1
|
Englund S, Frisell T, Qu Y, Gandhi K, Hultén A, Kierkegaard M, Piehl F, Longinetti E. Trajectories of self-reported fatigue following initiation of multiple sclerosis disease-modifying therapy. J Neurol Neurosurg Psychiatry 2024; 95:1012-1020. [PMID: 38744460 PMCID: PMC11503085 DOI: 10.1136/jnnp-2024-333595] [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: 02/09/2024] [Accepted: 04/02/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND We analysed the COMparison Between All immunoTherapies for Multiple Sclerosis (NCT03193866), a Swedish nationwide observational study in relapsing-remitting multiple sclerosis (RRMS), to identify trajectories of fatigue and their association with physical disability following start of disease-modifying therapy (DMT). METHODS Using a group-modelling approach, we assessed trajectories of fatigue with the Fatigue Scale for Motor and Cognitive Functions and physical disability with Expanded Disability Status Scale among 1587 and 1818 individuals who initiated a first DMT and had a first DMT switch, respectively, followed during 2011-2022. We investigated predictors of fatigue trajectories using group membership as a multinomial outcome and calculated conditional probabilities linking membership across the trajectories. RESULTS We identified five trajectories of fatigue in participants who initiated their first DMT: no fatigue (mean starting values=23.7; 18.2% of population), low (35.5; 23.9%), mild (49.0; 21.6%), moderate (61.3; 20.1%) and severe (78.7; 16.1%). While no, low, mild and severe fatigue trajectories remained stable, the moderate trajectory increased to severe fatigue. Similarly, we identified six fatigue trajectories among participants who did a DMT switch, all indicating stable values over time. Women initiating a first DMT were more likely than men to display a severe fatigue trajectory, relative to the no fatigue one. There was a strong association between fatigue and physical disability trajectories. CONCLUSIONS In this cohort of people with actively treated RRMS, self-reported fatigue remained stable or increased over the years following DMT start. There was a strong association between fatigue and disability after DMT start.
Collapse
Affiliation(s)
- Simon Englund
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Frisell
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Ying Qu
- GCSO, Janssen Pharmaceuticals, Stockholm, Sweden
| | - Kavita Gandhi
- Janssen Research & Development, LLC, Titusville, New Jersey, USA
| | | | - Marie Kierkegaard
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Academic Specialist Center, Center of Neurology, Stockholm Health Services, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Elisa Longinetti
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
2
|
Hu Y, Frisell T, Alping P, Song H, Pawitan Y, Fang F, Piehl F. Hospital-Treated Infections and Risk of Disability Worsening in Multiple Sclerosis. Ann Neurol 2024. [PMID: 38984615 DOI: 10.1002/ana.27026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 06/08/2024] [Accepted: 06/13/2024] [Indexed: 07/11/2024]
Abstract
OBJECTIVE To investigate the association between infections and disability worsening in people with multiple sclerosis (MS) treated with either B-cell depleting therapy (rituximab) or interferon-beta/glatiramer acetate (IFN/GA). METHODS This cohort study spanned from 2000 to 2021, using data from the Swedish MS Registry linked to national health care registries, comprising 8,759 rituximab and 7,561 IFN/GA treatment episodes. The risk of hospital-treated infection was estimated using multivariable Cox models. The association between infections and increase in Expanded Disability Status Scale (EDSS) scores was assessed using a doubly robust generalized estimating equations model. Additionally, a piece-wise exponential model analyzed events of increased disability beyond defined cut-off values, controlling for relapses, and MRI activity. RESULTS Compared with IFN/GA, rituximab displayed increased risk of both inpatient- and outpatient-treated infections (hazard ratio [HR], 2.08; 95% confidence interval [CI], 1.50-2.90 and HR, 1.37; 95% CI, 1.13-1.67, respectively). An inpatient-treated infection was associated with a 0.19-unit increase in EDSS (95% CI, 0.12-0.26). Degree of worsening was greatest for progressive MS, and under IFN/GA treatment, which unlike rituximab, was more commonly associated with MRI activity. After controlling for relapses and MRI activity, inpatient-treated infections were associated with disability worsening in people with relapsing-remitting MS treated with IFN/GA (HR, 2.01; 95% CI, 1.59-2.53), but not in those treated with rituximab. INTERPRETATION Compared to IFN/GA, rituximab doubled the infection risk, but reduced the risk of subsequent disability worsening. Further, the risk of worsening after hospital-treated infection was greater with progressive MS than with relapsing-remitting MS. Infection risk should be considered to improve long term outcomes. ANN NEUROL 2024.
Collapse
Affiliation(s)
- Yihan Hu
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Frisell
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Peter Alping
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Huan Song
- Med-X Center for Informatics, Sichuan University, Chengdu, China
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yudi Pawitan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Fang Fang
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Center for Neurology, Academic Specialist Center, Stockholm, Sweden
| |
Collapse
|
3
|
Piehl F, Alping P, Virtanen S, Englund S, Burman J, Fink K, Fogdell-Hahn A, Gunnarsson M, Hillert J, Langer-Gould A, Lycke J, Mellergård J, Nilsson P, Olsson T, Salzer J, Svenningsson A, Frisell T. COMBAT-MS: A Population-Based Observational Cohort Study Addressing the Benefit-Risk Balance of Multiple Sclerosis Therapies Compared with Rituximab. Ann Neurol 2024. [PMID: 38923558 DOI: 10.1002/ana.27012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE To assess comparative effectiveness, safety, and tolerability of off-label rituximab, compared with frequently used therapies approved for multiple sclerosis (MS). METHODS A Swedish cohort study of persons with relapsing-remitting MS, age 18 to 75 years at inclusion and with a first therapy start or a first therapy switch between 2011 and 2018. Low-dose rituximab was compared with MS-approved therapies. Primary outcomes were proportions with 12 months confirmed disability worsening and change in MS Impact Scale-29 (MSIS-29) scores, respectively. Secondary endpoints included relapses, therapy discontinuation, and serious adverse events. Analyses used an intention-to-treat approach and were adjusted for demographics, MS features, and health characteristics. RESULTS We included 2,449 participants as first therapy start and 2,463 as first therapy switch. Proportions with disability worsening at 3 years were 9.1% for rituximab as first therapy and 5.1% after therapy switch, with no differences to MS-approved comparators. Worsening on rituximab was mostly independent of relapses. MSIS-29 with rituximab at 3 years improved by 1.3/8.4 points (physical/psychological) for first disease-modifying therapy (DMT) and 0.4/3.6 for DMT switch, and was mostly similar across therapies. Rituximab had lower relapse rates and higher therapy persistence in both groups. The rate of hospital-treated infections was higher with rituximab after a therapy switch, but not as a first therapy. INTERPRETATION This population-based real-world cohort study found low rates of disability progression, mostly independent of relapses, and without significant differences between rituximab and MS-approved comparators. Rituximab led to lower rates of inflammatory activity and higher treatment persistence, but was associated with an increased rate of serious infections. ANN NEUROL 2024.
Collapse
Affiliation(s)
- Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Academic Specialist Center, Stockholm Health Services, Stockholm, Sweden
| | - Peter Alping
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Suvi Virtanen
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Simon Englund
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Joachim Burman
- Department of Neurology, Uppsala University Hospital, and Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Katharina Fink
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin Gunnarsson
- Department of Neurology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Annette Langer-Gould
- Clinical and Translational Neuroscience, Southern California Permanente Medical Group, Kaiser Permanente, Los Angeles, CA, USA
| | - Jan Lycke
- Department of Neurology, Sahlgrenska University Hospital, and Department of Clinical Neuroscience, University of Gothenburg, Gothenburg, Sweden
| | - Johan Mellergård
- Department of Neurology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Petra Nilsson
- Department of Neurology, Skåne University Hospital, and Department of Clinical Sciences/Neurology, Lund University, Lund, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jonatan Salzer
- Department of Neurology, Umeå University Hospital, and Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Anders Svenningsson
- Department of Neurology, Danderyd Hospital, and Department of Clinical Sciences, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Frisell
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Zaratin P, Samadzadeh S, Seferoğlu M, Ricigliano V, dos Santos Silva J, Tunc A, Brichetto G, Coetzee T, Helme A, Khan U, McBurney R, Peryer G, Weiland H, Baneke P, Battaglia MA, Block V, Capezzuto L, Carment L, Cortesi PA, Cutter G, Leocani L, Hartung HP, Hillert J, Hobart J, Immonen K, Kamudoni P, Middleton R, Moghames P, Montalban X, Peeters L, Sormani MP, van Tonder S, White A, Comi G, Vermersch P. The global patient-reported outcomes for multiple sclerosis initiative: bridging the gap between clinical research and care - updates at the 2023 plenary event. Front Neurol 2024; 15:1407257. [PMID: 38974689 PMCID: PMC11225898 DOI: 10.3389/fneur.2024.1407257] [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: 03/26/2024] [Accepted: 06/04/2024] [Indexed: 07/09/2024] Open
Abstract
Significant advancements have been achieved in delineating the progress of the Global PROMS (PROMS) Initiative. The PROMS Initiative, a collaborative endeavor by the European Charcot Foundation and the Multiple Sclerosis International Federation, strives to amplify the influence of patient input on MS care and establish a cohesive perspective on Patient-Reported Outcomes (PROs) for diverse stakeholders. This initiative has established an expansive, participatory governance framework launching four dedicated working groups that have made substantive contributions to research, clinical management, eHealth, and healthcare system reform. The initiative prioritizes the global integration of patient (For the purposes of the Global PROMS Initiative, the term "patient" refers to the people with the disease (aka People with Multiple Sclerosis - pwMS): any individual with lived experience of the disease. People affected by the disease/Multiple Sclerosis: any individual or group that is affected by the disease: E.g., family members, caregivers will be also engaged as the other stakeholders in the initiative). insights into the management of MS care. It merges subjective PROs with objective clinical metrics, thereby addressing the complex variability of disease presentation and progression. Following the completion of its second phase, the initiative aims to help increasing the uptake of eHealth tools and passive PROs within research and clinical settings, affirming its unwavering dedication to the progressive refinement of MS care. Looking forward, the initiative is poised to continue enhancing global surveys, rethinking to the relevant statistical approaches in clinical trials, and cultivating a unified stance among 'industry', regulatory bodies and health policy making regarding the application of PROs in MS healthcare strategies.
Collapse
Affiliation(s)
- Paola Zaratin
- Research Department, Italian Multiple Sclerosis Foundation, Genoa, Italy
| | - Sara Samadzadeh
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Institute of Regional Health Research and Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Neurology, The Center for Neurological Research, Næstved-Slagelse-Ringsted Hospitals, Slagelse, Denmark
| | - Meral Seferoğlu
- Department of Neurology, Bursa Faculty of Medicine, Bursa Yüksek İhtisas Training and Research Hospital, University of Health Sciences, Bursa, Türkiye
| | - Vito Ricigliano
- Sorbonne Université, Paris Brain Institute, ICM, CNRS, Inserm, Paris, France
- Neurology Department, Pitié-Salpêtrière Hospital, APHP, Paris, France
| | - Jonadab dos Santos Silva
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Programa de Pós Graduação Stricto Senso em Neurologia, Department of Neurology, Fluminense Federal University, Niterói, Brazil
| | - Abdulkadir Tunc
- Department of Neurology, Sakarya University Faculty of Medicine, Sakarya, Türkiye
| | | | - Timothy Coetzee
- National Multiple Sclerosis Society, New York, NY, United States
| | - Anne Helme
- Multiple Sclerosis International Federation, London, United Kingdom
| | - Usman Khan
- Institute for Healthcare Policy, KU Leuven, Leuven, Belgium
| | | | - Guy Peryer
- Multiple Sclerosis Society UK, London, United Kingdom
| | - Helga Weiland
- Multiple Sclerosis South Africa, Hermanus, Western Cape, South Africa
| | - Peer Baneke
- Multiple Sclerosis International Federation, London, United Kingdom
| | | | - Valerie Block
- University of California, San Francisco, San Francisco, CA, United States
| | | | | | - Paolo Angelo Cortesi
- Research Centre on Public Health (CESP), University of Milan-Bicocca, Milan, Italy
| | - Gary Cutter
- Department of Biostatistics, School of Public Health, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Letizia Leocani
- University Vita-Salute San Raffaele, Milan, Italy
- Department of Rehabilitation Sciences, Casa di Cura Igea, Milan, Italy
| | - Hans-Peter Hartung
- Department of Neurology, UKD, Medical Faculty, Heinrich Heine Universitat Düsseldorf, Düsseldorf, Germany
- Brain and Mind Center, University of Sydney, Camperdown, NSW, Australia
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Palacky University Olomouc, Olomouc, Czechia
| | - Jan Hillert
- Department of Clinical Neuroscience, Neurogenetics Multiple Sclerosis, Karolinska Institutet, Stockholm, Sweden
| | - Jeremy Hobart
- Plymouth University Peninsula Schools of Medicine and Dentistry Devon, Plymouth, United Kingdom
| | - Kaisa Immonen
- European Medicines Agency, Public and Stakeholder Engagement Department, Amsterdam, North Holland, Netherlands
| | | | - Rod Middleton
- Faculty of Medicine Health and Life-Sciences, Swansea University, Swansea, United Kingdom
| | | | - Xavier Montalban
- Hopital Vall d’Hebron, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Liesbet Peeters
- Hasselt University–Biomedical Research Institute (BIOMED), Hasselt, Belgium
| | | | - Susanna van Tonder
- European MS Platform, Brussels, Belgium
- MS Lëtzebuerg, Luxembourg, Belgium
| | - Angela White
- National Multiple Sclerosis Society, New York, NY, United States
| | | | - Patrick Vermersch
- Université de Lille, Inserm LilNCog, CHU Lille, FHU Precise, Lille, France
| |
Collapse
|
5
|
Machado A, Murley C, Dervish J, Teni FS, Friberg E. Work Adjustments by Types of Occupations Amongst People with Multiple Sclerosis: A Survey Study. JOURNAL OF OCCUPATIONAL REHABILITATION 2024; 34:461-471. [PMID: 37923959 PMCID: PMC11180149 DOI: 10.1007/s10926-023-10142-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/27/2023] [Indexed: 11/06/2023]
Abstract
PURPOSE To explore the occurrence of work adjustments for people with multiple sclerosis (MS) across types of occupations (managerial, office, and manual workers). METHODS All working-aged (20-50 years) residents in Sweden diagnosed with MS were invited to participate in a web-based survey in 2021. Responses were linked to individual-level nationwide registers. Descriptive analyses were conducted to compare sociodemographic and clinical variables across occupations as well as other responses. The odds ratio of having any adjustment at work was determined using multinomial logistic regression. RESULTS From all 4412 respondents (52% response rate), 3313 employees were included. The majority were women (72%) and had low (24.2%) or mild disease severity (44.7%). Nevertheless, different work adjustments across occupations were observed. Compared to the other occupations, office workers reported more invisible symptoms, more work adjustments and considered adapted schedules as the most important adjustment. On the contrary, more managers reported having no limiting symptoms and consequently, disclosed their diagnosis less often. They also reported having fewer work adjustments and more opportunities to modify their work than office and manual workers. Manual workers had a higher likelihood to report needing more support at work than office workers and managers. Further, a higher likelihood of having work adjustments was associated with progressive MS, higher MS severity, and invisible symptoms. CONCLUSION A more severe clinical profile of MS was associated with having work adjustments. The physical demands and responsibilities of an occupation play an important role when requesting and getting work adjustments amongst employees with MS.
