were decreased. However, the hepatic total protein levels were decreased only in the livers of MTX22 group. Results demonstrated the MTX genotoxic effects, haemato- and direct hepatotoxicity. While the haematological toxicity is ameliorated with time, the same was not observed in the hepatic injury.
Collapse
Affiliation(s)
- Luciana G Rossato
- REQUIMTE, Toxicology Laboratory, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Duggal NA, Upton J, Phillips AC, Sapey E, Lord JM. An age-related numerical and functional deficit in CD19(+) CD24(hi) CD38(hi) B cells is associated with an increase in systemic autoimmunity. Aging Cell 2013; 12:873-81. [PMID: 23755918 PMCID: PMC3814412 DOI: 10.1111/acel.12114] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2013] [Indexed: 12/23/2022] Open
Abstract
Autoimmunity increases with aging indicative of reduced immune tolerance, but the mechanisms involved are poorly defined. In recent years, subsets of B cells with immunoregulatory properties have been identified in murine models of autoimmune disorders, and these cells downregulate immune responses via secretion of IL10. In humans, immature transitional B cells with a CD19(+) CD24(hi) CD38(hi) phenotype have been reported to regulate immune responses via IL10 production. We found the frequency and numbers of CD19(+) CD24(hi) CD38(hi) cells were reduced in the PBMC pool with age. IL10 expression and secretion following activation via either CD40, or Toll-like receptors was also impaired in CD19(+) CD24(hi) CD38(hi) B cells from healthy older donors. When investigating the mechanisms involved, we found that CD19(+) CD24(hi) CD38(hi) B-cell function was compromised by age-related effects on both T cells and B cells: specifically, CD40 ligand expression was lower in CD4 T cells from older donors following CD3 stimulation, and signalling through CD40 was impaired in CD19(+) CD24(hi) CD38(hi) B cells from elders as evidenced by reduced phosphorylation (Y705) and activation of STAT3. However, there was no age-associated change in expression of costimulatory molecules CD80 and CD86 on CD19(+) CD24(hi) CD38(hi) cells, suggesting IL10-dependent immune suppression is impaired, but contact-dependent suppressive capacity is intact with age. Finally, we found a negative correlation between CD19(+) CD24(hi) CD38(hi) B-cell IL10 production and autoantibody (Rheumatoid factor) levels in older adults. We therefore propose that an age-related decline in CD19(+) CD24(hi) CD38(hi) B cell number and function may contribute towards the increased autoimmunity and reduced immune tolerance seen with aging.
Collapse
Affiliation(s)
- Niharika A. Duggal
- MRC-ARUK Centre for Musculoskeletal Ageing Research; School of Immunity and Infection; Birmingham University Medical School; Birmingham; B15 2TT; UK
| | - Jane Upton
- School of Sport and Exercise Sciences; Birmingham University Medical School; Birmingham; B15 2TT; UK
| | - Anna C. Phillips
- School of Sport and Exercise Sciences; Birmingham University Medical School; Birmingham; B15 2TT; UK
| | - Elizabeth Sapey
- School of Clinical and Experimental Medicine; Birmingham University Medical School; Birmingham; B15 2TT; UK
| | - Janet M. Lord
- MRC-ARUK Centre for Musculoskeletal Ageing Research; School of Immunity and Infection; Birmingham University Medical School; Birmingham; B15 2TT; UK
| |
Collapse
|
21
|
Jamroz-Wisniewska A, Beltowski J, Stelmasiak Z, Bartosik-Psujek H. Paraoxonase 1 activity in multiple sclerosis patients during mitoxantrone therapy. Acta Neurol Scand 2013; 127:e33-6. [PMID: 22881373 DOI: 10.1111/ane.12000] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2012] [Indexed: 11/30/2022]
Abstract
OBJECTIVES It has been implicated in many studies that reactive oxygen species play a role in the development of demyelination in multiple sclerosis (MS). Paraoxonase 1 (PON1) is an antioxidant enzyme that protects cell membranes against oxidative modification. Mitoxantrone is a cytotoxic drug approved for the treatment of MS with adverse effects associated potentially with an increased level of oxidative stress. The aim of this study was to assess the influence of mitoxantrone therapy on PON1 activity in patients with MS. METHODS A studied group included 26 patients with secondary progressive MS, 16 women and 10 men. The blood was collected before the beginning of the therapy as well as after 6 and 12 months. Patients were receiving mitoxantrone every 12 weeks. Serum PON1 activity was assayed using two synthetic substrates: paraoxon and phenyl acetate. RESULTS Paraoxonase 1 activity toward paraoxon and phenyl acetate and lipid profile did not change significantly in patients receiving mitoxantrone. CONCLUSIONS Mitoxantrone therapy does not influence PON1 activity.
