1
|
Kupjetz M, Patt N, Joisten N, Ueland PM, McCann A, Gonzenbach R, Bansi J, Zimmer P. Baseline Inflammation but not Exercise Modality Impacts Exercise-induced Kynurenine Pathway Modulation in Persons With Multiple Sclerosis: Secondary Results From a Randomized Controlled Trial. Int J Tryptophan Res 2024; 17:11786469241284423. [PMID: 39534856 PMCID: PMC11555752 DOI: 10.1177/11786469241284423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/29/2024] [Indexed: 11/16/2024] Open
Abstract
Background The kynurenine pathway (KP) is an important hub in neuroimmune crosstalk that is dysregulated in persons with multiple sclerosis (pwMS) and modulated by exercise in a modality-specific manner. Objectives To compare changes in the KP metabolite profile of pwMS (1) following combined treatments including either high-intensity interval training (HIIT) or moderate-intensity continuous training (MICT) during a 3-week multimodal rehabilitation, (2) to evaluate exercise response in relation to baseline systemic inflammation, and (3) to investigate associations of kynurenines with physical capacity and clinical outcomes. Methods For this secondary analysis of a randomized controlled trial, serum concentrations of kynurenines at baseline and after 3 weeks were determined using targeted metabolomics (LC-MS/MS). Exercise-induced changes in the KP metabolite profile according to treatment and baseline systemic inflammation (neutrophil-to-lymphocyte ratio (NLR) <3.12 versus ⩾3.12) were investigated using covariance analyses. Results Regardless of treatment, concentrations of tryptophan and most kynurenines decreased over time. Quinolinic acid concentration increased (p < .001). Participants with low and high NLR revealed differential exercise-induced changes in concentrations of kynurenines and NLR. The systemic inflammation markers neopterin (p = .015) and NLR (p < .001) decreased in the whole group and in participants with high NLR, respectively. Conclusions Combined treatments including HIIT or MICT do not differentially modulate the KP metabolite profile, with both reducing concentrations of most kynurenines. Baseline systemic inflammation may impact exercise-induced changes in the KP metabolite profile and anti-inflammatory effects of exercise in pwMS. Trial registration clinicaltrials.gov (identifier: NCT04356248).
Collapse
Affiliation(s)
- Marie Kupjetz
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund, Germany
| | - Nadine Patt
- Department of Neurology, Valens Rehabilitation Centre, Clinics of Valens, Valens, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Niklas Joisten
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund, Germany
- Division of Exercise and Movement Science, Institute for Sport Science, University of Göttingen, Göttingen, Germany
| | | | | | - Roman Gonzenbach
- Department of Neurology, Valens Rehabilitation Centre, Clinics of Valens, Valens, Switzerland
| | - Jens Bansi
- Department of Neurology, Valens Rehabilitation Centre, Clinics of Valens, Valens, Switzerland
- Department of Health, Physiotherapy, OST – Eastern Switzerland University of Applied Sciences, Sankt Gallen, Switzerland
| | - Philipp Zimmer
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund, Germany
| |
Collapse
|
2
|
Peternell A, Lechner C, Breu M, Preisel M, Schimmel M, Eisenkölbl A, Zobel J, Wendel EM, Reindl M, Rostásy K, Baumann M. Blood parameters in pediatric myelin oligodendrocyte glycoprotein antibody-associated disorders. Eur J Paediatr Neurol 2024; 50:86-95. [PMID: 38705015 DOI: 10.1016/j.ejpn.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND AND OBJECTIVES Patients with myelin oligodendrocyte glycoprotein antibody-associated disorders (MOGAD) clinically present e.g. with acute disseminated encephalomyelitis (ADEM), optic neuritis (ON), transverse myelitis (TM) or aquaporin-4-IgG (AQP4-IgG) negative neuromyelitis optica spectrum disorders (NMOSD)-like phenotypes. We aimed to analyze and compare blood parameters in children with MOGAD, AQP4-IgG-positive NMOSD (hence NMOSD), multiple sclerosis (MS) and healthy controls (HC). METHODS We evaluated differences in complete blood counts (CBC), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), monocyte-to-lymphocyte ratio (MLR) and C-reactive protein (CRP) between these four groups and within the groups between clinical attack, acute treatment and remission. RESULTS Our cohort consisted of 174 children and adolescents with a total of 550 timepoints: 66 patients had MOGAD (202 timepoints), 11 NMOSD (76 timepoints), 58 MS (219 timepoints) and 39 were HC (53 timepoints). At clinical attack, leukocyte counts were elevated in MOGAD compared to remission (p < 0.001) and compared to all other groups (p < 0.001). NLR was high in MOGAD and NMOSD, and PLR was high in NMOSD, however, after correction for multiple testing these findings did not remain significant. While glucocorticoids caused an increase of leukocyte counts and NLR in NMOSD and MS, these values remained stable during acute treatment in MOGAD. In remission, NLR normalized in MOGAD, while it stayed high in NMOSD. PLR increased in NMOSD and was significantly higher compared to all other groups. DISCUSSION Some blood parameters, mainly leukocyte and differential counts, might help clinicians to evaluate disease activity, differentiate relapses from pseudo-relapses and even distinguish between different disease entities.
Collapse
Affiliation(s)
- Alina Peternell
- Division of Pediatric Neurology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Lechner
- Division of Pediatric Neurology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Breu
- Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Martin Preisel
- Department of Pediatric Neurology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Mareike Schimmel
- Division of Pediatric Neurology, Children's Hospital, Medical University of Augsburg, Augsburg, Germany
| | - Astrid Eisenkölbl
- Department of Pediatrics and Adolescent Medicine, Johannes Kepler University Linz, Kepler University Hospital, Linz, Austria
| | - Joachim Zobel
- Division of Pediatric Neurology, Department of Pediatrics, Medical University of Graz, Graz, Austria
| | - Eva-Maria Wendel
- Department of Pediatric Neurology, Olgahospital Stuttgart, Stuttgart, Germany
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kevin Rostásy
- Department of Pediatric Neurology, Children's Hospital Datteln, University Witten/Herdecke, Datteln, Germany
| | - Matthias Baumann
- Division of Pediatric Neurology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
3
|
Yuan YG, Wang JL, Zhang YX, Li L, Reza AMMT, Gurunathan S. Biogenesis, Composition and Potential Therapeutic Applications of Mesenchymal Stem Cells Derived Exosomes in Various Diseases. Int J Nanomedicine 2023; 18:3177-3210. [PMID: 37337578 PMCID: PMC10276992 DOI: 10.2147/ijn.s407029] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/31/2023] [Indexed: 06/21/2023] Open
Abstract
Exosomes are nanovesicles with a wide range of chemical compositions used in many different applications. Mesenchymal stem cell-derived exosomes (MSCs-EXOs) are spherical vesicles that have been shown to mediate tissue regeneration in a variety of diseases, including neurological, autoimmune and inflammatory, cancer, ischemic heart disease, lung injury, and liver fibrosis. They can modulate the immune response by interacting with immune effector cells due to the presence of anti-inflammatory compounds and are involved in intercellular communication through various types of cargo. MSCs-EXOs exhibit cytokine storm-mitigating properties in response to COVID-19. This review discussed the potential function of MSCs-EXOs in a variety of diseases including neurological, notably epileptic encephalopathy and Parkinson's disease, cancer, angiogenesis, autoimmune and inflammatory diseases. We provided an overview of exosome biogenesis and factors that regulate exosome biogenesis. Additionally, we highlight the functions and potential use of MSCs-EXOs in the treatment of the inflammatory disease COVID-19. Finally, we covered a strategies and challenges of MSCs-EXOs. Finally, we discuss conclusion and future perspectives of MSCs-EXOs.