Collapse
Affiliation(s)
- Alejandra Machado
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, SE-171 77, Stockholm, Sweden.
| | - Chantelle Murley
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Jessica Dervish
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Fitsum Sebsibe Teni
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Emilie Friberg
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| |
Collapse
|
6
|
Alping P, Neovius M, Piehl F, Frisell T. Real-World Healthcare Cost Savings and Reduced Relapse Rate with Off-Label Rituximab versus Disease-Modifying Treatments Approved for Relapsing-Remitting Multiple Sclerosis: A Nationwide Cost-Effectiveness Study. Ann Neurol 2024; 95:1099-1111. [PMID: 38529711 DOI: 10.1002/ana.26914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024]
Abstract
OBJECTIVE Although off-label use of rituximab is a common alternative to disease-modifying therapies (DMTs) approved for multiple sclerosis (MS) in several countries, the impact of this on treatment cost-effectiveness is not well known. METHODS We evaluated the relative cost-effectiveness of rituximab and MS-approved DMTs in a register-based cohort study of Swedish residents with relapsing-remitting MS, aged 18-65 years, starting treatment with rituximab, natalizumab, fingolimod, or dimethyl fumarate between January 2010 and July 2016, and followed through July 2021 (n = 5,924). By linking the population-based Swedish MS register to several Swedish health care and demographic registers, we estimated health care costs in relation to number of relapses, over 5 years from treatment start. Differences between treatments were estimated in inverse probability of treatment-weighted regression models, adjusting for a broad range of potential confounders covering demographics, medical history, and MS-related clinical characteristics. RESULTS Off-label rituximab was associated with both lower total health care costs (mean cost savings ranged $35,000-$66,000 vs. each approved DMT), and fewer relapses (mean number of prevented relapses ranged 0.12-0.22), per started therapy over 5 years. Results were robust to variations in discounting and pricing of health care visits, with the main driver of cost-savings being the price of the index drug itself. INTERPRETATION The cost-effectiveness of rituximab dominated the MS-approved alternatives. Off-label, low-dose rituximab should be considered for persons with MS and could reduce barriers to treatment, especially in resource-limited settings. ANN NEUROL 2024;95:1099-1111.
Collapse
Affiliation(s)
- Peter Alping
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Martin Neovius
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Frisell
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
7
|
Murley C, Dervish J, Machado A, Svärd V, Wennman-Larsen A, Hillert J, Friberg E. Exploring the working life of people with multiple sclerosis during the COVID-19 pandemic in Sweden. BMC Public Health 2024; 24:1389. [PMID: 38783221 PMCID: PMC11119790 DOI: 10.1186/s12889-024-18844-9] [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: 02/13/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic led to vast changes in working life and conditions in which we work. These changes may affect people with multiple sclerosis (PwMS) differently. We aimed to describe the working situation of PwMS during the COVID-19 pandemic and the pandemic's impact on their working lives. METHODS All individuals aged 20-50 listed in the Swedish Multiple Sclerosis Registry were invited to participate in an online survey in 2021. Closed and open-ended responses linked to individual-level register data were used in this exploratory mixed-methods study. Differences in the proportions reporting specific impacts were assessed with chi-square tests by sex, MS severity, education, and profession. The open-ended answers were analysed through content analysis. RESULTS Over 8500 PwMS were invited (52% response rate). We included the 3887 respondents who answered questions about the impact of the pandemic on working life. Most (93.7%) reported being in paid work. An impact of the ongoing pandemic to one's daily occupation was reported by 26.2%, with different characteristics observed across the impacts. Four categories of type of answers were identified from the open-ended answers: Direct impact on one's occupation, Disclosing or concealing MS in the workplace, Worry and uncertainty, and Broader impact to life situation. CONCLUSIONS PwMS navigated the pandemic by interrupting as well as continuing their working lives. Many PwMS reported that the pandemic did not affect their work situation. However, the reported impacts differed among the participants and a sense of uncertainty and worry was often underlying their statements. Lessons from the pandemic may support future work participation.
Collapse
Affiliation(s)
- Chantelle Murley
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Jessica Dervish
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Alejandra Machado
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, SE-171 77, Sweden.
| | - Veronica Svärd
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, SE-171 77, Sweden
- Department of Social Work, School of Social Sciences, Huddinge, SE-141 89, Sweden
| | - Agneta Wennman-Larsen
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, SE-171 77, Sweden
- Department of Nursing Science, Sophiahemmet University, Stockholm, SE-114 86, Sweden
| | - Jan Hillert
- Division of Neurology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Emilie Friberg
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| |
Collapse
|
8
|
Virtanen S, Piehl F, Frisell T. Impact of previous treatment history and B-cell depletion treatment duration on infection risk in relapsing-remitting multiple sclerosis: a nationwide cohort study. J Neurol Neurosurg Psychiatry 2024:jnnp-2023-333206. [PMID: 38744458 DOI: 10.1136/jnnp-2023-333206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND B-cell depletion displays striking effectiveness in relapsing-remitting multiple sclerosis (RRMS), but is also associated with increased infection risk. To what degree previous treatment history, disease-modifying therapy (DMT) switching pattern and time on treatment modulate this risk is unknown. The objective here was to evaluate previous DMT use and treatment duration as predictors of infection risk with B-cell depletion. METHODS We conducted a nationwide RRMS cohort study leveraging data from the Swedish MS registry and national demographic and health registries recording all outpatient-treated and inpatient-treated infections and antibiotics prescriptions from 1 January 2012 to 30 June 2021. The risk of infection during treatment was compared by DMT, treatment duration, number and type of prior treatment and adjusted for a number of covariates. RESULTS Among 4694 patients with RRMS on B-cell depletion (rituximab), 6049 on other DMTs and 20 308 age-sex matched population controls, we found higher incidence rates of inpatient-treated infections with DMTs other than rituximab used in first line (10.4; 95% CI 8.1 to 12.9, per 1000 person-years), being further increased with rituximab (22.7; 95% CI 18.5 to 27.5), compared with population controls (6.6; 95% CI 6.0 to 7.2). Similar patterns were seen for outpatient infections and antibiotics prescriptions. Infection rates on rituximab did not vary between first versus later line treatment, type of DMT before switch or exposure time. CONCLUSION These findings underscore an important safety concern with B-cell depletion in RRMS, being evident also in individuals with shorter disease duration and no previous DMT exposure, in turn motivating the application of risk mitigation strategies.
Collapse
Affiliation(s)
- Suvi Virtanen
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Frisell
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
9
|
Sandesjö F, Tremlett H, Fink K, Marrie RA, Zhu F, Wickström R, McKay KA. Incidence rate and prevalence of pediatric-onset multiple sclerosis in Sweden: A population-based register study. Eur J Neurol 2024; 31:e16253. [PMID: 38369806 PMCID: PMC11236061 DOI: 10.1111/ene.16253] [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: 10/24/2023] [Revised: 01/18/2024] [Accepted: 02/04/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND AND PURPOSE Pediatric-onset multiple sclerosis (PoMS) is associated with high health care use. To plan resource allocation for this patient group, knowledge of the incidence rate and prevalence is important. However, such studies are scarce, few are population-based, and the methodology varies widely. We aimed to address this knowledge gap by performing a nationwide study of the incidence rate and prevalence of PoMS in Sweden, an area of high multiple sclerosis (MS) incidence and prevalence. METHODS MS cases were identified by linking two nationwide registers, the National Patient Register and the Swedish MS Registry. MS cases having their first central nervous system demyelinating event or MS clinical onset before age 18 years were classified as pediatric onset. Incidence rate and prevalence were estimated annually over the study period (2006-2016) for the total population and stratified by sex and age group (<12, 12-15, and 16-17 years). Temporal trends and ratios between sexes and age groups were estimated. RESULTS We identified 238 incident cases from 2006 to 2016, corresponding to an overall crude incidence rate of 1.12 per 100,000 person-years and an overall crude prevalence of 2.82 per 100,000 population. There was a higher incidence rate among females and the highest age category. The overall incidence rate and prevalence estimates remained stable during the study period. CONCLUSIONS Sweden exhibits a consistently high incidence rate and prevalence of PoMS that has remained stable over time. This knowledge serves as a tool to aid in planning resource allocation and health services for this patient population.
Collapse
Affiliation(s)
- Fredrik Sandesjö
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
| | - Helen Tremlett
- Division of Neurology, Department of Medicine, The Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Katharina Fink
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Ruth Ann Marrie
- Department of Internal MedicineDepartment of Community Health SciencesMax Rady College of Medicine, Rady Faculty of Health Sciences, University of ManitobaWinnipegManitobaCanada
| | - Feng Zhu
- Division of Neurology, Department of Medicine, The Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Ronny Wickström
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
| | - Kyla A. McKay
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| |
Collapse
|
10
|
Dervish J, Arfuch VM, Murley C, McKay KA, Machado A, Wennman-Larsen A, Friberg E. Disclosing or concealing multiple sclerosis in the workplace: two sides of the same coin-insights from a Swedish population-based survey. Front Public Health 2024; 12:1331746. [PMID: 38469271 PMCID: PMC10925750 DOI: 10.3389/fpubh.2024.1331746] [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: 11/01/2023] [Accepted: 02/16/2024] [Indexed: 03/13/2024] Open
Abstract
Background People with multiple sclerosis (PwMS) face health and social challenges of living with a chronic and potentially disabling condition. To disclose or conceal MS at work may critically affect individuals' work situation, career opportunities, and health. PwMS may experience a dilemma when assessing if the possible benefits of disclosing the diagnosis outweigh the possible risks. However, concealing in the long-term may have health implications and prevent opportunities for support and work adjustments. Few studies have examined what drives PwMS to disclose or conceal MS at work and the consequences of these ways of managing MS. Objectives To explore the reasons PwMS report for disclosing and/or concealing their MS diagnosis in the workplace, as well as the consequences they have experienced. Methods A web-based survey of PwMS was conducted in 2021. All individuals aged 20-50 listed in the Swedish MS registry were invited to participate. The response rate was 52% and among these participants, 3,810 (86%) completed questions regarding workplace disclosure and/or concealment of MS. Free-text responses on these topics were analyzed using inductive content analysis. Results It was common to disclose MS in the workplace (85%). Identified drivers for disclosure and concealment related to four categories: Work-related, Social, Personal and Circumstantial. Work-related drivers focused on employment or protecting one's career, and changing one's work situation versus maintaining it. Social drivers included the need for support, addressing or preventing stigma, and being considerate of others. Personal drivers were linked to moral values/personal beliefs and processing of the diagnosis. Circumstantial drivers related to involuntary or unforeseen events, timing factors, one's medical condition and external opinion/advice. Identified consequences for disclosure and concealment related to three categories: Work-life, Social, and Personal. Work-life consequences included work arrangements, and career opportunities. Social consequences were linked to MS awareness, stigma, interactions and social support, as well as dynamics of work relationships. Personal consequences involved levels of disease acceptance, and attitudes toward managing MS. Conclusion PwMS often described the question of disclosure as challenging and navigated it with caution, as both disclosure and concealment can yield favorable and unfavorable outcomes.
Collapse
Affiliation(s)
- Jessica Dervish
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Victoria Mailen Arfuch
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Psychiatry, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Chantelle Murley
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Kyla A. McKay
- Division of Neuro, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Alejandra Machado
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Wennman-Larsen
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Nursing Science, Sophiahemmet University, Stockholm, Sweden
| | - Emilie Friberg
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Longinetti E, Englund S, Burman J, Fink K, Fogdell-Hahn A, Gunnarsson M, Hillert J, Langer-Gould AM, Lycke J, Nilsson P, Salzer J, Svenningsson A, Mellergård J, Olsson T, Piehl F, Frisell T. Trajectories of cognitive processing speed and physical disability over 11 years following initiation of a first multiple sclerosis disease-modulating therapy. J Neurol Neurosurg Psychiatry 2024; 95:134-141. [PMID: 37558400 PMCID: PMC10850621 DOI: 10.1136/jnnp-2023-331784] [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: 05/08/2023] [Accepted: 07/30/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND We analysed the COMparison Between All immunoTherapies for Multiple Sclerosis (NCT03193866), a Swedish nationwide observational study in relapsing-remitting multiple sclerosis (RRMS), to identify trajectories of processing speed and physical disability after disease-modulating therapy (DMT) start. METHODS Using a group-modelling approach, we assessed trajectories of processing speed with oral Symbol Digit Modalities Test (SDMT) and physical disability with Expanded Disability Status Scale, from first DMT start among 1645 patients with RRMS followed during 2011-2022. We investigated predictors of trajectories using group membership as a multinomial outcome and calculated conditional probabilities linking membership across the trajectories. RESULTS We identified 5 stable trajectories of processing speed: low SDMT scores (mean starting values=29.9; 5.4% of population), low/medium (44.3; 25.3%), medium (52.6; 37.9%), medium/high (63.1; 25.8%) and high (72.4; 5.6%). We identified 3 physical disability trajectories: no disability/stable (0.8; 26.8%), minimal disability/stable (1.6; 58.1%) and moderate disability (3.2; 15.1%), which increased to severe disability. Older patients starting interferons were more likely than younger patients starting rituximab to be on low processing speed trajectories. Older patients starting teriflunomide, with more than one comorbidity, and a history of pain treatment were more likely to belong to the moderate/severe physical disability trajectory, relative to the no disability one. There was a strong association between processing speed and physical disability trajectories. CONCLUSIONS In this cohort of actively treated RRMS, patients' processing speed remained stable over the years following DMT start, whereas patients with moderate physical disability deteriorated in physical function. Nevertheless, there was a strong link between processing speed and disability after DMT start.
Collapse
Affiliation(s)
- Elisa Longinetti
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Simon Englund
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Joachim Burman
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Katharina Fink
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Jan Lycke
- Department of Clinical Neuroscience, University of Gothenburg, Goteborg, Sweden
| | - Petra Nilsson
- Department of Clinical Sciences, Division of Neurology, Lund University, Lund, Sweden
| | - Jonatan Salzer
- Department of Clinical Sciences, Neurosciences, Umeå University, Umeå, Sweden
| | - Anders Svenningsson
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Johan Mellergård
- Department of Neurology, Linköping University, Linkoping, Östergötland, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Frisell
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Murley C, Tinghög P, Teni FS, Machado A, Alexanderson K, Hillert J, Karampampa K, Friberg E. Excess costs of multiple sclerosis: a register-based study in Sweden. THE EUROPEAN JOURNAL OF HEALTH ECONOMICS : HEPAC : HEALTH ECONOMICS IN PREVENTION AND CARE 2023; 24:1357-1371. [PMID: 36418785 PMCID: PMC9685028 DOI: 10.1007/s10198-022-01547-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 11/07/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND AND OBJECTIVE Population-based estimates of the socioeconomic burden of multiple sclerosis (MS) are limited, especially regarding primary healthcare. This study aimed to estimate the excess costs of people with MS that could be attributed to their MS, including primary healthcare. METHODS An observational study was conducted of the 2806 working-aged people with MS in Stockholm, Sweden and 28,060 propensity score matched references without MS. Register-based resource use was quantified for 2018. Annual healthcare costs (primary, specialised outpatient, and inpatient healthcare visits along with prescribed drugs) and productivity losses (operationalised by sickness absence and disability pension days) were quantified using bottom-up costing. The costs of people with MS were compared with those of the references using independent t-tests with bootstrapped 95% confidence intervals (CIs) to isolate the excess costs of MS from the mean difference. RESULTS The mean annual excess costs of MS for healthcare were €7381 (95% CI 6991-7816) per person with MS with disease-modifying therapies as the largest component (€4262, 95% CI 4026-4497). There was a mean annual excess cost for primary healthcare of €695 (95% CI 585-832) per person with MS, comprising 9.4% of the excess healthcare costs of MS. The mean annual excess costs of MS for productivity losses were €13,173 (95% CI 12,325-14,019) per person with MS, predominately from disability pension (79.3%). CONCLUSIONS The socioeconomic burden of MS in Sweden from healthcare consumption and productivity losses was quantified, updating knowledge on the cost structure of the substantial excess costs of MS.