Collapse
Affiliation(s)
| | - J. Beltowski
- Department of Pathophysiology; Medical University of Lublin; Jaczewskiego; Poland
| | - Z. Stelmasiak
- Department of Neurology; Medical University of Lublin; Jaczewskiego; Poland
| | - H. Bartosik-Psujek
- Department of Neurology; Medical University of Lublin; Jaczewskiego; Poland
| |
Collapse
|
22
|
Mitoxantrone induces natural killer cell maturation in patients with secondary progressive multiple sclerosis. PLoS One 2012; 7:e39625. [PMID: 22768101 PMCID: PMC3387260 DOI: 10.1371/journal.pone.0039625] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 05/29/2012] [Indexed: 12/01/2022] Open
Abstract
Mitoxantrone is one of the few drugs approved for the treatment of progressive multiple sclerosis (MS). However, the prolonged use of this potent immunosuppressive agent is limited by the appearance of severe side effects. Apart from its general cytotoxic effect, the mode of action of mitoxantrone on the immune system is poorly understood. Thus, to develop safe therapeutic approaches for patients with progressive MS, it is essential to elucidate how mitoxantrone exerts it benefits. Accordingly, we initiated a prospective single-arm open-label study with 19 secondary progressive MS patients. We investigated long-term effects of mitoxantrone on patient peripheral immune subsets using flow cytometry. While we corroborate that mitoxantrone persistently suppresses B cells in vivo, we show for the first time that treatment led to an enrichment of neutrophils and immunomodulatory CD8low T cells. Moreover, sustained mitoxantrone applications promoted not only persistent NK cell enrichment but also NK cell maturation. Importantly, this mitoxantrone-induced NK cell maturation was seen only in patients that showed a clinical response to treatment. Our data emphasize the complex immunomodulatory role of mitoxantrone, which may account for its benefit in MS. In particular, these results highlight the contribution of NK cells to mitoxantrone efficacy in progressive MS.
Collapse
|
23
|
Dobson R, Meier UC, Giovannoni G. More to come: humoral immune responses in MS. J Neuroimmunol 2011; 240-241:13-21. [PMID: 22019113 DOI: 10.1016/j.jneuroim.2011.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 09/15/2011] [Accepted: 09/20/2011] [Indexed: 01/14/2023]
Abstract
Interest in the role of B-cells in multiple sclerosis (MS) pathogenesis has increased, and a number of B-cell targeted therapies are currently in clinical trials. B-cells are key mediators of the humoral immune response, with roles including antibody production and acting as antigen presenting cells. Whilst previously, the presence of B-cells within MS plaques has been thought to be secondary to T-cell dysregulation, it is now becoming clear that B-cells play an independent role in disease. In this review we will discuss the potential role of B-cells in MS, how this influences our understanding of the disease, and potential therapeutic implications.
Collapse
Affiliation(s)
- Ruth Dobson
- Centre for Neuroscience and Trauma, Blizard Institute of Cell and Molecular Science, London E1 2AT, UK.
| | | | | |
Collapse
|
24
|
Eckstein C, Saidha S, Levy M. A differential diagnosis of central nervous system demyelination: beyond multiple sclerosis. J Neurol 2011; 259:801-16. [DOI: 10.1007/s00415-011-6240-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 08/28/2011] [Accepted: 08/30/2011] [Indexed: 12/12/2022]
|
25
|
Li JM, Yang Y, Zhu P, Zheng F, Gong FL, Mei YW. Mitoxantrone exerts both cytotoxic and immunoregulatory effects on activated microglial cells. Immunopharmacol Immunotoxicol 2011; 34:36-41. [DOI: 10.3109/08923973.2011.572890] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
26
|
Duddy M, Haghikia A, Cocco E, Eggers C, Drulovic J, Carmona O, Zéphir H, Gold R. Managing MS in a changing treatment landscape. J Neurol 2011; 258:728-39. [DOI: 10.1007/s00415-011-6009-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 03/04/2011] [Accepted: 03/10/2011] [Indexed: 01/19/2023]
|
27
|
Abstract
Vitamin D is a potent immune modulator, keeping the T-cell compartment in a more tolerogenic state. Multiple sclerosis (MS), a disease in which an autoreactive T-cell response contributes to inflammation in the central nervous system, has been associated with vitamin D deficiency. The effects of vitamin D on the immune system are believed to be an important driver of this association. In this chapter, we elaborate on vitamin D as a modulator of the T-cell response. This discussion will be placed in the perspective of MS as a T-cell-mediated disease and in the perspective of the numerous association studies on vitamin D deficiency and multiple health outcomes. We conclude that there is a firm experimental and epidemiological basis supporting the model of vitamin D as a physiological immune modulator, on which intervention studies assessing clinical and immunological outcome measures should be designed.
