1
|
Zeydan B, Neyal N, Nathoo N, Rangachari M, Atkinson EJ, Son J, Conway BL, Tobin WO, Keegan BM, Weinshenker BG, Kantarci K, Oh J, Kantarci OH. Effects of androgen modifying therapies on disease activity in older men with multiple sclerosis. J Neuroimmunol 2025; 403:578589. [PMID: 40139128 PMCID: PMC12048229 DOI: 10.1016/j.jneuroim.2025.578589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Anti-inflammatory properties of androgens were assessed in animal models, but only several clinical studies investigated the effects of androgen on multiple sclerosis (MS). Inflammatory activity in MS often attenuates with aging, and androgen modifying therapies (AMT), which mimic decreased androgen levels, are frequently used in older men. We aimed to investigate if the number of disease activity events would increase in older (≥55 years) male persons with MS (MPwMS) who are on AMT. METHODS MPwMS with AMT initiation ≥55 years (AMT; n = 60) and without AMT history (no-AMT; n = 80) were included from a clinical-based observational study cohort. Clinical (relapses), Radiological (new lesions), and Disease (relapses and/or new lesions) activity events were evaluated before and after the age at AMT initiation in AMT and no-AMT groups. RESULTS Age at MS onset, progressive MS rate and treatment frequencies were similar between the groups. When events before and after the age at AMT initiation (mean = 65.0 ± 7.0 years) were compared, there was a drop in the percentage of individuals with Clinical (38.5 % vs. 10 %), Radiological (46.2 % vs. 40 %) and Disease (62.8 % vs. 40 %) activity events in the no-AMT group. In the AMT group, the percentage of individuals with Clinical activity events was not as dramatically decreased (33.3 % vs. 22.2 %), Radiological activity events was increased (35.2 % vs. 46.7 %), and Disease activity events was sustained (51.9 % vs. 51.1 %). The probability of Disease activity was higher in the AMT group at 3 years of AMT initiation (40 % vs. 29 %) compared to the no-AMT group matched for disease duration, but the difference was not significant (p = 0.582). CONCLUSIONS Rather than the expected decrease in disease activity with age, MPwMS receiving AMT experienced sustained or increased disease activity, particularly at >65 years. Close clinical monitoring of MPwMS starting on these medications is necessary.
Collapse
Affiliation(s)
- Burcu Zeydan
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Radiology, Mayo Clinic, Rochester, MN, USA; Women's Health Research Center, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA.
| | - Nur Neyal
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Nabeela Nathoo
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Manu Rangachari
- Centre de recherche du CHU de Québec, axe Neurosciences, Université Laval, Québec City, QC, Canada; Faculté de médecine, Université Laval, Québec City, QC, Canada
| | | | - Jiye Son
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Brittani L Conway
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - W Oliver Tobin
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - B Mark Keegan
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - Brian G Weinshenker
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, University of Virginia, VA, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA; Women's Health Research Center, Mayo Clinic, Rochester, MN, USA
| | - Jiwon Oh
- Department of Neurology, University of Toronto, Toronto, ON, Canada
| | - Orhun H Kantarci
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
2
|
Simonsen CS, Moen MH, Celius EG. Lower threshold for hormone therapy in women with multiple sclerosis. TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2025; 145:25-0070. [PMID: 40272242 DOI: 10.4045/tidsskr.25.0070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025] Open
|
3
|
Lorefice L, Ferraro E. Does menopause affect MS progression? Evidence and ongoing debates. Neurodegener Dis Manag 2025; 15:117-119. [PMID: 40194957 PMCID: PMC12118413 DOI: 10.1080/17582024.2025.2489878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025] Open
Affiliation(s)
- L Lorefice
- Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, Cagliari, Italy
| | - E Ferraro
- Multiple Sclerosis Center, S. Filippo Neri Hospital, Rome, Italy
| |
Collapse
|
4
|
Silverman HE, Bostrom A, Nylander AN, Akula A, Lazar AA, Gomez R, Santaniello A, Renschen A, Harms MM, Cooper TP, Lincoln R, Poole S, Abdelhak A, Henry RG, Oksenberg J, Hauser SL, Cree BAC, Bove R. Association of Menopause With Functional Outcomes and Disease Biomarkers in Women With Multiple Sclerosis. Neurology 2025; 104:e210228. [PMID: 39715474 PMCID: PMC11666275 DOI: 10.1212/wnl.0000000000210228] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/30/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVE The impact of menopause on the brain is not well understood. Hormonal changes, including puberty and pregnancy, influence the onset and course of multiple sclerosis (MS). After menopause, a worsening of MS disease trajectory measured on the clinician-rated Expanded Disability Status Scale (EDSS) was reported in some, but not all, studies. Evaluating the association between menopause and more objective measures of CNS injury is warranted. This study sought to assess the trajectory of objective functional outcomes and disease biomarkers in women with MS before and after menopause in a longitudinal prospective observational cohort. METHODS Data were collected prospectively from a longitudinally followed MS cohort, including the performance-based Multiple Sclerosis Functional Composite (MSFC) as the primary functional outcome and the paraclinical marker of neuronal injury serum neurofilament light chain (sNfL) as the primary biomarker outcome. Outcomes were analyzed using segmented linear mixed model regressions adjusted for age, BMI, and tobacco use, with a change in slope at the time of menopause, as the a priori inflection point. RESULTS One hundred and eighty-four postmenopausal women met inclusion criteria. Participants were followed for a median of 13 years (interquartile range [IQR] = 4, range: 1-17). The median MS duration was 24 years (IQR = 13, range: 3-64), and the median EDSS score was 2.5 (IQR = 2, range: 0-8). The median age at natural menopause was 50 years (IQR = 5, range: 33-60); 17% of participants used any systemic menopausal hormone therapy. Menopause reflected an inflection point in MSFC worsening (slope difference 0.08, 95% CI 0.01, 0.14, p = 0.0163) and increase in serum neurofilament light chain (slope difference -0.95, 95% CI -1.74 to -0.16, p = 0.0194) while the opposite was found for EDSS (slope difference 0.05, 95% CI 0.01-0.09, p = 0.0200). Findings remained significant after adjustment for multiple covariates. When using additional nonlinear regression modeling, similar inflection points were found (within 3 years of the final menstrual period) for sNfL and EDSS but not MSFC. DISCUSSION The menopausal transition may represent an inflection in accumulation of neuronal injury and functional decline in MS.
Collapse
Affiliation(s)
- Hannah E Silverman
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Alan Bostrom
- Division of Oral Epidemiology, Department of Preventive and Restorative Dental Sciences, University of California, San Francisco;and
| | - Alyssa N Nylander
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Amit Akula
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Ann A Lazar
- Division of Oral Epidemiology, Department of Preventive and Restorative Dental Sciences, University of California, San Francisco;and
- Division of Biostatistics, Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Refujia Gomez
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Adam Santaniello
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Adam Renschen
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Meagan Michaela Harms
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Tiffany P Cooper
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Robin Lincoln
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Shane Poole
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Ahmed Abdelhak
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Roland G Henry
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Jorge Oksenberg
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Stephen L Hauser
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| | | | - Riley Bove
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco
| |
Collapse
|
5
|
Traiffort E, Kassoussi A, Zahaf A. Revisiting the role of sexual hormones in the demyelinated central nervous system. Front Neuroendocrinol 2025; 76:101172. [PMID: 39694337 DOI: 10.1016/j.yfrne.2024.101172] [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: 07/03/2024] [Revised: 12/09/2024] [Accepted: 12/15/2024] [Indexed: 12/20/2024]
Abstract
Sex-related differences characterize multiple sclerosis, an autoimmune, inflammatory and neurodegenerative disease displaying higher incidence in females as well as discrepancies in susceptibility and progression. Besides clinical specificities, molecular and cellular differences related to sex hormones were progressively uncovered improving our understanding of the mechanisms involved in this disabling disease. The most recent findings may give rise to the identification of novel therapeutic perspectives that could meet the urgent need for a treatment preventing the transition from the recurrent- to the progressive form of the disease. The present review is an update of our current knowledge about progestagens, androgens and estrogens in the context of CNS demyelination including their synthesis, the impact of their dysregulation, the preclinical and clinical data presently available, the main molecular dimorphisms related to these hormones and their age-related changes and relationship with failure of spontaneous remyelination, likely impacting the inexorable progression of multiple sclerosis towards irreversible disabilities.
Collapse
Affiliation(s)
| | | | - Amina Zahaf
- U1195 Inserm, Paris-Saclay University, Kremlin-Bicêtre, France
| |
Collapse
|
6
|
Simonsen CS, Flemmen HØ, Broch L, Myklebust H, Berg‐Hansen P, Brunborg C, Celius EG. The influence of menopause on multiple sclerosis. Eur J Neurol 2025; 32:e16566. [PMID: 39601490 PMCID: PMC11625929 DOI: 10.1111/ene.16566] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/25/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
INTRODUCTION One third of the multiple sclerosis (MS) population consists of peri- or postmenopausal women. Many symptoms of menopause overlap those of MS. Some studies show increased speed of disability progression after menopause, while others indicate an unaltered trajectory. OBJECTIVE Determine the association between menopause and MS disease course. METHODS Cohort study with clinical data collected prospectively. Self-reported age of menopause, smoking and parity collected retrospectively. RESULTS We included 559 peri-/postmenopausal women and 386 similarly aged men. There was no significant difference in EDSS progression (slope coef 0.03, 0.02-0.08, p = 0.208) or annual relapse rate (ARR) (0.10, 0.29-0.10, p = 0.324) in the 5 years before and after menopause. Women's EDSS progressed slower than men's after menopause (coef -0.02, 95% CI -0.04 to -0.002, p = 0.032), but there was no difference in ARR [coef. -0.01, 95% CI -0.04 to -0.01, p = 0.262]. Women who reached menopause before MS onset had shorter time to diagnosis than women who reached menopause after onset (3.1 years (3.1) vs. 7.4 years (8.1), p < 0.001). Women who reached menopause before diagnosis had an even longer time to diagnosis (8.8 (9.3) vs. 5.5 (6.3), p < 0.001). CONCLUSIONS There were no significant differences in EDSS progression or ARR before and after menopause. In fact, men progressed faster than women, suggesting there is no negative impact of menopause on disease progression, as measured by EDSS and relapses.
Collapse
Affiliation(s)
| | | | - Line Broch
- Department of NeurologyVestre Viken Hospital TrustDrammenNorway
- Department of NeurologyOslo University HospitalOsloNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Harald Myklebust
- Department of NeurologyVestre Viken Hospital TrustDrammenNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| | | | - Cathrine Brunborg
- Oslo Centre for Biostatistics and Epidemiology, Research Support ServicesOslo University HospitalOsloNorway
| | - Elisabeth Gulowsen Celius
- Department of NeurologyOslo University HospitalOsloNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| |
Collapse
|
7
|
Magyari M. All women with multiple sclerosis should start hormone replacement therapy at menopause unless contraindicated: No. Mult Scler 2024; 30:1109-1111. [PMID: 38907634 DOI: 10.1177/13524585241254987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Affiliation(s)
- Melinda Magyari
- The Danish Multiple Sclerosis Registry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
DiMauro KA, Swetlik C, Cohen JA. Management of multiple sclerosis in older adults: review of current evidence and future perspectives. J Neurol 2024; 271:3794-3805. [PMID: 38689068 PMCID: PMC11233312 DOI: 10.1007/s00415-024-12384-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/02/2024] [Accepted: 04/14/2024] [Indexed: 05/02/2024]
Abstract
IMPORTANCE The prevalence of multiple sclerosis (MS) and aging MS patients is increasing worldwide. There is a need to better understand this MS sub-population, which historically is underrepresented in the literature. This narrative review examines the evolving demographics, disease course, and treatments for older adults with MS (OAMS) to address current knowledge gaps and highlight areas critical for future research. OBSERVATIONS OAMS populations require special consideration by clinicians. Older individuals have different care needs than individuals with adult onset MS who are mid-life or younger. Comorbidities, an aging immune system, increasing neurodegeneration, decreasing neurologic reserve, changing benefit/risk relationship for disease modifying therapies (DMTs), and wellness require special attention to provide holistic comprehensive care. Active areas of research include potential cessation of DMTs and novel disease targets. CONCLUSIONS AND RELEVANCE This review highlights both the current knowledge and information gaps in the literature that are critical to understanding and properly managing OAMS. The aims are to inform MS clinicians in their current practice, as well as inspire future studies which are critical to providing quality and evidence-based care for OAMS.