Collapse
Affiliation(s)
- Yu-Guo Yuan
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center of Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Jia-Lin Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center of Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Ya-Xin Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center of Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Ling Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center of Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Abu Musa Md Talimur Reza
- Department of Molecular Biology and Genetics, Faculty of Science, Gebze Technical University, Gebze, Kocaeli, Türkiye
| | | |
Collapse
|
4
|
Heydari Z, Peshkova M, Gonen ZB, Coretchi I, Eken A, Yay AH, Dogan ME, Gokce N, Akalin H, Kosheleva N, Galea-Abdusa D, Ulinici M, Vorojbit V, Shpichka A, Groppa S, Vosough M, Todiras M, Butnaru D, Ozkul Y, Timashev P. EVs vs. EVs: MSCs and Tregs as a source of invisible possibilities. J Mol Med (Berl) 2023; 101:51-63. [PMID: 36527475 PMCID: PMC9759062 DOI: 10.1007/s00109-022-02276-2] [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] [Revised: 11/11/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022]
Abstract
Extracellular vesicles (EVs) are produced by various cells and exist in most biological fluids. They play an important role in cell-cell signaling, immune response, and tumor metastasis, and also have theranostic potential. They deliver many functional biomolecules, including DNA, microRNAs (miRNA), messenger RNA (mRNA), long non-coding RNA (lncRNA), lipids, and proteins, thus affecting different physiological processes in target cells. Decreased immunogenicity compared to liposomes or viral vectors and the ability to cross through physiological barriers such as the blood-brain barrier make them an attractive and innovative option as diagnostic biomarkers and therapeutic carriers. Here, we highlighted two types of cells that can produce functional EVs, namely, mesenchymal stem/stromal cells (MSCs) and regulatory T cells (Tregs), discussing MSC/Treg-derived EV-based therapies for some specific diseases including acute respiratory distress syndrome (ARDS), autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Zahra Heydari
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Maria Peshkova
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia
| | | | - Ianos Coretchi
- Department of Pharmacology and Clinical Pharmacology, Nicolae Testemitanu State University of Medicine and Pharmacy, Chisinau, Moldova
| | - Ahmet Eken
- Betül-Ziya Eren Genome and Stem Cell Center (GENKOK), Kayseri, Turkey.,Department of Medical Biology, Erciyes University School of Medicine, Kayseri, Turkey
| | - Arzu Hanım Yay
- Betül-Ziya Eren Genome and Stem Cell Center (GENKOK), Kayseri, Turkey.,Department of Histology and Embryology, Erciyes University School of Medicine, Kayseri, Turkey
| | - Muhammet Ensar Dogan
- Department of Medical Genetic, Erciyes University School of Medicine, Kayseri, Turkey
| | - Nuriye Gokce
- Department of Medical Genetic, Erciyes University School of Medicine, Kayseri, Turkey
| | - Hilal Akalin
- Department of Medical Genetic, Erciyes University School of Medicine, Kayseri, Turkey
| | - Nastasia Kosheleva
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,FSBSI Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Daniela Galea-Abdusa
- Genetics Laboratory, Nicolae Testemitanu State University of Medicine and Pharmacy, Chisinau, Moldova
| | - Mariana Ulinici
- Department of Microbiology and Immunology, Nicolae Testemitanu State University of Medicine and Pharmacy, Chisinau, Moldova
| | - Valentina Vorojbit
- Department of Microbiology and Immunology, Nicolae Testemitanu State University of Medicine and Pharmacy, Chisinau, Moldova
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia.,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Stanislav Groppa
- Department of Neurology, Nicolae Testemițanu State University of Medicine and Pharmacy, Chisinau, Moldova.,Laboratory of Neurobiology and Medical Genetics, Nicolae Testemițanu State University of Medicine and Pharmacy, Chisinau, Moldova.,Department of Neurology, Institute of Emergency Medicine, Chisinau, Moldova
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran.
| | - Mihail Todiras
- Drug Research Center, Nicolae Testemitanu State University of Medicine and Pharmacy, Chisinau, Moldova
| | | | - Yusuf Ozkul
- Betül-Ziya Eren Genome and Stem Cell Center (GENKOK), Kayseri, Turkey. .,Department of Medical Genetic, Erciyes University School of Medicine, Kayseri, Turkey.
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia. .,World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia. .,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
5
|
Baharlooi H, Mansourabadi AH, Minbashi Moeini M, Mohamed Khosroshahi L, Azimi M. Nucleic Acids as Novel Therapeutic Modalities to Address Multiple Sclerosis Onset and Progression. Cell Mol Neurobiol 2022; 42:2611-2627. [PMID: 34694513 PMCID: PMC11421605 DOI: 10.1007/s10571-021-01158-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 10/17/2021] [Indexed: 02/07/2023]
Abstract
The issue of treating Multiple Sclerosis (MS) begins with disease-modifying treatments (DMTs) which may cause lymphopenia, dyspnea, and many other adverse effects. Consequently, further identification and evaluation of alternative treatments are crucial to monitoring their long-term outcomes and hopefully, moving toward personalized approaches that can be translated into clinical treatments. In this article, we focused on the novel therapeutic modalities that alter the interaction between the cellular constituents contributing to MS onset and progression. Furthermore, the studies that have been performed to evaluate and optimize drugs' efficacy, and particularly, to show their limitations and strengths are also presented. The preclinical trials of novel approaches for multiple sclerosis treatment provide promising prospects to cure the disease with pinpoint precision. Considering the fact that not a single treatment could be effective enough to cover all aspects of MS treatment, additional researches and therapies need to be developed in the future. Since the pathophysiology of MS resembles a jigsaw puzzle, researchers need to put a host of pieces together to create a promising window towards MS treatment. Thus, a combination therapy encompassing all these modules is highly likely to succeed in dealing with the disease. The use of different therapeutic approaches to re-induce self-tolerance in autoreactive cells contributing to MS pathogenesis is presented. A Combination therapy using these tools may help to deal with the clinical disabilities and symptoms of the disease in the future.
Collapse
Affiliation(s)
- Hussein Baharlooi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Mansourabadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Moein Minbashi Moeini
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Faculty of Pharmacy, Université Laval, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec, Canada
| | | | - Maryam Azimi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Hemmat highway, Tehran, Iran.
| |
Collapse
|
6
|
Barzegar M, Houshi S, Sadeghi E, Hashemi MS, Pishgahi G, Bagherieh S, Afshari-Safavi A, Mirmosayyeb O, Shaygannejad V, Zabeti A. Association of Disease-Modifying Therapies with COVID-19 Susceptibility and Severity in Patients with Multiple Sclerosis: A Systematic Review and Network Meta-Analysis. Mult Scler Int 2022; 2022:9388813. [PMID: 36187599 PMCID: PMC9519336 DOI: 10.1155/2022/9388813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/03/2022] [Indexed: 12/24/2022] Open
Abstract
Background We conducted this study to assess the effect of disease-modifying therapies (DMTs) on coronavirus disease (COVID-19) susceptibility and severity in people with multiple sclerosis (MS). Methods Available studies from PubMed, Scopus, EMBASE, Web of Science, and gray literature, including reference lists and conference abstracts, were searched from December 1, 2019, to July 26, 2021. We included cross-sectional, case-control, and cohort studies assessing the association of DMTs with risk of contracting COVID-19 or its outcomes in MS patients on univariate or multivariate regression analyses. We conducted a network meta-analysis (NMA) to compare the risk of COVID-19 and developing severe infection across DMTs. Results Out of the initial 3893 records and 1883 conference abstracts, a total of 10 studies were included. Pairwise comparisons showed that none of the DMTs meaningfully affect the risk of acquiring infection. There was significant total heterogeneity and inconsistency across this NMA. In comparison with no DMT, dimethyl fumarate (0.62 (0.42, 0.93)), fingolimod (0.55 (0.32, 0.94)), natalizumab (0.50 (0.31, 0.81)), and interferon (0.42 (0.22, 0.79)) were associated with a decreased risk of severe COVID-19; but, rituximab was observed to increase the risk (1.94 (1.20, 3.12)). Compared to rituximab or ocrelizumab, all DMTs were associated with a decreased risk. Pairwise comparisons showed no differences across other DMTs. Interferon and rituximab were associated with the lowest and highest risks of severe COVID-19. Conclusion Our study showed an increased risk of severe COVID-19 in patients on rituximab and ocrelizumab. No association with COVID-19 severity across other DMTs was observed.
Collapse
Affiliation(s)
- Mahdi Barzegar
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shakiba Houshi
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Erfan Sadeghi
- Department of Biostatistics and Epidemiology, Faculty of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mozhgan Sadat Hashemi
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasem Pishgahi
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Bagherieh
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Afshari-Safavi
- Department of Biostatistics and Epidemiology, Faculty of Health, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Omid Mirmosayyeb
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vahid Shaygannejad
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Aram Zabeti
- Department of Neurology and Rehabilitation Medicine, Waddell center in Multiple Sclerosis, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
7
|
Rowles WM, Hsu WY, McPolin K, Li A, Merrill S, Guo CY, Green AJ, Gelfand JM, Bove RM. Transitioning From S1P Receptor Modulators to B Cell-Depleting Therapies in Multiple Sclerosis: Clinical, Radiographic, and Laboratory Data. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:e1183. [PMID: 35581005 PMCID: PMC9128034 DOI: 10.1212/nxi.0000000000001183] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 03/29/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND OBJECTIVES Patients with multiple sclerosis (MS) transition from oral sphingosine-1-receptor (S1P) modulators to anti-CD20 therapies for several circumstances. Optimal timing of this transition is uncertain, given competing concerns of rebound disease activity and ensuring immune reconstitution. The objective of this study was to evaluate the relationship between inflammatory activity and the transition period from fingolimod to anti-CD20 therapies in a real-world MS cohort. METHODS Medical records were reviewed for all patients at our center transitioning from fingolimod to rituximab or ocrelizumab between 2010 and October 2020. Time periods reviewed were the following: before fingolimod discontinuation, interval between fingolimod and anti-CD20 treatments, and after the first anti-CD20 infusion. The primary outcome was clinical relapses; MRI activity, time to absolute lymphocyte count (ALC) recovery, and infections were secondary. Clinical and demographic factors significant in univariable analyses were included in multivariable analyses. RESULTS Transition data were available for 108 patients (68.5% women, 68.5% relapsing-remitting MS, mean age 44.6 years). The median (interquartile range) interval between fingolimod and anti-CD20 therapy was 28 (1-115.2) days. Six of 51 patients (11.8%) with intervals >1 month and 0/57 patients with shorter intervals experienced a relapse (MRI confirmed) within 6 months of fingolimod discontinuation. In the year following anti-CD20 initiation, 4/108 patients (3.7%) experienced a relapse (median 214.5 days after infusion). An additional 7% of those undergoing contrast-enhanced MRIs developed Gd+ lesions. ALC normalized following treatment switch in 89/92; the interval between treatments was unrelated to ALC recovery or infection. DISCUSSION Delaying anti-CD20 start to monitor ALC after S1P modulator discontinuation may not be necessary and could increase rebound risk. ALC monitoring could instead occur after a rapid switch to anti-CD20 treatment.