Collapse
Affiliation(s)
- Chantelle Murley
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Petter Tinghög
- Department of Health Sciences, Swedish Red Cross University, 141 21, Huddinge, Sweden
- Division of Psychology, Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Fitsum Sebsibe Teni
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Alejandra Machado
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Kristina Alexanderson
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Jan Hillert
- Division of Neurology, Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Korinna Karampampa
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Emilie Friberg
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| |
Collapse
|
13
|
Mosconi P, Guerra T, Paletta P, D'Ettorre A, Ponzio M, Battaglia MA, Amato MP, Bergamaschi R, Capobianco M, Comi G, Gasperini C, Patti F, Pugliatti M, Ulivelli M, Trojano M, Lepore V. Data monitoring roadmap. The experience of the Italian Multiple Sclerosis and Related Disorders Register. Neurol Sci 2023; 44:4001-4011. [PMID: 37311951 PMCID: PMC10264214 DOI: 10.1007/s10072-023-06876-9] [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: 02/20/2023] [Accepted: 05/24/2023] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Over the years, disease registers have been increasingly considered a source of reliable and valuable population studies. However, the validity and reliability of data from registers may be limited by missing data, selection bias or data quality not adequately evaluated or checked. This study reports the analysis of the consistency and completeness of the data in the Italian Multiple Sclerosis and Related Disorders Register. METHODS The Register collects, through a standardized Web-based Application, unique patients. Data are exported bimonthly and evaluated to assess the updating and completeness, and to check the quality and consistency. Eight clinical indicators are evaluated. RESULTS The Register counts 77,628 patients registered by 126 centres. The number of centres has increased over time, as their capacity to collect patients. The percentages of updated patients (with at least one visit in the last 24 months) have increased from 33% (enrolment period 2000-2015) to 60% (enrolment period 2016-2022). In the cohort of patients registered after 2016, there were ≥ 75% updated patients in 30% of the small centres (33), in 9% of the medium centres (11), and in all the large centres (2). Clinical indicators show significant improvement for the active patients, expanded disability status scale every 6 months or once every 12 months, visits every 6 months, first visit within 1 year and MRI every 12 months. CONCLUSIONS Data from disease registers provide guidance for evidence-based health policies and research, so methods and strategies ensuring their quality and reliability are crucial and have several potential applications.
Collapse
Affiliation(s)
- Paola Mosconi
- Laboratorio di Ricerca per il Coinvolgimento dei Cittadini in Sanità, Dipartimento di Salute Pubblica, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, 20156, Italy.
| | - Tommaso Guerra
- Dipartimento Scienze Mediche di Base, Neuroscienze ed Organi di Senso, Università degli Studi Aldo Moro, Bari, Italy
| | - Pasquale Paletta
- Laboratorio di Ricerca per il Coinvolgimento dei Cittadini in Sanità, Dipartimento di Salute Pubblica, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, 20156, Italy
| | - Antonio D'Ettorre
- Laboratorio di Ricerca per il Coinvolgimento dei Cittadini in Sanità, Dipartimento di Salute Pubblica, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, 20156, Italy
| | - Michela Ponzio
- Scientific Research Area, Italian Multiple Sclerosis Foundation, Genoa, Italy
| | - Mario Alberto Battaglia
- Scientific Research Area, Italian Multiple Sclerosis Foundation, Genoa, Italy
- Department of Physiopathology, Experimental Medicine and Public Health, University of Siena, Siena, Italy
| | | | - Roberto Bergamaschi
- Centro Interdipartimentale Sclerosi Multipla, Fondazione Istituto Neurologico C. Mondino, Pavia, Italy
| | - Marco Capobianco
- Centro Sclerosi Multipla, SC Neurologia, AO Santa Croce E Carle, Cuneo, Italy
| | - Giancarlo Comi
- Casa di Cura del Policlinico, Università Vita Salute San Raffaele, Milan, Italy
| | - Claudio Gasperini
- UOC di Neurologia e Neurofisiopatologia Azienda Ospedaliera S. Camillo-Forlanini, Rome, Italy
| | - Francesco Patti
- Centro Sclerosi Multipla AOU Policlinico Vittorio Emanuele, Catania, Italy
| | - Maura Pugliatti
- Centro di Servizio e Ricerca sulla Sclerosi Multipla, AOU di Ferrara, Ferrara, Italy
| | - Monica Ulivelli
- Dipartimento di Scienze Mediche Chirurgiche e Neuroscienze, Università degli Studi di Siena, Siena, Italy
| | - Maria Trojano
- Dipartimento Scienze Mediche di Base, Neuroscienze ed Organi di Senso, Università degli Studi Aldo Moro, Bari, Italy
| | - Vito Lepore
- Laboratorio di Ricerca per il Coinvolgimento dei Cittadini in Sanità, Dipartimento di Salute Pubblica, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, 20156, Italy
| |
Collapse
|
14
|
Boles GS, Hillert J, Ramanujam R, Westerlind H, Olsson T, Kockum I, Manouchehrinia A. The familial risk and heritability of multiple sclerosis and its onset phenotypes: A case-control study. Mult Scler 2023; 29:1209-1215. [PMID: 37435869 PMCID: PMC10503245 DOI: 10.1177/13524585231185258] [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: 03/16/2023] [Revised: 06/05/2023] [Accepted: 06/13/2023] [Indexed: 07/13/2023]
Abstract
BACKGROUND The two main phenotypes of multiple sclerosis (MS), primary progressive (PPMS) and relapsing Onset (ROMS), show clinical and demographic differences suggesting possible differential risk mechanisms. Understanding the heritable features of these phenotypes could provide aetiological insight. OBJECTIVES To evaluate the magnitude of familial components in PPMS and ROMS and to estimate the heritability of disease phenotypes. METHODS We used data from 25,186 MS patients of Nordic ancestry from the Swedish MS Registry between 1987 and 2019 with known disease phenotype (1593 PPMS and 16,718 ROMS) and 251,881 matched population-based controls and 3,364,646 relatives of cases and controls. Heritability was calculated using threshold-liability models. For familial odds ratios (ORs), logistic regression with robust sandwich estimator was utilized. RESULTS The OR of MS diagnosis in those with a first-degree family member with ROMS was 7.00 and 8.06 in those with PPMS. The corresponding ORs for having a second-degree family member with ROMS was 2.16 and 2.18 in PPMS. The additive genetic effect in ROMS was 0.54 and 0.22 in PPMS. CONCLUSION Risk of MS increases by several folds in those with a relative with MS. The likelihood of developing either disease phenotype appears independent of genetic predisposition.
Collapse
Affiliation(s)
| | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Ryan Ramanujam
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Helga Westerlind
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Centre for Molecular Medicine (CMM), Department of Clinical Neurosciences, Karolinska Institute, Stockholm, Sweden
| | - Ingrid Kockum
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Centre for Molecular Medicine (CMM), Department of Clinical Neurosciences, Karolinska Institute, Stockholm, Sweden
| | - Ali Manouchehrinia
- The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
- Centre for Molecular Medicine (CMM), Department of Clinical Neurosciences, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
15
|
Oddsson A, Sulem P, Sveinbjornsson G, Arnadottir GA, Steinthorsdottir V, Halldorsson GH, Atlason BA, Oskarsson GR, Helgason H, Nielsen HS, Westergaard D, Karjalainen JM, Katrinardottir H, Fridriksdottir R, Jensson BO, Tragante V, Ferkingstad E, Jonsson H, Gudjonsson SA, Beyter D, Moore KHS, Thordardottir HB, Kristmundsdottir S, Stefansson OA, Rantapää-Dahlqvist S, Sonderby IE, Didriksen M, Stridh P, Haavik J, Tryggvadottir L, Frei O, Walters GB, Kockum I, Hjalgrim H, Olafsdottir TA, Selbaek G, Nyegaard M, Erikstrup C, Brodersen T, Saevarsdottir S, Olsson T, Nielsen KR, Haraldsson A, Bruun MT, Hansen TF, Steingrimsdottir T, Jacobsen RL, Lie RT, Djurovic S, Alfredsson L, Lopez de Lapuente Portilla A, Brunak S, Melsted P, Halldorsson BV, Saemundsdottir J, Magnusson OT, Padyukov L, Banasik K, Rafnar T, Askling J, Klareskog L, Pedersen OB, Masson G, Havdahl A, Nilsson B, Andreassen OA, Daly M, Ostrowski SR, Jonsdottir I, Stefansson H, Holm H, Helgason A, Thorsteinsdottir U, Stefansson K, Gudbjartsson DF. Deficit of homozygosity among 1.52 million individuals and genetic causes of recessive lethality. Nat Commun 2023; 14:3453. [PMID: 37301908 PMCID: PMC10257723 DOI: 10.1038/s41467-023-38951-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Genotypes causing pregnancy loss and perinatal mortality are depleted among living individuals and are therefore difficult to find. To explore genetic causes of recessive lethality, we searched for sequence variants with deficit of homozygosity among 1.52 million individuals from six European populations. In this study, we identified 25 genes harboring protein-altering sequence variants with a strong deficit of homozygosity (10% or less of predicted homozygotes). Sequence variants in 12 of the genes cause Mendelian disease under a recessive mode of inheritance, two under a dominant mode, but variants in the remaining 11 have not been reported to cause disease. Sequence variants with a strong deficit of homozygosity are over-represented among genes essential for growth of human cell lines and genes orthologous to mouse genes known to affect viability. The function of these genes gives insight into the genetics of intrauterine lethality. We also identified 1077 genes with homozygous predicted loss-of-function genotypes not previously described, bringing the total set of genes completely knocked out in humans to 4785.
Collapse
Affiliation(s)
| | | | | | - Gudny A Arnadottir
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | | | | | - Henriette Svarre Nielsen
- Deptartment of Obstetrics and Gynecology, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - David Westergaard
- Deptartment of Obstetrics and Gynecology, Copenhagen University Hospital, Hvidovre, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Methods and Analysis, Statistics Denmark, Copenhagen, Denmark
| | - Juha M Karjalainen
- Institute for Molecular Medicine, Finland, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | - Kristjan H S Moore
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland
- Department of Anthropology, University of Iceland, Reykjavik, Iceland
| | - Helga B Thordardottir
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | - Ida Elken Sonderby
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- NORMENT Centre, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - Maria Didriksen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Pernilla Stridh
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center of Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Bergen Center of Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Laufey Tryggvadottir
- Icelandic Cancer Registry, Icelandic Cancer Society, Reykjavik, Iceland
- Faculty of Medicine, BMC, Laeknagardur, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Oleksandr Frei
- NORMENT Centre, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Centre for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
| | | | - Ingrid Kockum
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center of Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Hjalgrim
- Department of Clinical Medicine, Faculty of Health, University of Copenhagen, Copenhagen, Denmark
- Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | | | - Geir Selbaek
- Norwegian National Centre of Ageing and Health, Vestfold Hospital Trust, Tonsberg, Norway
- Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Mette Nyegaard
- Deptartment of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Thorsten Brodersen
- Department of Clinical Immunology, Zealand University Hospital, Koge, Denmark
| | - Saedis Saevarsdottir
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center of Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Kaspar Rene Nielsen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Asgeir Haraldsson
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Children's Hospital Iceland, Landspitali University Hospital, Reykjavik, Iceland
| | - Mie Topholm Bruun
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | - Thomas Folkmann Hansen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Copenhagen University Hospital, Rigshospitalet, Glostrup, Denmark
| | - Thora Steingrimsdottir
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Rikke Louise Jacobsen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Rolv T Lie
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- NORMENT Centre, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - Lars Alfredsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Soren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pall Melsted
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Bjarni V Halldorsson
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland
- School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
| | | | | | - Leonid Padyukov
- Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Johan Askling
- Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Lars Klareskog
- Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Ole Birger Pedersen
- Department of Clinical Medicine, Faculty of Health, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Zealand University Hospital, Koge, Denmark
| | | | - Alexandra Havdahl
- Department of Mental Disorders, Norwegian Institute of Public Health, Oslo, Norway
- Nic Waals Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
- PROMENTA Research Center, Department of Psychology, University of Oslo, Oslo, Norway
| | - Bjorn Nilsson
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund, Sweden
| | - Ole A Andreassen
- NORMENT Centre, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Mark Daly
- Institute for Molecular Medicine, Finland, University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Deptartment of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ingileif Jonsdottir
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Hilma Holm
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland
| | - Agnar Helgason
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland
- Department of Anthropology, University of Iceland, Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland.
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland
- School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
| |
Collapse
|
16
|
Ayoubi S, Asadigandomani H, Bafrani MA, Shirkoohi A, Nasiri M, Sahraian MA, Eskandarieh S. The National Multiple Sclerosis Registry System of Iran (NMSRI): aspects and methodological dimensions. Mult Scler Relat Disord 2023; 72:104610. [PMID: 36931079 DOI: 10.1016/j.msard.2023.104610] [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: 01/10/2023] [Revised: 02/26/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023]
Abstract
INTRODUCTION Multiple Sclerosis (MS) as one of the most common causes of disability around the world requires a uniform standardized information registry system to help policy-makers systematically plan for care quality improvements. The aim of this study is to verify aspects and methodological scopes of MS registry system in Iran. METHODS The National MS Registry System in Iran (NMSRI) is a population-based registry system that systemically identifies and collects all MS patients' data in a specific geographical area. It supports 22 medical science universities and 13 MS societies in 18 provinces of Iran. The information items taken from each patient to collect the data set and data are gathered from all available sources including public and private hospitals, clinics, neurologists' offices, and all MS societies. They are recorded in District Health Information System 2 (DHIS2) software. DISCUSSION The NMSRI is a successful system of collecting MS patients' data. It can lead to positive results, such as updating patients' data to receive new treatments, fair allocation of treatment budgets, and providing researchers with novel ideas to carry out research projects.
Collapse
Affiliation(s)
- Saeideh Ayoubi
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Asadigandomani
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Melika Arab Bafrani
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Aryan Shirkoohi
- School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
| | - Mohamadreza Nasiri
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Sahraian
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sharareh Eskandarieh
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Englund S, Kierkegaard M, Burman J, Fink K, Fogdell-Hahn A, Gunnarsson M, Hillert J, Langer-Gould A, Lycke J, Nilsson P, Salzer J, Svenningsson A, Mellergård J, Olsson T, Longinetti E, Frisell T, Piehl F. Predictors of patient-reported fatigue symptom severity in a nationwide multiple sclerosis cohort. Mult Scler Relat Disord 2023; 70:104481. [PMID: 36603296 DOI: 10.1016/j.msard.2022.104481] [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: 04/05/2022] [Revised: 11/30/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Fatigue is a debilitating symptom of multiple sclerosis (MS), but its relation to sociodemographic and disease-related characteristics has not been investigated in larger studies. The objectives of this study were to evaluate predictors of self-reported fatigue in a Swedish nationwide register-based MS cohort. METHODS Using a repeated cross-sectional design, we included 2,165 persons with relapsing- remitting and secondary progressive MS with one or multiple Fatigue Scale for Motor and Cognitive Functions (FSMC) scores, which was modelled using multivariable linear regressions for multiple predictors. RESULTS Only associations to expanded disability status scale (EDSS) and Symbol Digit Modalities Test (SDMT) were considered clinically meaningful among MS-associated characteristics in our main model; compared to mild disability (EDSS 0-2.5), those with severe disability (EDSS ≥6) scored 17.6 (95% CI 13.1-22.2) FSMC points higher, while the difference was 10.7 (95% CI 8.0-13.4) points for the highest and lowest quartiles of SDMT. Differences between highest and lowest quartiles of health-related quality of life (HRQoL) instruments were even greater and considered clinically meaningful; EuroQoL Visual Analogue Scale (EQ-VAS) 31.9 (95% CI 29.9-33.8), Multiple Sclerosis Impact Scale (MSIS-29) psychological component 35.6 (95% CI 33.8-37.4) and MSIS-29 physical component 45.5 (95% CI 43.7-47.4). CONCLUSION Higher self-reported fatigue is associated with higher disability level and worse cognitive processing speed, while associations to other MS-associated characteristics including MS type, line of disease modifying therapy (DMT), MS duration, relapse and new cerebral lesions are weak. Furthermore, we found a strong correlation between high fatigue rating and lower ratings on health-related quality of life instruments.