Collapse
Affiliation(s)
- Kimberly A DiMauro
- Mellen Center for MS Treatment and Research, Cleveland Clinic, Neurological Institute, Cleveland, OH, USA
| | - Carol Swetlik
- Mellen Center for MS Treatment and Research, Cleveland Clinic, Neurological Institute, Cleveland, OH, USA
| | - Jeffrey A Cohen
- Mellen Center for MS Treatment and Research, Cleveland Clinic, Neurological Institute, Cleveland, OH, USA.
| |
Collapse
|
9
|
Winschel I, Willing A, Engler JB, Walkenhorst M, Meurs N, Binkle-Ladisch L, Woo MS, Pfeffer LK, Sonner JK, Borgmeyer U, Hagen SH, Grünhagel B, Claussen JM, Altfeld M, Friese MA. Sex- and species-specific contribution of CD99 to T cell costimulation during multiple sclerosis. Biol Sex Differ 2024; 15:41. [PMID: 38750588 PMCID: PMC11097467 DOI: 10.1186/s13293-024-00618-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Differences in immune responses between women and men are leading to a strong sex bias in the incidence of autoimmune diseases that predominantly affect women, such as multiple sclerosis (MS). MS manifests in more than twice as many women, making sex one of the most important risk factor. However, it is incompletely understood which genes contribute to sex differences in autoimmune incidence. To address that, we conducted a gene expression analysis in female and male human spleen and identified the transmembrane protein CD99 as one of the most significantly differentially expressed genes with marked increase in men. CD99 has been reported to participate in immune cell transmigration and T cell regulation, but sex-specific implications have not been comprehensively investigated. METHODS In this study, we conducted a gene expression analysis in female and male human spleen using the Genotype-Tissue Expression (GTEx) project dataset to identify differentially expressed genes between women and men. After successful validation on protein level of human immune cell subsets, we assessed hormonal regulation of CD99 as well as its implication on T cell regulation in primary human T cells and Jurkat T cells. In addition, we performed in vivo assays in wildtype mice and in Cd99-deficient mice to further analyze functional consequences of differential CD99 expression. RESULTS Here, we found higher CD99 gene expression in male human spleens compared to females and confirmed this expression difference on protein level on the surface of T cells and pDCs. Androgens are likely dispensable as the cause shown by in vitro assays and ex vivo analysis of trans men samples. In cerebrospinal fluid, CD99 was higher on T cells compared to blood. Of note, male MS patients had lower CD99 levels on CD4+ T cells in the CSF, unlike controls. By contrast, both sexes had similar CD99 expression in mice and Cd99-deficient mice showed equal susceptibility to experimental autoimmune encephalomyelitis compared to wildtypes. Functionally, CD99 increased upon human T cell activation and inhibited T cell proliferation after blockade. Accordingly, CD99-deficient Jurkat T cells showed decreased cell proliferation and cluster formation, rescued by CD99 reintroduction. CONCLUSIONS Our results demonstrate that CD99 is sex-specifically regulated in healthy individuals and MS patients and that it is involved in T cell costimulation in humans but not in mice. CD99 could potentially contribute to MS incidence and susceptibility in a sex-specific manner.
Collapse
Affiliation(s)
- Ingo Winschel
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Willing
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mark Walkenhorst
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina Meurs
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lena Kristina Pfeffer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Uwe Borgmeyer
- Center of Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sven Hendrik Hagen
- Research Department Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Benjamin Grünhagel
- Research Department Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Janna M Claussen
- Research Department Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Marcus Altfeld
- Research Department Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
10
|
Juutinen L, Ahinko K, Hagman S, Basnyat P, Jääskeläinen O, Herukka SK, Sumelahti ML. The association of menopausal hormone levels with progression-related biomarkers in multiple sclerosis. Mult Scler Relat Disord 2024; 85:105517. [PMID: 38442501 DOI: 10.1016/j.msard.2024.105517] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/06/2024] [Accepted: 02/24/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) progression coincides temporally with menopause. However, it remains unclear whether the changes in disease course are related to the changes in reproductive hormone concentrations. We assessed the association of menopausal hormonal levels with progression-related biomarkers of MS and evaluated the changes in serum neurofilament light chain (sNfL) and glial fibrillary acidic protein (sGFAP) levels during menopausal hormone therapy (MHT) in a prospective baseline-controlled design. METHODS The baseline serum estradiol, follicle stimulating hormone, and luteinizing hormone levels were measured from menopausal women with MS (n = 16) and healthy controls (HC, n = 15). SNfL and sGFAP were measured by single-molecule array. The associations of hormone levels with sNfL and sGFAP, and with Expanded Disability Status Scale (EDSS) and lesion load and whole brain volumes (WBV) in MRI were analyzed with Spearman's rank correlation and age-adjusted linear regression model. Changes in sNfL and sGFAP during one-year treatment with estradiol hemihydrate combined with cyclic dydrogesterone were assessed with Wilcoxon Signed Ranks Test. RESULTS In MS group, baseline estradiol had a positive correlation with WBV in MRI and an inverse correlation with lesion load, sNfL and sGFAP, but no correlation with EDSS. The associations of low estradiol with high sGFAP and low WBV were independent of age. During MHT, there was no significant change in sNfL and sGFAP levels in MS group while in HC, sGFAP slightly decreased at three months but returned to baseline at 12 months. CONCLUSION Our preliminary findings suggest that low estradiol in menopausal women with MS has an age-independent association with more pronounced brain atrophy and higher sGFAP and thus advanced astrogliosis which could partially explain the more rapid progression of MS after menopause. One year of MHT did not alter the sGFAP or sNfL levels in women with MS.
Collapse
Affiliation(s)
- Laura Juutinen
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere University, Finland; Department of Neurosciences and Rehabilitation, Tampere University Hospital, P.O. Box 2000, FI, 33521, Tampere, Finland.
| | - Katja Ahinko
- Department of Obstetrics and Gynecology, Tampere University Hospital, P.O. Box 2000, FI, 33521 Tampere, Finland
| | - Sanna Hagman
- Neuroimmunology Research Group, Faculty of Medicine and Health Technology, Tampere University, FI, 33014 Tampere University, Finland
| | - Pabitra Basnyat
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere University, Finland
| | - Olli Jääskeläinen
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
| | - Sanna-Kaisa Herukka
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland; Department of Neurology, Kuopio University Hospital, P.O. Box 1711, 70211, Kuopio, Finland
| | - Marja-Liisa Sumelahti
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere University, Finland
| |
Collapse
|
11
|
Yaghoobpoor S, Fathi M, Vakili K, Sayehmiri F, Alipour M, Miriran ZS, Ghayyem H, Tutunchian Z, Hajibeygi R, Batool Z, Mirzadeh M, Aghazadeh MH, Hajiesmaeili M. Insulin-like growth factor-1 (IGF-1) levels in multiple sclerosis patients: A systematic review and meta-analysis. PLoS One 2024; 19:e0297091. [PMID: 38630771 PMCID: PMC11023272 DOI: 10.1371/journal.pone.0297091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 12/23/2023] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Multiple sclerosis (MS) is a chronic progressive autoimmune disorder of the central nervous system (CNS) that can cause inflammation, demyelination, and axon degeneration. Insulin-like growth factor-1 (IGF-1) is a single-chain polypeptide mainly synthesized in the liver and brain. IGF-1 causes neuronal and non-neuronal cell proliferation, survival, and differentiation. Therefore, it can be used in treating neuro-demyelinating diseases such as MS. The current systematic review and meta-analysis aims to compare the levels of IGF-1 in MS patients and healthy controls and also investigates IGF binding proteins (IGF-BP) and growth hormone (GH) levels between MS patients and healthy controls. METHODS In this study, we systematically searched electronic databases of PubMed, Scopus, Web of Science (WOS), and Google Scholar, up to December 2022. Studies that measured IGF-1, GH, IGFBP-1, IGFBP-2, or IGFBP-3 in MS patients and healthy controls in either blood or cerebral spinal fluid (CSF) were identified. We calculated Standardized mean differences (SMD) to compare levels of IGF-1, GH, IGFBP-1, IGFBP-2, or IGFBP-3 in MS patients and controls. RESULTS Finally, we included 11 eligible studies from 1998 to 2018. The sample size of included studies varied from 20 to 200 resulting in a total sample size of 1067 individuals, 531 MS patients, and 536 healthy controls. The mean age of the patient and control groups were 38.96 and 39.38, respectively. The average EDSS among patients was 4.56. We found that blood levels of IGF-1 (SMD = 0.20, 95% CI = -0.20 to 0.59, I2 = 82.4%, K = 8, n = 692), CSF level of IGF-1 (SMD = 0.25, 95% CI = -0.06 to 0.56, I2 = 0.0%, K = 3 n = 164) and blood levels of GH were not significantly higher in MS patients than controls (SMD = 0.08, 95% CI = -0.33 to 0.49, I2 = 77.0% K = 3, n = 421). Moreover, the blood levels of IGFBP-1 (SMD = 0.70, 95% CI = 0.01 to 1.40, I2 = 77%, K = 4, n = 255) were significantly higher in MS cases than in controls. However, the blood levels of IGFBP-2 (SMD = 0.43, 95% CI = -0.34 to 1.21, I2 = 64.2%, K = 3, n = 78) and blood levels of IGFBP-3 (SMD = 1.04, 95% CI = -0.09 to 2.17, I2 = 95.6%, K = 6, n = 443) were not significantly higher in patients than controls. CONCLUSION Our meta-analysis revealed no significant difference in serum levels of IGF-1, GH, IGFBP-2, and IGFBP-3 between the MS group and healthy controls, except for IGFBP1. However, our systematic review showed that the studies were controversial for IGFBP-3 serum levels. Some studies found an increase in serum level of IGFBP-3 in MS patients compared to the healthy group, while others showed a decrease.
Collapse
Affiliation(s)
- Shirin Yaghoobpoor
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sayehmiri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Milad Alipour
- Medical Student, Department of Medicine, Islamic Azad University Tehran Medical Sciences, Tehran, Iran
| | | | - Hani Ghayyem
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zohreh Tutunchian
- Advanced Diagnostic and Interventional Radiology Research Center(ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Ramtin Hajibeygi
- Advanced Diagnostic and Interventional Radiology Research Center(ADIR), Tehran University of Medical Science, Tehran, Iran
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Moein Mirzadeh
- Department of Surgery, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mohammadreza Hajiesmaeili
- Anesthesia and Critical Care Department, Critical Care Quality Improvement Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Goyne CE, Fair AE, Sumowski PE, Graves JS. The Impact of Aging on Multiple Sclerosis. Curr Neurol Neurosci Rep 2024; 24:83-93. [PMID: 38416310 DOI: 10.1007/s11910-024-01333-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/29/2024]
Abstract
PURPOSE OF REVIEW Multiple sclerosis (MS) is a chronic, immune-mediated demyelinating disorder of the central nervous system. Age is one of the most important factors in determining MS phenotype. This review provides an overview of how age influences MS clinical characteristics, pathology, and treatment. RECENT FINDINGS New methods for measuring aging have improved our understanding of the aging process in MS. New studies have characterized the molecular and cellular composition of chronic active or smoldering plaques in MS. These lesions are important contributors to disability progression in MS. These studies highlight the important role of immunosenescence and the innate immune system in sustaining chronic inflammation. Given these changes in immune function, several studies have assessed optimal treatment strategies in aging individuals with MS. MS phenotype is intimately linked with chronologic age and immunosenescence. While there are many unanswered questions, there has been much progress in understanding this relationship which may lead to more effective treatments for progressive disease.
Collapse
Affiliation(s)
- Christopher E Goyne
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA
| | - Ashley E Fair
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA
| | - Paige E Sumowski
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA
| | - Jennifer S Graves
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA.
| |
Collapse
|
13
|
Ross L, Finlayson M, Amato MP, Cohen JA, Hellwig K, Tintore M, Vukusic S, Salter A, Marrie RA. Priority setting: women's health topics in multiple sclerosis. Front Neurol 2024; 15:1355817. [PMID: 38440114 PMCID: PMC10910071 DOI: 10.3389/fneur.2024.1355817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/06/2024] [Indexed: 03/06/2024] Open
Abstract
Background A scoping review found that most studies on women's health in multiple sclerosis (MS) focused on pregnancy, fetal/neonatal outcomes and sexual dysfunction. Few studies addressed menopause, contraception, gynecologic cancers/cancer screening. However, the perceived relative importance of these knowledge gaps to people living with MS and other partners is unknown. We engaged a range of partners, including people living with MS, health care providers, researchers, and patient advocacy groups, to set priorities for future research in women's health in MS. Methods We employed a three-step global engagement process. First, we identified which broad research topics relevant to women's health in MS were of highest priority using two surveys. Second, we developed specific research questions within these topics using focus groups. Finally, we prioritized the research questions with a third survey. Results Overall, 5,266 individuals responded to the initial surveys [n = 1,430 global survey, mean (SD) age 50.0 (12.6), all continents; n = 3,836 North American Research Committee on Multiple Sclerosis survey, mean (SD) age 64.8 (9.6), United States]. Menopause, sexual dysfunction, pregnancy, gynecologic cancer/cancer screening, hormones and parenthood were identified as the most important topics. Focus groups generated 80 potential research questions related to these topics. In the final survey 712 individuals prioritized these questions. The highest priority questions in each research topic were: (i) How do perimenopause and menopause affect disease activity, course, response to disease-modifying treatment and quality of life in MS; (ii) What are the most effective strategies for managing issues around sexual intimacy, including related to low sexual desire, changes in physical function, and MS symptoms; (iii) Are there long-term effects of disease-modifying therapies on the children of persons with MS; (iv) What are the short and long-term effects of disease-modifying drugs on gynecologic cancer risk, particularly for high efficacy disease-modifying drugs and hematopoietic stem cell transplantation; (v) Are there hormone related treatments that can stabilize fluctuations in MS symptoms; and (vi) How does MS fatigue impact parenting strategies. Conclusion Priorities for research relating to women's health issues for persons with MS have been delineated using a collaborative process with key partners. Alignment of future research with these priorities should be monitored.