Collapse
Affiliation(s)
- Joost Smolders
- School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | | |
Collapse
|
28
|
Mix E, Meyer-Rienecker H, Hartung HP, Zettl UK. Animal models of multiple sclerosis--potentials and limitations. Prog Neurobiol 2010; 92:386-404. [PMID: 20558237 PMCID: PMC7117060 DOI: 10.1016/j.pneurobio.2010.06.005] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 06/01/2010] [Accepted: 06/07/2010] [Indexed: 12/17/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is still the most widely accepted animal model of multiple sclerosis (MS). Different types of EAE have been developed in order to investigate pathogenetic, clinical and therapeutic aspects of the heterogenic human disease. Generally, investigations in EAE are more suitable for the analysis of immunogenetic elements (major histocompatibility complex restriction and candidate risk genes) and for the study of histopathological features (inflammation, demyelination and degeneration) of the disease than for screening of new treatments. Recent studies in new EAE models, especially in transgenic ones, have in connection with new analytical techniques such as microarray assays provided a deeper insight into the pathogenic cellular and molecular mechanisms of EAE and potentially of MS. For example, it was possible to better delineate the role of soluble pro-inflammatory (tumor necrosis factor-α, interferon-γ and interleukins 1, 12 and 23), anti-inflammatory (transforming growth factor-β and interleukins 4, 10, 27 and 35) and neurotrophic factors (ciliary neurotrophic factor and brain-derived neurotrophic factor). Also, the regulatory and effector functions of distinct immune cell subpopulations such as CD4+ Th1, Th2, Th3 and Th17 cells, CD4+FoxP3+ Treg cells, CD8+ Tc1 and Tc2, B cells and γδ+ T cells have been disclosed in more detail. The new insights may help to identify novel targets for the treatment of MS. However, translation of the experimental results into the clinical practice requires prudence and great caution.
Collapse
Key Words
- apc, antigen-presenting cell
- at-eae, adoptive transfer eae
- bbb, blood–brain barrier
- bdnf, brain-derived neurotrophic factor
- cd, cluster of differentiation
- cns, central nervous system
- cntf, ciliary neurotrophic factor
- eae, experimental autoimmune encephalomyelitis
- hla, human leukocyte antigen
- ig, immunoglobulin
- il, interleukin
- ifn, interferon
- ivig, intravenous immunoglobulin
- mab, monoclonal antibody
- mbp, myelin basic protein
- mhc, major histocompatibility complex
- mog, myelin oligodendrocyte glycoprotein
- mp, methylprednisolone
- mri, magnetic resonance imaging
- ms, multiple sclerosis
- nk, natural killer
- odc, oligodendrocyte
- qtl, quantitative trait locus
- plp, proteolipid protein
- tc, cytotoxic t cell
- tcr, t cell receptor
- tgf, transforming growth factor
- th cell, helper t cell
- tnf, tumor necrosis factor
- animal model
- autoimmunity
- experimental autoimmune encephalomyelitis
- immunogenetics
- immunomodulatory therapy
- multiple sclerosis
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Clinical Trials as Topic
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Gene Expression Profiling
- History, 19th Century
- History, 20th Century
- History, 21st Century
- Humans
- Microarray Analysis
- Multiple Sclerosis/genetics
- Multiple Sclerosis/immunology
- Multiple Sclerosis/physiopathology
- Multiple Sclerosis/therapy
Collapse
Affiliation(s)
- Eilhard Mix
- Department of Neurology, University of Rostock, Germany
| | | | - Hans-Peter Hartung
- Department of Neurology, Heinrich-Heine-University, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Uwe K. Zettl
- Department of Neurology, University of Rostock, Germany
| |
Collapse
|
29
|
Vogelgesang A, Rosenberg S, Skrzipek S, Bröker BM, Dressel A. Mitoxantrone treatment in multiple sclerosis induces TH2-type cytokines. Acta Neurol Scand 2010; 122:237-43. [PMID: 19925529 DOI: 10.1111/j.1600-0404.2009.01295.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Mitoxantrone is a cytotoxic drug with immune modulatory properties used in the treatment of progressive forms of multiple sclerosis (MS). We explored the effect of mitoxantrone treatment in MS patients on cytokine patterns induced in peripheral blood mononuclear cells (PBMC) and T-cell subsets ex vivo. MATERIALS AND METHODS Blood was obtained before mitoxantrone infusion and 6, 12 and 18 days thereafter. Proliferation and prototypic TH1-, TH17- and TH2-type cytokines were determined following in vitro stimulation of PBMC, CD4+ and CD8+ T cells. In addition, a patient cohort receiving its first mitoxantrone treatment was cross-sectionally compared with a cohort of patients with more than 1 year of treatment. RESULTS Mitoxantrone treatment increased the ex vivo production of the TH2 cytokines interleukin-4 (IL-4; P < 0.05) and IL-5 (P < 0.001) in phytohemagglutinin-stimulated CD4+ T cells within 18 days of treatment. The cross-sectional study revealed that long-term treatment with mitoxantrone increased the inducibility of IL-4 and IL-5 secretion by PBMCs and CD4+ T cells even further. No significant changes were observed for interferon-γ, tumour necrosis factor-α, IL-17 and IL-10. Mitoxantrone did not alter the proliferative capacity of ex vivo-stimulated T cells. CONCLUSION Mitoxantrone treatment in MS enhances the inducibility of TH2-type cytokines, which may contribute to its beneficial effects in MS.