Collapse
Affiliation(s)
- Simon Englund
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Marie Kierkegaard
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Joachim Burman
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Katharina Fink
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin Gunnarsson
- Department of Neurology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Annette Langer-Gould
- Clinical and Translational Neuroscience, Southern California Permanente Medical Group, Kaiser Permanente, Pasadena, United States
| | - Jan Lycke
- Department of Clinical Neuroscience, University of Gothenburg, Gothenburg, Sweden
| | - Petra Nilsson
- Department of Clinical Sciences, Division of Neurology, Lund University, Lund, Sweden
| | - Jonatan Salzer
- Department of Pharmacology and Clinical Neuroscience, Umea University, Umeå, Sweden
| | | | - Johan Mellergård
- Department of Biomedical and Clinical Sciences, Division of Neurobiology, Linköping University, Linköping, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Elisa Longinetti
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Frisell
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
18
|
Zondra Revendova K, Starvaggi Cucuzza C, Manouchehrinia A, Khademi M, Bar M, Leppert D, Sandberg E, Ouellette R, Granberg T, Piehl F. Demographic and disease-related factors impact on cerebrospinal fluid neurofilament light chain levels in multiple sclerosis. Brain Behav 2023; 13:e2873. [PMID: 36573731 PMCID: PMC9847611 DOI: 10.1002/brb3.2873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/07/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Neurofilament light (NfL) levels reflect inflammatory disease activity in multiple sclerosis (MS), but it is less clear if NfL also can serve as a biomarker for MS progression in treated patients without relapses and focal lesion accrual. In addition, it has not been well established if clinically effective treatment re-establishes an age and sex pattern for cerebrospinal fluid NfL (cNfL) as seen in controls, and to what degree levels are affected by disability level and magnetic resonance imaging (MRI) atrophy metrics. METHODS We included subjects for whom cNfL levels had been determined as per clinical routine or in clinical research, classified as healthy controls (HCs, n = 89), MS-free disease controls (DCs, n = 251), untreated MS patients (uMS; n = 296), relapse-free treated MS patients (tMS; n = 78), and ProTEct-MS clinical trial participants (pMS; n = 41). RESULTS Using linear regression, we found a positive association between cNfL and age, as well as lower concentrations among women, in all groups, except for uMS patients. In contrast, disability level in the entire MS cohort, or T1 and T2 lesion volumes, brain parenchymal fraction, thalamic fraction, and cortical thickness in the pMS trial cohort, did not correlate with cNfL concentrations. Furthermore, the cNfL levels in tMS and pMS groups did not differ. CONCLUSIONS In participants with MS lacking signs of inflammatory disease activity, disease modulatory therapy reinstates an age and sex cNfL pattern similar to that of control subjects. No significant association was found between cNfL levels and clinical worsening, disability level, or MRI metrics.
Collapse
Affiliation(s)
- Kamila Zondra Revendova
- Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Centre for Neurology, Academic Specialist Center, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Neurosciences, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Chiara Starvaggi Cucuzza
- Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Centre for Neurology, Academic Specialist Center, Karolinska University Hospital, Stockholm, Sweden
| | - Ali Manouchehrinia
- Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mohsen Khademi
- Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Michal Bar
- Department of Clinical Neurosciences, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - David Leppert
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Basel, Switzerland
| | - Elisabeth Sandberg
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Russell Ouellette
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Tobias Granberg
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Fredrik Piehl
- Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Centre for Neurology, Academic Specialist Center, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
19
|
Starvaggi Cucuzza C, Longinetti E, Ruffin N, Evertsson B, Kockum I, Jagodic M, Al Nimer F, Frisell T, Piehl F. Sustained Low Relapse Rate With Highly Variable B-Cell Repopulation Dynamics With Extended Rituximab Dosing Intervals in Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200056. [PMID: 36411076 PMCID: PMC9749930 DOI: 10.1212/nxi.0000000000200056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 09/16/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVES B cell-depleting therapies are highly effective in relapsing-remitting multiple sclerosis (RRMS) but are associated with increased infection risk and blunted humoral vaccination responses. Extension of dosing intervals may mitigate such negative effects, but its consequences on MS disease activity are yet to be ascertained. The objective of this study was to determine clinical and neuroradiologic disease activity, as well as B-cell repopulation dynamics, after implementation of extended rituximab dosing in RRMS. METHODS We conducted a prospective observational study in a specialized-care, single-center setting, including patients with RRMS participating in the COMBAT-MS and MultipleMS observational drug trials, who had received at least 2 courses of rituximab (median follow-up 4.2 years, range 0.1-8.9 years). Using Cox regression, hazard ratios (HRs) of clinical relapse and/or contrast-enhancing lesions on MRI were calculated in relation to time since last dose of rituximab. RESULTS A total of 3,904 dose intervals were accumulated in 718 patients and stratified into 4 intervals: <8, ≥8 to 12, ≥12 to 18, and ≥18 months. We identified 24 relapses of which 20 occurred within 8 months since previous infusion and 4 with intervals over 8 months. HRs for relapse when comparing ≥8 to 12, ≥12 to 18, and ≥18 months with <8 months since last dose were 0.28 (95% CI 0.04-2.10), 0.38 (95% CI 0.05-2.94), and 0.89 (95% CI 0.20-4.04), respectively, and thus nonsignificant. Neuroradiologic outcomes mirrored relapse rates. Dynamics of total B-cell reconstitution varied considerably, but median total B-cell counts reached lower level of normal after 12 months and median memory B-cell counts after 16 months. DISCUSSION In this prospective cohort of rituximab-treated patients with RRMS exposed to extended dosing intervals, we could not detect a relation between clinical or neuroradiologic disease activity and time since last infusion. Total B- and memory B-cell repopulation kinetics varied considerably. These findings, relevant for assessing risk-mitigation strategies with anti-CD20 therapies in RRMS, suggest that relapse risk remains low with extended infusion intervals. Further studies are needed to investigate the relation between B-cell repopulation dynamics and adverse event risks associated with B-cell depletion.
Collapse
Affiliation(s)
- Chiara Starvaggi Cucuzza
- From the Department of Clinical Neuroscience (C.S.C., E.L., N.R., B.E., I.K., M.J., F.A.N., F.P.), Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine (C.S.C., N.R., I.K., M.J., F.A.N., F.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (B.E., F.P.), Karolinska University Hospital, Stockholm, Sweden; Center for Neurology (C.S.C., I.K., M.J., F.A.N., F.P.), Academic Specialist Center, Stockholm, Sweden; and Clinical Epidemiology Division (T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Elisa Longinetti
- From the Department of Clinical Neuroscience (C.S.C., E.L., N.R., B.E., I.K., M.J., F.A.N., F.P.), Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine (C.S.C., N.R., I.K., M.J., F.A.N., F.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (B.E., F.P.), Karolinska University Hospital, Stockholm, Sweden; Center for Neurology (C.S.C., I.K., M.J., F.A.N., F.P.), Academic Specialist Center, Stockholm, Sweden; and Clinical Epidemiology Division (T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Nicolas Ruffin
- From the Department of Clinical Neuroscience (C.S.C., E.L., N.R., B.E., I.K., M.J., F.A.N., F.P.), Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine (C.S.C., N.R., I.K., M.J., F.A.N., F.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (B.E., F.P.), Karolinska University Hospital, Stockholm, Sweden; Center for Neurology (C.S.C., I.K., M.J., F.A.N., F.P.), Academic Specialist Center, Stockholm, Sweden; and Clinical Epidemiology Division (T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Björn Evertsson
- From the Department of Clinical Neuroscience (C.S.C., E.L., N.R., B.E., I.K., M.J., F.A.N., F.P.), Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine (C.S.C., N.R., I.K., M.J., F.A.N., F.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (B.E., F.P.), Karolinska University Hospital, Stockholm, Sweden; Center for Neurology (C.S.C., I.K., M.J., F.A.N., F.P.), Academic Specialist Center, Stockholm, Sweden; and Clinical Epidemiology Division (T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid Kockum
- From the Department of Clinical Neuroscience (C.S.C., E.L., N.R., B.E., I.K., M.J., F.A.N., F.P.), Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine (C.S.C., N.R., I.K., M.J., F.A.N., F.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (B.E., F.P.), Karolinska University Hospital, Stockholm, Sweden; Center for Neurology (C.S.C., I.K., M.J., F.A.N., F.P.), Academic Specialist Center, Stockholm, Sweden; and Clinical Epidemiology Division (T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Maja Jagodic
- From the Department of Clinical Neuroscience (C.S.C., E.L., N.R., B.E., I.K., M.J., F.A.N., F.P.), Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine (C.S.C., N.R., I.K., M.J., F.A.N., F.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (B.E., F.P.), Karolinska University Hospital, Stockholm, Sweden; Center for Neurology (C.S.C., I.K., M.J., F.A.N., F.P.), Academic Specialist Center, Stockholm, Sweden; and Clinical Epidemiology Division (T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Faiez Al Nimer
- From the Department of Clinical Neuroscience (C.S.C., E.L., N.R., B.E., I.K., M.J., F.A.N., F.P.), Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine (C.S.C., N.R., I.K., M.J., F.A.N., F.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (B.E., F.P.), Karolinska University Hospital, Stockholm, Sweden; Center for Neurology (C.S.C., I.K., M.J., F.A.N., F.P.), Academic Specialist Center, Stockholm, Sweden; and Clinical Epidemiology Division (T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Frisell
- From the Department of Clinical Neuroscience (C.S.C., E.L., N.R., B.E., I.K., M.J., F.A.N., F.P.), Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine (C.S.C., N.R., I.K., M.J., F.A.N., F.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (B.E., F.P.), Karolinska University Hospital, Stockholm, Sweden; Center for Neurology (C.S.C., I.K., M.J., F.A.N., F.P.), Academic Specialist Center, Stockholm, Sweden; and Clinical Epidemiology Division (T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- From the Department of Clinical Neuroscience (C.S.C., E.L., N.R., B.E., I.K., M.J., F.A.N., F.P.), Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine (C.S.C., N.R., I.K., M.J., F.A.N., F.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (B.E., F.P.), Karolinska University Hospital, Stockholm, Sweden; Center for Neurology (C.S.C., I.K., M.J., F.A.N., F.P.), Academic Specialist Center, Stockholm, Sweden; and Clinical Epidemiology Division (T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
20
|
Longinetti E, Bower H, McKay KA, Englund S, Burman J, Fink K, Fogdell-Hahn A, Gunnarsson M, Hillert J, Langer-Gould A, Lycke J, Nilsson P, Salzer J, Svenningsson A, Mellergård J, Olsson T, Piehl F, Frisell T. COVID-19 clinical outcomes and DMT of MS patients and population-based controls. Ann Clin Transl Neurol 2022; 9:1449-1458. [PMID: 35993445 PMCID: PMC9463950 DOI: 10.1002/acn3.51646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 07/20/2022] [Indexed: 11/10/2022] Open
Abstract
Objective To estimate risks for all‐cause mortality and for severe COVID‐19 in multiple sclerosis patients and across relapsing–remitting multiple sclerosis patients exposed to disease‐modifying therapies. Methods We conducted a Swedish nationwide population‐based multi‐register linkage cohort study and followed all multiple sclerosis patients (n = 17,692 in March 2020), individually age‐, sex‐, and region‐matched to five population‐based controls (n = 86,176 in March 2020) during March 2020–June 2021. We compared annual all‐cause mortality within and across cohorts, and assessed incidence rates and relative risks for hospitalization, intensive care admission, and death due to COVID‐19 in relation to disease‐modifying therapy use, using Cox regression. Results Absolute all‐cause mortality among multiple sclerosis patients was higher from March to December 2020 than in previous years, but relative risks versus the population‐based controls were similar to preceding years. Incidence rates of hospitalization, intensive care admission, and death due to COVID‐19 remained in line with those for all‐cause hospitalization, intensive care admission, and mortality. Among relapsing–remitting patients on rituximab, trends for differences in risk of hospitalization due to COVID‐19 remained in the demographics‐, socioeconomic status‐, comorbidity‐, and multiple sclerosis severity‐adjusted model. Interpretation Risks of severe COVID‐19‐related outcomes were increased among multiple sclerosis patients as a whole compared to population controls, but risk increases were also seen for non‐COVID‐19 hospitalization, intensive care admission, and mortality, and did not significantly differ during the pandemic compared to pre‐pandemic years. The risk conveyed by disease‐modifying therapies was smaller than previously assumed, likely as a consequence of the possibility to better control for confounders.
Collapse
Affiliation(s)
- Elisa Longinetti
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Hannah Bower
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Kyla A McKay
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Simon Englund
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Joachim Burman
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Katharina Fink
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Annette Langer-Gould
- Clinical and Translational Neuroscience, Southern California Permanente Medical Group, Kaiser Permanente, Los Angeles, USA
| | - Jan Lycke
- Department of Clinical Neuroscience, University of Gothenburg, Gothenburg, Sweden
| | - Petra Nilsson
- Department of Clinical Sciences, Division of Neurology, Lund University, Lund, Sweden
| | - Jonatan Salzer
- Department of Clinical Sciences, Neurosciences, Umeå University, Umeå, Sweden
| | - Anders Svenningsson
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Johan Mellergård
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Frisell
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
21
|
Brand JS, Smith KA, Piehl F, Olsson T, Montgomery S. Risk of serious infections in multiple sclerosis patients by disease course and disability status: Results from a Swedish register-based study. Brain Behav Immun Health 2022; 22:100470. [PMID: 35607517 PMCID: PMC9123212 DOI: 10.1016/j.bbih.2022.100470] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/26/2022] [Accepted: 05/06/2022] [Indexed: 12/31/2022] Open
Abstract
Background and objectives Serious infections are an emerging concern with increasing use of potent immunomodulation in multiple sclerosis (MS), but the extent to which MS disease features influence infectious susceptibility is poorly characterized. The objective of this study was to assess the associations of MS disease course and disability status with risk of serious infections. Methods A cohort of 8660 MS patients was individually matched on age, sex and region of residence with 86,600 people without MS from the general population using national registers in Sweden. The study period was from 1996 to 2012, with follow-up until December 31, 2014. The main outcomes were infection as the underlying or contributory cause of death or infection-related hospital admission identified in the Cause of Death and Patient registers. MS disease course (relapsing-remitting or progressive disease) and Expanded Disability Status Scale (EDSS) score (six and over or below six) were extracted from the MS Register Hazard ratios (HRs) for any serious infection were estimated using flexible parametric models. Results During a median follow-up of 9.6 years (interquartile range = 5.5-13.5 years), 1337 MS patients experienced a serious infection. Compared with individually matched people without MS, risk of serious infection was greater for progressive disease (HR = 3.80; 95% CI 3.52: 4.09) than relapsing-remitting disease (HR = 1.77; 95% CI: 1.62:1.93). A similar pattern of risk was seen for dichotomised EDSS score (HR = 4.26; 95% CI 3.87: 4.70 for EDSS 6.0-9.5 and HR = 1.30; 95% CI 1.1853: 1.43 for EDSS 0.0-5.5). Overall, associations with greater disability did not notably differ by immunomodulatory therapy use, but associations with lower disability were more pronounced in patients receiving these therapies. Conclusions Disease course or EDSS score (which may be more readily available than MS course in some patients) should be considered in individual management and monitoring of MS patients, including assessing benefit-risk of therapies that influence general immune function.