Collapse
Affiliation(s)
- Lindsay Ross
- Department of Neurology, Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Marcia Finlayson
- School of Rehabilitation Therapy, Queen’s University, Kingston, ON, Canada
| | - Maria Pia Amato
- Department of NEUROFARBA, Section of Neurosciences, University of Florence, Florence, Italy
- IRCCS Fondazione Don Carol Gnocchi, Florence, Italy
| | - Jeffrey Alan Cohen
- Department of Neurology, Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Kerstin Hellwig
- Department of Neurology, Katholische Klinikum, Ruhr University, Bochum, Germany
| | - Mar Tintore
- Multiple Sclerosis Centre of Catalonia, Department of Neurology, Hospital Universitari Vall d’Hebron, Universitat Autonoma de Barcelona, Universitat de Vic-Universitat Central de Catalunya, Barcelona, Spain
| | - Sandra Vukusic
- Service de Neurologie, sclérose en plaques, pathologies de la myéline et neuro-inflammation-et Fondation Eugène Devic EDMUS pour la Sclérose en Plaques, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Lyon, France
- Centre des Neurosciences de Lyon, INSERM 1028 et CNRS UMR5292, Observatoire Français de la Sclérose en Plaque, Lyon, France
- Université Claude Bernard Lyon 1, Lyon, France
| | - Amber Salter
- Department of Neurology, Section on Statistical Planning and Analysis, UT Southwestern Medical Center, Dallas, TX, United States
| | - Ruth Ann Marrie
- Department of Neurology, Section on Statistical Planning and Analysis, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Department of Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
14
|
Abstract
Multiple sclerosis has a 3:1 female-to-male predominance and commonly presents in young adult women. The hormonal changes in women throughout their lifetime do affect the underlying pathology of multiple sclerosis, and the needs of women therefore change with age. Although multiple sclerosis does not adversely affect fertility or pregnancy, there are many factors to consider when caring for women throughout family planning, pregnancy, and the postpartum period. The care of these women and complex decisions regarding disease-modifying therapy use in family planning should be individualized and comprehensive.
Collapse
Affiliation(s)
- Riley Bove
- UCSF Weill Institute for Neurosciences, 1651 Fourth Street, San Francisco, CA 94158, USA
| | - Paige Sutton
- OhioHealth Multiple Sclerosis Center, 3535 Olentangy River Road, Columbus, OH 43214, USA.
| | - Jacqueline Nicholas
- OhioHealth Multiple Sclerosis Center, 3535 Olentangy River Road, Columbus, OH 43214, USA
| |
Collapse
|
15
|
Siddiqui A, Yang JH, Hua LH, Graves JS. Clinical and Treatment Considerations for the Pediatric and Aging Patients with Multiple Sclerosis. Neurol Clin 2024; 42:255-274. [PMID: 37980118 DOI: 10.1016/j.ncl.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2023]
Abstract
Chronologic aging is associated with multiple pathologic and immunologic changes that impact the clinical course of multiple sclerosis (MS). Clinical phenotypes evolve across the lifespan, from a highly inflammatory course in the very young to a predominantly neurodegenerative phenotype in older patients. Thus, unique clinical considerations arise for the diagnosis and management of the two age extremes of pediatric and geriatric MS populations. This review covers epidemiology, diagnosis, and treatment strategies for these populations with nuanced discussions on therapeutic approaches to effectively care for patients living with MS at critical transition points during their lifespan.
Collapse
Affiliation(s)
- Areeba Siddiqui
- Cleveland Clinic Lou Ruvo Center for Brain Health, 888 W. Bonneville Avenue, Las Vegas, NV 89106, USA
| | - Jennifer H Yang
- Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, Mail Code 0662, La Jolla, CA 92093, USA; Division of Pediatric Neurology, Rady Children's Hospital, 3020 Children's Way MC 5009, San Diego, CA 92123, USA
| | - Le H Hua
- Cleveland Clinic Lou Ruvo Center for Brain Health, 888 W. Bonneville Avenue, Las Vegas, NV 89106, USA.
| | - Jennifer S Graves
- Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, Mail Code 0662, La Jolla, CA 92093, USA; Division of Pediatric Neurology, Rady Children's Hospital, 3020 Children's Way MC 5009, San Diego, CA 92123, USA
| |
Collapse
|
16
|
Giovannoni G, Ford HL, Schmierer K, Middleton R, Stennett AM, Pomeroy I, Fisniku L, Scalfari A, Bannon C, Stross R, Hughes S, Williams A, Josephs S, Peel C, Straukiene A. MS care: integrating advanced therapies and holistic management. Front Neurol 2024; 14:1286122. [PMID: 38351950 PMCID: PMC10862341 DOI: 10.3389/fneur.2023.1286122] [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: 08/30/2023] [Accepted: 11/23/2023] [Indexed: 02/16/2024] Open
Abstract
Lifestyle and environmental factors are key determinants in disease causality and progression in neurological conditions, including multiple sclerosis (MS). Lack of exercise, poor diet, tobacco smoking, excessive alcohol intake, social determinants of health, concomitant medications, poor sleep and comorbidities can exacerbate MS pathological processes by impacting brain health and depleting neurological reserves, resulting in more rapid disease worsening. In addition to using disease-modifying therapies to alter the disease course, therapeutic strategies in MS should aim to preserve as much neurological reserve as possible by promoting the adoption of a "brain-healthy" and "metabolically-healthy" lifestyle. Here, we recommend self-regulated lifestyle modifications that have the potential to improve brain health, directly impact on disease progression and improve outcomes in people with MS. We emphasise the importance of self-management and adopting a multidisciplinary, collaborative and person-centred approach to care that encompasses the healthcare team, family members and community support groups.
Collapse
Affiliation(s)
- Gavin Giovannoni
- Centre for Neuroscience, Surgery and Trauma, Faculty of Medicine and Dentistry, The Blizard Institute, Queen Mary University of London, London, United Kingdom
- Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Helen L. Ford
- Leeds Teaching Hospitals, University of Leeds, Leeds, United Kingdom
| | - Klaus Schmierer
- Centre for Neuroscience, Surgery and Trauma, Faculty of Medicine and Dentistry, The Blizard Institute, Queen Mary University of London, London, United Kingdom
- Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Rod Middleton
- Disease Registers & Data Research in Health Data Science, Swansea University Medical School, Swansea, United Kingdom
| | - Andrea M. Stennett
- Centre for Neuroscience, Surgery and Trauma, Faculty of Medicine and Dentistry, The Blizard Institute, Queen Mary University of London, London, United Kingdom
- Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Ian Pomeroy
- The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
- Department of Neurology, University of Liverpool, Liverpool, United Kingdom
| | - Leonora Fisniku
- Department of Neurosciences (Addenbrooke’s), Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Antonio Scalfari
- Centre of Neuroscience, Department of Medicine, Imperial College London, Charing Cross Hospital, London, United Kingdom
| | | | - Ruth Stross
- Neurology Academy, Sheffield, United Kingdom
- Kingston Hospitals NHS Foundation Trust, Surrey, United Kingdom
| | - Sarah Hughes
- Torbay and South Devon NHS Foundation Trust, Torquay, United Kingdom
| | - Adam Williams
- Devon Partnership NHS Trust, Paignton, United Kingdom
| | | | | | - Agne Straukiene
- Torbay and South Devon NHS Foundation Trust, Torquay, United Kingdom
- University of Plymouth, Plymouth, United Kingdom
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Given the potential for exogenous hormones to influence risk and course of MS, this narrative review aims to summarize current knowledge from observational and interventional studies of exogenous hormones in humans with MS. RECENT FINDINGS Large randomized clinical trials for combined oral contraceptives and estriol both show modest effect on inflammatory activity, with the latter showing potential neuroprotective effect. After fertility treatment, large actively treated cohorts have not confirmed any elevated risk of relapse. Preclinical data suggest that androgens, selective estrogen receptor modulators (SERMs), and selective androgen receptor modulators (SARMs) may be neuroprotective but clinical data are lacking. Gender affirming treatment, particularly estrogen in trans-women, could possibly be associated with elevated risk of inflammation. For women with MS entering menopause, hormone therapy appears safe during the appropriate menopausal window, but its long-term effects on neuroprotection are unknown. Exogenous hormones, used in varied doses and for diverse indications, have variable effects on MS risk, inflammatory activity, and neuroprotection. Large randomized trials are needed before it is possible to determine the true effect of exogenous hormones in a condition as complex as MS.
Collapse
Affiliation(s)
- Stephanie Hsu
- UCSF Weill Institute for Neuroscience, Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Riley Bove
- UCSF Weill Institute for Neuroscience, Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
18
|
Neyal N, Atkinson EJ, Smith CY, Weis DM, Rocca LG, Rocca WA, Kantarci K, Kantarci OH, Zeydan B. Smoking, early menopause and multiple sclerosis disease course. Climacteric 2023; 26:560-564. [PMID: 37387356 PMCID: PMC10756919 DOI: 10.1080/13697137.2023.2221381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/10/2023] [Accepted: 05/27/2023] [Indexed: 07/01/2023]
Abstract
Smoking is associated with an increased risk of multiple sclerosis (MS), and smoking and early menopause are related to poor outcomes in MS. Smoking is also associated with early menopause. To explore this intricate relationship between smoking status, age at menopause and disease course in MS, 137 women with MS and 396 age-matched controls were included in this case-control study. Age at menopause (median 49.0 vs. 50.0 years; p = 0.79) and smoking status (40.3% vs. 47.6%; p = 0.15) were similar among MS and control women. Relapsing MS onset was earlier in ever-smoker women with early menopause compared to the rest of the women (median 30.4 vs. 37.0 years; p = 0.02) and also compared to ever-smoker women with normal age at menopause (median 30.4 vs. 41.0 years; p = 0.008) and never-smoker women with early menopause (median 30.4 vs. 41.5 years; p = 0.004). Progressive MS onset was also earlier in ever-smoker women with early menopause compared to ever-smoker women with normal age at menopause (median 41.1 vs. 49.4 years; p = 0.05) and never-smoker women with early menopause (median 41.1 vs. 50.1 years; p = 0.12). Our results suggest that smoking and menopause associate with MS disease course, including the onset of relapsing and progressive MS in women.
Collapse
Affiliation(s)
- Nur Neyal
- Mayo Clinic, Department of Neurology, Rochester, MN, United States
- Mayo Clinic, Department of Radiology, Rochester, MN, United States
| | - Elizabeth J. Atkinson
- Mayo Clinic, Department of Quantitative Health Sciences, Rochester, MN, United States
| | - Carin Y. Smith
- Mayo Clinic, Department of Quantitative Health Sciences, Rochester, MN, United States
| | - Delana M. Weis
- Mayo Clinic, Center for Multiple Sclerosis and Autoimmune Neurology, Rochester, MN, United States
| | | | - Walter A. Rocca
- Mayo Clinic, Department of Neurology, Rochester, MN, United States
- Mayo Clinic, Department of Quantitative Health Sciences, Rochester, MN, United States
- Mayo Clinic, Women’s Health Research Center, Rochester, MN, United States
| | - Kejal Kantarci
- Mayo Clinic, Department of Radiology, Rochester, MN, United States
- Mayo Clinic, Women’s Health Research Center, Rochester, MN, United States
| | - Orhun H. Kantarci
- Mayo Clinic, Department of Neurology, Rochester, MN, United States
- Mayo Clinic, Center for Multiple Sclerosis and Autoimmune Neurology, Rochester, MN, United States
| | - Burcu Zeydan
- Mayo Clinic, Department of Neurology, Rochester, MN, United States
- Mayo Clinic, Department of Radiology, Rochester, MN, United States
- Mayo Clinic, Center for Multiple Sclerosis and Autoimmune Neurology, Rochester, MN, United States
- Mayo Clinic, Women’s Health Research Center, Rochester, MN, United States
| |
Collapse
|
19
|
Wang H, Zhang X, Li H, Sun Z, Zhong Y. Gender differences in the burden of multiple sclerosis in China from 1990 to 2019 and its 25-year projection: An analysis of the Global Burden of Diseases Study. Health Sci Rep 2023; 6:e1738. [PMID: 38033712 PMCID: PMC10685393 DOI: 10.1002/hsr2.1738] [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: 07/09/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
Background and Aims Multiple sclerosis (MS) is a crippling, chronic, gender-related disease that causes burdens to individuals and society. China has a considerable and increasing population of MS. We aim to analyze the gender disparities in the burden of MS in China and predict the trends. Methods The study was conducted based on the Global Burden of Disease Study 2019. Data on incidence, prevalence, deaths, and disability-adjusted life years (DALYs) of MS in China from 1990 to 2019 was descriptively analyzed by year, gender, and age group. The Nordpred package in R (version 4.2.2) was used for age-period-cohort analysis to predict the all-ages numbers and age-standardized rates of incidence, prevalence, deaths, and DALYs in China from 2020 to 2044. Results The number of prevalent cases of MS in 2019 reached 18,143.56 (95% uncertainty intervals [UI]: 13,997.71-22,658.60) in males and 24,427.11 (95% UI: 18,906.02-30,530.21) in females in China. The peak age of prevalence was shifted from 40-44 years in 1990 to 45-49 years in 2019 in females but remained unchanged in males. In contrast to the increased age-standardized prevalence rate, the age-standardized death rate (ASDR) and age-standardized DALYs rate showed downward trends, which were more significant in females. Different from the global, Chinese males showed lower prevalence but higher deaths and DALYs than females for age-standardized rates and numbers. In the next 25 years, the patient population will remain large and peak around 44,599.78 in 2025-2029. The ASDR, age-standardized DALYs rate, and DALYs number were expected to decrease. The improvements in deaths and DALYs will be more significant in females. Conclusion Males with MS had a lower prevalence but higher deaths and DALYs than females in China. The ASDR and age-standardized DALYs rate have reduced over the past 30 years and were expected to continue decreasing, especially in females. The burden of MS will remain notable in the next 25 years.