Collapse
Affiliation(s)
- William M. Rowles
- From the UCSF Weill Institute for Neurosciences (W.M.R., W.-Y.H., K.M., A.L., C.-Y.G., A.J.G., J.M.G., R.M.B.), Division of Neuroimmunology and Glial Biology, Department of Neurology, Department of Clinical Pharmacy (S.M.), and UCSF Department of Ophthalmology (A.J.G.), University of California, San Francisco
| | - Wan-Yu Hsu
- From the UCSF Weill Institute for Neurosciences (W.M.R., W.-Y.H., K.M., A.L., C.-Y.G., A.J.G., J.M.G., R.M.B.), Division of Neuroimmunology and Glial Biology, Department of Neurology, Department of Clinical Pharmacy (S.M.), and UCSF Department of Ophthalmology (A.J.G.), University of California, San Francisco
| | - Kira McPolin
- From the UCSF Weill Institute for Neurosciences (W.M.R., W.-Y.H., K.M., A.L., C.-Y.G., A.J.G., J.M.G., R.M.B.), Division of Neuroimmunology and Glial Biology, Department of Neurology, Department of Clinical Pharmacy (S.M.), and UCSF Department of Ophthalmology (A.J.G.), University of California, San Francisco
| | - Alyssa Li
- From the UCSF Weill Institute for Neurosciences (W.M.R., W.-Y.H., K.M., A.L., C.-Y.G., A.J.G., J.M.G., R.M.B.), Division of Neuroimmunology and Glial Biology, Department of Neurology, Department of Clinical Pharmacy (S.M.), and UCSF Department of Ophthalmology (A.J.G.), University of California, San Francisco
| | - Steven Merrill
- From the UCSF Weill Institute for Neurosciences (W.M.R., W.-Y.H., K.M., A.L., C.-Y.G., A.J.G., J.M.G., R.M.B.), Division of Neuroimmunology and Glial Biology, Department of Neurology, Department of Clinical Pharmacy (S.M.), and UCSF Department of Ophthalmology (A.J.G.), University of California, San Francisco
| | - Chu-Yueh Guo
- From the UCSF Weill Institute for Neurosciences (W.M.R., W.-Y.H., K.M., A.L., C.-Y.G., A.J.G., J.M.G., R.M.B.), Division of Neuroimmunology and Glial Biology, Department of Neurology, Department of Clinical Pharmacy (S.M.), and UCSF Department of Ophthalmology (A.J.G.), University of California, San Francisco
| | - Ari J. Green
- From the UCSF Weill Institute for Neurosciences (W.M.R., W.-Y.H., K.M., A.L., C.-Y.G., A.J.G., J.M.G., R.M.B.), Division of Neuroimmunology and Glial Biology, Department of Neurology, Department of Clinical Pharmacy (S.M.), and UCSF Department of Ophthalmology (A.J.G.), University of California, San Francisco
| | - Jeffrey Marc Gelfand
- From the UCSF Weill Institute for Neurosciences (W.M.R., W.-Y.H., K.M., A.L., C.-Y.G., A.J.G., J.M.G., R.M.B.), Division of Neuroimmunology and Glial Biology, Department of Neurology, Department of Clinical Pharmacy (S.M.), and UCSF Department of Ophthalmology (A.J.G.), University of California, San Francisco
| | - Riley M. Bove
- From the UCSF Weill Institute for Neurosciences (W.M.R., W.-Y.H., K.M., A.L., C.-Y.G., A.J.G., J.M.G., R.M.B.), Division of Neuroimmunology and Glial Biology, Department of Neurology, Department of Clinical Pharmacy (S.M.), and UCSF Department of Ophthalmology (A.J.G.), University of California, San Francisco
| |
Collapse
|
8
|
Mao-Draayer Y, Cohen JA, Bar-Or A, Han MH, Singer B, Williams IM, Meng X, Elam C, Weiss JL, Cox GM, Ziehn M, Cree BAC. Immune cell subset profiling in multiple sclerosis after fingolimod initiation and continued treatment: The FLUENT study. Mult Scler J Exp Transl Clin 2022; 8:20552173221115023. [PMID: 35936922 PMCID: PMC9346260 DOI: 10.1177/20552173221115023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 07/05/2022] [Indexed: 11/19/2022] Open
Abstract
Background Fingolimod is a sphingosine 1-phosphate receptor modulator approved for
relapsing MS. Long-term effects on the immunological profile are not fully
understood. Objective Investigate fingolimod's temporal effects on immune cell subsets, and safety
outcomes. Methods In FLUENT, a 12-month, prospective, non-randomized, open-label, phase IV
study, adult participants received fingolimod 0.5 mg/day. Changes in immune
cell subsets, anti-John Cunningham virus (JCV) antibody index, and serum
neurofilament levels were assessed. Results 165 fingolimod-naive and 217 participants treated for 2–12 years in routine
clinical practice were enrolled. Levels of all monitored peripheral
lymphocyte subsets were reduced from month 3 in fingolimod-naive
participants. Greatest reductions occurred in naive and central memory
CD4+ and CD8+ T cells, and in naive and memory B cells. Most lymphocyte
subset levels remained stable in the continuous fingolimod group. Components
of the innate immune system remained within reference ranges. No increase in
JCV seropositivity was observed. No single cellular subset correlated with
anti-JCV antibody index at any time point. Neurofilament levels remained
within healthy adult reference limits throughout. No opportunistic
infections were reported; no new or unexpected safety signals were
observed. Conclusion FLUENT provides insights into the utility of immunological profiling to
evaluate therapy response and potential infection risk.
Collapse
Affiliation(s)
- Yang Mao-Draayer
- Autoimmunity Center of Excellence, Multiple Sclerosis Center, University of Michigan, Ann Arbor, MI, USA
| | | | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics, and the Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - May H Han
- Department of Neurology, Stanford University, Stanford, CA, USA
| | - Barry Singer
- Missouri Baptist Medical Center, St Louis, MO, USA
| | | | | | | | | | | | - Marina Ziehn
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Bruce AC Cree
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | | |
Collapse
|
9
|
Darazam IA, Rabiei MM, Moradi O, Gharehbagh FJ, Roozbeh M, Nourinia R, Hatami F, Shojaei M, Lotfollahi L. A Case of Fingolimod-associated Cryptococcal Meningitis. Curr HIV Res 2022; 20:337-342. [PMID: 35770404 DOI: 10.2174/1570162x20666220629124225] [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: 01/20/2022] [Revised: 04/10/2022] [Accepted: 05/05/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Leukopenia, a rare adverse effect of Fingolimod therapy, paves the way for opportunistic infections. In this study, we reported rare fingolimod associated cryptococcal meningitis. CASE PRESENTATION A 39-year-old woman with RRMS was referred to the emergency department. The patient's major complaints were headache, fever, weakness, and progressive loss of consciousness within the last two days prior to the referral. The patient had a history of hospitalization due to RRMS [two times]. In the second hospitalization, interferon Beta-1a was replaced with Fingolimod. Using polymerase chain reaction, Cryptococcus neoformans was detected in CSF. Liposomal amphotericin B and fluconazole [800 mg per day] were started. Six weeks later, the patient was discharged without any major complaints. CONCLUSION Albeit fingolimod associated cryptococcal meningitis is a rare event, Fingolimod therapy in patients with MS should be performed cautiously. Regular follow-ups may give rise to a timely diagnosis of probable fingolimod associated cryptococcal meningitis. Fingolimod therapy can lead to lymphocytopenia and various infections. We, therefore, suggest that intermittent blood lymphocyte counts as well as monitoring of clinical manifestations among MS patients treated with Fingolimod to avoid additional neurological and physical disabilities in these patients.