Collapse
Affiliation(s)
- A Vogelgesang
- Department of Neurology, University of Greifswald, Greifswald, Germany
| | | | | | | | | |
Collapse
|
30
|
Abstract
The likely pathogenic mechanisms of multiple sclerosis (MS) provide a sound rationale for investigating the efficacy of drugs possessing immunosuppressive or immunomodulatory properties. With proven efficacy, safety and tolerability, interferon beta formulations and glatiramer acetate have become the mainstay of initial treatment for patients with relapsing forms of MS. More recently, natalizumab, a humanized monoclonal antibody (mAb) against the cellular adhesion molecule α4-integrin, has been employed for patients with an inadequate response or lack of tolerability to an alternate MS therapy, or as initial therapy for patients with severe disease. Various agents initially developed for oncological indications, either as chemotherapeutics or mAbs, may also have current or future uses in MS treatment. Mitoxantrone is currently the only chemotherapeutic agent approved for treatment of MS in the United States, while in parts of Europe azathioprine is approved and widely used for MS treatment. Other chemotherapeutics that have been tested in MS to date include cyclophosphamide, methotrexate, cladribine, and the mAbs alemtuzumab and rituximab. While there has been varying evidence of efficacy for these compounds, each appears to be associated with serious risks that require careful consideration and management. Given the risks that have been demonstrated for available chemotherapeutic agents and while long-term postmarketing safety data are still not available for those agents in development, it seems prudent to carefully assess the possible use of chemotherapeutics in the treatment of MS. A thorough risk-benefit analysis is becoming increasingly important in the assessment of therapeutic options for this disabling disease.
Collapse
Affiliation(s)
- Bernd C. Kieseier
- Department of Neurology, Heinrich-Heine University, Moorenstrasse 5, 40225 Duesseldorf, Germany
| |
Collapse
|
31
|
Bar-Or A, Oger J, Gibbs E, Niino M, Aziz T, Renoux C, Alatab S, Shi FD, Campagnolo D, Jalili F, Rhodes S, Yamashita T, Fan B, Freedman MS, Panitch H, Arnold DL, Vollmer T. Serial combination therapy: is immune modulation in multiple sclerosis enhanced by initial immune suppression? Mult Scler 2009; 15:959-64. [DOI: 10.1177/1352458509106230] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background Although the concept that an initial course of immune-suppression facilitates subsequent immune-modulation (such as Th1 to Th2 deviation) is attractive for several autoimmune diseases, such a mechanism for serial-combination therapy has never been formally demonstrated. Recently, brief mitoxantrone induction-chemotherapy followed by immune-modulation with glatiramer acetate (GA) was significantly more effective at reducing multiple sclerosis disease activity than with GA alone. Objective To examine whether the benefit of initial immune suppression with mitoxantrone before GA treatment is associated with more efficient immune modulation. Methods IgG1/IgG4 GA-reactive antibody profiles, previously established as markers of GA-induced Th2 immune-deviation, were prospectively measured in vivo in patients treated with GA alone or with mitoxantrone induction therapy followed by GA. Results Significant and sustained increase in IgG4 antibodies (and the anticipated reversal of the IgG1/IgG4 ratio) was seen in patients treated with GA alone. Combination therapy resulted in lesser IgG4 induction (and no reversal of IgG1/IgG4 ratio). Thus, the enhanced efficacy of mitoxantrone–GA combination regimen was associated with decreased, rather than increased, efficiency of shifting the GA-reactive IgG1/IgG4 antibody profile. Conclusion These results provide important insights into mechanisms of combination therapy and therapeutic strategies for autoimmune diseases.
Collapse
Affiliation(s)
- A Bar-Or
- Montreal Neurological Institute Montreal, Quebec, Canada
| | - J Oger
- Division of Neurology, Department of Medicine, Multiple Sclerosis Clinic and Brain Research Centre, The University of British Columbia, Vancouver, British Columbia, Canada
| | - E Gibbs
- Division of Neurology, Department of Medicine, Multiple Sclerosis Clinic and Brain Research Centre, The University of British Columbia, Vancouver, British Columbia, Canada
| | - M Niino
- Montreal Neurological Institute Montreal, Quebec, Canada
| | - T Aziz
- Division of Neurology, Department of Medicine, Multiple Sclerosis Clinic and Brain Research Centre, The University of British Columbia, Vancouver, British Columbia, Canada
| | - C Renoux
- Montreal Neurological Institute Montreal, Quebec, Canada
| | - S Alatab
- Montreal Neurological Institute Montreal, Quebec, Canada
| | - FD Shi
- Barrow Neurology Institute of St Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - D Campagnolo
- Barrow Neurology Institute of St Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - F Jalili
- Montreal Neurological Institute Montreal, Quebec, Canada
| | - S Rhodes
- Barrow Neurology Institute of St Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - T Yamashita
- Montreal Neurological Institute Montreal, Quebec, Canada
| | - B Fan
- Montreal Neurological Institute Montreal, Quebec, Canada
| | - MS Freedman
- Multiple Sclerosis Research Unit, The Ottawa Hospital General Campus, University of Ottawa, Ottawa, Canada
| | - H Panitch
- Department of Neurology, Multiple Sclerosis Center, University of Vermont, College of Medical, Burlington, Vermont, USA
| | - DL Arnold
- Montreal Neurological Institute Montreal, Quebec, Canada
| | - T Vollmer
- Department of Neurology, Rocky Mountain MS Center, Anschutz Medical Center, University of Colorado, Denver, Colorado, USA
| |
Collapse
|
32
|
Chan A, Stüve O, von Ahsen N. Immunosuppression in clinical practice: approaches to individualized therapy. J Neurol 2009; 255 Suppl 6:22-7. [PMID: 19300956 DOI: 10.1007/s00415-008-6005-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite novel immunoactive agents, immunosuppressants still play a considerable role in the treatment of MS, especially in rapidly progressive cases. Given the limited tolerability and potentially severe side effects of most immunosuppressive drugs, identification of patients with a favorable benefit-risk profile is essential. A narrow therapeutic index, with sometimes high interindividual variability in terms of response and side effects may partially be explained by genetic factors affecting different metabolic pathways. Here, we will review practical aspects in the clinical use of immunosuppressants in MS and discuss approaches to individualized treatment schemes, including novel pharmacogenetic strategies.