Collapse
Affiliation(s)
- Heng Wang
- Department of Ophthalmology, Peking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Xia Zhang
- Department of Ophthalmology, Peking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Heyan Li
- Beijing Tongren Eye Center, Beijing Tongren HospitalCapital Medical UniversityBeijingChina
| | - Zixi Sun
- Department of Ophthalmology, Peking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Yong Zhong
- Department of Ophthalmology, Peking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| |
Collapse
|
20
|
Itoh N, Itoh Y, Stiles L, Voskuhl R. Sex differences in the neuronal transcriptome and synaptic mitochondrial function in the cerebral cortex of a multiple sclerosis model. Front Neurol 2023; 14:1268411. [PMID: 38020654 PMCID: PMC10654219 DOI: 10.3389/fneur.2023.1268411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Multiple sclerosis (MS) affects the cerebral cortex, inducing cortical atrophy and neuronal and synaptic pathology. Despite the fact that women are more susceptible to getting MS, men with MS have worse disability progression. Here, sex differences in neurodegenerative mechanisms are determined in the cerebral cortex using the MS model, chronic experimental autoimmune encephalomyelitis (EAE). Methods Neurons from cerebral cortex tissues of chronic EAE, as well as age-matched healthy control, male and female mice underwent RNA sequencing and gene expression analyses using RiboTag technology. The morphology of mitochondria in neurons of cerebral cortex was assessed using Thy1-CFP-MitoS mice. Oxygen consumption rates were determined using mitochondrial respirometry assays from intact as well as permeabilized synaptosomes. Results RNA sequencing of neurons in cerebral cortex during chronic EAE in C57BL/6 mice showed robust differential gene expression in male EAE compared to male healthy controls. In contrast, there were few differences in female EAE compared to female healthy controls. The most enriched differential gene expression pathways in male mice during EAE were mitochondrial dysfunction and oxidative phosphorylation. Mitochondrial morphology in neurons showed significant abnormalities in the cerebral cortex of EAE males, but not EAE females. Regarding function, synaptosomes isolated from cerebral cortex of male, but not female, EAE mice demonstrated significantly decreased oxygen consumption rates during respirometry assays. Discussion Cortical neuronal transcriptomics, mitochondrial morphology, and functional respirometry assays in synaptosomes revealed worse neurodegeneration in male EAE mice. This is consistent with worse neurodegeneration in MS men and reveals a model and a target to develop treatments to prevent cortical neurodegeneration and mitigate disability progression in MS men.
Collapse
Affiliation(s)
- Noriko Itoh
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yuichiro Itoh
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Linsey Stiles
- Department of Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rhonda Voskuhl
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
21
|
Morales-Rodriguez D, Anderson A, Nylander A, Hsu S, Singh J, Rowles W, Walsh CM, Braley TJ, Bove R. Well-being at midlife: Correlates of mental health in ambulatory menopausal women with multiple sclerosis. Mult Scler 2023; 29:1493-1502. [PMID: 37715710 PMCID: PMC10580672 DOI: 10.1177/13524585231197056] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/20/2023] [Accepted: 07/29/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND A majority of women with multiple sclerosis (MS) are diagnosed prior to menopause, yet their experiences during this transition are not well characterized. OBJECTIVES To explore associations between mental health, sleep, and other quality of life metrics, and vasomotor symptoms (VMSs) in ambulatory, menopausal women with MS. METHODS A secondary analysis was performed of baseline data from two trials enrolling ambulatory peri/postmenopausal women with MS: NCT02710214 (N = 24, bothersome VMS) and NCT04002934 (ongoing, N = 35, myelin repair). Measures analyzed were 36-Item Short-Form Survey (SF-36) (primary scale: general mental health), subjective sleep quality (Pittsburg Sleep Quality Index), VMS (daily diary, interference), mood (Center for Epidemiologist Studies-Depression Scale (CES-D)), walking impairment (timed 25-foot walk (T25FW)), and global disability (Expanded Disability Status Scale (EDSS)). RESULTS Participants' characteristics (N = 59) were: mean age 51.8 years (SD = 3.4), mean disease duration 11.3 years (SD = 7.6), median EDSS 3.0 (IQR = 2.0-4.0). Mental health was associated with better sleep quality (rho = -0.41, p = 0.019) and better mood (rho = -0.75, p < 0.001), but not with EDSS or T25FW (rho < 0.20, p > 0.10). Worse sleep quality also correlated with more frequent VMS (rho = 0.41, p = 0.02) and VMS interference (rho = 0.59, p < 0.001). CONCLUSIONS Findings suggest that optimizing sleep quality, mood, and hot flash quantity/interference could substantially improve mental health in menopausal women with MS-and highlight an important care gap in this population.
Collapse
Affiliation(s)
- Denisse Morales-Rodriguez
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Annika Anderson
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Alyssa Nylander
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Stephanie Hsu
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jessica Singh
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Will Rowles
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Christine M Walsh
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Tiffany J Braley
- Division of Multiple Sclerosis and Clinical Neuroimmunology, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Riley Bove
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
22
|
Itoh N, Itoh Y, Meyer CE, Suen TT, Cortez-Delgado D, Rivera Lomeli M, Wendin S, Somepalli SS, Golden LC, MacKenzie-Graham A, Voskuhl RR. Estrogen receptor beta in astrocytes modulates cognitive function in mid-age female mice. Nat Commun 2023; 14:6044. [PMID: 37758709 PMCID: PMC10533869 DOI: 10.1038/s41467-023-41723-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Menopause is associated with cognitive deficits and brain atrophy, but the brain region and cell-specific mechanisms are not fully understood. Here, we identify a sex hormone by age interaction whereby loss of ovarian hormones in female mice at midlife, but not young age, induced hippocampal-dependent cognitive impairment, dorsal hippocampal atrophy, and astrocyte and microglia activation with synaptic loss. Selective deletion of estrogen receptor beta (ERβ) in astrocytes, but not neurons, in gonadally intact female mice induced the same brain effects. RNA sequencing and pathway analyses of gene expression in hippocampal astrocytes from midlife female astrocyte-ERβ conditional knock out (cKO) mice revealed Gluconeogenesis I and Glycolysis I as the most differentially expressed pathways. Enolase 1 gene expression was increased in hippocampi from both astrocyte-ERβ cKO female mice at midlife and from postmenopausal women. Gain of function studies showed that ERβ ligand treatment of midlife female mice reversed dorsal hippocampal neuropathology.
Collapse
Affiliation(s)
- Noriko Itoh
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yuichiro Itoh
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Cassandra E Meyer
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Timothy Takazo Suen
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Diego Cortez-Delgado
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - Sophia Wendin
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sri Sanjana Somepalli
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Lisa C Golden
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Allan MacKenzie-Graham
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Rhonda R Voskuhl
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
23
|
Lorefice L, Fenu G, Fronza M, Murgia F, Frau J, Coghe G, Barracciu MA, Atzori L, Angioni S, Cocco E. Menopausal transition in multiple sclerosis: relationship with disease activity and brain volume measurements. Front Neurol 2023; 14:1251667. [PMID: 37602270 PMCID: PMC10434500 DOI: 10.3389/fneur.2023.1251667] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Background Recent evidence has shown a significant association between menopause and multiple sclerosis (MS) progression. This study investigated the possible role of menopause in influencing MS from clinical and neuroradiological perspectives. Notably, the possible association between menopause and brain atrophy has been evaluated. Materials and methods This study included women with MS whose ages ranged from 45 to 55 years. Demographic and clinical characteristics were collected, and the reproductive phase was defined as non-menopausal or menopausal based on the final menstrual period. Thus, MS activity over the past year was reported as the annualised relapse rate (ARR), and MRI activity (defined as new T2 lesions and/or the presence of gadolinium-enhancing lesions at the last MRI assessment in comparison with the MRI performed within the previous 12 months) were compared between non-menopausal women (non-MW) and menopausal women (MW). Volume measurements of the whole brain (WB), white matter (WM), grey matter (GM), and cortical GM were estimated using the SIENAX software, and the possible relationship with menopausal status was assessed by regression analysis. Results The study included 147 women with MS. Eighty-four (57.1%) were MW, with a mean age of 48.5 ± 4.3 years at menopause onset and a mean duration of menopause of 4.1 ± 1.1 years. When compared for ARR, MW reported a lower rate than the non-MW (ARR of 0.29 ± 0.4 vs. 0.52 ± 0.5; p < 0.01). MRI activity was observed in 13.1% of MW and 20.6% of non-MW (p = 0.03). Lower cortical GM volumes (578.1 ± 40.4 mL in MW vs. 596.9 ± 35.8 mL in non-MW; p < 0.01) have also been reported. Finally, multivariate analysis showed a significant association of lower ARR (p = 0.001) and cortical GM volume (p = 0.002) with menopausal status after correction for chronological age and other variables. Discussion Menopause may be an adverse prognostic factor of MS. Our preliminary results suggest that menopause may facilitate cortical GM atrophy, probably due to a decline in the neuroprotective effects of estrogen, with negative effects on MS evolution.
Collapse
Affiliation(s)
- Lorena Lorefice
- Department of Medical Sciences and Public Health, Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, University of Cagliari, Cagliari, Italy
| | - Giuseppe Fenu
- Department of Neurosciences, ARNAS Brotzu, Cagliari, Italy
| | - Marzia Fronza
- Department of Medical Sciences and Public Health, Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, University of Cagliari, Cagliari, Italy
| | - Federica Murgia
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Jessica Frau
- Department of Medical Sciences and Public Health, Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, University of Cagliari, Cagliari, Italy
| | - Giancarlo Coghe
- Department of Medical Sciences and Public Health, Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, University of Cagliari, Cagliari, Italy
| | | | - Luigi Atzori
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Stefano Angioni
- Division of Gynecology and Obstetrics, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | - Eleonora Cocco
- Department of Medical Sciences and Public Health, Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, University of Cagliari, Cagliari, Italy
| |
Collapse
|
24
|
Bridge F, Butzkueven H, Van der Walt A, Jokubaitis VG. The impact of menopause on multiple sclerosis. Autoimmun Rev 2023; 22:103363. [PMID: 37230311 DOI: 10.1016/j.autrev.2023.103363] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/21/2023] [Indexed: 05/27/2023]
Abstract
Menopause, defined as the permanent cessation of ovarian function, represents a period of significant fluctuation in sex hormone concentrations. Sex hormones including oestrogen, progesterone, testosterone and anti-Mullerian hormone are thought have neuroinflammatory effects and are implicated in both neuroprotection and neurodegeneration. Sex hormones are thought to have a role in modifying clinical trajectory in multiple sclerosis (MS) throughout the lifespan. Multiple sclerosis predominantly effects women and is typically diagnosed early in a woman's reproductive life. Most women with MS will undergo menopause. Despite this, the effect of menopause on MS disease course remains unclear. This review examines the relationship between sex hormones and MS disease activity and clinical course, particularly around the time of menopause. It will consider the role of interventions such as exogenous hormone replacement therapy in modulating clinical outcomes in this period. Understanding the impact of menopause on multiple sclerosis is fundamental for delivering optimal care to women with MS as they age and will inform treatment decisions with the aim of minimising relapses, disease accrual and improving quality of life.
Collapse
Affiliation(s)
- Francesca Bridge
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia.
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Anneke Van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Vilija G Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|
25
|
Graham EL, Bakkensen JB, Anderson A, Lancki N, Davidson A, Perez Giraldo G, Jungheim ES, Vanderhoff AC, Ostrem B, Mok-Lin E, Huang D, Bevan CJ, Jacobs D, Kaplan TB, Houtchens MK, Bove R. Inflammatory Activity After Diverse Fertility Treatments: A Multicenter Analysis in the Modern Multiple Sclerosis Treatment Era. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:10/3/e200106. [PMID: 36922025 PMCID: PMC10018493 DOI: 10.1212/nxi.0000000000200106] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/06/2023] [Indexed: 03/17/2023]
Abstract
BACKGROUND AND OBJECTIVES Patients with multiple sclerosis (MS) may seek fertility treatment (FT)-including in vitro fertilization (IVF). Variable relapse risk after IVF has been reported in small historical cohorts, with more recent studies suggesting no change in annualized relapse rate (ARR). The objective of this study was to evaluate ARR 12 months pre-FT and 3 months post-FT in a multicenter cohort and identify factors associated with an increased risk of relapse. METHODS Patients with clinically isolated syndrome (CIS) or MS aged 18-45 years with at least 1 FT from January 1, 2010, to October 14, 2021, were retrospectively identified at 4 large academic MS centers. The exposed period of 3 months after FT was compared with the unexposed period of 12 months before FT. FTs included controlled ovarian stimulation followed by fresh embryo transfer (COS-ET), COS alone, embryo transfer (ET) alone, and oral ovulation induction (OI). The Wilcoxon signed rank test and mixed Poisson regression models with random effects were used to compare ARR pre-FT vs post-FT, with the incidence rate ratio (IRR) and 95% CI reported. RESULTS One hundred twenty-four FT cycles among 65 patients with MS (n = 56) or CIS (n = 9) were included: 61 COS-ET, 19 COS alone, 30 ET alone, and 14 OI. The mean age at FT was 36.5 ± 3.8 years, and the mean disease duration was 8.2 ± 5.0 years. Across 80 cycles with COS, only 5 relapses occurred among 4 unique patients within 3 months of treatment. The mean ARR after COS and before was not different (0.26 vs 0.25, p = 0.37), and the IRR was 0.95 (95% CI: 0.52-1.76, p = 0.88). No cycles with therapeutic disease-modifying therapies (DMTs) during COS had 3 months relapse (ARR 0 post-COS vs 0.18 pre-COS, p = 0.02, n = 34). Relapse rates did not vary by COS protocol. Among COS-ET cycles that achieved pregnancy (n = 43), ARR decreased from 0.26 to 0.09 (p = 0.04) within the first trimester of pregnancy. There were no relapses 3 months after ET alone and 1 relapse after OI. DISCUSSION In this modern multicenter cohort of patients with MS undergoing diverse FTs, which included 43% on DMTs, we did not observe an elevated relapse risk after FT.