Collapse
Affiliation(s)
- Ilad Alavi Darazam
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdi Rabiei
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omid Moradi
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farid Javandoust Gharehbagh
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad Roozbeh
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramin Nourinia
- Ophthalmic Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Firouze Hatami
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maziar Shojaei
- Department of Neurology, Loghman Hakim Hospital, Kamali, Iran
| | - Legha Lotfollahi
- Department of Nephrology, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Reynolds A, Gaughan M, Holden D, Redenbaugh V, Dunne J, Redmond J, Conlon N. The effects of dimethyl fumarate and fingolimod on T-cell lymphocyte proliferation in patients with multiple sclerosis. Ir J Med Sci 2022; 191:2759-2762. [PMID: 35028898 DOI: 10.1007/s11845-021-02913-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 12/25/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The disease-modifying therapies (DMT), dimethyl fumarate (DMF) and fingolimod (FTY) improve the outcomes in multiple sclerosis (MS) by reducing relapses and numbers and volume of lesions. They mediate their effects through reduction of immune reactivation, which may potentially lead to lymphopaenia and increased risk of infections. Previous studies have examined the effects of these therapies on lymphocyte subsets; however, the in vivo effects on circulating lymphocyte proliferation require further elucidation. The aim of this study was to determine the effects of DMF and FTY on T-cell proliferation in patients with MS. METHOD We examined T-cell lymphocyte proliferation and lymphocyte subsets in ten patients (five on DMF, five on FTY) before starting DMT and again 4 to 11 months after being maintained on DMT. RESULTS In the FTY-treated group, the mean percentage proliferation was significantly lower using both assays (PHA assay mean percentage change - 51.2 ± 25.97, p < 0.05; anti-CD3/CD28 assay mean percentage change - 39.74 ± 27.85, p < 0.05). There was no statistical difference in T-cell lymphocyte proliferation in the DMF-treated group for either assay (PHA, p = 0.316; anti-CD3/CD28, p = 0.373). CONCLUSIONS This pilot study suggests that the T-lymphocytes of patients on FTY have an abnormal proliferation response as well as being reduced in the circulation.
Collapse
Affiliation(s)
- Audrey Reynolds
- Department of Neurology, St James's Hospital, Dublin 8, Ireland.
| | - Maria Gaughan
- Department of Neurology, St James's Hospital, Dublin 8, Ireland
| | - Dean Holden
- Department of Immunology, St James's Hospital, Dublin 8, Ireland
| | | | - Jean Dunne
- Department of Immunology, St James's Hospital, Dublin 8, Ireland
| | - Janice Redmond
- Department of Neurology, St James's Hospital, Dublin 8, Ireland
| | - Niall Conlon
- Department of Immunology, St James's Hospital, Dublin 8, Ireland
| |
Collapse
|
11
|
Lymphocyte Counts and Multiple Sclerosis Therapeutics: Between Mechanisms of Action and Treatment-Limiting Side Effects. Cells 2021; 10:cells10113177. [PMID: 34831400 PMCID: PMC8625745 DOI: 10.3390/cells10113177] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 01/18/2023] Open
Abstract
Although the detailed pathogenesis of multiple sclerosis (MS) is not completely understood, a broad range of disease-modifying therapies (DMTs) are available. A common side effect of nearly every MS therapeutic agent is lymphopenia, which can be both beneficial and, in some cases, treatment-limiting. A sound knowledge of the underlying mechanism of action of the selected agent is required in order to understand treatment-associated changes in white blood cell counts, as well as monitoring consequences. This review is a comprehensive summary of the currently available DMTs with regard to their effects on lymphocyte count. In the first part, we describe important general information about the role of lymphocytes in the course of MS and the essentials of lymphopenic states. In the second part, we introduce the different DMTs according to their underlying mechanism of action, summarizing recommendations for lymphocyte monitoring and definitions of lymphocyte thresholds for different therapeutic regimens.
Collapse
|
12
|
Baeva ME, Baev PB, Nelson J, Kazimirchik A, Vorobeychik G. A retrospective analysis of changes in lymphocyte levels in patients with multiple sclerosis during and after Tecfidera® treatment. Mult Scler J Exp Transl Clin 2021; 7:20552173211029674. [PMID: 34345437 PMCID: PMC8283074 DOI: 10.1177/20552173211029674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/14/2021] [Indexed: 11/16/2022] Open
Abstract
Background There are currently no best practice recommendations for lymphocyte subset monitoring for patients with multiple sclerosis (pwMS) on disease-modifying therapies including Tecfidera® (dimethyl fumarate, DMF). However, there have been several cases of pwMS on DMF without severe lymphopenia who had high CD4:CD8 T cell ratios and went on to develop progressive multifocal leukoencephalopathy. Objective Our objective was to characterize the changes in immune profile during and after DMF treatment in pwMS. Methods A retrospective analysis of longitudinal data from 299 pwMS who have been treated with DMF at the Fraser Health Multiple Sclerosis Clinic in British Columbia, Canada. The blood test results were taken from January 1, 2013 to April 1, 2020. Results Our results suggest that CD8+ T cells had the highest proportional decrease compared to other lymphocyte subset populations and overall lymphocyte count in response to DMF treatment. CD56+ Natural Killer cells were similarly decreased in response to DMF treatment. CD4:CD8 T cell ratio was the measurement that had the highest rate of change in response to DMF initiation and discontinuation. Conclusion CD8+ T cell count and CD4:CD8 T cell ratio may be a more sensitive measurement of the immune landscape of patients with MS on DMF.
Collapse
Affiliation(s)
- Maria-Elizabeth Baeva
- Fraser Health Multiple Sclerosis Clinic, Burnaby Hospital, Burnaby Hospital, Burnaby, British Columbia, Canada
| | - Philip Boris Baev
- Fraser Health Multiple Sclerosis Clinic, Burnaby Hospital, Burnaby Hospital, Burnaby, British Columbia, Canada
| | - Jill Nelson
- Fraser Health Multiple Sclerosis Clinic, Burnaby Hospital, Burnaby Hospital, Burnaby, British Columbia, Canada
| | - Anna Kazimirchik
- Fraser Health Multiple Sclerosis Clinic, Burnaby Hospital, Burnaby Hospital, Burnaby, British Columbia, Canada
| | - Galina Vorobeychik
- Fraser Health Multiple Sclerosis Clinic, Burnaby Hospital, Burnaby Hospital, Burnaby, British Columbia, Canada
| |
Collapse
|
13
|
Lee CH, Jiang B, Nakhaei-Nejad M, Barilla D, Blevins G, Giuliani F. Cross-sectional analysis of peripheral blood mononuclear cells in lymphopenic and non-lymphopenic relapsing-remitting multiple sclerosis patients treated with dimethyl fumarate. Mult Scler Relat Disord 2021; 52:103003. [PMID: 34118574 DOI: 10.1016/j.msard.2021.103003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/24/2021] [Accepted: 04/29/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Relapsing-remitting multiple sclerosis (RRMS) is an autoimmune disorder of the central nervous system. Dimethyl Fumarate is a disease-modifying medication used to treat RRMS patients that can induce lymphopenia. We aimed to immunophenotype peripheral blood mononuclear cells (PBMC) in RRMS patients cross-sectionally and examine the characteristics and modifications of lymphopenia over time. METHODS Characterization of PBMC was done by multiparametric flow cytometry. Patients had been on treatment for up to 4 years and were grouped into lymphopenic (DMF-L) and non-lymphopenic (DMF-N) patients. RESULTS Lymphopenia affected the cell population changes over time, with other patient characteristics (gender, age, and previous treatment status) also having significant effects. In both lymphopenic and non-lymphopenic patients, PBMC percentages were reduced over time. While overall T and B cells frequencies were not affected, males, older patients and untreated patients had significant changes in B cell subpopulations over time. CD4+ to CD8+T cell ratio increased significantly in lymphopenic patients over time. CD4-CD8-T cell population was similarly reduced in both lymphopenic and non-lymphopenic patients, over time. While the monocyte and NK overall populations were not changed, non-classical monocyte subpopulation decreased over time in lymphopenic patients. We also found CD56-CD16+ and CD56-CD16- NK cells frequencies changed over time in lymphopenic patients. Immune populations showed correlations with clinical outcomes measured by EDSS and relapse rate. Analysis of the overall immunophenotype showed that, while groups divided by other patient characteristics showed differences, the lymphopenia status overrode these differences, resulting in similar immunophenotype within DMF-L. CONCLUSIONS Our data provide evidence that under the same therapy, lymphopenia affects how the immunophenotype changes over time and can override the differences associated with other patient characteristics and possibly mask other significant changes in the immune profile of patients.