Collapse
Affiliation(s)
- Andrew Chan
- Department of Neurology, Ruhr University Bochum, St. Josef Hospital, Gudrunstr. 56, 44791 Bochum, Germany.
| | | | | |
Collapse
|
33
|
Menge T, Weber MS, Hemmer B, Kieseier BC, von Büdingen HC, Warnke C, Zamvil SS, Boster A, Khan O, Hartung HP, Stüve O. Disease-modifying agents for multiple sclerosis: recent advances and future prospects. Drugs 2009; 68:2445-68. [PMID: 19016573 DOI: 10.2165/0003495-200868170-00004] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the CNS. Currently, six medications are approved for immunmodulatory and immunosuppressive treatment of the relapsing disease course and secondary-progressive MS. In the first part of this review, the pathogenesis of MS and its current treatment options are discussed. During the last decade, our understanding of autoimmunity and the pathogenesis of MS has advanced substantially. This has led to the development of a number of compounds, several of which are currently undergoing clinical testing in phase II and III studies. While current treatment options are only available for parenteral administration, several oral compounds are now in clinical trials, including the immunosuppressive agents cladribine and laquinimod. A novel mode of action has been described for fingolimod, another orally available agent, which inhibits egress of activated lymphocytes from draining lymph nodes. Dimethylfumarate exhibits immunomodulatory as well as immunosuppressive activity when given orally. All of these compounds have successfully shown efficacy, at least in regards to the surrogate marker contrast-enhancing lesions on magnetic resonance imaging. Another class of agents that is highlighted in this review are biological agents, namely monoclonal antibodies (mAb) and recombinant fusion proteins. The humanized mAb daclizumab inhibits T-lymphocyte activation via blockade of the interleukin-2 receptor. Alemtuzumab and rituximab deplete leukocytes and B cells, respectively; the fusion protein atacicept inhibits specific B-cell growth factors resulting in reductions in B-cells and plasma cells. These compounds are currently being tested in phase II and III studies in patients with relapsing MS. The concept of neuro-protection and -regeneration has not advanced to a level where specific compounds have entered clinical testing. However, several agents approved for conditions other than MS are highlighted. Finally, with the advent of these highly potent novel therapies, rare, but potentially serious adverse effects have been noted, namely infections and malignancies. These are critically reviewed and put into perspective.
Collapse
Affiliation(s)
- Til Menge
- Department of Neurology, Heinrich Heine-University, Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Piao WH, Wong R, Bai XF, Huang J, Campagnolo DI, Dorr RT, Vollmer TL, Shi FD. Therapeutic Effect of Anthracene-Based Anticancer Agent Ethonafide in an Animal Model of Multiple Sclerosis. THE JOURNAL OF IMMUNOLOGY 2007; 179:7415-23. [DOI: 10.4049/jimmunol.179.11.7415] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
36
|
Hemmer B, Hartung HP. Toward the development of rational therapies in multiple sclerosis: what is on the horizon? Ann Neurol 2007; 62:314-26. [DOI: 10.1002/ana.21289] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
37
|
Treatment of Multiple Sclerosis with Methylprednisolone and Mitoxantrone Modulates the Expression of CXC Chemokine Receptors in PBMC. J Clin Immunol 2007; 28:122-30. [DOI: 10.1007/s10875-007-9142-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Accepted: 10/02/2007] [Indexed: 01/21/2023]
|
38
|
Abstract
Immunosuppressive agents have been used in multiple sclerosis (MS) for decades. The approval of several immunomodulatory agents against MS beginning in the 1990s, whose putative mechanisms of action appeared "more MS-specific," curtailed the importance of immunosuppressants, which made them treatment options of second choice. However, with the recent approval of mitoxantrone for treatment of patients with active forms of relapsing-remitting or secondary progressive MS and with a number of oral immunosuppressive agents being assessed in phase II and III clinical trials, a "renaissance" of this type of agents is currently occurring. This review provides an outline of the most important clinical studies and discusses relevant side effects of the major immunosuppressants (i.e., mitoxantrone, azathioprine, cyclophosphamide, methotrexate, mycophenolate mofetil, cladribine, and sirolimus/temsirolimus). The current knowledge of the putative mechanisms of action of these compounds is discussed.