Collapse
Affiliation(s)
- Edith L Graham
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA.
| | - Jennifer B Bakkensen
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - Annika Anderson
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - Nicola Lancki
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - Anne Davidson
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - Gina Perez Giraldo
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - Emily S Jungheim
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - Anna C Vanderhoff
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - Bridget Ostrem
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - Evelyn Mok-Lin
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - David Huang
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - Carolyn J Bevan
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - Dina Jacobs
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - Tamara B Kaplan
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - Maria K Houtchens
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| | - Riley Bove
- From the Department of Neurology (E.L.G., G.P.G., C.J.B.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (J.B.B., E.S.J.), Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL; UCSF Weill Institute for the Neurosciences (A.A., B.O., R.B.), Department of Neurology, University of California, San Francisco (UCSF); Division of Biostatistics (N.L.), Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; Feinberg School of Medicine (A.D.), Northwestern University, Chicago, IL; Department of Obstetrics and Gynecology (A.C.V.), Division of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, Boston, MA; UCSF Center for Reproductive Health (E.M.-L., D.H.), Mission Bay Campus, San Francisco, CA; Department of Neurology (D.J.), University of Pennsylvania, Philadelphia; and Department of Neurology (T.B.K., M.K.H.), Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
26
|
Lorefice L, D’Alterio MN, Firinu D, Fenu G, Cocco E. Impact of Menopause in Patients with Multiple Sclerosis: Current Perspectives. Int J Womens Health 2023; 15:103-109. [PMID: 36721498 PMCID: PMC9884461 DOI: 10.2147/ijwh.s334719] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Given the aging population, with a peak age-specific prevalence that is shifting beyond the age of 50, several women currently living with MS are very close to menopause. Menopause is usually characterized by several specific symptoms with adverse impacts on different aspects of a woman's quality of life, such as fatigue, and cognitive, mood and bladder disorders, which overlap with symptoms of MS. Generally, after this biological transition, women with MS appear to be subject to less inflammatory activity. However, several studies have reported an increase of disability accumulation after menopause, suggesting that it is a turning point to a more progressive phase of the disease. This may be attributable to the hormonal and immunological changes associated with menopause, with several effects on neuroinflammation and neurodegeneration increasing due to the immunosenescence of aging. This review summarizes the hormonal and immunological changes associated with menopause, detailing the effects on MS symptoms, outcomes, and the aging process. Furthermore, possible interventions to improve patients' quality of life are evaluated. In fact, it is increasingly necessary to improve the global management of MS women, as well as their lives, at this multifaceted turning point.
Collapse
Affiliation(s)
- Lorena Lorefice
- Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Maurizio Nicola D’Alterio
- Division of Gynecology and Obstetrics, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | - Davide Firinu
- Clinical Immunology, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Giuseppe Fenu
- Department of Neurosciences, ARNAS Brotzu, Cagliari, Italy
| | - Eleonora Cocco
- Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
27
|
Boziki M, Theotokis P, Kesidou E, Karafoulidou E, Konstantinou C, Michailidou I, Bahar Y, Altintas A, Grigoriadis N. Sex, aging and immunity in multiple sclerosis and experimental autoimmune encephalomyelitis: An intriguing interaction. Front Neurol 2023; 13:1104552. [PMID: 36698908 PMCID: PMC9869255 DOI: 10.3389/fneur.2022.1104552] [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: 11/21/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) with a profound neurodegenerative component early in the disease pathogenesis. Age is a factor with a well-described effect on the primary disease phenotype, namely, the relapsing-remitting vs. the primary progressive disease. Moreover, aging is a prominent factor contributing to the transition from relapsing-remitting MS (RRMS) to secondary progressive disease. However, sex also seems to, at least in part, dictate disease phenotype and evolution, as evidenced in humans and in animal models of the disease. Sex-specific gene expression profiles have recently elucidated an association with differential immunological signatures in the context of experimental disease. This review aims to summarize current knowledge stemming from experimental autoimmune encephalomyelitis (EAE) models regarding the effects of sex, either independently or as a factor combined with aging, on disease phenotype, with relevance to the immune system and the CNS.
Collapse
Affiliation(s)
- Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Karafoulidou
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Chrystalla Konstantinou
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Iliana Michailidou
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Ayse Altintas
- School of Medicine, Koç University, Istanbul, Turkey
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece,*Correspondence: Nikolaos Grigoriadis ✉
| |
Collapse
|
28
|
Graves JS, Krysko KM, Hua LH, Absinta M, Franklin RJM, Segal BM. Ageing and multiple sclerosis. Lancet Neurol 2023; 22:66-77. [PMID: 36216015 DOI: 10.1016/s1474-4422(22)00184-3] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/09/2022] [Accepted: 04/20/2022] [Indexed: 11/07/2022]
Abstract
The factor that is most relevant and strongly associated with the clinical course of multiple sclerosis is chronological age. Very young patients exclusively have relapsing remitting disease, whereas those with later onset disease face a more rapid development of permanent disability. For people with progressive multiple sclerosis, the poor response to current disease modifying therapies might be related to ageing in the immune system and CNS. Ageing is also associated with increased risks of side-effects caused by some multiple sclerosis therapies. Both somatic and reproductive ageing processes might contribute to development of progressive multiple sclerosis. Understanding the role of ageing in immune and neural cell function in patients with multiple sclerosis might be key to halting non-relapse-related progression. The growing literature on potential therapies that target senescent cells and ageing processes might provide effective strategies for remyelination and neuroprotection.
Collapse
Affiliation(s)
- Jennifer S Graves
- Department of Neurosciences, University of California, San Diego, CA, USA; Pediatric Multiple Sclerosis Center, Rady Children's Hospital, San Diego, CA, USA; Department of Neurology, San Diego VA Hospital, San Diego, CA, USA.
| | - Kristen M Krysko
- Division of Neurology, Department of Medicine, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Le H Hua
- Department of Neurology, Cleveland Clinic, Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Martina Absinta
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA; Division of Neuroscience, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, Italy
| | - Robin J M Franklin
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Benjamin M Segal
- Department of Neurology and the Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
29
|
Alvarez-Sanchez N, Dunn SE. Potential biological contributers to the sex difference in multiple sclerosis progression. Front Immunol 2023; 14:1175874. [PMID: 37122747 PMCID: PMC10140530 DOI: 10.3389/fimmu.2023.1175874] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/03/2023] [Indexed: 05/02/2023] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated disease that targets the myelin sheath of central nervous system (CNS) neurons leading to axon injury, neuronal death, and neurological progression. Though women are more highly susceptible to developing MS, men that develop this disease exhibit greater cognitive impairment and accumulate disability more rapidly than women. Magnetic resonance imaging and pathology studies have revealed that the greater neurological progression seen in males correlates with chronic immune activation and increased iron accumulation at the rims of chronic white matter lesions as well as more intensive whole brain and grey matter atrophy and axon loss. Studies in humans and in animal models of MS suggest that male aged microglia do not have a higher propensity for inflammation, but may become more re-active at the rim of white matter lesions as a result of the presence of pro-inflammatory T cells, greater astrocyte activation or iron release from oligodendrocytes in the males. There is also evidence that remyelination is more efficient in aged female than aged male rodents and that male neurons are more susceptible to oxidative and nitrosative stress. Both sex chromosome complement and sex hormones contribute to these sex differences in biology.
Collapse
Affiliation(s)
- Nuria Alvarez-Sanchez
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
- Department of Immunology, 1 King’s College Circle, Toronto, ON, Canada
| | - Shannon E. Dunn
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
- Department of Immunology, 1 King’s College Circle, Toronto, ON, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
- *Correspondence: Shannon E. Dunn,
| |
Collapse
|
30
|
Voskuhl R, Itoh Y. The X factor in neurodegeneration. J Exp Med 2022; 219:e20211488. [PMID: 36331399 PMCID: PMC9641640 DOI: 10.1084/jem.20211488] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/22/2022] [Accepted: 10/12/2022] [Indexed: 07/25/2023] Open
Abstract
Given the aging population, it is important to better understand neurodegeneration in aging healthy people and to address the increasing incidence of neurodegenerative diseases. It is imperative to apply novel strategies to identify neuroprotective therapeutics. The study of sex differences in neurodegeneration can reveal new candidate treatment targets tailored for women and men. Sex chromosome effects on neurodegeneration remain understudied and represent a promising frontier for discovery. Here, we will review sex differences in neurodegeneration, focusing on the study of sex chromosome effects in the context of declining levels of sex hormones during aging.
Collapse
Affiliation(s)
- Rhonda Voskuhl
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Yuichiro Itoh
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
31
|
Nytrova P, Dolezal O. Sex bias in multiple sclerosis and neuromyelitis optica spectrum disorders: How it influences clinical course, MRI parameters and prognosis. Front Immunol 2022; 13:933415. [PMID: 36016923 PMCID: PMC9396644 DOI: 10.3389/fimmu.2022.933415] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
This review is a condensed summary of representative articles addressing the sex/gender bias in multiple sclerosis (MS) and neuromyelitis optica spectrum disorders (NMOSD). The strong effects of sex on the incidence and possibly also the activity and progression of these disorders should be implemented in the evaluation of any phase of clinical research and also in treatment choice consideration in clinical practice and evaluation of MRI parameters. Some relationships between clinical variables and gender still remain elusive but with further understanding of sex/gender-related differences, we should be able to provide appropriate patient-centered care and research.
Collapse
Affiliation(s)
- Petra Nytrova
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czechia
- *Correspondence: Petra Nytrova,
| | - Ondrej Dolezal
- Department of Neurology, Dumfries and Galloway Royal Infirmary, NHS Scotland, Dumfries, United Kingdom
| |
Collapse
|
32
|
Diem L, Hammer H, Hoepner R, Pistor M, Remlinger J, Salmen A. Sex and gender differences in autoimmune demyelinating CNS disorders: Multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD) and myelin-oligodendrocyte-glycoprotein antibody associated disorder (MOGAD). INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 164:129-178. [PMID: 36038203 DOI: 10.1016/bs.irn.2022.06.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Multiple sclerosis (MS), Neuromyelitis optica spectrum disorder (NMOSD) and Myelin-Oligodendrocyte-Glycoprotein antibody associated disorder (MOGAD) are demyelinating disorders of the central nervous system (CNS) of autoimmune origin. Here, we summarize general considerations on sex-specific differences in the immunopathogenesis and hormonal influences as well as key clinical and epidemiological elements. Gender-specific issues are widely neglected starting with the lacking separation of sex as a biological variable and gender comprising the sociocultural components. As for other autoimmune diseases, female preponderance is common in MS and NMOSD. However, sex distribution in MOGAD seems equal. As in MS, immunotherapy in NMOSD and MOGAD is crucial to prevent further disease activity. Therefore, we assessed data on sex differences of the currently licensed disease-modifying treatments for efficacy and safety. This topic seems widely neglected with only fragmented information resulting from post-hoc analyses of clinical trials or real-world post-marketing studies afflicted with lacking power and/or inherent sources of bias. In summary, biological hypotheses of sex differences including genetic factors, the constitution of the immune system and hormonal influences are based upon human and preclinical data, especially for the paradigmatic disease of MS whereas specific data for NMOSD and MOGAD are widely lacking. Epidemiological and clinical differences between men and women are well described for MS and to some extent for NMOSD, yet, with remaining contradictory findings. MOGAD needs further detailed investigation. Sex-specific analyses of safety and efficacy of long-term immunotherapies need to be addressed in future studies designed and powered to answer the pressing questions and to optimize and individualize treatment.
Collapse
Affiliation(s)
- Lara Diem
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Freiburgstrasse, Bern, Switzerland
| | - Helly Hammer
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Freiburgstrasse, Bern, Switzerland
| | - Robert Hoepner
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Freiburgstrasse, Bern, Switzerland
| | - Max Pistor
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Freiburgstrasse, Bern, Switzerland
| | - Jana Remlinger
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Freiburgstrasse, Bern, Switzerland; Department of Biomedical Research and Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Anke Salmen
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Freiburgstrasse, Bern, Switzerland.
| |
Collapse
|
33
|
Juutinen L, Ahinko K, Tinkanen H, Rosti-Otajärvi E, Sumelahti ML. Menopausal symptoms and hormone therapy in women with multiple sclerosis: A baseline-controlled study. Mult Scler Relat Disord 2022; 67:104098. [PMID: 35994896 DOI: 10.1016/j.msard.2022.104098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/20/2022] [Accepted: 08/07/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND Depression, sleep disturbances, and cognitive difficulties impair the quality of life in people with multiple sclerosis (MS). Similar symptoms are also frequent during the menopausal transition. In clinical practice, it is important to consider the multifactorial causes of these overlapping symptoms and the potential benefits of menopausal hormone therapy (MHT). The objective of this study was to evaluate vasomotor symptoms (VMS), mood, sleep, and cognition of menopausal women with and without MS at baseline and during one year of MHT. METHODS In this prospective baseline-controlled study, peri- and early postmenopausal participants with (n=14) and without (n=13) MS received MHT containing 1 or 2 mg of estradiol and cyclical 10 mg dydrogesterone for one year. VMS frequency, depressive symptoms (measured by Beck Depression Inventory), insomnia severity (Insomnia Severity Index), and cognitive performance (Paced Auditory Serial Addition Test; PASAT, Symbol Digit Modalities Test; SDMT) were evaluated at baseline and at 3 and 12 months of treatment. Differences in the outcome measures between groups at baseline were assessed using the Mann-Whitney U test. Changes during follow-up compared to baseline within groups were evaluated by Wilcoxon Signed Ranks Test. P < 0.05 was considered for statistical significance. MS activity was monitored by clinical assessment and brain MRI at baseline and at 12 months. RESULTS Depressive symptoms were more common in MS group, while vasomotor and insomnia symptoms were equally common. During follow-up with MHT, VMS frequency decreased in both groups. Depressive symptoms decreased at 3 months (p = 0.031 with MS; p = 0.024 without MS) and the reduction was sustained at 12 months (p = 0.017; p = 0.042, respectively). Alleviation in insomnia symptoms was seen in participants without MS at 3 months (p = 0.029) and in those participants with MS suffering insomnia at baseline (p = 0.016 at 3 months; p = 0.047 at 12 months). Both groups improved their performance in PASAT, but no significant change was observed in SDMT. MS activity at baseline was mainly stable, and no increase in activity was detected during MHT. CONCLUSION Improvements in vasomotor, depressive, and insomnia symptoms observed during one year of MHT are encouraging and suggest that larger placebo-controlled studies of MHT in women with MS are warranted. Cognitive implications were inconclusive because the findings in PASAT likely result from practice effect. MHT did not show any adverse effect on MS activity and increasing safety data will hopefully facilitate patient recruitment for future studies.