Collapse
Affiliation(s)
- Chieh-Hsin Lee
- Division of Neurology, Department of Medicine, University of Alberta, Alberta Canada T6G 2M8, Edmonton, Canada
| | - Bei Jiang
- Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, Canada
| | - Maryam Nakhaei-Nejad
- Division of Neurology, Department of Medicine, University of Alberta, Alberta Canada T6G 2M8, Edmonton, Canada
| | - David Barilla
- Division of Neurology, Department of Medicine, University of Alberta, Alberta Canada T6G 2M8, Edmonton, Canada
| | - Gregg Blevins
- Division of Neurology, Department of Medicine, University of Alberta, Alberta Canada T6G 2M8, Edmonton, Canada
| | - Fabrizio Giuliani
- Division of Neurology, Department of Medicine, University of Alberta, Alberta Canada T6G 2M8, Edmonton, Canada.
| |
Collapse
|
14
|
Aslanyan A, Anwar-Hashim Z, Siripurapu R, Mihalova T. Hepatitis C during disease modifying therapy with Fingolimod for Relapsing Remitting Multiple Sclerosis: diagnosis and treatment. BMJ Case Rep 2021; 14:14/2/e238167. [PMID: 33637491 PMCID: PMC7919550 DOI: 10.1136/bcr-2020-238167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
We present a case of 48-year-old woman with relapsing remitting multiple sclerosis (MS), who switched disease modifying therapy from Copaxone to Fingolimod due to her clinical and radiological MS disease progression. Unexpectedly after 2.5 years of stable MS symptoms and liver function tests (LFTs), we noted deranged LFTs during routine testing. Additional investigations showed hepatitis C positivity, genotype 3. It is likely a case of hepatitis C reactivation secondary to prolonged immunosuppressive effects of Fingolimod. Although the increased risk of viral reactivation related to varicella zoster virus is known to occur with Fingolimod treatment, to our knowledge, this is only the second case of hepatitis C disease activity reported with Fingolimod treatment. We would like to raise the awareness of hepatitis C viral reactivation as a possible complication of prolonged immunosuppression with Fingolimod.
Collapse
Affiliation(s)
- Aram Aslanyan
- Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Salford, UK
| | - Zoheb Anwar-Hashim
- Gastroenterology Department, Blackpool Teaching Hospitals NHS Foundation Trust, Blackpool, UK
| | - Rekha Siripurapu
- Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Salford, UK
| | - Tatiana Mihalova
- Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Salford, UK
| |
Collapse
|
15
|
Camara-Lemarroy CR, Silva C, Metz LM, Cerchiaro G, Greenfield J, Dowlatabadi R, Vogel HJ, Lee CH, Giuliani F, Nakhaei-Nejad M, Li DKB, Traboulsee A, Yong VW. Multimodal peripheral fluid biomarker analysis in clinically isolated syndrome and early multiple sclerosis. Mult Scler Relat Disord 2021; 50:102809. [PMID: 33581614 DOI: 10.1016/j.msard.2021.102809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/06/2021] [Accepted: 01/31/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Increasing evidence suggests that various inflammatory, immunological and metabolic pathways are altered in the clinically isolated syndrome (CIS) of multiple sclerosis (MS). Moreover, recent diagnostic criteria have made possible the very early diagnosis of MS. We evaluated multiple fluid biomarkers in people with early MS and CIS. METHODS We measured blood levels of cytokines, matrix metalloproteinases (MMPs), serum metabolomics and immune cell immunophenotyping in participants in the Trial of Minocycline in a Clinically Isolated Syndrome of Multiple Sclerosis. RESULTS When compared with healthy controls, people with early MS/CIS had higher levels of eotaxin, MCP-3, IL-1 receptor antagonist, IL-1β, IL-9 and IP-10, as well as MMPs 1, 8 and 9. In metabolomics analysis, the alanine, aspartate and glutamate metabolism and the synthesis and degradation of ketone bodies pathways were altered compared to healthy controls. There were no differences in lymphocyte subpopulation numbers. Out of all these biomarkers, only MMP-1 was able to differentiate between early MS and CIS, and was found to correlate with lesion volume and gadolinium enhancing lesions on MRI. CONCLUSION The immunological and metabolic profile of CIS and early MS is remarkably similar, supporting that these are a continuum of a common underlying pathophysiological process.
Collapse
Affiliation(s)
- Carlos R Camara-Lemarroy
- Departments of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Claudia Silva
- Departments of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Luanne M Metz
- Departments of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Graziela Cerchiaro
- Departments of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jamie Greenfield
- Departments of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Reza Dowlatabadi
- Department of Biological Science, Bio-NMR-metabolomics Research center, University of Calgary, Calgary, Canada
| | - Hans J Vogel
- Department of Biological Science, Bio-NMR-metabolomics Research center, University of Calgary, Calgary, Canada
| | - Chieh-Hsin Lee
- Department of Medicine, University of Alberta, Alberta, Canada
| | | | | | - David K B Li
- Department of Medicine, University of British Columbia, Canada
| | | | - V Wee Yong
- Departments of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
16
|
Dello Russo C, Scott KA, Pirmohamed M. Dimethyl fumarate induced lymphopenia in multiple sclerosis: A review of the literature. Pharmacol Ther 2020; 219:107710. [PMID: 33091427 DOI: 10.1016/j.pharmthera.2020.107710] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2020] [Indexed: 12/13/2022]
Abstract
Dimethyl fumarate (DMF) is a first line medication for multiple sclerosis. It has a favourable safety profile, however, there is concern regarding the occurrence of moderate-severe and sustained lymphopenia and the associated risk of progressive multifocal leukoencephalopathy. We carried out an extensive literature review to understand the molecular mechanisms underlying this adverse reaction. Dynamic changes in certain components of the immune system are likely to be important for the therapeutic effects of DMF, including depletion of memory T cells and decrease in activated T cells together with expansion of naïve T cells. Similar modifications were reported for the B cell components. CD8+ T cells are particularly susceptible to DMF-induced cell death, with marked reductions observed in lymphopenic subjects. The reasons underlying such increased sensitivity are not known, nor it is known how expansion of other lymphocyte subsets occurs. Understanding the molecular mechanisms underlying DMF action is challenging: in vivo DMF is rapidly metabolized to monomethyl fumarate (MMF), a less potent immunomodulator in vitro. Pharmacokinetics indicate that MMF is the main active species in vivo. However, the relative importance of DMF and MMF in toxicity remains unclear, with evidence presented in favour of either of the compounds as toxic species. Pharmacogenetic studies to identify genetic predictors of DMF-induced lymphopenia are limited, with inconclusive results. A role of the gut microbiome in the pharmacological effects of DMF is emerging. It is clear that further investigations are necessary to understand the mechanisms of DMF-induced lymphopenia and devise preventive strategies. Periodic monitoring of absolute lymphocyte counts, currently performed in clinical practise, allows for the early detection of lymphopenia as a risk-minimization strategy.
Collapse
Affiliation(s)
- Cinzia Dello Russo
- MRC Centre for Drug Safety Science and Wolfson Centre for Personalised Medicine, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, UK; Dept. of Healthcare Surveillance and Bioethics, Section of Pharmacology, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Kathryn Anne Scott
- MRC Centre for Drug Safety Science and Wolfson Centre for Personalised Medicine, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, UK
| | - Munir Pirmohamed
- MRC Centre for Drug Safety Science and Wolfson Centre for Personalised Medicine, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, UK.
| |
Collapse
|
17
|
Huber JE, Chang Y, Meinl I, Kümpfel T, Meinl E, Baumjohann D. Fingolimod Profoundly Reduces Frequencies and Alters Subset Composition of Circulating T Follicular Helper Cells in Multiple Sclerosis Patients. THE JOURNAL OF IMMUNOLOGY 2020; 204:1101-1110. [PMID: 32034063 DOI: 10.4049/jimmunol.1900955] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 12/12/2019] [Indexed: 12/22/2022]
Abstract
Fingolimod is an effective treatment for relapsing-remitting multiple sclerosis. It is well established that fingolimod, a modulator of the sphingosine-1-phosphate pathway, restrains the egress of CCR7+ lymphocytes from lymphatic tissues into the blood, thus resulting in reduced lymphocyte counts in peripheral blood. CXCR5+ T follicular helper (Tfh) cells provide help to B cells, are essential for the generation of potent Ab responses, and have been shown to be critically involved in the pathogenesis of several autoimmune diseases. Besides lymphoid tissue-resident Tfh cells, CXCR5+ circulating Tfh (cTfh) cells have been described in the blood, their numbers correlating with the magnitude of Tfh cells in lymphoid tissues. Although the effect of fingolimod on circulating lymphocyte subsets has been established, its effect on cTfh cells remains poorly understood. In this study, we found that although fingolimod strongly and disproportionally reduced cTfh cell frequencies, frequencies of activated cTfh cells were increased, and the composition of the cTfh cell pool was skewed toward a cTfh1 cell phenotype. The circulating T follicular regulatory cell subset and CXCR5+ CD8+ T cell frequencies were also strongly and disproportionally decreased after fingolimod treatment. In contrast, relative frequencies of CXCR5- memory Th cells as well as regulatory T and B cells were increased. In summary, these data provide new insights into fingolimod-induced compositional changes of lymphocyte populations in the blood, in particular cTfh cells, and thus contribute to a better understanding of the mechanism of action of fingolimod in multiple sclerosis patients.