Collapse
Affiliation(s)
- Judith S. Brand
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Örebro University, Sweden
| | - Kelsi A. Smith
- Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Scott Montgomery
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Örebro University, Sweden
- Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
- Department of Epidemiology and Public Health, University College London, UK
| |
Collapse
|
22
|
Ezabadi SG, Sahraian MA, Maroufi H, Shahrbaf MA, Eskandarieh S. Global assessment of characteristics of multiple sclerosis registries; A systematic review. Mult Scler Relat Disord 2022; 63:103928. [DOI: 10.1016/j.msard.2022.103928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/14/2022] [Accepted: 05/27/2022] [Indexed: 11/30/2022]
|
23
|
Benkert P, Meier S, Schaedelin S, Manouchehrinia A, Yaldizli Ö, Maceski A, Oechtering J, Achtnichts L, Conen D, Derfuss T, Lalive PH, Mueller C, Müller S, Naegelin Y, Oksenberg JR, Pot C, Salmen A, Willemse E, Kockum I, Blennow K, Zetterberg H, Gobbi C, Kappos L, Wiendl H, Berger K, Sormani MP, Granziera C, Piehl F, Leppert D, Kuhle J, Aeschbacher S, Barakovic M, Buser A, Chan A, Disanto G, D'Souza M, Du Pasquier R, Findling O, Galbusera R, Hrusovsky K, Khalil M, Lorscheider J, Mathias A, Orleth A, Radue EW, Rahmanzadeh R, Sinnecker T, Subramaniam S, Vehoff J, Wellmann S, Wuerfel J, Zecca C. Serum neurofilament light chain for individual prognostication of disease activity in people with multiple sclerosis: a retrospective modelling and validation study. Lancet Neurol 2022; 21:246-257. [DOI: 10.1016/s1474-4422(22)00009-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 11/08/2021] [Accepted: 12/20/2021] [Indexed: 12/17/2022]
|
24
|
Brusini I, Platten M, Ouellette R, Piehl F, Wang C, Granberg T. Automatic deep learning multicontrast corpus callosum segmentation in multiple sclerosis. J Neuroimaging 2022; 32:459-470. [PMID: 35083815 PMCID: PMC9304261 DOI: 10.1111/jon.12972] [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: 11/22/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Corpus callosum (CC) atrophy is predictive of future disability in multiple sclerosis (MS). However, current segmentation methods are either labor- or computationally intensive. We therefore developed an automated deep learning-based CC segmentation tool and hypothesized that its output would correlate with disability. METHODS A cohort of 631 MS patients (449 females, baseline age 41 ± 11 years) with both 3-dimensional T1-weighted and T2-weighted fluid-attenuated inversion recovery (FLAIR) MRI was used for the development. Data from 204 patients were manually segmented to train convolutional neural networks in extracting the midsagittal intracranial and CC areas. Remaining data were used to compare segmentations with FreeSurfer and benchmark the outputs with regard to clinical correlations. A 1.5 and 3 Tesla reproducibility cohort of 9 MS patients evaluated the segmentation robustness. RESULTS The deep learning-based tool was accurate in selecting the appropriate slice for segmentation (98% accuracy within 3 mm of the manual ground truth) and segmenting the CC (Dice coefficient .88-.91) and intracranial areas (.97-.98). The accuracy was lower with higher atrophy. Reproducibility was excellent (intraclass correlation coefficient > .90) for T1-weighted scans and moderate-good for FLAIR (.74-.75). Segmentations were associated with baseline and future (average follow-up time 6-7 years) Expanded Disability Status Scale (ρ = -.13 to -.24) and Symbol Digit Modalities Test (r = .18-.29) scores. CONCLUSIONS We present a fully automatic deep learning-based CC segmentation tool optimized to modern imaging in MS with clinical correlations on par with computationally expensive alternatives.
Collapse
Affiliation(s)
- Irene Brusini
- School of Chemistry, Biotechnology, and Health, Royal Institute of Technology, Stockholm, Sweden.,Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Michael Platten
- School of Chemistry, Biotechnology, and Health, Royal Institute of Technology, Stockholm, Sweden.,Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Russell Ouellette
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.,Center for Neurology, Academic Specialist Center, Stockholm Health Services, Stockholm, Sweden
| | - Chunliang Wang
- School of Chemistry, Biotechnology, and Health, Royal Institute of Technology, Stockholm, Sweden
| | - Tobias Granberg
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
McKay KA, Bedri SK, Manouchehrinia A, Stawiarz L, Olsson T, Hillert J, Fink K. Reduction in cognitive processing speed surrounding multiple sclerosis relapse. Ann Neurol 2022; 91:417-423. [PMID: 34984719 PMCID: PMC9303402 DOI: 10.1002/ana.26301] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/02/2022] [Accepted: 01/03/2022] [Indexed: 11/21/2022]
Abstract
Objective The purpose of this study was to explore the longitudinal relationship between multiple sclerosis (MS) relapses and information processing efficiency among persons with relapsing–remitting MS. Methods We conducted a Swedish nationwide cohort study of persons with incident relapsing–remitting MS (2001–2019). Relapse information and symbol digit modalities test (SDMT) scores were obtained from the Swedish MS Registry. Follow‐up was categorized into 2 periods based on relapse status: “relapse” (90 days pre‐relapse to 730 days post‐relapse, subdivided into 10 periods) and “remission.” Linear mixed models compared SDMT scores during the relapse periods to SDMT scores recorded during remission (reference) with results reported as β‐coefficients and 95% confidence intervals (CIs), adjusted for age, sex, SDMT type (written vs oral), time‐varying, disease‐modifying therapy exposure and sequence of SDMT. Results Over a mean (SD) follow‐up of 10.7 (4.3) years, 31,529 distinct SDMTs were recorded among 3,877 persons with MS. There was a significant decline in information processing efficiency that lasted from 30 days pre‐relapse up to 550 days post‐relapse, with the largest decline occurring 0 to 30 days post‐relapse (β‐coefficient: −4.00 (95% CI = −4.61 to −3.39), relative to the period of remission. Interpretation We found evidence of cognitive change up to 1 month prior to relapse onset. The reduction in SDMT lasted 1.5 years and was clinically significant up to 3 months post‐relapse. These results suggest that the effects of a relapse on cognition are longer than previously thought and highlight the importance of reducing relapse rates as a potential means of preserving cognitive function. ANN NEUROL 2022;91:417–423
Collapse
Affiliation(s)
- Kyla A McKay
- Department of Clinical Neuroscience Neuro Division, Karolinska Institutet Stockholm Sweden
- Centre for Molecular Medicine Karolinska University Hospital Stockholm Sweden
| | - Sahl K Bedri
- Department of Clinical Neuroscience Neuro Division, Karolinska Institutet Stockholm Sweden
- Centre for Molecular Medicine Karolinska University Hospital Stockholm Sweden
| | - Ali Manouchehrinia
- Department of Clinical Neuroscience Neuro Division, Karolinska Institutet Stockholm Sweden
- Centre for Molecular Medicine Karolinska University Hospital Stockholm Sweden
| | - Leszek Stawiarz
- Department of Clinical Neuroscience Neuro Division, Karolinska Institutet Stockholm Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience Neuro Division, Karolinska Institutet Stockholm Sweden
- Centre for Molecular Medicine Karolinska University Hospital Stockholm Sweden
| | - Jan Hillert
- Department of Clinical Neuroscience Neuro Division, Karolinska Institutet Stockholm Sweden
| | - Katharina Fink
- Department of Clinical Neuroscience Neuro Division, Karolinska Institutet Stockholm Sweden
- Centrum for Neurology, Academical Specialist Center Stockholm Sweden
| |
Collapse
|
26
|
Teljas C, Boström I, Marrie RA, Landtblom A, Manouchehrinia A, Hillert J, McKay KA. Validating the diagnosis of multiple sclerosis using Swedish administrative data in Värmland County. Acta Neurol Scand 2021; 144:680-686. [PMID: 34357597 DOI: 10.1111/ane.13514] [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: 04/19/2021] [Revised: 07/09/2021] [Accepted: 07/25/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Multiple sclerosis (MS) is a chronic neurodegenerative disease of the central nervous system. Identifying MS at the population level is important for disease surveillance and allocation of resources. The Swedish National Patient Registry (NPR) has been used to study the epidemiology of MS, but the accuracy of this resource is not known. We aimed to validate a definition of MS using the Swedish NPR in Värmland County using a longitudinal cohort design. MATERIALS AND METHODS Data were extracted from the NPR, the Total Population Register, the Swedish MS Register, and medical records for the years 2001-2013. Fifteen algorithms of hospitalizations and clinic visits for MS were developed and compared with findings in medical records, which acted as the "gold standard" definition. Sensitivity, specificity, and positive and negative predictive values (PPV, NPV) were estimated. RESULTS Of 805 eligible persons identified in the NPR, 763 had MS (94.8%) according to medical records. Of these, 544 (71.3%) were also registered in the SMSreg. The case definition that had a well-balanced sensitivity and specificity required three or more clinic or hospital visits for MS (sensitivity of 85.3% (95% CI: 82.6-87.8) and specificity of 81.0% (95%CI: 65.9-91.4). CONCLUSIONS Multiple case definitions with high sensitivity and moderate specificity were found, suggesting that the NPR can be used to accurately identify persons with MS.
Collapse
Affiliation(s)
- Cecilia Teljas
- Department of Clinical Neuroscience Karolinska Institutet Stockholm Sweden
| | - Inger Boström
- Department of Biomedical and Clinical Sciences Faculty of Medicine and Health Sciences Linköping University Sweden
| | - Ruth Ann Marrie
- Rady Faculty of Health Sciences Max Rady College of MedicineUniversity of Manitoba Winnipeg MB Canada
| | - Anne‐Marie Landtblom
- Department of Biomedical and Clinical Sciences Faculty of Medicine and Health Sciences Linköping University Sweden
- Department of Neuroscience Uppsala University Uppsala Sweden
| | - Ali Manouchehrinia
- Department of Clinical Neuroscience Karolinska Institutet Stockholm Sweden
- Center for Molecular Medicine Karolinska University Hospital Stockholm Sweden
| | - Jan Hillert
- Department of Clinical Neuroscience Karolinska Institutet Stockholm Sweden
| | - Kyla A. McKay
- Department of Clinical Neuroscience Karolinska Institutet Stockholm Sweden
- Center for Molecular Medicine Karolinska University Hospital Stockholm Sweden
| |
Collapse
|
27
|
McKay KA, Piehl F, Englund S, He A, Langer-Gould A, Hillert J, Frisell T. Rituximab Infusion Timing, Cumulative Dose, and Hospitalization for COVID-19 in Persons With Multiple Sclerosis in Sweden. JAMA Netw Open 2021; 4:e2136697. [PMID: 34851401 PMCID: PMC8637249 DOI: 10.1001/jamanetworkopen.2021.36697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/05/2021] [Indexed: 11/23/2022] Open
Affiliation(s)
- Kyla A. McKay
- Neuro Division, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Centre for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Fredrik Piehl
- Neuro Division, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- The Karolinska University Hospital and Academic Specialist Centre, Stockholm Health Services, Stockholm, Sweden
| | - Simon Englund
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Anna He
- Neuro Division, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Centre for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Annette Langer-Gould
- Department of Neurology, Los Angeles Medical Center, Southern California Permanente Medical Group, Los Angeles
| | - Jan Hillert
- Neuro Division, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Frisell
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
28
|
Abstract
BACKGROUND Multiple sclerosis (MS) is the most common neurological cause of disability in young adults. Off-label rituximab for MS is used in most countries surveyed by the International Federation of MS, including high-income countries where on-label disease-modifying treatments (DMTs) are available. OBJECTIVES: To assess beneficial and adverse effects of rituximab as 'first choice' and as 'switching' for adults with MS. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, CINAHL, and trial registers for completed and ongoing studies on 31 January 2021. SELECTION CRITERIA We included randomised controlled trials (RCTs) and controlled non-randomised studies of interventions (NRSIs) comparing rituximab with placebo or another DMT for adults with MS. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. We used the Cochrane Collaboration's tool for assessing risk of bias. We rated the certainty of evidence using GRADE for: disability worsening, relapse, serious adverse events (SAEs), health-related quality of life (HRQoL), common infections, cancer, and mortality. We conducted separate analyses for rituximab as 'first choice' or as 'switching', relapsing or progressive MS, comparison versus placebo or another DMT, and RCTs or NRSIs. MAIN RESULTS We included 15 studies (5 RCTs, 10 NRSIs) with 16,429 participants of whom 13,143 were relapsing MS and 3286 progressive MS. The studies were one to two years long and compared rituximab as 'first choice' with placebo (1 RCT) or other DMTs (1 NRSI), rituximab as 'switching' against placebo (2 RCTs) or other DMTs (2 RCTs, 9 NRSIs). The studies were conducted worldwide; most originated from high-income countries, six from the Swedish MS register. Pharmaceutical companies funded two studies. We identified 14 ongoing studies. Rituximab as 'first choice' for relapsing MS Rituximab versus placebo: no studies met eligibility criteria for this comparison. Rituximab versus other DMTs: one NRSI compared rituximab with interferon beta or glatiramer acetate, dimethyl fumarate, natalizumab, or fingolimod in active relapsing MS at 24 months' follow-up. Rituximab likely results in a large reduction in relapses compared with interferon beta or glatiramer acetate (hazard ratio (HR) 0.14, 95% confidence interval (CI) 0.05 to 0.39; 335 participants; moderate-certainty evidence). Rituximab may reduce relapses compared with dimethyl fumarate (HR 0.29, 95% CI 0.08 to 1.00; 206 participants; low-certainty evidence) and natalizumab (HR 0.24, 95% CI 0.06 to 1.00; 170 participants; low-certainty evidence). It may make little or no difference on relapse compared with fingolimod (HR 0.26, 95% CI 0.04 to 1.69; 137 participants; very low-certainty evidence). The study reported no deaths over 24 months. The study did not measure disability worsening, SAEs, HRQoL, and common infections. Rituximab as 'first choice' for progressive MS One RCT compared rituximab with placebo in primary progressive MS at 24 months' follow-up. Rituximab likely results in little to no difference in the number of participants who have disability worsening compared with placebo (odds ratio (OR) 0.71, 95% CI 0.45 to 1.11; 439 participants; moderate-certainty evidence). Rituximab may result in little to no difference in recurrence of relapses (OR 0.60, 95% CI 0.18 to 1.99; 439 participants; low-certainty evidence), SAEs (OR 1.25, 95% CI 0.71 to 2.20; 439 participants; low-certainty evidence), common infections (OR 1.14, 95% CI 0.75 to 1.73; 439 participants; low-certainty evidence), cancer (OR 0.50, 95% CI 0.07 to 3.59; 439 participants; low-certainty evidence), and mortality (OR 0.25, 95% CI 0.02 to 2.77; 439 participants; low-certainty evidence). The study did not measure HRQoL. Rituximab versus other DMTs: no studies met eligibility criteria for this comparison. Rituximab as 'switching' for relapsing MS One RCT compared rituximab with placebo in relapsing MS at 12 months' follow-up. Rituximab may decrease recurrence of relapses compared with placebo (OR 0.38, 95% CI 0.16 to 0.93; 104 participants; low-certainty evidence). The data did not confirm or exclude a beneficial or detrimental effect of rituximab relative to placebo on SAEs (OR 0.90, 95% CI 0.28 to 2.92; 104 participants; very low-certainty evidence), common infections (OR 0.91, 95% CI 0.37 to 2.24; 104 participants; very low-certainty evidence), cancer (OR 1.55, 95% CI 0.06 to 39.15; 104 participants; very low-certainty evidence), and mortality (OR 1.55, 95% CI 0.06 to 39.15; 104 participants; very low-certainty evidence). The study did not measure disability worsening and HRQoL. Five NRSIs compared rituximab with other DMTs in relapsing MS at 24 months' follow-up. The data did not confirm or exclude a beneficial or detrimental effect of rituximab relative to interferon beta or glatiramer acetate on disability worsening (HR 0.86, 95% CI 0.52 to 1.42; 1 NRSI, 853 participants; very low-certainty evidence). Rituximab likely results in a large reduction in relapses compared with interferon beta or glatiramer acetate (HR 0.18, 95% CI 0.07 to 0.49; 1 NRSI, 1383 participants; moderate-certainty evidence); and fingolimod (HR 0.08, 95% CI 0.02 to 0.32; 1 NRSI, 256 participants; moderate-certainty evidence). The data did not confirm or exclude a beneficial or detrimental effect of rituximab relative to natalizumab on relapses (HR 1.0, 95% CI 0.2 to 5.0; 1 NRSI, 153 participants; very low-certainty evidence). Rituximab likely increases slightly common infections compared with interferon beta or glatiramer acetate (OR 1.71, 95% CI 1.11 to 2.62; 1 NRSI, 5477 participants; moderate-certainty evidence); and compared with natalizumab (OR 1.58, 95% CI 1.08 to 2.32; 2 NRSIs, 5001 participants; moderate-certainty evidence). Rituximab may increase slightly common infections compared with fingolimod (OR 1.26, 95% CI 0.90 to 1.77; 3 NRSIs, 5187 participants; low-certainty evidence). It may make little or no difference compared with ocrelizumab (OR 0.02, 95% CI 0.00 to 0.40; 1 NRSI, 472 participants; very low-certainty evidence). The data did not confirm or exclude a beneficial or detrimental effect of rituximab on mortality compared with fingolimod (OR 5.59, 95% CI 0.22 to 139.89; 1 NRSI, 136 participants; very low-certainty evidence) and natalizumab (OR 6.66, 95% CI 0.27 to 166.58; 1 NRSI, 153 participants; very low-certainty evidence). The included studies did not measure SAEs, HRQoL, and cancer. AUTHORS' CONCLUSIONS For preventing relapses in relapsing MS, rituximab as 'first choice' and as 'switching' may compare favourably with a wide range of approved DMTs. A protective effect of rituximab against disability worsening is uncertain. There is limited information to determine the effect of rituximab for progressive MS. The evidence is uncertain about the effect of rituximab on SAEs. They are relatively rare in people with MS, thus difficult to study, and they were not well reported in studies. There is an increased risk of common infections with rituximab, but absolute risk is small. Rituximab is widely used as off-label treatment in people with MS; however, randomised evidence is weak. In the absence of randomised evidence, remaining uncertainties on beneficial and adverse effects of rituximab for MS might be clarified by making real-world data available.