Collapse
Affiliation(s)
- Laura Juutinen
- Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Building, Arvo Ylpön katu 34, 33520 Tampere, Finland; Department of Neurosciences and Rehabilitation, Tampere University Hospital, P.O. Box 2000, FI-33521 Tampere, Finland.
| | - Katja Ahinko
- Department of Obstetrics and Gynecology, Tampere University Hospital, P.O. Box 2000, FI-33521 Tampere, Finland
| | - Helena Tinkanen
- Department of Obstetrics and Gynecology, Tampere University Hospital, P.O. Box 2000, FI-33521 Tampere, Finland
| | - Eija Rosti-Otajärvi
- Department of Neurosciences and Rehabilitation, Tampere University Hospital, P.O. Box 2000, FI-33521 Tampere, Finland; Department of Rehabilitation and psychosocial support, Tampere University Hospital, P.O. Box 2000, FI-33521 Tampere, Finland
| | - Marja-Liisa Sumelahti
- Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Building, Arvo Ylpön katu 34, 33520 Tampere, Finland
| |
Collapse
|
34
|
Magyari M, Koch-Henriksen N. Quantitative effect of sex on disease activity and disability accumulation in multiple sclerosis. J Neurol Neurosurg Psychiatry 2022; 93:716-722. [PMID: 35393340 PMCID: PMC9279846 DOI: 10.1136/jnnp-2022-328994] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/16/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To quantify sex differences in activity and severity of multiple sclerosis (MS) and how it depends on disease duration and time since clinical onset. METHODS All Danish citizens with onset of relapsing MS since 1996 who have received disease-modifying therapy have been followed with annual or biannual control visits with mandatory notification of the Danish Multiple Sclerosis Registry. Men and women were compared by the inverse probability of being female. Relapse rates and changes in the Expanded Disability Status Scale (EDSS) scores were analysed with weighted general linear models, and we used weighted Cox regression for HRs between men and women for different EDSS endpoints. RESULTS We included 3028 men and 6619 women. The weighted female:male relapse rate ratio was 1.16 (95% CI: 1.10 to 1.22) but after age 50 years, the difference disappeared. The annualised increase in EDSS was 0.07 in men (95% CI: 0.05 to 0.08) and 0.05 in women (95% CI: 0.04 to 0.06); p=0.017. With women as reference, the HR for reaching EDSS 4 was 1.34 (95% CI: 1.23 to 1.45; p<0.001), and for reaching EDSS 6 it was 1.43 (95% CI: 1.28 to 1.61; p<0.001). The diagnostic delay did not differ significantly between the sexes. CONCLUSION Women have more inflammatory disease activity in terms of relapses than men up to the age of menopause indicating that sex hormones may play a role. Men are more subject to the neurodegenerative component of MS than women, particularly after the age of 45 years.
Collapse
Affiliation(s)
- Melinda Magyari
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, Copenhagen, Denmark .,The Danish Multiple Sclerosis Registry, Rigshospitalet, Copenhagen, Denmark
| | - Nils Koch-Henriksen
- The Danish Multiple Sclerosis Registry, Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
35
|
Kaplan TB, Gopal A, Block VJ, Suskind AM, Zhao C, Polgar-Turcsanyi M, Saraceno TJ, Gomez R, Santaniello A, Consortium SUMMIT, Ayoubi NE, Cree BA, Hauser SL, Weiner H, Chitnis T, Khoury S, Bove R. Challenges to Longitudinal Characterization of Lower Urinary Tract Dysfunction in Multiple Sclerosis. Mult Scler Relat Disord 2022; 62:103793. [DOI: 10.1016/j.msard.2022.103793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/01/2022] [Accepted: 04/03/2022] [Indexed: 11/24/2022]
|
36
|
Bove R, Anderson A, Rowles W, Rankin KA, Hills NK, Carleton M, Cooper J, Cree BA, Gelfand JM, Graves J, Henry RG, Krysko KM, Rush G, Zamvil SS, Joffe H, Chan JR, Green A. A Hormonal therapy for menopausal women with MS: A Phase Ib/IIa Randomized Controlled Trial. Mult Scler Relat Disord 2022; 61:103747. [DOI: 10.1016/j.msard.2022.103747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/07/2022] [Accepted: 03/17/2022] [Indexed: 12/27/2022]
|
37
|
Kopp TI, Lidegaard Ø, Magyari M. Hormone therapy and disease activity in Danish women with multiple sclerosis - a population-based cohort study. Eur J Neurol 2022; 29:1753-1762. [PMID: 35196406 PMCID: PMC9314629 DOI: 10.1111/ene.15299] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 02/03/2022] [Accepted: 02/15/2022] [Indexed: 11/30/2022]
Abstract
Background and purpose Sex differences in multiple sclerosis (MS) prevalence and disease course are thought to be driven by hormones. Exogenous exposure to estrogens may affect MS disease course. Thus, our aim was to investigate the association between hormone therapy (HT) and disease activity and disability accrual among women with MS. Methods A register‐based cohort study was conducted with prospectively enrolled cases from the Danish MS registry. Information on hormone exposure was retrieved from the National Prescription Registry. Outcomes were relapse rate, relapse rate ratio, recurrent relapses, 6‐month confirmed and sustained Expanded Disability Status Scale (EDSS) milestones 4 and 6, and recurrent EDSS worsening. Results In all, 3325 women were eligible for analyses, of whom 333 (10%) were ever on HT at some time during follow‐up. We found no association between HT and disability accrual, although a trend for increasing risk with increasing length of use was seen. The risk of reaching 6‐month confirmed and sustained EDSS 4 among users was 0.6 (95% confidence interval [CI] = 0.3–1.2) after <1 year of use and 1.4 (95% CI = 0.9–2.2) after >5 years of HT compared to never use. The risk of recurrent relapse was increased by 20% (95% CI = 1.0–1.4) among current users of HT compared to nonusers. However, the risk of recurrent relapses was driven by the first calendar period (1996–2005) before the introduction of high‐efficacy disease‐modifying therapy. Conclusions Our findings from this nationwide MS population suggest that HT does not affect disability accrual in women with MS, especially if used for <5 years.
Collapse
Affiliation(s)
- Tine Iskov Kopp
- The Danish Multiple Sclerosis Registry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Øjvind Lidegaard
- Department of Gynaecology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Melinda Magyari
- The Danish Multiple Sclerosis Registry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,The Danish Multiple Sclerosis Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
38
|
Miner AE, Graves JS. What telomeres teach us about MS. Mult Scler Relat Disord 2021; 54:103084. [PMID: 34371369 DOI: 10.1016/j.msard.2021.103084] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/14/2021] [Accepted: 06/09/2021] [Indexed: 02/03/2023]
Abstract
While the precise mechanisms driving progressive forms of MS are not fully understood, patient age has clear impact on disease phenotype. The very young with MS have high relapse rates and virtually no progressive disease, whereas older patients tend to experience more rapid disability accumulation with few relapses. Defining a patient's biological age may offer more precision in determining the role of aging processes in MS phenotype and pathophysiology than just working with an individual's birthdate. The most well recognized measurement of an individual's "biological clock" is telomere length (TL). While TL may differ across tissue types in an individual, most cells TL correlate well with leukocyte TL (LTL), which is the most common biomarker used for aging. LTL has been associated with risk for aging related diseases and most recently with higher levels of disability and brain atrophy in people living with MS. LTL explains 15% of the overall association of chronological age with MS disability level. While LTL may be used just as a biomarker of overall somatic aging processes, triggering of the DNA damage response by telomere attrition leads to senescence pathways that are likely highly relevant to a chronic autoimmune disease. Considering reproductive aging factors, particularly ovarian aging in women, which correlates with LTL and oocyte telomere length, may complement measurements of somatic aging in understanding MS progression. The key to stopping non-relapse related progression in MS might lie in targeting pathways related to biological aging effects on the immune and nervous systems.
Collapse
Affiliation(s)
- Annalise E Miner
- Department of Neurosciences, University of California, San Diego, USA
| | - Jennifer S Graves
- Department of Neurosciences, University of California, San Diego, USA.
| |
Collapse
|
39
|
Roeder HJ, Leira EC. Effects of the Menstrual Cycle on Neurological Disorders. Curr Neurol Neurosci Rep 2021; 21:34. [PMID: 33970361 DOI: 10.1007/s11910-021-01115-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW The menstrual cycle involves recurrent fluctuations in hormone levels and temperature via neuroendocrine feedback loops. This paper reviews the impact of the menstrual cycle on several common neurological conditions, including migraine, seizures, multiple sclerosis, stroke, and Parkinson's disease. RECENT FINDINGS The ovarian steroid hormones, estrogen and progesterone, have protean effects on central nervous system functioning that can impact the likelihood, severity, and presentation of many neurological diseases. Hormonal therapies have been explored as a potential treatment for many neurological diseases with varying degrees of evidence and success. Neurological conditions also impact women's reproductive health, and the cessation of ovarian function with menopause may also alter the course of neurological diseases. Medication selection must consider hormonal effects on metabolism and the potential for adverse drug reactions related to menstruation, fertility, and pregnancy outcomes. Novel medications with selective affinity for hormonal receptors are desirable. Neurologists and gynecologists must collaborate to provide optimal care for women with neurological disorders.
Collapse
Affiliation(s)
- Hannah J Roeder
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Enrique C Leira
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA. .,Department of Neurosurgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA. .,Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
40
|
Bove R, Okai A, Houtchens M, Elias-Hamp B, Lugaresi A, Hellwig K, Kubala Havrdová E. Effects of Menopause in Women With Multiple Sclerosis: An Evidence-Based Review. Front Neurol 2021; 12:554375. [PMID: 33815241 PMCID: PMC8017266 DOI: 10.3389/fneur.2021.554375] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 02/17/2021] [Indexed: 12/20/2022] Open
Abstract
Over two thirds of all individuals who develop multiple sclerosis (MS) will be women prior to the age of menopause. Further, an estimated 30% of the current MS population consists of peri- or postmenopausal women. The presence of MS does not appear to influence age of menopausal onset. In clinical practice, symptoms of MS and menopause can frequently overlap, including disturbances in cognition, mood, sleep, and bladder function, which can create challenges in ascertaining the likely cause of symptoms to be treated. A holistic and comprehensive approach to address these common physical and psychological changes is often suggested to patients during menopause. Although some studies have suggested that women with MS experience reduced relapse rates and increased disability progression post menopause, the data are not consistent enough for firm conclusions to be drawn. Mechanisms through which postmenopausal women with MS may experience disability progression include neuroinflammation and neurodegeneration from age-associated phenomena such as immunosenescence and inflammaging. Additional effects are likely to result from reduced levels of estrogen, which affects MS disease course. Following early retrospective studies of women with MS receiving steroid hormones, more recent interventional trials of exogenous hormone use, albeit as oral contraceptive, have provided some indications of potential benefit on MS outcomes. This review summarizes current research on the effects of menopause in women with MS, including the psychological impact and symptoms of menopause on disease worsening, and the treatment options. Finally, we highlight the need for more inclusion of MS patients from underrepresented racial and geographic groups in clinical trials, including among menopausal women.