Collapse
Affiliation(s)
- Johanna E Huber
- Institute for Immunology, Biomedical Center, Faculty of Medicine, LMU Munich, Planegg-Martinsried 82152, Germany; and
| | - Yinshui Chang
- Institute for Immunology, Biomedical Center, Faculty of Medicine, LMU Munich, Planegg-Martinsried 82152, Germany; and
| | - Ingrid Meinl
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, LMU Munich, Planegg-Martinsried 82152, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, LMU Munich, Planegg-Martinsried 82152, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, LMU Munich, Planegg-Martinsried 82152, Germany
| | - Dirk Baumjohann
- Institute for Immunology, Biomedical Center, Faculty of Medicine, LMU Munich, Planegg-Martinsried 82152, Germany; and
| |
Collapse
|
18
|
Inshasi JS, Almadani A, Fahad SA, Noori SI, Alsaadi T, Shakra M, Shatila AO, Zein TM, Boshra A. High-efficacy therapies for relapsing-remitting multiple sclerosis: implications for adherence. An expert opinion from the United Arab Emirates. Neurodegener Dis Manag 2020; 10:257-266. [PMID: 32438857 DOI: 10.2217/nmt-2020-0016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/05/2020] [Indexed: 12/31/2022] Open
Abstract
The number of disease-modifying treatments (DMDs) for relapsing-remitting multiple sclerosis has increased. DMDs differ not only in their efficacy and safety/tolerability, but also in the treatment burden of, associated with their initiation, route/frequency of administration, maintenance treatment and monitoring. High-efficacy DMDs bring the prospect of improved suppression of relapses and progression of disability, but may have serious safety issues, and burdensome long-term monitoring. Studies of patient preferences in this area have focused on side effects, efficacy and route of administration. Adherence to DMDs is often suboptimal in relapsing-remitting multiple sclerosis and there is a need to understand more about how the complex therapeutic and administration profiles of newer DMDs interact with these barriers to support optimal adherence to therapy.
Collapse
Affiliation(s)
- Jihad Said Inshasi
- MS Section, Rashid Hospital & Dubai Medical College, Dubai Health Authority (DHA), Dubai, UAE
| | - Abubaker Almadani
- Rashid Hospital & Dubai Medical College, Dubai health Authority (DHA), Dubai, UAE
| | - Sarmad Al Fahad
- Neurology Department, Neurospinal Hospital, Baghdad Medical College, Dubai, UAE
| | | | - Taoufik Alsaadi
- Neurology Department, American Center for Psychiatry & Neurology, Dubai, UAE
| | - Mustafa Shakra
- Department of Neurology, Sheikh Khalifa Medical City, Abu Dhabi, UAE
| | | | | | | |
Collapse
|
19
|
Haile Y, Adegoke A, Laribi B, Lin J, Anderson CC. Anti-CD52 blocks EAE independent of PD-1 signals and promotes repopulation dominated by double-negative T cells and newly generated T and B cells. Eur J Immunol 2020; 50:1362-1373. [PMID: 32388861 DOI: 10.1002/eji.201948288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 04/02/2020] [Accepted: 05/06/2020] [Indexed: 01/23/2023]
Abstract
Lymphocyte depletion using anti-CD52 antibody effectively reduces relapses of multiple sclerosis (MS). To begin to understand what mechanisms might control this outcome, we examined the effect of a murine-CD52-specific mAb on the depletion and repopulation of immune cells in mice with experimental autoimmune encephalomyelitis (EAE), a model of MS. We tested whether the tolerance-promoting receptor programmed cell death protein-1 (PD-1) is required for disease remission post anti-CD52, and found that PD-1-deficient mice with a more severe EAE were nevertheless effectively treated with anti-CD52. Anti-CD52 increased the proportions of newly generated T cells and double-negative (DN) T cells while reducing newly generated B cells; the latter effect being associated with a higher expression of CD52 by these cells. In the longer term, anti-CD52 caused substantial increases in the proportion of newly generated lymphocytes and DN T cells in mice with EAE. Thus, the rapid repopulation of lymphocytes from central lymphoid organs post anti-CD52 may limit further disease. Furthermore, these data identify DN T cells, a subset with immunoregulatory potential, as a significant hyperrepopulating subset following CD52-mediated depletion.
Collapse
Affiliation(s)
- Yohannes Haile
- Department of Surgery, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes and Transplant Institutes, University of Alberta, Edmonton, AB, Canada
| | - Adeolu Adegoke
- Department of Surgery, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes and Transplant Institutes, University of Alberta, Edmonton, AB, Canada
| | - Bahareh Laribi
- Alberta Diabetes and Transplant Institutes, University of Alberta, Edmonton, AB, Canada
| | - Jiaxin Lin
- Department of Surgery, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes and Transplant Institutes, University of Alberta, Edmonton, AB, Canada
| | - Colin C Anderson
- Department of Surgery, University of Alberta, Edmonton, AB, Canada.,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes and Transplant Institutes, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
20
|
Boffa G, Bruschi N, Cellerino M, Lapucci C, Novi G, Sbragia E, Capello E, Uccelli A, Inglese M. Fingolimod and Dimethyl-Fumarate-Derived Lymphopenia is not Associated with Short-Term Treatment Response and Risk of Infections in a Real-Life MS Population. CNS Drugs 2020; 34:425-432. [PMID: 32193826 DOI: 10.1007/s40263-020-00714-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The association between treatment-related lymphopenia in multiple sclerosis, drug efficacy and the risk of infections is not yet fully understood. OBJECTIVE The objective of this study was to assess whether lymphopenia is associated with short-term treatment response and infection rate in a real-life multiple sclerosis population treated with fingolimod and dimethyl-fumarate. We assessed the associations between baseline absolute lymphocyte count and the lymphocyte mean percentage decrease at 6 and 12 months with treatment response and the occurrence of adverse events over 12 months in the entire cohort of patients and in the two treatment groups separately. METHODS This is a retrospective observational real-world study of patients with multiple sclerosis treated with fingolimod and dimethyl-fumarate at the MS Center of the University of Genoa between 2011 and 2018. Patients with at least 12 months of follow-up were eligible if [1] they had an Expanded Disability Status Scale assessment at baseline and 12 months after treatment onset, [2] they had undergone brain magnetic resonance imaging at baseline and after 12 months, and [3] absolute lymphocyte counts were available at baseline, 6 and 12 months. Patients shifting from dimethyl-fumarate to fingolimod or vice versa were excluded from the analysis. RESULTS In total, 137 and 75 patients treated with fingolimod and dimethyl-fumarate, respectively, were included in the analysis. At 12 months, fingolimod-treated patients were more likely to experience grade II and grade III lymphopenia compared with dimethyl-fumarate patients (p < 0.001, χ2 = 94) and had a higher lymphocyte mean percentage decrease (p < 0.001, U = 540). A higher number of previous therapies and a lower baseline absolute lymphocyte count were predictors of lymphopenia at 6 months (p = 0.047, odds ratio = 1.60 and p = 0.014, odds ratio = 1.1) and 12 months (p = 0.003, odds ratio = 1.97 and p = 0.023, odds ratio = 1.1). In fingolimod-treated patients only, female sex and a higher Expanded Disability Status Scale score were predictors of lymphopenia at 12 months (p = 0.006, odds ratio = 7.58 and p = 0.03, odds ratio = 1.56). Neither absolute lymphocyte count at 6 and 12 months nor the mean percentage decrease at 6 and 12 months predicted No Evidence of Disease Activity (NEDA-3) status at 1 year, the occurrence of relapses, disease activity on MRI or disability progression. CONCLUSIONS Our findings suggest that peripheral blood lymphocyte changes are not associated with short-term treatment response and with the rate of infections during fingolimod and dimethyl-fumarate treatment in real-world patients. Higher treatment exposure and a lower baseline absolute lymphocyte count are risk factors for lymphopenia development during fingolimod and dimethyl-fumarate therapy.