Collapse
Affiliation(s)
- Graziella Filippini
- Scientific Director's Office, Carlo Besta Foundation and Neurological Institute, Milan, Italy
| | - Jera Kruja
- Neurology, UHC Mother Theresa, University of Medicine, Tirana, Albania
| | - Cinzia Del Giovane
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
- Population Health Laboratory (#PopHealthLab), University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
29
|
McKay KA, Wickström R, Hillert J, Karrenbauer VD. Cerebrospinal fluid markers in incident pediatric-onset multiple sclerosis: a nationwide study. Sci Rep 2021; 11:18528. [PMID: 34535701 PMCID: PMC8448868 DOI: 10.1038/s41598-021-97543-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 08/26/2021] [Indexed: 01/12/2023] Open
Abstract
To investigate whether cerebrospinal fluid (CSF) markers differ between pediatric-onset multiple sclerosis (PoMS, onset < 18 years) and adult-onset (AoMS), and whether these markers are associated with clinical outcomes among PoMS. Prospective nationwide registry study of incident MS, including persons with a CSF sample < 3 years post-MS onset. We compared CSF oligoclonal band (OCB) status, immunoglobulin G (IgG) index levels, and mononuclear cell count between PoMS and AoMS. Within the PoMS cohort we analyzed the association between CSF markers, relapse rate and Expanded Disability Status Scale (EDSS) score, using negative binomial regression and generalized estimating equations, respectively. The cohort consisted of 130 PoMS and 3228 AoMS cases. The PoMS group had higher odds of OCB-positivity (odds ratio: 2.70; 95% CI 1.21–7.67). None of the CSF markers were associated with relapse rate in the PoMS cohort; however, OCB-positivity was associated with higher EDSS scores. This study suggested that PoMS more commonly display CSF evidence for intrathecal IgG production than AoMS. Further, we found evidence of a relationship between OCB-positivity and subsequent disability, suggesting that they could play a role in the prognostication of MS in children.
Collapse
Affiliation(s)
- Kyla A McKay
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden. .,Center for Molecular Medicine (CMM) L8:05, Karolinska University Hospital, 171 76, Stockholm, Sweden.
| | - Ronny Wickström
- Neuropediatric Unit, Astrid Lindgren's Childrens's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Virginija Danylaite Karrenbauer
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Theme Neuro, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
30
|
Spelman T, Magyari M, Piehl F, Svenningsson A, Rasmussen PV, Kant M, Sellebjerg F, Joensen H, Hillert J, Lycke J. Treatment Escalation vs Immediate Initiation of Highly Effective Treatment for Patients With Relapsing-Remitting Multiple Sclerosis: Data From 2 Different National Strategies. JAMA Neurol 2021; 78:1197-1204. [PMID: 34398221 DOI: 10.1001/jamaneurol.2021.2738] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Importance Treatment strategies for relapsing-remitting multiple sclerosis (RRMS) vary markedly between Denmark and Sweden. The difference in the association of these national strategies with clinical outcomes is unknown. Objective To investigate the association of national differences in disease-modifying treatment (DMT) strategies for RRMS with disability outcomes. Design, Setting, and Participants This cohort study used data on 4861 patients from the Danish and Swedish national multiple sclerosis (MS) registries from the date of index DMT initiation (between January 1, 2013, and December 31, 2016) until the last recorded visit at time of data extraction (October 2, 2019). Exposures All MS-specific DMTs initiated during the observation period were included in the analysis. Main Outcomes and Measures The primary study outcome was time to 24-week confirmed disability worsening. Secondary outcomes were 24-week confirmed disability improvement, milestone Expanded Disability Status Scale scores of 3 and 4, annualized relapse rate, time to first relapse, and treatment switching. Data were analyzed using inverse probability of treatment weighting-based models using a propensity score to weight and correct the comparison for the imbalance of confounders observed at baseline between the 2 countries. Results A total of 2700 patients from the Swedish MS registry (1867 women [69.2%]; mean [SD] age, 36.1 [9.5] years) and 2161 patients from the Danish MS registry (1472 women [68.1%]; mean [SD] age, 37.3 [9.4 years]) started a first DMT between 2013 and 2016, were included in the analysis, and were observed for a mean (SD) of 4.1 (1.5) years. A total of 1994 Danish patients (92.3%) initiated a low to moderately effective DMT (teriflunomide, 907 [42.0%]) and 165 (7.6%) initiated a highly effective DMT, whereas a total of 1769 Swedish patients (65.5%) initiated a low to moderately effective DMT (teriflunomide, 64 [2.4%]) and 931 (34.5%) initiated a highly effective DMT. The Swedish treatment strategy was associated with a 29% reduction in the rate of postbaseline 24-week confirmed disability worsening relative to the Danish treatment strategy (hazard ratio, 0.71; 95% CI, 0.57-0.90; P = .004). The Swedish treatment strategy was also associated with a 24% reduction in the rate of reaching an expanded disability status scale score of 3 (hazard ratio, 0.76; 95% CI, 0.60-0.97; P = .03) and a 25% reduction in the rate of reaching an expanded disability status scale score of 4 (hazard ratio, 0.75; 95% CI, 0.61-0.96; P = .01) relative to Danish patients. Conclusions and Relevance The findings of this study suggest that there is an association between differences in treatment strategies for RRMS and disability outcomes at a national level. Escalation of treatment efficacy was inferior to using more efficacious DMT as initial treatment.
Collapse
Affiliation(s)
- Tim Spelman
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Melinda Magyari
- The Danish Multiple Sclerosis Registry, Department of Neurology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.,Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Anders Svenningsson
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | | | - Matthias Kant
- Multiple Sclerosis Clinic Southern Denmark, Department of Brain and Nerve Diseases, University of Southern Denmark, Odense, Denmark.,Department of Brain & Nerve Diseases, Hospital of Southern Jutland, Odense, Denmark
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Hanna Joensen
- The Danish Multiple Sclerosis Registry, Department of Neurology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Jan Lycke
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Gothenburg, Sweden.,Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
31
|
Longinetti E, Frisell T, Englund S, Reutfors J, Fang F, Piehl F. Risk of depression in multiple sclerosis across disease-modifying therapies. Mult Scler 2021; 28:632-641. [PMID: 34264143 PMCID: PMC8961249 DOI: 10.1177/13524585211031128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background: Depression and use of antidepressants are more common among patients with
multiple sclerosis (MS) compared to the general population, but the relation
of psychiatric comorbidity to use of different disease-modifying therapies
(DMTs) is less clear. Objective: To determine whether risk of incident depression or antidepressant use
differed across DMTs, and to assess whether depression and antidepressants
affected risk of DMT discontinuation and MS relapses. Methods: We prospectively followed for 8 years a register-based nationwide cohort of
3803 relapsing-remitting MS patients. Results: Patients on rituximab had a lower risk of being diagnosed with depression or
initiating antidepressants compared with the reference group treated with
interferons (hazard ratio (HR) = 0.72, 95% confidence interval (CI) =
0.54–0.96). Patients diagnosed with depression discontinued interferon
treatment to a higher extent than patients without depression (HR = 1.51;
95% CI = 1.15–1.98), as did patients on fingolimod initiating an
antidepressant compared to patients who did not initiate an antidepressant
(HR = 1.47; 95% CI = 1.04–2.08). Conclusions: Our results indicate that the choice of DMT is associated with subsequent
risk of depression in MS, but further studies are needed to establish
whether there is a causal link. Overall, depression and use of
antidepressants displayed limited associations with DMT discontinuation and
MS relapse.
Collapse
Affiliation(s)
- Elisa Longinetti
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Frisell
- Department of Medicine Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
| | - Simon Englund
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Johan Reutfors
- Department of Medicine Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
| | - Fang Fang
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
32
|
Hillert J, Tsai JA, Nouhi M, Glaser A, Spelman T. A comparative study of teriflunomide and dimethyl fumarate within the Swedish MS Registry. Mult Scler 2021; 28:237-246. [PMID: 34080926 PMCID: PMC8795225 DOI: 10.1177/13524585211019649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background: Teriflunomide and dimethyl fumarate (DMF) are first-line disease-modifying treatments for multiple sclerosis with similar labels that are used in comparable populations. Objectives: The objective of this study was to compare the effectiveness and persistence of teriflunomide and DMF in a Swedish real-world setting. Methods: All relapsing-remitting multiple sclerosis (RRMS) patients in the Swedish MS registry initiating teriflunomide or DMF were included in the analysis. The primary endpoint was treatment persistence. Propensity score matching was used to adjust comparisons for baseline confounders. Results: A total of 353 teriflunomide patients were successfully matched to 353 DMF. There was no difference in the rate of overall treatment discontinuation by treatment group across the entire observation period (hazard ratio (HR) = 1.12; 95% confidence interval (CI) = 0.91–1.39; p = 0.277; reference = teriflunomide). Annualised relapse rate (ARR) was comparable (p = 0.237) between DMF (0.07; 95% CI = 0.05–0.10) and teriflunomide (0.09; 95% CI = 0.07–0.12). There was no difference in time to first on-treatment relapse (HR = 0.78; 95% CI = 0.50–1.21), disability progression (HR = 0.55; 95% CI = 0.27–1.12) or confirmed improvement (HR = 1.17; 95% CI = 0.57–2.36). Conclusion: This population-based real-world study reports similarities in treatment persistence, clinical effectiveness and quality of life outcomes between teriflunomide and dimethyl fumarate.
Collapse
Affiliation(s)
- Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | | | | - Anna Glaser
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Tim Spelman
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
33
|
Stenberg E, Forsberg L, Hedström A, Hillert J, Näslund E. Bariatric and metabolic surgery in patients with morbid obesity and multiple sclerosis - a nationwide, matched cohort study. Surg Obes Relat Dis 2021; 17:1108-1114. [PMID: 33753006 DOI: 10.1016/j.soard.2021.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Despite an association between obesity and multiple sclerosis (MS), very little is known regarding the safety and efficacy outcomes for patients with MS and severe obesity undergoing metabolic surgery. OBJECTIVES The aim of the present study was to evaluate early complications and efficacy outcomes of metabolic surgery in patients with severe obesity and MS. SETTING Nationwide, Sweden. METHODS In this, matched cohort study, 196 patients with an MS diagnosis in the Swedish MS register who were undergoing metabolic surgery (gastric bypass or sleeve gastrectomy) with a registration in the Scandinavian Obesity Surgery Registry (SOReg) were matched 1:10 with a control group without MS diagnosis from the SOReg. A 2-stage matching procedure was used (exact match by surgical method, followed by propensity Score matching, including age, sex, preoperative BMI, surgical center, surgical access, year of surgery, hypertension, diabetes, sleep apnea, and dyslipidemia). RESULTS Weight loss at 2 years after surgery was similar for patients with MS and controls (total weight loss 31.6 ± 9.1 versus 31.8 ± 9.2, P = .735). No significant differences were seen in either the overall postoperative complication rate (7.9% versus 7.2%, P = .778), or serious postoperative complications (3.7% versus 2.8%, P = .430). All aspects of health-related quality of life (HRQoL) improved in both groups but less so for the physical aspects of HRQoL in patients with MS. CONCLUSION Metabolic surgery is a safe and efficient treatment for severe obesity in patients with MS, and it leads to subsequent improvements in HRQoL. Further studies addressing the effects of metabolic surgery on MS-related symptoms are needed.
Collapse
Affiliation(s)
- Erik Stenberg
- Department of Surgery, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
| | - Lars Forsberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Hedström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Erik Näslund
- Division of Surgery, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Alping P, Burman J, Lycke J, Frisell T, Piehl F. Safety of Alemtuzumab and Autologous Hematopoietic Stem Cell Transplantation Compared to Noninduction Therapies for Multiple Sclerosis. Neurology 2021; 96:e1574-e1584. [PMID: 33514645 PMCID: PMC8032381 DOI: 10.1212/wnl.0000000000011545] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/07/2020] [Indexed: 11/15/2022] Open
Abstract
Objective To assess safety outcomes for the induction therapies alemtuzumab and autologous hematopoietic stem cell transplantation (AHSCT) compared to noninduction disease-modifying therapies. Methods We performed a population-based cohort study linking the Swedish Multiple Sclerosis Register to national health care registers. Alemtuzumab, AHSCT, and a matched reference group of noninduction therapies (natalizumab, dimethyl fumarate, rituximab, fingolimod) were included if started between 2008 and 2017. Main outcomes were death, thyroid disease, nonthyroid autoimmune disease, and infection. Results We identified 132 alemtuzumab-treated and 139 AHSCT-treated (68% high-dose cyclophosphamide and anti-thymocyte globulin [ATG], 32% BCNU, etoposide, cytosine-arabinoside, and melphalan/ATG) patients, together with 2,486 matched patients treated with noninduction therapies. Four patients in the alemtuzumab group died (incidence rate [IR] per 1,000 person-years 8.6, 95% confidence interval [CI] 2.3–22.0) compared to 1 patient in the AHSCT group (IR 1.7, 95% CI 0.0–9.6), and the mortality rate in the reference group was 0.7 (95% CI 0.3–1.3). Thyroid disease was most frequent in the alemtuzumab group (IR 109, 95% CI 75–154) but also occurred more often for AHSCT (IR 34, 95% CI 18–56) compared to the reference (IR 5.3 95% CI 3.9–7.1). The incidence of nonthyroid autoimmune disease was similar in all groups. IR for infection diagnosed ≥6 months from therapy initiation was 53 (95% CI 30–87) for alemtuzumab, 108 (95% CI 75–150) for AHSCT, and 51 (95% CI 46–57) for the reference. Conclusion We confirmed a high incidence of thyroid disease in alemtuzumab- and, to a smaller extent, AHSCT-treated patients and found a higher incidence of infection for AHSCT compared to both alemtuzumab and noninduction therapies. The incidence of nonthyroid autoimmune disease was low for both therapies. Classification of Evidence This study provides Class III evidence of an increased risk of thyroid disease with alemtuzumab and an increased risk of infection with AHSCT treatment.