Collapse
Affiliation(s)
- Oliver Neuhaus
- Department of Neurology, Kliniken Landkreis Sigmaringen GmbH, D-72488 Sigmaringen, Germany
| | - Bernd C. Kieseier
- Department of Neurology, Heinrich Heine University, Moorenstraβe 5, D-40225 Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Heinrich Heine University, Moorenstraβe 5, D-40225 Düsseldorf, Germany
| |
Collapse
|
39
|
Neuhaus O, Kieseier BC, Hartung HP. Pharmacokinetics and pharmacodynamics of the interferon-betas, glatiramer acetate, and mitoxantrone in multiple sclerosis. J Neurol Sci 2007; 259:27-37. [PMID: 17391705 DOI: 10.1016/j.jns.2006.05.071] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Revised: 04/25/2006] [Accepted: 05/01/2006] [Indexed: 11/15/2022]
Abstract
Five disease-modifying agents are currently approved for long-term treatment of multiple sclerosis (MS), namely three interferon-beta preparations, glatiramer acetate, and mitoxantrone(1). Pharmacokinetics describes the fate of drugs in the human body by studying their absorption, distribution, metabolism and excretion. Pharmacodynamics is dedicated to the mechanisms of action of drugs. The understanding of the pharmacokinetics and pharmacodynamics of the approved disease-modifying agents against MS is of importance as it might contribute to the development of future derivatives with a potentially higher efficacy and a more favourable safety profile. This article reviews data thus far present both on the pharmacokinetics as well as on the putative mechanisms of action of the interferon-betas, glatiramer acetate, and mitoxantrone in the immunopathogenesis of MS.
Collapse
Affiliation(s)
- Oliver Neuhaus
- Department of Neurology, Heinrich Heine University, Düsseldorf, Germany.
| | | | | |
Collapse
|
40
|
Duddy M, Niino M, Adatia F, Hebert S, Freedman M, Atkins H, Kim HJ, Bar-Or A. Distinct effector cytokine profiles of memory and naive human B cell subsets and implication in multiple sclerosis. THE JOURNAL OF IMMUNOLOGY 2007; 178:6092-9. [PMID: 17475834 DOI: 10.4049/jimmunol.178.10.6092] [Citation(s) in RCA: 502] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although recent animal studies have fuelled growing interest in Ab-independent functions of B cells, relatively little is known about how human B cells and their subsets may contribute to the regulation of immune responses in either health or disease. In this study, we first confirm that effector cytokine production by normal human B cells is context dependent and demonstrate that this involves the reciprocal regulation of proinflammatory and anti-inflammatory cytokines. We further report that this cytokine network is dysregulated in patients with the autoimmune disease multiple sclerosis, whose B cells exhibit a decreased average production of the down-regulatory cytokine IL-10. Treatment with the approved chemotherapeutic agent mitoxantrone reciprocally modulated B cell proinflammatory and anti-inflammatory cytokines, establishing that the B cell cytokine network can be targeted in vivo. Prospective studies of human B cells reconstituting following in vivo depletion suggested that different B cell subsets produced distinct effector cytokines. We confirmed in normal human B cell subsets that IL-10 is produced almost exclusively by naive B cells while the proinflammatory cytokines lymphotoxin and TNF-alpha are largely produced by memory B cells. These results point to an in vivo switch in the cytokine "program" of human B cells transitioning from the naive pool to the memory pool. We propose a model that ascribes distinct and proactive roles to memory and naive human B cell subsets in the regulation of memory immune responses and in autoimmunity. Our findings are of particular relevance at a time when B cell directed therapies are being applied to clinical trials of several autoimmune diseases.
Collapse
Affiliation(s)
- Martin Duddy
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, and Department of Neurology, Ottawa General Hospital, Canada
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Gonsette RE. Compared benefit of approved and experimental immunosuppressive therapeutic approaches in multiple sclerosis. Expert Opin Pharmacother 2007; 8:1103-16. [PMID: 17516874 DOI: 10.1517/14656566.8.8.1103] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
An important amount has been learnt about the mechanisms of action, efficacy and long-term toxicities of mitoxantrone. Importantly, recent observations strongly suggest that early administration of potent immunosuppressants (mitoxantrone and alemtuzumab) is definitely more effective than approved immunomodulators to delay or even reverse disability progression. Given the cardiotoxicity of mitoxantrone, restricting exposure to the drug to 2 or 3 years, the benefits and risks of immunosuppressants previously used as off-label treatments (cyclophosphamide and cladribine) have been revisited, and the potential efficacy in multiple sclerosis of recent immunosuppressants used in other autoimmune diseases, organ transplantation and cancer therapy has received increasing attention. Those immunosuppressants comprise monoclonal antibodies targeting B cells, lymphocytes and monocytes, IL-2 receptor and alpha4 integrin, as well as new molecules (pixantrone and isoxazole derivatives) and a new generation of immunosuppressants (fingolimod), which modulate lymphocyte re-circulation. This review addresses the most recent data concerning the efficacy and safety of mitoxantrone and of new experimental therapies that are presently in progress.