Collapse
Affiliation(s)
- Riley Bove
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Annette Okai
- Multiple Sclerosis Treatment Center of Dallas, Dallas, TX, United States
| | - Maria Houtchens
- Partners Multiple Sclerosis Center, Brigham and Women's Hospital, Boston, MA, United States
| | - Birte Elias-Hamp
- Neurological Private Practice, Institute of Neuroimmunology and Multiple Sclerosis, Hamburg, Germany
| | - Alessandra Lugaresi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Kerstin Hellwig
- Department of Neurology, Ruhr University Bochum and St. Josef-Hospital, Bochum, Germany
| | - Eva Kubala Havrdová
- Department of Neurology and Center of Clinical Neuroscience, First Medical Faculty, General University Hospital, Charles University, Prague, Czechia
| |
Collapse
|
41
|
Otero-Romero S, Midaglia L, Carbonell-Mirabent P, Zuluaga M, Galán I, Río J, Arrambide G, Rodríguez-Barranco M, Vidal-Jordana A, Castillo J, Rodríguez-Acevedo B, Zabalza A, Nos C, Comabella-Lopez M, Mulero P, Auger C, Sastre-Garriga J, Pérez-Hoyos S, Rovira A, Montalban X, Tintoré M. Menopause does not modify disability trajectories in a longitudinal cohort of women with clinically isolated syndrome and multiple sclerosis followed from disease onset. Eur J Neurol 2021; 29:1075-1081. [PMID: 33609298 DOI: 10.1111/ene.14782] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND PURPOSE To evaluate the effect of menopause on disability accumulation in women followed from their clinically isolated syndrome (CIS). METHODS We examined the longitudinal changes in Expanded Disability Status Scale (EDSS) scores from CIS until the last follow-up in women belonging to the Barcelona CIS prospective cohort, followed through their menopausal transition. The analysis is based on 13,718 EDSS measurements, with an average of 28 EDSS measurements per patient. Differences in EDSS trajectories between menopausal and nonmenopausal women, controlling for age and disease duration, were evaluated. We performed two sensitivity analyses in women with confirmed MS and in those experiencing early menopause. RESULTS From 764 eligible women, 496 (65%) responded to the questionnaire, and 74 (14.9%) reached menopause over the follow-up. We did not find a significant inflection point in EDSS trajectories around menopause (slope change -0.009; 95% CI -0.066; 0.046). The annual increase in EDSS over the complete course of the disease was significantly higher in menopausal women (0.049; 95% CI, 0.026-0.074) versus nonmenopausal (0.019; 95% CI, 0.008-0.031; interaction p value 0.025). This difference was lost when controlling for age and disease duration (EDSS annual increase of 0.059; 95% CI, 0.025-0.094 vs. 0.038; 95% CI, 0.021-0.057, respectively; interaction p value 0.321). No inflection point was detected when the analysis was restricted to women with confirmed MS or with earlier menopause. CONCLUSIONS Menopause is not associated with an increased risk of disability in a CIS population, considering EDSS trajectories throughout the course of the disease together with age and disease duration.
Collapse
Affiliation(s)
- Susana Otero-Romero
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain.,Preventive Medicine and Epidemiology Department, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Luciana Midaglia
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Pere Carbonell-Mirabent
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María Zuluaga
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ingrid Galán
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jordi Río
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Georgina Arrambide
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta Rodríguez-Barranco
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Angela Vidal-Jordana
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joaquin Castillo
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Breogán Rodríguez-Acevedo
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ana Zabalza
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carlos Nos
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Comabella-Lopez
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Patricia Mulero
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Auger
- Neuroradiology Section, Department of Radiology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jaume Sastre-Garriga
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Santiago Pérez-Hoyos
- Statistics and bioinformatics unit, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Rovira
- Neuroradiology Section, Department of Radiology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Montalban
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mar Tintoré
- Neurology-Neuroimmunology Department, Centro de Esclerosis Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
42
|
Jakimovski D, Eckert SP, Zivadinov R, Weinstock-Guttman B. Considering patient age when treating multiple sclerosis across the adult lifespan. Expert Rev Neurother 2021; 21:353-364. [PMID: 33595379 DOI: 10.1080/14737175.2021.1886082] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: The successful development of anti-inflammatory disease-modifying treatments (DMT) significantly improved disease outcomes and longevity of persons with multiple sclerosis (pwMS). However, the shift toward an elderly MS population has resulted with new concerns regarding DMT efficacy and safety.Areas covered: This review summarizes the evidence of an age-based decrease in the efficacy of MS DMTs and increase in pharmacovigilance concerns. The age effects on pathophysiological MS processes, immunosenescence and its relevance to DMT selection or discontinuation are also reviewed. Lastly, the authors discuss the influence of age-associated comorbidities on DMT initiation and drug-induced events.Expert opinion: There is an age discrepancy between pwMS included in regulatory drug trials and an aging real-world MS population. Most trials demonstrate significantly diminished anti-inflammatory efficacy in patients older than 40 years old. Older age is associated with a greater risk for adverse events including serious infections. Age-associated comorbidities influence the risk-benefit analysis and sometimes cause patients to discontinue DMTs. Instead of chronological age cutoffs, therefore, studies should aim at promoting biologically-based age biomarkers.
Collapse
Affiliation(s)
- Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.,Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| | - Svetlana P Eckert
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.,Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA.,Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Bianca Weinstock-Guttman
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
43
|
Sparaco M, Bonavita S. The role of sex hormones in women with multiple sclerosis: From puberty to assisted reproductive techniques. Front Neuroendocrinol 2021; 60:100889. [PMID: 33189769 DOI: 10.1016/j.yfrne.2020.100889] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/02/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Multiple Sclerosis is a multifactorial chronic autoimmune disease, affecting predominantly females in the fertile age. Sex hormones changes during a woman's life, from puberty to menopause, including pregnancy and puerperium, may influence the onset and course of Multiple Sclerosis. The effect of estrogen levels on immune, clinical and radiological aspects of Multiple Sclerosis, also stimulated investigation on the effect of sexual hormones therapies, such as oral contraceptives and assisted reproductive technique, on the Multiple Sclerosis course. SEARCH STRATEGY AND SELECTION CRITERIA A literature search for original articles and reviews was conducted in the databases, including PubMed, Scopus, and ClinicalTrials.gov of the U.S. National Library of Medicine site from 1988 to 2020. RESULTS AND CONCLUSION This review reports the effects of the physiological and iatrogenic hormonal changes either on immune or clinical or paraclinical features in the different life stages of women affected by Multiple Sclerosis.
Collapse
Affiliation(s)
- Maddalena Sparaco
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Miraglia, 2, 80138 Naples, Italy
| | - Simona Bonavita
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Miraglia, 2, 80138 Naples, Italy.
| |
Collapse
|
44
|
Zeydan B, Atkinson EJ, Weis DM, Smith CY, Gazzuola Rocca L, Rocca WA, Keegan BM, Weinshenker BG, Kantarci K, Kantarci OH. Reproductive history and progressive multiple sclerosis risk in women. Brain Commun 2020; 2:fcaa185. [PMID: 33409489 PMCID: PMC7772117 DOI: 10.1093/braincomms/fcaa185] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/23/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
Being a woman is one of the strongest risk factors for multiple sclerosis. The natural reproductive period from menarche to natural menopause corresponds to the active inflammatory disease period in multiple sclerosis. The fifth decade marks both the peri-menopausal transition in the reproductive aging and a transition from the relapsing-remitting to the progressive phase in multiple sclerosis. A short reproductive period with premature/early menopause and/or low number of pregnancies may be associated with an earlier onset of the progressive multiple sclerosis phase. A cross-sectional study of survey-based reproductive history in a multiple sclerosis clinical series enriched for patients with progressive disease, and a case–control study of multiple sclerosis and age/sex matched controls from a population-based cohort were conducted. Menarche age, number of complete/incomplete pregnancies, menopause type and menopause age were compared between 137 cases and 396 control females. Onset of relapsing-remitting phase of multiple sclerosis, progressive disease onset and reaching severe disability (expanded disability status scale 6) were studied as multiple sclerosis-related outcomes (n = 233). Menarche age was similar between multiple sclerosis and control females (P = 0.306). Females with multiple sclerosis had fewer full-term pregnancies than the controls (P < 0.001). Non-natural menopause was more common in multiple sclerosis (40.7%) than in controls (30.1%) (P = 0.030). Age at natural menopause was similar between multiple sclerosis (median, interquartile range: 50 years, 48–52) and controls (median, interquartile range: 51 years, 49–53) (P = 0.476). Nulliparous females had earlier age at progressive multiple sclerosis onset (mean ± standard deviation: 41.9 ± 12.5 years) than females with ≥1 full-term pregnancies (mean ± standard deviation: 47.1 ± 9.7 years) (P = 0.069) with a pregnancy-dose effect [para 0 (mean ± standard deviation: 41.9 ± 12.5 years), para 1–3 (mean ± standard deviation: 46.4 ± 9.2 years), para ≥4 (mean ± standard deviation: 52.6 ± 12.9 years) (P = 0.005)]. Menopause age was associated with progressive multiple sclerosis onset age (R2 = 0.359, P < 0.001). Duration from onset of relapses to onset of progressive multiple sclerosis was shorter for females with premature/early menopause (n = 26; mean ± standard deviation: 12.9 ± 9.0 years) than for females with normal menopause age (n = 39; mean ± standard deviation: 17.8 ± 10.3 years) but was longer than for males (mean ±standard deviation: 10.0 ± 9.4 years) (P = 0.005). There was a pregnancy-dose effect of age at expanded disability status scale 6 (para 0: 43.0 ± 13.2 years, para 1–3: 51.7 ± 11.3 years, para ≥4: 53.5 ± 4.9 years) (P = 0.013). Age at menopause was associated with age at expanded disability status scale 6 (R2 = 0.229, P < 0.003). Premature/early menopause or nulliparity was associated with earlier onset of progressive multiple sclerosis with a ‘dose effect’ of pregnancies on delaying progressive multiple sclerosis and severe disability. Although causality remains uncertain, our results suggest a beneficial impact of oestrogen in delaying progressive multiple sclerosis. If confirmed in prospective studies, our findings have implications for counselling women with multiple sclerosis about pregnancy, surgical menopause and menopausal hormone therapy.
Collapse
Affiliation(s)
- Burcu Zeydan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA.,Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA.,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN 55905, USA.,Women's Health Research Center, Mayo Clinic, Rochester, MN 55905, USA
| | - Elizabeth J Atkinson
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Delana M Weis
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA.,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Carin Y Smith
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Liliana Gazzuola Rocca
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Walter A Rocca
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA.,Women's Health Research Center, Mayo Clinic, Rochester, MN 55905, USA.,Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Brian Mark Keegan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA.,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Brian G Weinshenker
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA.,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA.,Women's Health Research Center, Mayo Clinic, Rochester, MN 55905, USA
| | - Orhun H Kantarci
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA.,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
45
|
Amato MP, Fonderico M, Portaccio E, Pastò L, Razzolini L, Prestipino E, Bellinvia A, Tudisco L, Fratangelo R, Comi G, Patti F, De Luca G, Brescia Morra V, Cocco E, Pozzilli C, Sola P, Bergamaschi R, Salemi G, Inglese M, Millefiorini E, Galgani S, Zaffaroni M, Ghezzi A, Salvetti M, Lus G, Florio C, Totaro R, Granella F, Vianello M, Gatto M, Di Battista G, Aguglia U, Logullo FO, Simone M, Lucisano G, Iaffaldano P, Trojano M. Disease-modifying drugs can reduce disability progression in relapsing multiple sclerosis. Brain 2020; 143:3013-3024. [DOI: 10.1093/brain/awaa251] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 05/30/2020] [Accepted: 06/29/2020] [Indexed: 11/13/2022] Open
Abstract
Abstract
An ever-expanding number of disease-modifying drugs for multiple sclerosis have become available in recent years, after demonstrating efficacy in clinical trials. In the real-world setting, however, disease-modifying drugs are prescribed in patient populations that differ from those included in pivotal studies, where extreme age patients are usually excluded or under-represented. In this multicentre, observational, retrospective Italian cohort study, we evaluated treatment exposure in three cohorts of patients with relapsing-remitting multiple sclerosis defined by age at onset: paediatric-onset (≤18 years), adult-onset (18–49 years) and late-onset multiple sclerosis (≥50 years). We included patients with a relapsing-remitting phenotype, ≥5 years follow-up, ≥3 Expanded Disability Status Scale (EDSS) evaluations and a first neurological evaluation within 3 years from the first demyelinating event. Multivariate Cox regression models (adjusted hazard ratio with 95% confidence intervals) were used to assess the risk of reaching a first 12-month confirmed disability worsening and the risk of reaching a sustained EDSS of 4.0. The effect of disease-modifying drugs was assessed as quartiles of time exposure. We found that disease-modifying drugs reduced the risk of 12-month confirmed disability worsening, with a progressive risk reduction in different quartiles of exposure in paediatric-onset and adult-onset patients [adjusted hazard ratios in non-exposed versus exposed >62% of the follow-up time: 8.0 (3.5–17.9) for paediatric-onset and 6.3 (4.9–8.0) for adult-onset, P < 0.0001] showing a trend in late-onset patients [adjusted hazard ratio = 1.9 (0.9–4.1), P = 0.07]. These results were confirmed for a sustained EDSS score of 4.0. We also found that relapses were a risk factor for 12-month confirmed disability worsening in all three cohorts, and female sex exerted a protective role in the late-onset cohort. This study provides evidence that sustained exposure to disease-modifying drugs decreases the risk of disability accumulation, seemingly in a dose-dependent manner. It confirms that the effectiveness of disease-modifying drugs is lower in late-onset patients, although still detectable.