Collapse
Affiliation(s)
- Peter Alping
- From the Department of Clinical Neuroscience (P.A., F.P.) and Clinical Epidemiology Division (P.A., T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm; Department of Neuroscience (J.B.), Uppsala University; Department of Clinical Neuroscience (J.L.), Institute of Neuroscience and Physiology, University of Gothenburg; and Academic Specialist Centre (F.P.), Stockholm Health Services, Sweden.
| | - Joachim Burman
- From the Department of Clinical Neuroscience (P.A., F.P.) and Clinical Epidemiology Division (P.A., T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm; Department of Neuroscience (J.B.), Uppsala University; Department of Clinical Neuroscience (J.L.), Institute of Neuroscience and Physiology, University of Gothenburg; and Academic Specialist Centre (F.P.), Stockholm Health Services, Sweden
| | - Jan Lycke
- From the Department of Clinical Neuroscience (P.A., F.P.) and Clinical Epidemiology Division (P.A., T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm; Department of Neuroscience (J.B.), Uppsala University; Department of Clinical Neuroscience (J.L.), Institute of Neuroscience and Physiology, University of Gothenburg; and Academic Specialist Centre (F.P.), Stockholm Health Services, Sweden
| | - Thomas Frisell
- From the Department of Clinical Neuroscience (P.A., F.P.) and Clinical Epidemiology Division (P.A., T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm; Department of Neuroscience (J.B.), Uppsala University; Department of Clinical Neuroscience (J.L.), Institute of Neuroscience and Physiology, University of Gothenburg; and Academic Specialist Centre (F.P.), Stockholm Health Services, Sweden
| | - Fredrik Piehl
- From the Department of Clinical Neuroscience (P.A., F.P.) and Clinical Epidemiology Division (P.A., T.F.), Department of Medicine Solna, Karolinska Institutet, Stockholm; Department of Neuroscience (J.B.), Uppsala University; Department of Clinical Neuroscience (J.L.), Institute of Neuroscience and Physiology, University of Gothenburg; and Academic Specialist Centre (F.P.), Stockholm Health Services, Sweden
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW To outline recent applications of e-health data and digital tools for improving the care and management of healthcare for people with multiple sclerosis. RECENT FINDINGS The digitization of most clinical data, along with developments in communication technologies, miniaturization of sensors and computational advances are enabling aggregation and clinically meaningful analyses of real-world data from patient registries, digital patient-reported outcomes and electronic health records (EHR). These data are allowing more confident descriptions of prognoses for multiple sclerosis patients and the long-term relative benefits and safety of disease-modifying treatments (DMT). Registries allow detailed, multiple sclerosis-specific data to be shared between clinicians more easily, provide data needed to improve the impact of DMT and, with EHR, characterize clinically relevant interactions between multiple sclerosis and other diseases. Wearable sensors provide continuous, long-term measures of performance dynamics in relevant ecological settings. In conjunction with telemedicine and online apps, they promise a major expansion of the scope for patients to manage aspects of their own care. Advances in disease understanding, decision support and self-management using these Big Data are being accelerated by machine learning and artificial intelligence. SUMMARY Both health professionals and patients can employ e-health approaches and tools for development of a more patient-centred learning health system.
Collapse
|
36
|
Murley C, Tinghög P, Karampampa K, Hillert J, Alexanderson K, Friberg E. Types of working-life sequences among people recently diagnosed with multiple sclerosis in Sweden: a nationwide register-based cohort study. BMJ Open 2020; 10:e039228. [PMID: 33376161 PMCID: PMC7778766 DOI: 10.1136/bmjopen-2020-039228] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES To explore sequences of annual states of activity and sickness absence (SA) or disability pension (DP) (SA/DP) among working-aged people with multiple sclerosis (PwMS) as well as characteristics associated with the identified types of working-life sequences. DESIGN Nationwide Swedish register-based cohort study from 1 year prior to 5 years after the year of multiple sclerosis (MS) diagnosis. SETTING Sweden. PARTICIPANTS PwMS diagnosed in 2008-2011 when aged 20-55 (n=2652, 69.9% women). PRIMARY AND SECONDARY OUTCOME MEASURES Individual-level sequences spanning 7 years were constructed with annual states regarding activity (income from paid work, student allowances, parental leave or unemployment compensation) and/or SA/DP. Types of working-life sequences were identified among the individuals' sequences using hierarchical cluster analysis with optimal matching dissimilarity measures. RESULTS Six types of working-life sequences were identified. The largest cluster, Stable High Activity, represented 48.4% of the cohort. Other types were: Stable High SA/DP (14.5%); Other (4.5%); and three types with mixed activity and varying SA/DP regarding the number of days/year and timing (32.6%). Characteristics of the different identified types of sequences were subsequently investigated. All types of sequences had lower odds for university education (OR range: 0.18-0.72) compared with Stable High Activity. Increasingly higher odds of having anxiety/depression compared with Stable High Activity were observed across the types of sequences, by increasing proportions of SA/DP. Stable High SA/DP sequences were less likely than Stable High Activity to be prescribed MS drugs in the MS diagnosis year (OR 0.61; 95% CI 0.47 to 0.78). All types of sequences had higher disposable income in the final study year than the first, except for Stable High SA/DP sequences (Swedish Krona 4669, 95% CI -1892 to 11 230). CONCLUSIONS Diversity in working life was influenced by sociodemographic and clinical characteristics resulting in different activity and SA/DP patterns across the six identified types of working-life sequences.
Collapse
Affiliation(s)
- Chantelle Murley
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171-77 Stockholm, Sweden
| | - Petter Tinghög
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171-77 Stockholm, Sweden
- Department of Health Sciences, Swedish Red Cross University College, 141-21 Huddinge, Sweden
| | - Korinna Karampampa
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171-77 Stockholm, Sweden
| | - Jan Hillert
- Division of Neurology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, 171-77 Sweden
| | - Kristina Alexanderson
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171-77 Stockholm, Sweden
| | - Emilie Friberg
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171-77 Stockholm, Sweden
| |
Collapse
|
37
|
Razaz N, Piehl F, Frisell T, Langer-Gould AM, McKay KA, Fink K. Disease activity in pregnancy and postpartum in women with MS who suspended rituximab and natalizumab. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 7:7/6/e903. [PMID: 33087582 PMCID: PMC7641107 DOI: 10.1212/nxi.0000000000000903] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 08/24/2020] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To evaluate risks of disease reactivity during pregnancy and postpartum following rituximab (RTX) and natalizumab (NTZ) suspension in women with MS. METHODS An observational cohort study of all women with MS disease onset before childbirth between 2006 and 2017. Women were identified through the Swedish MS Registry, a nationwide clinical register, with substratification into 3 groups: women who suspended RTX and NTZ within 6 months before conception and women who were not treated with any disease-modifying treatment (DMT) within 1 year of conception. The primary outcome was the annualized relapse rate (ARR) during pregnancy and 1 year postpartum. RESULTS We identified 2,386 women with MS onset before a live birth; of these, 76 women suspended RTX and 53 suspended NTZ, and 457 were untreated within 1 year before conception. In all women, regardless of the treatment type, the ARR declined from 0.05-0.04 prepregnancy to 0.03-0.02 during pregnancy, returning to prepregnancy rates at 3-6 months (0.05) postpartum. In the suspended cohort, 76% (98/129) of women resumed a DMT after delivery. The relapse rate 1 year postpartum was significantly higher in the suspended NTZ women compared with the suspended RTX women (adjusted rate ratio [aRR] 7.65, 95% CI 2.47-23.6) and was lower in the suspended RTX women compared with the untreated women (aRR 0.21, 95% CI 0.08-0.61). CONCLUSION Disease reactivity during the postpartum period was lower among women with MS who suspended RTX before pregnancy, relative to those who suspended NTZ and untreated women. These findings suggest that RTX may exert long-acting effects on MS disease activity that encompass pregnancy and postpartum periods. CLASSIFICATION OF EVIDENCE This study provides Class IV evidence that in patients with MS who were on treatment before pregnancy, RTX reduces clinical disease activity compared with NTZ in the postpartum period.
Collapse
Affiliation(s)
- Neda Razaz
- From the Department of Medicine (N.R., T.F.), Solna, Clinical Epidemiology Division, Karolinska Institutet; Department of Clinical Neuroscience (F.P., K.A.M., K.F.), Karolinska Institutet; Centrum for Neurology (F.P., K.F.), Academical Specialist Center, Stockholm, Sweden; Southern California Permanente Medical Group/Kaiser Permanente (A.M.L.-G.), Pasadena, CA; and Centre for Molecular Medicine (K.A.M.), Karolinska University Hospital, Stockholm, Sweden.
| | - Fredrik Piehl
- From the Department of Medicine (N.R., T.F.), Solna, Clinical Epidemiology Division, Karolinska Institutet; Department of Clinical Neuroscience (F.P., K.A.M., K.F.), Karolinska Institutet; Centrum for Neurology (F.P., K.F.), Academical Specialist Center, Stockholm, Sweden; Southern California Permanente Medical Group/Kaiser Permanente (A.M.L.-G.), Pasadena, CA; and Centre for Molecular Medicine (K.A.M.), Karolinska University Hospital, Stockholm, Sweden
| | - Thomas Frisell
- From the Department of Medicine (N.R., T.F.), Solna, Clinical Epidemiology Division, Karolinska Institutet; Department of Clinical Neuroscience (F.P., K.A.M., K.F.), Karolinska Institutet; Centrum for Neurology (F.P., K.F.), Academical Specialist Center, Stockholm, Sweden; Southern California Permanente Medical Group/Kaiser Permanente (A.M.L.-G.), Pasadena, CA; and Centre for Molecular Medicine (K.A.M.), Karolinska University Hospital, Stockholm, Sweden
| | - Annette M Langer-Gould
- From the Department of Medicine (N.R., T.F.), Solna, Clinical Epidemiology Division, Karolinska Institutet; Department of Clinical Neuroscience (F.P., K.A.M., K.F.), Karolinska Institutet; Centrum for Neurology (F.P., K.F.), Academical Specialist Center, Stockholm, Sweden; Southern California Permanente Medical Group/Kaiser Permanente (A.M.L.-G.), Pasadena, CA; and Centre for Molecular Medicine (K.A.M.), Karolinska University Hospital, Stockholm, Sweden
| | - Kyla A McKay
- From the Department of Medicine (N.R., T.F.), Solna, Clinical Epidemiology Division, Karolinska Institutet; Department of Clinical Neuroscience (F.P., K.A.M., K.F.), Karolinska Institutet; Centrum for Neurology (F.P., K.F.), Academical Specialist Center, Stockholm, Sweden; Southern California Permanente Medical Group/Kaiser Permanente (A.M.L.-G.), Pasadena, CA; and Centre for Molecular Medicine (K.A.M.), Karolinska University Hospital, Stockholm, Sweden
| | - Katharina Fink
- From the Department of Medicine (N.R., T.F.), Solna, Clinical Epidemiology Division, Karolinska Institutet; Department of Clinical Neuroscience (F.P., K.A.M., K.F.), Karolinska Institutet; Centrum for Neurology (F.P., K.F.), Academical Specialist Center, Stockholm, Sweden; Southern California Permanente Medical Group/Kaiser Permanente (A.M.L.-G.), Pasadena, CA; and Centre for Molecular Medicine (K.A.M.), Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
38
|
Dunn N, Fogdell-Hahn A, Hillert J, Spelman T. Long-Term Consequences of High Titer Neutralizing Antibodies to Interferon-β in Multiple Sclerosis. Front Immunol 2020; 11:583560. [PMID: 33178215 PMCID: PMC7593513 DOI: 10.3389/fimmu.2020.583560] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/07/2020] [Indexed: 11/22/2022] Open
Abstract
Background Neutralizing anti-drug antibodies (NAbs) to interferon beta (IFNβ) develop in up to 47% of multiple sclerosis (MS) treated patients inhibiting treatment effect of IFNβ. However, the long-term effect of NAbs remain unknown. Objective To investigate the long-term consequences of high titer NAbs to IFNβ on disease activity and progression in MS patients. Methods An observational study including data from all IFNβ treated relapsing remitting MS patients with sufficient NAb test results from the Swedish MS registry. Patients were classified into either confirmed ‘high titer’ or ‘persistent negative’ groups and analyzed for differences in disease activity and progression over time. Results A total of 197 high-titer and 2907 persistent negative patients with 19969.6 follow up years of data were included. High titer NAbs were associated with a higher degree of disease activity at baseline. However, even when accounting for this, the presence of high titer NAbs were also associated with higher disease activity during IFNβ treatment. This persisted even after the next DMT start, suggesting that earlier high titers may partially reduce the effect of later treatments. No difference was found in confirmed disability progression. Conclusion High titer NAbs to IFNβ are associated with higher disease activity, persisting even after IFNβ discontinuation or switch. These results support use of highly efficient treatment earlier in patients with active disease, to avoid these complications.
Collapse
Affiliation(s)
- Nicky Dunn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Clinical Neuroimmunology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Clinical Neuroimmunology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tim Spelman
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
39
|
Evertsson B, Hoyt T, Christensen A, Nimer FA, Foley J, Piehl F. A comparative study of tolerability and effects on immunoglobulin levels and CD19 cell counts with ocrelizumab vs low dose of rituximab in multiple sclerosis. Mult Scler J Exp Transl Clin 2020; 6:2055217320964505. [PMID: 33110619 PMCID: PMC7556182 DOI: 10.1177/2055217320964505] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/16/2020] [Indexed: 12/24/2022] Open
Abstract
Background Rituximab (RTX) and ocrelizumab (OCR) are two anti-CD20 biologics used in MS; however, comparisons on safety and efficacy are rare. Objective To compare treatment outcomes over the first year with RTX and OCR. Methods Retrospective cohort study comprising MS patients initiating RTX at the Karolinska University Hospital (Sweden; n = 311) and OCR at Rocky Mountain MS Clinic (Utah, USA; n = 161), respectively. Results Levels of immunoglobulin G measured in blood dropped 0.16 g/L (95% confidence interval 0.01 to 0.31) with each OCR infusion, but remained stable with RTX. In contrast, levels of immunoglobulin M decreased to a similar extent with both drugs. Ten and 15% of patients discontinued treatment with RTX and OCR, respectively (n.s), however, adverse events leading to treatment discontinuation were more common with OCR (6.8% vs 2.6%; p = 0.026). Only 3.1 and 1.6% discontinued OCR and RTX, respectively, due to lack of effect (n.s). The degree of B cell depletion was superior with OCR. Conclusion Overall, differences between the two treatments were small. Although the study design precludes robust conclusions regarding the risk-benefit with the studied therapies, our findings indicate that the tolerability and safety with RTX is not inferior to OCR.