Collapse
|
42
|
Debouverie M, Taillandier L, Pittion-Vouyovitch S, Louis S, Vespignani H. Clinical follow-up of 304 patients with multiple sclerosis three years after mitoxantrone treatment. Mult Scler 2007; 13:626-31. [PMID: 17548442 DOI: 10.1177/1352458506072543] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The objectives of this study were to assess the benefits of 1) mitoxantrone after three years of follow-up and 2) disease-modifying treatment (DMT) after stopping mitoxantrone. A retrospective analysis was performed on 304 patients with active relapsing-remitting (RR) or progressive multiple sclerosis (PMS) who were treated with mitoxantrone. After mitoxantrone therapy, some patients received DMT (interferon-beta or glatiramer acetate) while others did not. The disease course of the two groups was evaluated by the Expanded Disability Status Scale (EDSS) before and after mitoxantrone and then every year for three years. The mean EDSS score at starting mitoxantrone and three years after stopping mitoxantrone respectively, were: 3.3 (1.3) and 3.2 (1.7) for the RRMS patients and 5.9 (1.2) and 6.4 (1.4) for the PMS patients. Before starting mitoxantrone, demographic and clinical parameters of predictive disability were not significantly different between patients who received DMT or not. The variation of EDSS between time of stopping mitoxantrone and three years later was significantly different (+0.9 versus +0.3; P=0.03) for patients with RRMS. We found that mitoxantrone treatment induces stable disease up to two years after discontinuation of mitoxantrone therapy. In the third year, patients without DMT deteriorated.
Collapse
Affiliation(s)
- M Debouverie
- Department of Neurology, Central Hospital, 54000 Nancy, France.
| | | | | | | | | |
Collapse
|
43
|
Zenovich AG, Davis BH, Taylor DA. Comparison of intracardiac cell transplantation: autologous skeletal myoblasts versus bone marrow cells. Handb Exp Pharmacol 2007:117-65. [PMID: 17554507 DOI: 10.1007/978-3-540-68976-8_6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
An increasing number of patients living with cardiovascular disease (CVD) and still unacceptably high mortality created an urgent need to effectively treat and prevent disease-related events. Within the past 5 years, skeletal myoblasts (SKMBs) and bone marrow (or blood)-derived mononuclear cells (BMNCs) have demonstrated preclinical efficacy in reducing ischemia and salvaging already injured myocardium, and in preventing left ventricular (LV) remodeling, respectively. These findings have been translated into clinical trials, so far totaling over 200 patients for SKMBs and over 800 patients for BMNCs. These safety/feasibility and early phase II studies showed promising but somewhat conflicting symptomatic and functional improvements, and some safety concerns have arisen. However, the patient population, cell type, dose, time and mode of delivery, and outcome measures differed, making comparisons problematic. In addition, the mechanisms through which cells engraft and deliver their beneficial effects remain to be fully elucidated. It is now time to critically evaluate progress made and challenges encountered in order to select not only the most suitable cells for cardiac repair but also to define appropriate patient populations and outcome measures. Reiterations between bench and bedside will increase the likelihood of cell therapy success, reduce the time to development of combined of drug- and cell-based disease management algorithms, and offer these therapies to patients to achieve a greater reduction of symptoms and allow for a sustained improvement of quality of life.
Collapse
Affiliation(s)
- A G Zenovich
- Center for Cardiovascular Repair, 312 Church Street SE, NHH 7-105A, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
44
|
Antel J, Bar-Or A. Roles of immunoglobulins and B cells in multiple sclerosis: from pathogenesis to treatment. J Neuroimmunol 2006; 180:3-8. [PMID: 16934338 DOI: 10.1016/j.jneuroim.2006.06.032] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2006] [Accepted: 06/23/2006] [Indexed: 12/23/2022]
Abstract
Immunoglobulins (Igs) have long been implicated in contributing to the disease course of multiple sclerosis (MS). The earliest and perhaps still most consistent abnormal immunologic laboratory finding in MS is the increased concentration of Ig in the CSF, representing intrathecal antibody synthesis. Analysis of CSF Ig in terms of rate of production and restricted diversity (oligoclonal bands) remains a supportive diagnostic criteria for MS. Despite large-scale studies such as the analysis of 1000 cases reported by Ebers and Paty [Ebers, G.C., Paty, D.W., 1980. CSF electrophoresis in one thousand patients. Can. J. Neurol. Sci. 7 (4) 275-280], the challenge of correlating CSF Ig profiles and specific disease phenotypes remains. More recently, evidence from animal models and several human studies suggests that antibody-independent functions of B cells may also be implicated in the pathogenesis of MS. This presentation considers what roles Ig and/or B cells can play in mediating or regulating disease-relevant immune responses in MS. A timely corollary is whether B cell/Ig-directed therapeutic strategies can be effective in MS.