Collapse
Affiliation(s)
- Giacomo Boffa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Nicolò Bruschi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Maria Cellerino
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Caterina Lapucci
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Giovanni Novi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Elvira Sbragia
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Elisabetta Capello
- Ospedale Policlinico San Martino IRCCS, Largo Daneo 3, 16100, Genoa, Italy
| | - Antonio Uccelli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy.,Ospedale Policlinico San Martino IRCCS, Largo Daneo 3, 16100, Genoa, Italy
| | - Matilde Inglese
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy. .,Ospedale Policlinico San Martino IRCCS, Largo Daneo 3, 16100, Genoa, Italy.
| |
Collapse
|
21
|
Baharlooi H, Azimi M, Salehi Z, Izad M. Mesenchymal Stem Cell-Derived Exosomes: A Promising Therapeutic Ace Card to Address Autoimmune Diseases. Int J Stem Cells 2020; 13:13-23. [PMID: 31887849 PMCID: PMC7119210 DOI: 10.15283/ijsc19108] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/03/2019] [Indexed: 12/12/2022] Open
Abstract
With the development of novel treatments for autoimmune disorders, it has become a popular research focus which mesenchymal stem cells (MSCs) have the capacity to counteract with autoimmune diseases progression. One of the underlying mechanisms behind their activities is the release of extracellular vesicles especially exosomes. MSC-derived exosomes are hypoimmunogenic nanocarriers which contain numerous immunoregulatory factors and similar to other exosomes, are able to pass through boundaries like the blood-brain barrier (BBB). Accumulating evidence provided by animal studies has demonstrated that MSC-derived exosomes, as a novel therapy, can re-induce self-tolerance, without subsequent complications reported for other treatments. Therefore, therapeutic applications of MSC-derived exosomes are contributing to core advances in the field of autoimmune diseases. Here, we briefly describe the biological characteristics of MSC-derived exosomes and review the experimentally verified outcomes for autoimmune disease therapy purposes.
Collapse
Affiliation(s)
- Hussein Baharlooi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Azimi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Izad
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Zanghì A, D'Amico E, Patti F. Immunosuppression in relapsing remitting multiple sclerosis: moving towards personalized treatment. Expert Rev Neurother 2020; 20:771-782. [PMID: 31971026 DOI: 10.1080/14737175.2020.1721282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Therapeutic armamentarium in Multiple Sclerosis (MS) has radically changed in the last few decades due to the development of disease modifying treatments (DMTs) with highly selective mechanisms of action. AREAS COVERED In this review, the authors will focus on the current role of immunosuppressive DMTs in the management of the relapsing-remitting form of MS (RRMS), moving from the rationale of its use and looking at the possibility to design an idealistic scenario of a personalized approach for each single patient. EXPERT OPINION Questions remain open about whether initial high-efficacy immunosuppressive DMTs improve long-term outcomes, whether prolonged exposure to these agents increases adverse events and what the strongest early surrogate markers are for predicting long-term treatment responses to high-efficacy drugs. In this way, the immunosuppressive DMTs, are used to hit the immune system early and hard with the idealistic goal of striking the autoimmune activities before the neurological damage becomes irreversible.
Collapse
Affiliation(s)
- Aurora Zanghì
- Department "G.F. Ingrassia", MS Center, University of Catania , Catania, Italy
| | - Emanuele D'Amico
- Department "G.F. Ingrassia", MS Center, University of Catania , Catania, Italy
| | - Francesco Patti
- Department "G.F. Ingrassia", MS Center, University of Catania , Catania, Italy
| |
Collapse
|
23
|
Förster M, Küry P, Aktas O, Warnke C, Havla J, Hohlfeld R, Mares J, Hartung HP, Kremer D. Managing Risks with Immune Therapies in Multiple Sclerosis. Drug Saf 2020; 42:633-647. [PMID: 30607830 DOI: 10.1007/s40264-018-0782-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Since the introduction of the interferons in the 1990s, a multitude of different immunomodulatory and immunosuppressant disease-modifying therapies for multiple sclerosis (MS) have been developed. They have all shown positive effects on clinical endpoints such as relapse rate and disease progression and are a heterogeneous group of therapeutics comprising recombinant pegylated and non-pegylated interferon-β variants, peptide combinations, monoclonal antibodies, and small molecules. However, they have relevant side effect profiles, which necessitate thorough monitoring and straightforward patient education. In individual cases, side effects can be severe and potentially life-threatening, which is why knowledge about (neurological and non-neurological) adverse drug reactions is essential for prescribing neurologists as well as general practitioners. This paper aims to provide an overview of currently available MS therapies, their modes of action and safety profiles, and the necessary therapy monitoring.
Collapse
Affiliation(s)
- Moritz Förster
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Clemens Warnke
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Joachim Havla
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilian-Universität München, Munich, Germany
| | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilian-Universität München, Munich, Germany.,The Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jan Mares
- Department of Neurology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
| | - David Kremer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
24
|
Kürtüncü M, Yılmaz V, Akçay Hİ, Türkoğlu R, Altunrende B, Çınar SA, Ulusoy C, Gündüz T, İçöz S, Kasap M, Çalışkan Z, Ötünç G, Eraksoy M, Tüzün E. Impact of fingolimod on CD4+ T cell subset and cytokine profile of relapsing remitting multiple sclerosis patients. J Neuroimmunol 2019; 337:577065. [PMID: 31526917 DOI: 10.1016/j.jneuroim.2019.577065] [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] [Received: 07/25/2019] [Revised: 09/07/2019] [Accepted: 09/10/2019] [Indexed: 10/26/2022]
Abstract
Fingolimod inhibits the egress of lymphocytes from lymphatic tissues and also directly affects their functions by modulation of the sphingosine-1-phosphate receptor 1 (S1P1). Our aim was to evaluate the impact of fingolimod on diverse CD4+ T cell subsets, and cytokines. Sixty-six relapsing remitting multiple sclerosis (RRMS) patients were treated with oral fingolimod (0.5 mg) for 6 months, and blood samples were collected at baseline, 3 months, and 6 months. Serum levels of seven cytokines and five chemokines were measured by multiplex immunoassay, and frequencies of peripheral blood mononuclear cell subsets were assessed by flow cytometry, and compared with those of 60 healthy controls. CCL2 (p = 0.039), and CCL5 (p = 0.001) levels were significantly higher in fingolimod-treated patients than healthy controls, whereas end-of-study serum levels of IL-6, IL-8, IL-17A, IL-22, IL-23, TNF-α, CXCL10, and CXCL13 were comparable to the baseline levels. Six months of fingolimod treatment reduced CD3+ T cell (mean ± standard deviation, 72.9% ± 5.5 vs. 60.1% ± 11.1, p < 0.001), CD4+ T cell (62.2% ± 8.5 vs. 24.6% ± 12.9, p < 0.001), CD4+CD25hi regulatory T cell (Treg) (3.4% ± 1.3 vs. 2.0% ± 1.4, p < 0.01), and CD19+ B cell (13.2% ± 5.8 vs. 5.3% ± 2.7, p < 0.001) frequencies, while CD8+ T cells (31.8% ± 7.8 vs. 57.8% ± 13.2, p < 0.001) were increased, and NK and NKT cells remained unchanged. The proportions of intracytoplasmic IL-4, IL-10, IFN-γ, and TNF-α-producing T cells were increased, whereas IL-17-producing cells remained relatively constant as measured by flow cytometry. Fingolimod appears to primarily diminish lymphocyte subsets involved in antigen presentation (CD19+ B and CD4+ T cells) rather than immune cells (CD8+ T, NK, and NKT cells) in charge of host defense against pathogens. In contrast, a relative increase is observed in pro- and anti-inflammatory cytokine-producing T helper subsets (IFN-γ, TNF-α, IL-4, and IL-10-producing CD4+ T cells), suggesting that effector T cells are suppressed to a lesser degree by S1P1 modulation.
Collapse
Affiliation(s)
- Murat Kürtüncü
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.
| | - Vuslat Yılmaz
- Department of Neuroscience, Aziz Sancar Institute for Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Halil İbrahim Akçay
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Recai Türkoğlu
- Department of Neurology, Saglik Bilimleri University, Istanbul, Turkey
| | - Burcu Altunrende
- Department of Neurology, Faculty of Medicine, Istanbul Bilim University, Istanbul, Turkey
| | - Suzan Adın Çınar
- Department of Immunology, Aziz Sancar Institute for Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Canan Ulusoy
- Department of Neuroscience, Aziz Sancar Institute for Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Tuncay Gündüz
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sema İçöz
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | | | | | | | - Mefküre Eraksoy
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Erdem Tüzün
- Department of Neuroscience, Aziz Sancar Institute for Experimental Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
25
|
Hemond CC, Glanz BI, Bakshi R, Chitnis T, Healy BC. The neutrophil-to-lymphocyte and monocyte-to-lymphocyte ratios are independently associated with neurological disability and brain atrophy in multiple sclerosis. BMC Neurol 2019; 19:23. [PMID: 30755165 PMCID: PMC6371437 DOI: 10.1186/s12883-019-1245-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 01/30/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Serum hematological indices such as the neutrophil-lymphocyte ratio (NLR) or monocyte-lymphocyte ratio (MLR) have been used as biomarkers of pathogenic inflammation and prognostication in multiple areas of medicine; recent evidence shows correlation with psychological parameters as well. OBJECTIVES/AIMS To characterize clinical, neuroimaging, and psycho-neuro-immunological associations with NLR and MLR in persons with multiple sclerosis (MS). METHODS We identified a large cohort of clinically well-defined patients from our longitudinal database that included MS-related outcomes, disease-modifying therapy, patient-reported outcome (PRO) measures, and quantified cerebral MRI at 1.5 T. We queried hospital records for complete blood counts within 2 months of each clinic visit and excluded those obtained during clinical relapses. Four hundred eighty-three patients, with a mean of 3 longitudinal observations each, were identified who met these criteria. Initial analyses assessed the association between NLR and MLR as the outcomes, and psychological and demographic predictors in univariable and multivariable models controlling for age, gender and treatment. The second set of analyses assessed the association between clinical and MRI outcomes including whole brain atrophy and T2-hyperintense lesion volume, with NLR and MLR as predictors in univariable and multivariable models. All analyses used a mixed effects linear or logistic regression model with repeated measures. RESULTS Unadjusted analyses demonstrated significant associations between higher (log-transformed) NLR (but not MLR) and PRO measures including increasing depression (p = 0.01), fatigue (p < 0.01), and decreased physical quality of life (p < 0.01). Higher NLR and MLR strongly predicted increased MS-related disability as assessed by the Expanded Disability Status Scale, independent of all demographic, clinical, treatment-related, and psychosocial variables (p < 0.001). Lastly, higher NLR and MLR significantly discriminated progressive from relapsing status (p ≤ 0.01 for both), and higher MLR correlated with increased whole-brain atrophy (p < 0.05) but not T2 hyperintense lesion volume (p > 0.05) even after controlling for all clinical and demographic covariates. Sensitivity analyses using a subset of untreated patients (N = 146) corroborated these results. CONCLUSIONS Elevated NLR and MLR may represent hematopoetic bias toward increased production and pro-inflammatory priming of the myeloid innate immune system (numerator) in conjunction with dysregulated adaptive immune processes (denominator), and consequently reflect a complementary and independent marker for severity of MS-related neurological disability and MRI outcomes.