Collapse
Affiliation(s)
- Maria Pia Amato
- Department NEUROFARBA, University of Florence, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | | | - Emilio Portaccio
- SOC Neurologia, Ospedale San Giovanni di Dio, AUSL Toscana Centro1, Florence, Italy
| | - Luisa Pastò
- Department NEUROFARBA, University of Florence, Florence, Italy
| | | | - Elio Prestipino
- Department NEUROFARBA, University of Florence, Florence, Italy
| | | | - Laura Tudisco
- Department NEUROFARBA, University of Florence, Florence, Italy
| | | | - Giancarlo Comi
- San Raffaele Hospital - INSPE; Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Patti
- Dipartimento di Scienze Mediche e Chirurgiche e Tecnologie Avanzate, GF Ingrassia, Sez. Neuroscienze, Centro Sclerosi Multipla, University of Catania, Catania, Sicily, Italy
| | - Giovanna De Luca
- Centro Sclerosi Multipla, Clinica Neurologica, Policlinico SS Annunziata, Università ‘G. d'Annunzio’, Chieti-Pescara, Italy
| | - Vincenzo Brescia Morra
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University, Napoli, Italy
| | - Eleonora Cocco
- Centro Sclerosi Multipla, ASSL Cagliari (ATS Sardegna); Dipartimento di Scienze Mediche e Sanità Pubblica, University of Cagliari, Cagliari, Italy
| | - Carlo Pozzilli
- Multiple Sclerosis Center, S. Andrea Hospital, Dept. of Human Neuroscience, Sapienza University, Rome, Italy
| | - Patrizia Sola
- Centro Malattie Demielinizzanti - Dipartimento di Neuroscienze, Azienda Ospedaliero-Universitaria/OCSAE, UO Neurologia, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Giuseppe Salemi
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Sicily, Italy
| | - Matilde Inglese
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Ospedale Policlinico San Martino, IRCCS, Genoa, Italy
| | - Enrico Millefiorini
- Multiple Sclerosis Center, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - Simonetta Galgani
- multiple sclerosis Centre, Department of Neurosciences, S. Camillo - Forlanini Hospital, Rome, Italy
| | - Mauro Zaffaroni
- ASST della Valle Olona, Multiple Sclerosis Center, S. Antonio Abate Hospital of Gallarate, Gallarate, Italy
| | - Angelo Ghezzi
- ASST della Valle Olona, Multiple Sclerosis Center, S. Antonio Abate Hospital of Gallarate, Gallarate, Italy
| | - Marco Salvetti
- Department of Neuroscience, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Centre for Experimental Neurological Therapies, S. Andrea Hospital/Sapienza University, Rome, Italy
- IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Rome, Italy
| | - Giacomo Lus
- Università della Campania Luigi Vanvitelli, Naples, Italy
| | - Ciro Florio
- Multiple Sclerosis Center, Cardarelli Hospital, Naples, Italy
| | - Rocco Totaro
- Demyelinating Diseases Center, Department of Neurology, San Salvatore Hospital, L'Aquila, Italy
| | - Franco Granella
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, Italy
| | - Marika Vianello
- Centro Sclerosi Multipla - Ospedale Regionale ‘Ca’ Foncello', Neurology Unit, Treviso, Italy
| | - Maurizia Gatto
- Ospedale Generale Regionale ‘F. Miulli’, Neurology Unit, Acquaviva delle Fonti (BA), Italy
| | | | - Umberto Aguglia
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, Neurology Unit, Catanzaro, Italy
| | | | - Marta Simone
- Child Neuropsychiatric Unit, Department of Biomedical Sciences and Human Oncology, University ‘Aldo Moro’ of Bari, Policlinico Piazza G. Cesare, 11, 70121, Bari, Italy
| | - Giuseppe Lucisano
- Center for Outcomes Research and Clinical Epidemiology, Pescara, Italy
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari ‘Aldo Moro’ Policlinico, Bari, Italy
| | - Pietro Iaffaldano
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari ‘Aldo Moro’ Policlinico, Bari, Italy
| | - Maria Trojano
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari ‘Aldo Moro’ Policlinico, Bari, Italy
| |
Collapse
|
46
|
Souza KM, Diniz IM, de Lemos LLP, Junior NGR, Zuppo IDF, Teodoro JA, Acurcio FDA, Atallah ÁN, Júnior AAG. Effectiveness of first-line treatment for relapsing-remitting multiple sclerosis in Brazil: A 16-year non-concurrent cohort study. PLoS One 2020; 15:e0238476. [PMID: 32877451 PMCID: PMC7467258 DOI: 10.1371/journal.pone.0238476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/17/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Relapsing-remitting multiple sclerosis (RRMM) is a chronic, progressive, inflammatory and immune-mediated disease that affects the central nervous system and is characterized by episodes of neurological dysfunction followed by a period of remission. The pharmacological strategy aims to delay the progression of the disease and prevent relapse. Interferon beta and glatiramer are commonly used in the Brazilian public health system and are available to patients who meet the guideline criteria. The scenario of multiple treatments available and in development brings the need for discussion and evaluation of the technologies already available before the incorporation of new drugs. This study analyses the effectiveness of first-line treatment of RRMS measured by real-world evidence data, from the Brazilian National Health System (SUS). METHODS AND FINDINGS We conducted a non-concurrent national cohort between 2000 and 2015. The study population consisted of 22,722 patients with RRMS using one of the following first-line drugs of interest: glatiramer or one of three presentations of interferon beta. Kaplan-Meier analysis was used to estimate the time to treatment failure. A univariate and multivariate Cox proportional hazard model was used to evaluate factors associated with treatment failure. In addition, patients were propensity score-matched (1:1) in six groups of comparative first-line treatments to evaluate the effectiveness among them. The analysis indicated a higher risk of treatment failure in female patients (HR = 1.08; P = 0,01), those with comorbidities at baseline (HR = 1.20; P<0,0001), in patients who developed comorbidities after starting treatment (i.e., rheumatoid arthritis-HR = 1.65; P<0,0001), those exclusive SUS patients (HR = 1.31; P<0,0001) and among patients using intramuscular interferon beta (IM βINF-1a) (28% to 60% compared to the other three treatments; P<0,0001). Lower risk of treatment failure was found among patients treated with glatiramer. CONCLUSIONS This retrospective cohort suggests that glatiramer is associated with greater effectiveness compared to the three presentations of interferon beta. When evaluating beta interferons, the results suggest that the intramuscular presentation is not effective in the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Kathiaja Miranda Souza
- Programa de Pós-Graduação em Saúde Baseada em Evidências, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Isabela Maia Diniz
- SUS Collaborating Centre for Technology Assessment & Excellence in Health, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Medicamentos e Assistência Farmacêutica, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lívia Lovato Pires de Lemos
- SUS Collaborating Centre for Technology Assessment & Excellence in Health, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Saúde Pública, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Nélio Gomes Ribeiro Junior
- SUS Collaborating Centre for Technology Assessment & Excellence in Health, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Isabella de Figueiredo Zuppo
- SUS Collaborating Centre for Technology Assessment & Excellence in Health, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana Alvares Teodoro
- SUS Collaborating Centre for Technology Assessment & Excellence in Health, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Medicamentos e Assistência Farmacêutica, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Francisco de Assis Acurcio
- SUS Collaborating Centre for Technology Assessment & Excellence in Health, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Medicamentos e Assistência Farmacêutica, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Saúde Pública, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Álvaro Nagib Atallah
- Programa de Pós-Graduação em Saúde Baseada em Evidências, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
- Cochrane Brazil, Centro de Estudos de Saúde Baseada em Evidências e Avaliação Tecnológica em Saúde, São Paulo, São Paulo, Brazil
| | - Augusto Afonso Guerra Júnior
- SUS Collaborating Centre for Technology Assessment & Excellence in Health, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Medicamentos e Assistência Farmacêutica, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
47
|
Midaglia L, Otero S, Baró F, Montalban X, Tintoré M. Menopause and multiple sclerosis: Influence on prognosis and role of disease-modifying drugs and hormonal replacement therapy. Mult Scler 2020; 28:173-182. [DOI: 10.1177/1352458520952022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background:Sex hormones play a role in both the risk and the prognosis of multiple sclerosis (MS). Considering all stages of women’s reproductive life, data regarding the influence of menopause on MS and vice versa are scarce.Objective:The aim of this study was to review the evidence addressing the relationship between menopause and MS.Methods:A literature search through PubMed was conducted, selecting studies that assessed (1) the influence of menopause in the MS course, (2) the influence of MS and disease-modifying drugs (DMD) on the development of menopause and (3) the effect of hormone replacement therapy (HRT) on symptoms of menopausal MS patients.Results:(1) Most studies suggest menopause may transitorily aggravate MS symptoms. Two studies found an inflexion point on the Expanding Disability Status Scale (EDSS) with clinical worsening during the menopausal transition. Another study considering full EDSS trajectories from clinically isolated syndrome to postmenopause did not find such an EDSS inflection; (2) MS and DMD do not seem to alter the age of menopause onset; and (3) HRT in menopausal MS patients has not shown consistent benefits.Conclusion:Menopause seems to be associated with transient symptom worsening, but the existence of an inflection in disability progression is still controversial. Properly designed studies are necessary to achieve conclusive results.
Collapse
Affiliation(s)
- Luciana Midaglia
- Department of Neurology-Neuroimmunology, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Susana Otero
- Department of Preventive Medicine and Epidemiology, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francesc Baró
- Department of Gynecology, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Montalban
- Department of Neurology-Neuroimmunology, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mar Tintoré
- Department of Neurology-Neuroimmunology, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
48
|
Gilli F, DiSano KD, Pachner AR. SeXX Matters in Multiple Sclerosis. Front Neurol 2020; 11:616. [PMID: 32719651 PMCID: PMC7347971 DOI: 10.3389/fneur.2020.00616] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is the most common chronic inflammatory and neurodegenerative disease of the central nervous system (CNS). An interesting feature that this debilitating disease shares with many other inflammatory disorders is that susceptibility is higher in females than in males, with the risk of MS being three times higher in women compared to men. Nonetheless, while men have a decreased risk of developing MS, many studies suggest that males have a worse clinical outcome. MS exhibits an apparent sexual dimorphism in both the immune response and the pathophysiology of the CNS damage, ultimately affecting disease susceptibility and progression differently. Overall, women are predisposed to higher rates of inflammatory relapses than men, but men are more likely to manifest signs of disease progression and worse CNS damage. The observed sexual dimorphism in MS may be due to sex hormones and sex chromosomes, acting in parallel or combination. In this review, we outline current knowledge on the sexual dimorphism in MS and discuss the interplay of sex chromosomes, sex hormones, and the immune system in driving MS disease susceptibility and progression.
Collapse
Affiliation(s)
- Francesca Gilli
- Department of Neurology, Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | | | | |
Collapse
|
49
|
Krysko KM, Graves JS, Dobson R, Altintas A, Amato MP, Bernard J, Bonavita S, Bove R, Cavalla P, Clerico M, Corona T, Doshi A, Fragoso Y, Jacobs D, Jokubaitis V, Landi D, Llamosa G, Longbrake EE, Maillart E, Marta M, Midaglia L, Shah S, Tintore M, van der Walt A, Voskuhl R, Wang Y, Zabad RK, Zeydan B, Houtchens M, Hellwig K. Sex effects across the lifespan in women with multiple sclerosis. Ther Adv Neurol Disord 2020; 13:1756286420936166. [PMID: 32655689 PMCID: PMC7331774 DOI: 10.1177/1756286420936166] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating central nervous system disorder that is more common in women, with onset often during reproductive years. The female:male sex ratio of MS rose in several regions over the last century, suggesting a possible sex by environmental interaction increasing MS risk in women. Since many with MS are in their childbearing years, family planning, including contraceptive and disease-modifying therapy (DMT) counselling, are important aspects of MS care in women. While some DMTs are likely harmful to the developing fetus, others can be used shortly before or until pregnancy is confirmed. Overall, pregnancy decreases risk of MS relapses, whereas relapse risk may increase postpartum, although pregnancy does not appear to be harmful for long-term prognosis of MS. However, ovarian aging may contribute to disability progression in women with MS. Here, we review sex effects across the lifespan in women with MS, including the effect of sex on MS susceptibility, effects of pregnancy on MS disease activity, and management strategies around pregnancy, including risks associated with DMT use before and during pregnancy, and while breastfeeding. We also review reproductive aging and sexual dysfunction in women with MS.
Collapse
Affiliation(s)
- Kristen M Krysko
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco, 675 Nelson Rising Lane, Suite 221, San Francisco, CA 94158, USA
| | - Jennifer S Graves
- Department of Neurosciences, University of California San Diego, UCSD ACTRI, La Jolla, CA, USA
| | - Ruth Dobson
- Preventive Neurology Unit, Wolfson Institute of Preventive Neurology, Queen Mary University of London, London, UK
| | - Ayse Altintas
- Department of Neurology, School of Medicine, Koc University, Istanbul, Turkey
| | - Maria Pia Amato
- Department NEUROFARBA, Section of Neurosciences, University of Florence, Florence, Italy
| | - Jacqueline Bernard
- Department of Neurology, Oregon Health Science University, Portland, OR, USA
| | - Simona Bonavita
- Department of Advanced Medical and Surgical Sciences, University of Campania, "Luigi Vanvitelli", Naples, Italy
| | - Riley Bove
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco CA, USA
| | - Paola Cavalla
- Department of Neuroscience and Mental Health, City of Health and Science University Hospital of Torino, Turin, Italy
| | - Marinella Clerico
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Teresa Corona
- Clinical Laboratory of Neurodegenerative Disease, National Institute of Neurology and Neurosurgery of Mexico, Mexico City, Mexico
| | - Anisha Doshi
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, University College London (UCL) Institute of Neurology, London, UK
| | - Yara Fragoso
- Multiple Sclerosis & Headache Research Institute, Santos, SP, Brazil
| | - Dina Jacobs
- Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Vilija Jokubaitis
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Doriana Landi
- Department of Systems Medicine, Multiple Sclerosis Center and Research Unit, Tor Vergata University and Hospital, Rome, Italy
| | | | | | | | - Monica Marta
- Neurosciences and Trauma Centre, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Luciana Midaglia
- Department of Neurology-Neuroimmunology, Multiple Sclerosis Centre of Catalonia, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Suma Shah
- Department of Neurology, Duke University, Durham, NC, USA
| | - Mar Tintore
- Department of Neurology-Neuroimmunology, Multiple Sclerosis Centre of Catalonia, Vall d'Hebron University Hospital, Barcelona, Spain
| | | | - Rhonda Voskuhl
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Yujie Wang
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Rana K Zabad
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Burcu Zeydan
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Maria Houtchens
- Department of Neurology, Partners MS Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kerstin Hellwig
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
50
|
|