Collapse
Affiliation(s)
- Jack Antel
- Neuroimmunology Unit, Montreal Neurological Institute and McGill University, 3801 University Street, Room # 111, Montreal, Quebec, Canada H3A2B4
| | | |
Collapse
|
45
|
Pelfrey CM, Cotleur AC, Zamor N, Lee JC, Fox RJ. Immunological studies of mitoxantrone in primary progressive MS. J Neuroimmunol 2006; 175:192-9. [PMID: 16644023 DOI: 10.1016/j.jneuroim.2006.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2005] [Revised: 03/02/2006] [Accepted: 03/08/2006] [Indexed: 11/20/2022]
Abstract
Mitoxantrone (MX) has demonstrated efficacy in multiple sclerosis (MS), but its immunologic mechanisms of action are poorly understood. Furthermore, no study has examined the immunological effects of MX in primary progressive MS (PPMS). This study investigated the immunological effects of MX therapy in PPMS patients. Lymphocyte phenotypes and chemokine receptor (CCR) expression were evaluated by flow cytometry on fresh PBMC from 20 PPMS patients enrolled in a placebo (PLC)-controlled trial of MX. Longitudinal data were collected at weeks 0, 12, 24, 36 and short-term data (pre-/post-infusion) were collected at weeks 0 and 36. CXCR3, CCR1, CCR2 and CCR5 on CD4 and CD8 T cells and CD14 monocytes were evaluated. MX therapy induced a significant increase in the proportion of CD8 T cells (CD45RO(-)) over 9 months. Expression of several CCR increased following MX treatment. Two of the eight MX-treated patients demonstrated dramatic upregulation (70-76%) of CCR2 on monocytes. These two patients were the only MX-treated patients to demonstrate active inflammation by magnetic resonance imaging (MRI). PLC patients did not show significant changes in CCR expression. MX therapy was not associated with selective loss of CD4, CD8 or CD14 cells 1 month after treatment or over 9 months. These results suggest that MX therapy leads to a longitudinal increase in CD8 T cells and may increase CCR in patients with inflammation on MRI. Overall, MX did not show extensive immune changes in PPMS, although patients with active disease (gadolinium enhancing lesions) may identify a subset of PPMS subjects who respond immunologically to MX therapy. An improved understanding of MX may help identify markers of disease activity and response to therapy.
Collapse
Affiliation(s)
- Clara M Pelfrey
- Department of Neurosciences NC30, Lerner Research Institute, Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Ave., Cleveland, OH, USA.
| | | | | | | | | |
Collapse
|
46
|
Angelucci F, Batocchi AP, Caggiula M, Frisullo G, Patanella K, Sancricca C, Nociti V, Tonali PA, Mirabella M. In vivo effects of mitoxantrone on the production of pro- and anti-inflammatory cytokines by peripheral blood mononuclear cells of secondary progressive multiple sclerosis patients. Neuroimmunomodulation 2006; 13:76-81. [PMID: 16974110 DOI: 10.1159/000095762] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2006] [Accepted: 07/19/2006] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Mitoxantrone is an antineoplastic agent also used for the treatment of multiple sclerosis (MS). However, despite its efficacy, few data are available on its mechanism of action. The current study was designed to evaluate the short-term (1 month) and long-term (12 months) in vivo effects of mitoxantrone on pro- and anti-inflammatory cytokine production by the peripheral blood mononuclear cells (PBMC) of secondary progressive MS patients. METHODS Eighteen patients with secondary progressive MS underwent mitoxantrone therapy (at a dose of 12 mg/m(2) once every 3 months) over a 1-year period. Blood samples were obtained at baseline, after 1 month and after 12 months of treatment. The production of cytokines in the PBMC was measured by enzyme-linked immunosorbent assay. RESULTS There were no significant effects of mitoxantrone on proinflammatory cytokines [interleukin (IL) 6 and IL-12p40] and anti-inflammatory cytokines (IL-10 and transforming growth factor-beta) in our patients. Patients who showed no signs of therapeutic response were characterized by a higher basal PBMC production of IL-6 compared with that of the responding patients (p < 0.05) and mitoxantrone reduced this production after 12 months of treatment (p < 0.05). In the responding patients, IL-10 was significantly increased by mitoxantrone after 12 months of treatment (p < 0.05). CONCLUSION These findings provide additional information useful in the selection of the patient population suitable for mitoxantrone treatment and suggest that most probably the therapeutic action of mitoxantrone in MS is not entirely mediated by its immunosuppressant effects.
Collapse
Affiliation(s)
- Francesco Angelucci
- Department of Neuroscience, Institute of Neurology, Catholic University, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Neuhaus O, Kieseier BC, Hartung HP. Therapeutic role of mitoxantrone in multiple sclerosis. Pharmacol Ther 2005; 109:198-209. [PMID: 16095713 DOI: 10.1016/j.pharmthera.2005.07.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Accepted: 07/07/2005] [Indexed: 11/26/2022]
Abstract
Mitoxantrone is approved by several health authorities for treatment of active forms of relapsing-remitting or secondary progressive multiple sclerosis (SPMS). This review provides an outline on relevant preclinical as well as clinical studies, places mitoxantrone in the context of other therapeutic approaches against multiple sclerosis (MS), and discusses relevant side effects. The current knowledge of the putative mechanisms of action of the compound is discussed.
Collapse
Affiliation(s)
- Oliver Neuhaus
- Department of Neurology, Heinrich Heine University, Moorenstrasse 5, D-40225 Düsseldorf, Germany.
| | | | | |
Collapse
|