Collapse
Affiliation(s)
- Christopher C. Hemond
- Department of Neurology, University of Massachusetts Medical Center, 55 Lake Ave North, Worcester, MA 01655 USA
- Department of Neurology, Harvard Medical School, Boston, MA USA
- Partners Multiple Sclerosis Center, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital, Boston, MA USA
| | - Bonnie I. Glanz
- Department of Neurology, Harvard Medical School, Boston, MA USA
- Partners Multiple Sclerosis Center, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital, Boston, MA USA
| | - Rohit Bakshi
- Department of Neurology, Harvard Medical School, Boston, MA USA
- Partners Multiple Sclerosis Center, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital, Boston, MA USA
- Department of Radiology, Harvard Medical School, Boston, MA USA
| | - Tanuja Chitnis
- Department of Neurology, Harvard Medical School, Boston, MA USA
- Partners Multiple Sclerosis Center, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital, Boston, MA USA
| | - Brian C. Healy
- Department of Neurology, Harvard Medical School, Boston, MA USA
- Partners Multiple Sclerosis Center, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital, Boston, MA USA
| |
Collapse
|
26
|
Kaufmann M, Haase R, Proschmann U, Ziemssen T, Akgün K. Real World Lab Data: Patterns of Lymphocyte Counts in Fingolimod Treated Patients. Front Immunol 2018; 9:2669. [PMID: 30524432 PMCID: PMC6256977 DOI: 10.3389/fimmu.2018.02669] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022] Open
Abstract
Objective: Fingolimod is approved for the treatment of highly active relapsing remitting multiple sclerosis (MS) patients and acts by its unique mechanism of action via sphingosine-1-phosphate receptor-modulation. Although fingolimod-associated lymphopenia is a well-known phenomenon, the exact cause for the intra- and inter-individual differences of the fluctuation of lymphocyte count and its subtypes is still subject of debate. In this analysis, we aim to estimate the significance of the individual variation of distinct lymphocyte subsets for differences in absolute lymphocyte decrease in fingolimod treated patients and discuss how different lymphocyte subset patterns are related to clinical presentation in a long-term real life setting. Methods/Design: One hundred and thirteen patients with MS were characterized by complete blood cell count and immune cell phentopying of peripheral lymphocyte subsets before, at month 1 and every 3 months up to 36 months of fingolimod treatment. In addition, patients were monitored regarding clinical parameters (relapses, disability, MRI). Results: There was no significant association of baseline lymphocyte count and lymphocyte subtypes with lymphocyte decrease after fingolimod start. The initial drop of the absolute lymphocyte count could not predict the level of lymphocyte count during steady state on fingolimod. Variable CD8+ T cell and NK cell counts account for the remarkable intra- and inter-individual differences regarding initial drop and steady state level of lymphocyte count during fingolimod treatment, whereas CD4+ T cells and B cells mostly present a quite uniform decrease in all treated patients. Selected patients with lymphocyte count >1.0 GPT/l differed by higher CD8+ T cells and NK cell counts compared to lymphopenic patients but presented comparable clinical effectiveness during treatment. Conclusion: Monitoring of the absolute lymphocyte count at steady state seems to be a rough estimate of fingolimod induced lymphocyte redistribution. Our results suggest, that evaluation of distinct lymphocyte subsets as CD4+ T cells allow a more detailed evaluation to weigh and interpret degree of lymphopenia and treatment response in fingolimod treated patients.
Collapse
Affiliation(s)
- Maxi Kaufmann
- MS Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
| | - Rocco Haase
- MS Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
| | - Undine Proschmann
- MS Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
| | - Tjalf Ziemssen
- MS Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
| | - Katja Akgün
- MS Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
| |
Collapse
|
27
|
Rasche L, Paul F. Ozanimod for the treatment of relapsing remitting multiple sclerosis. Expert Opin Pharmacother 2018; 19:2073-2086. [PMID: 30407868 DOI: 10.1080/14656566.2018.1540592] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Ozanimod is a selective sphingosine 1-phosphate receptor 1 and 5 modulator under development by Celgene, for the treatment of relapsing remitting multiple sclerosis. Extensive clinical experience has become available for the related compound fingolimod, favoring the sphingosine 1-phosphate therapeutic concept. Off-target effects have been attributed to its low receptor specificity and have prompted the development of next generation sphingosine 1-phosphate receptor modulators. Areas covered: The authors evaluate the literature of ozanimod, using the PubMed database as well as repositories of the European Committee for Treatment and Research in Multiple Sclerosis and the American and European Academy of Neurology. Specifically, the authors cover and discuss the preclinical data on ozanimod, pharmacokinetics and dynamics, and data on efficacy and safety from the pivotal trials. Expert opinion: Superiority of ozanimod over intramuscular interferon β-1a with regard to reduction in annualized relapse rate and magnetic resonance imaging outcomes has been shown in two phase III trials. The beneficial effect on brain volume and gray matter loss are encouraging and in line with data on other newer immunomodulators. Ozanimod is a valuable contribution to the therapeutic armamentarium in MS, although the effect on disability progression is unclear and requires further investigations.
Collapse
Affiliation(s)
- Ludwig Rasche
- a NeuroCure Clinical Research Center , Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health , Berlin , Germany
| | - Friedemann Paul
- a NeuroCure Clinical Research Center , Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health , Berlin , Germany.,b Experimental and Clinical Research Center , Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health , Berlin , Germany.,c Department of Neurology , Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin , Germany
| |
Collapse
|
28
|
Høglund RA, Polak J, Vartdal F, Holmøy T, Lossius A. B-cell composition in the blood and cerebrospinal fluid of multiple sclerosis patients treated with dimethyl fumarate. Mult Scler Relat Disord 2018; 26:90-95. [PMID: 30243235 DOI: 10.1016/j.msard.2018.08.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/22/2018] [Accepted: 08/30/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND B cells may contribute to the immunopathogenesis of multiple sclerosis (MS). Dimethyl fumarate (DMF) has recently been shown to reduce the frequency of memory B cells in blood, but it is not known whether the drug influences the cellular composition in the cerebrospinal fluid (CSF). METHODS A cross-sectional study examining the cellular composition in blood and cerebrospinal fluid (CSF) from 10 patients treated with DMF and 18 patients receiving other disease modifying drugs or no treatment. RESULTS Patients treated with DMF had reduced proportions of memory B cells in blood compared to other MS patients (p = 0.0007), and the reduction correlated with treatment duration (rs = -0.75, p = 0.021). In the CSF, the absolute number of mononuclear cells were significantly lower in DMF-treated patients compared to the other patients (p = 0.023), and there was a disproportionate decrease of plasmablasts (p = 0.031). CONCLUSION The results of this exploratory study support a B-cell mediated mechanism of action for DMF in both blood and CSF.
Collapse
Affiliation(s)
- Rune A Høglund
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Justyna Polak
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology and Transfusion Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Frode Vartdal
- Department of Immunology and Transfusion Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trygve Holmøy
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andreas Lossius
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway; Department of Immunology and Transfusion Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
29
|
Characterization of lymphopenia in patients with MS treated with dimethyl fumarate and fingolimod. NEUROLOGY - NEUROIMMUNOLOGY NEUROINFLAMMATION 2018; 5:e445. [PMID: 29417951 PMCID: PMC5795901 DOI: 10.1212/nxi.0000000000000445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|