Collapse
Affiliation(s)
- Björn Evertsson
- Neuroimmunology Unit, Karolinska Institutet, Clinical Neuroscience, Karolinska University Hospital, Stockholm, Sweden
| | - Tammy Hoyt
- Neuroimmunology Unit, Karolinska Institutet, Clinical Neuroscience, Karolinska University Hospital, Stockholm, Sweden
| | - Angel Christensen
- Rocky Mountain Multiple Sclerosis Clinic, Neurology, Salt Lake City, UT, USA
| | - Faiez Al Nimer
- Neuroimmunology Unit, Karolinska Institutet, Clinical Neuroscience, Karolinska University Hospital, Stockholm, Sweden
| | - John Foley
- Rocky Mountain Multiple Sclerosis Clinic, Neurology, Salt Lake City, UT, USA
| | - Fredrik Piehl
- Neuroimmunology Unit, Karolinska Institutet, Clinical Neuroscience, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
40
|
Kavaliunas A, Manouchehrinia A, Gyllensten H, Alexanderson K, Hillert J. Importance of early treatment decisions on future income of multiple sclerosis patients. Mult Scler J Exp Transl Clin 2020; 6:2055217320959116. [PMID: 33110615 PMCID: PMC7564625 DOI: 10.1177/2055217320959116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 07/27/2020] [Indexed: 11/30/2022] Open
Abstract
Background Early initiation of disease-modifying treatment (DMT) is associated with
better disability outcomes in multiple sclerosis (MS). However, little is
known of how treatment decisions affect socio-economic outcomes. Objective To estimate the long-term impact of early initiation of DMT on the income of
MS patients. Methods In total, 3610 MS patients were included in this register-based cohort study.
We measured the association between the time to treatment and the outcome,
defined as time from treatment initiation to a 95% decrease in annual
earnings compared to each patient´s baseline level. Additionally, the
association between time to treatment and increase of social benefits
(sickness absence, disability pension) was investigated. A Cox model was
adjusted for sex, onset age, education, family situation, country of birth,
living area, and disability. Results MS patients initiating treatment later had a higher risk of reaching the
outcome- those who started treatment after 2 years from MS onset lost 95% of
their earnings sooner (HR, 1.19; 95% CI, 1.04–1.37). Furthermore, risk to
receive an annual compensation of SEK 100,000 (≈EUR 10,500) was higher for
the delayed treatment group. Conclusion Early treatment initiation in MS is associated with better socioeconomic
outcome, adding to previous studies showing benefits regarding
disability.
Collapse
Affiliation(s)
- Andrius Kavaliunas
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Hospital, Stockholm, Sweden
| | - Ali Manouchehrinia
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Hanna Gyllensten
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
41
|
Manouchehrinia A, Stridh P, Khademi M, Leppert D, Barro C, Michalak Z, Benkert P, Lycke J, Alfredsson L, Kappos L, Piehl F, Olsson T, Kuhle J, Kockum I. Plasma neurofilament light levels are associated with risk of disability in multiple sclerosis. Neurology 2020; 94:e2457-e2467. [PMID: 32434867 PMCID: PMC7455371 DOI: 10.1212/wnl.0000000000009571] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 12/27/2019] [Indexed: 12/31/2022] Open
Abstract
Objective To investigate the association between plasma neurofilament light chain (pNfL) levels and the risk of developing sustained disability worsening. Methods Concentrations of pNfL were determined in 4,385 persons with multiple sclerosis (MS) and 1,026 randomly selected population-based sex- and age-matched controls using the highly sensitive Single Molecule Array (SimoaTM) NF-Light Advantage Kit. We assessed the impact of age-stratified pNfL levels above the 80th, 95th, and 99th percentiles among controls on the risk of Expanded Disability Status Scale (EDSS) worsening within the following year and reaching sustained EDSS scores of 3.0, 4.0, and 6.0 and conversion to secondary progressive multiple sclerosis (SPMS). Results The median (interquartile range [IQR]) pNfL was 7.5 (4.1) pg/mL in controls and 11.4 (9.6) pg/mL in MS (p < 0.001). The median (IQR) duration of follow-up was 5 (5.1) years. High pNfL was associated with increased adjusted rates of EDSS worsening ranging between 1.4 (95% confidence intervals [CIs]: 1.1–1.8) and 1.7 (95% CI: 1.4–2.3). High pNfL was also associated with the risk of reaching a sustained EDSS score of 3.0, with adjusted rates ranging between 1.5 (95% CI: 1.2–1.8) and 1.55 (95% CI: 1.3–1.8) over all percentile cutoffs (all p < 0.001). Similar increases were observed for the risk of sustained EDSS score 4.0. In contrast, the risk of reaching sustained EDSS score 6.0 and conversion to SPMS was not consistently significant. Conclusions Elevated pNfL levels at early stages of MS are associated with an increased risk of reaching sustained disability worsening. Hence, pNfL may serve as a prognostic tool to assess the risk of developing permanent disability in MS.
Collapse
Affiliation(s)
- Ali Manouchehrinia
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden.
| | - Pernilla Stridh
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| | - Mohsen Khademi
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| | - David Leppert
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| | - Christian Barro
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| | - Zuzanna Michalak
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| | - Pascal Benkert
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| | - Jan Lycke
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| | - Lars Alfredsson
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| | - Ludwig Kappos
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| | - Fredrik Piehl
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| | - Tomas Olsson
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| | - Jens Kuhle
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| | - Ingrid Kockum
- From the Department of Clinical Neuroscience (A.M., P.S., M.K., F.P., T.O., I.K.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Karolinska Institutet; Centre for Molecular Medicine (A.M., P.S., M.K., F.P., T.O., I.K.), Karolinska University Hospital, Stockholm, Sweden; Departments of Medicine, Biomedicine, and Clinical Research (D.L., C.B., Z.M., L.K., J.K.), Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Switzerland; Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Gothenburg; Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| |
Collapse
|
42
|
Wallin MT, Whitham R, Maloni H, Jin S, Duckart J, Haselkorn J, Culpepper WJ. The Multiple Sclerosis Surveillance Registry: A Novel Interactive Database Within the Veterans Health Administration. Fed Pract 2020; 37:S18-S23. [PMID: 32341633 PMCID: PMC7182245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
OBJECTIVE To demonstrate the infrastructure and utility of an interactive health system database for multiple sclerosis (MS), we present the MS Surveillance Registry (MSSR) within the US Department of Veterans Affairs (VA). BACKGROUND Disease specific databases can be helpful in the management of neurologic conditions but few are fully integrated into the electronic health record and linked to health system data. Creating a consistent information technology (IT) architecture and with ongoing support within disease specific registries has been a challenge. METHODS Building the MSSR was initiated by an iterative process with an IT team and MS health care providers. A common registry platform shared by other VA disease specific registries (eg, traumatic brain injury and cancer) was used to develop the IT infrastructure. MS cases were entered online into the MS Assessment Tool at selected MS Centers of Excellence (MSCoE) clinics in the US. Other large VA databases linked to MSSR are reviewed. Patient demographic and clinical characteristics were compared and contrasted with the broader VA population and other US registry populations. RESULTS We have enrolled 1,743 patients with MS in the MSSR through fiscal year 2019 from selected MS regional programs in the VA MSCoE network. The mean age of patients was 56.0 years, with a 2.7 male:female ratio. Among those with definite MS, the mean European Database for MS Disability Score was 4.7 and 75% had ever used an MS disease modifying therapy. A summary electronic dashboard was developed for health care providers to easily access demographic and clinical data for individuals and groups of patients. Data on comorbid conditions, pharmacy and prosthetics utilization, outpatient clinic visits, and inpatient admission were documented for each patient. CONCLUSIONS The MSSR is a unique electronic database that has enhanced clinical management of MS and serves as a national source for clinical outcomes.
Collapse
Affiliation(s)
- Mitchell T Wallin
- is the Director of the VA Multiple Sclerosis Center of Excellence (MSCoE)-East and Associate Professor of Neurology, George Washington University School of Medicine in Washington, DC. is Professor Emeritus of Neurology at Oregon Health and Science University in Portland. is the Clinical Director of the VA MSCoE-East in Washington, DC. is a Statistician and Data Analyst at VA MSCoE-East in Baltimore, Maryland. is a Health System Specialist at the VA Office of Inspector General in Portland. is the Director of the VA MSCoE-West and a Professor of Physical Medicine and Rehabilitation at the University of Washington School of Medicine and Public Health in Seattle. is the Director of the Veterans Health Administration Epidemiology Program and Director of Epidemiology and Informatics at VA MSCoE-East and an Adjunct Associate Professor of Neurology at the University of Maryland School of Medicine in Baltimore
| | - Ruth Whitham
- is the Director of the VA Multiple Sclerosis Center of Excellence (MSCoE)-East and Associate Professor of Neurology, George Washington University School of Medicine in Washington, DC. is Professor Emeritus of Neurology at Oregon Health and Science University in Portland. is the Clinical Director of the VA MSCoE-East in Washington, DC. is a Statistician and Data Analyst at VA MSCoE-East in Baltimore, Maryland. is a Health System Specialist at the VA Office of Inspector General in Portland. is the Director of the VA MSCoE-West and a Professor of Physical Medicine and Rehabilitation at the University of Washington School of Medicine and Public Health in Seattle. is the Director of the Veterans Health Administration Epidemiology Program and Director of Epidemiology and Informatics at VA MSCoE-East and an Adjunct Associate Professor of Neurology at the University of Maryland School of Medicine in Baltimore
| | - Heidi Maloni
- is the Director of the VA Multiple Sclerosis Center of Excellence (MSCoE)-East and Associate Professor of Neurology, George Washington University School of Medicine in Washington, DC. is Professor Emeritus of Neurology at Oregon Health and Science University in Portland. is the Clinical Director of the VA MSCoE-East in Washington, DC. is a Statistician and Data Analyst at VA MSCoE-East in Baltimore, Maryland. is a Health System Specialist at the VA Office of Inspector General in Portland. is the Director of the VA MSCoE-West and a Professor of Physical Medicine and Rehabilitation at the University of Washington School of Medicine and Public Health in Seattle. is the Director of the Veterans Health Administration Epidemiology Program and Director of Epidemiology and Informatics at VA MSCoE-East and an Adjunct Associate Professor of Neurology at the University of Maryland School of Medicine in Baltimore
| | - Shan Jin
- is the Director of the VA Multiple Sclerosis Center of Excellence (MSCoE)-East and Associate Professor of Neurology, George Washington University School of Medicine in Washington, DC. is Professor Emeritus of Neurology at Oregon Health and Science University in Portland. is the Clinical Director of the VA MSCoE-East in Washington, DC. is a Statistician and Data Analyst at VA MSCoE-East in Baltimore, Maryland. is a Health System Specialist at the VA Office of Inspector General in Portland. is the Director of the VA MSCoE-West and a Professor of Physical Medicine and Rehabilitation at the University of Washington School of Medicine and Public Health in Seattle. is the Director of the Veterans Health Administration Epidemiology Program and Director of Epidemiology and Informatics at VA MSCoE-East and an Adjunct Associate Professor of Neurology at the University of Maryland School of Medicine in Baltimore
| | - Jonathan Duckart
- is the Director of the VA Multiple Sclerosis Center of Excellence (MSCoE)-East and Associate Professor of Neurology, George Washington University School of Medicine in Washington, DC. is Professor Emeritus of Neurology at Oregon Health and Science University in Portland. is the Clinical Director of the VA MSCoE-East in Washington, DC. is a Statistician and Data Analyst at VA MSCoE-East in Baltimore, Maryland. is a Health System Specialist at the VA Office of Inspector General in Portland. is the Director of the VA MSCoE-West and a Professor of Physical Medicine and Rehabilitation at the University of Washington School of Medicine and Public Health in Seattle. is the Director of the Veterans Health Administration Epidemiology Program and Director of Epidemiology and Informatics at VA MSCoE-East and an Adjunct Associate Professor of Neurology at the University of Maryland School of Medicine in Baltimore
| | - Jodie Haselkorn
- is the Director of the VA Multiple Sclerosis Center of Excellence (MSCoE)-East and Associate Professor of Neurology, George Washington University School of Medicine in Washington, DC. is Professor Emeritus of Neurology at Oregon Health and Science University in Portland. is the Clinical Director of the VA MSCoE-East in Washington, DC. is a Statistician and Data Analyst at VA MSCoE-East in Baltimore, Maryland. is a Health System Specialist at the VA Office of Inspector General in Portland. is the Director of the VA MSCoE-West and a Professor of Physical Medicine and Rehabilitation at the University of Washington School of Medicine and Public Health in Seattle. is the Director of the Veterans Health Administration Epidemiology Program and Director of Epidemiology and Informatics at VA MSCoE-East and an Adjunct Associate Professor of Neurology at the University of Maryland School of Medicine in Baltimore
| | - William J Culpepper
- is the Director of the VA Multiple Sclerosis Center of Excellence (MSCoE)-East and Associate Professor of Neurology, George Washington University School of Medicine in Washington, DC. is Professor Emeritus of Neurology at Oregon Health and Science University in Portland. is the Clinical Director of the VA MSCoE-East in Washington, DC. is a Statistician and Data Analyst at VA MSCoE-East in Baltimore, Maryland. is a Health System Specialist at the VA Office of Inspector General in Portland. is the Director of the VA MSCoE-West and a Professor of Physical Medicine and Rehabilitation at the University of Washington School of Medicine and Public Health in Seattle. is the Director of the Veterans Health Administration Epidemiology Program and Director of Epidemiology and Informatics at VA MSCoE-East and an Adjunct Associate Professor of Neurology at the University of Maryland School of Medicine in Baltimore
| |
Collapse
|
43
|
He A, Merkel B, Brown JWL, Zhovits Ryerson L, Kister I, Malpas CB, Sharmin S, Horakova D, Kubala Havrdova E, Spelman T, Izquierdo G, Eichau S, Trojano M, Lugaresi A, Hupperts R, Sola P, Ferraro D, Lycke J, Grand'Maison F, Prat A, Girard M, Duquette P, Larochelle C, Svenningsson A, Petersen T, Grammond P, Granella F, Van Pesch V, Bergamaschi R, McGuigan C, Coles A, Hillert J, Piehl F, Butzkueven H, Kalincik T. Timing of high-efficacy therapy for multiple sclerosis: a retrospective observational cohort study. Lancet Neurol 2020; 19:307-316. [DOI: 10.1016/s1474-4422(20)30067-3] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 02/04/2020] [Accepted: 02/24/2020] [Indexed: 11/30/2022]
|
44
|
Delcoigne B, Manouchehrinia A, Barro C, Benkert P, Michalak Z, Kappos L, Leppert D, Tsai JA, Plavina T, Kieseier BC, Lycke J, Alfredsson L, Kockum I, Kuhle J, Olsson T, Piehl F. Blood neurofilament light levels segregate treatment effects in multiple sclerosis. Neurology 2020; 94:e1201-e1212. [PMID: 32047070 PMCID: PMC7387108 DOI: 10.1212/wnl.0000000000009097] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/21/2019] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE To determine factors (including the role of specific disease modulatory treatments [DMTs]) associated with (1) baseline, (2) on-treatment, and (3) change (from treatment start to on-treatment assessment) in plasma neurofilament light chain (pNfL) concentrations in relapsing-remitting multiple sclerosis (RRMS). METHODS Data including blood samples analyses and long-term clinical follow-up information for 1,261 Swedish patients with RRMS starting novel DMTs were analyzed using linear regressions to model pNfL and changes in pNfL concentrations as a function of clinical variables and DMTs (alemtuzumab, dimethyl fumarate, fingolimod, natalizumab, rituximab, and teriflunomide). RESULTS The baseline pNfL concentration was positively associated with relapse rate, Expanded Disability Status Scale score, Age-Related MS Severity Score, and MS Impact Score (MSIS-29), and negatively associated with Symbol Digit Modalities Test performance and the number of previously used DMTs. All analyses, which used inverse propensity score weighting to correct for differences in baseline factors at DMT start, highlighted that both the reduction in pNfL concentration from baseline to on-treatment measurement and the on-treatment pNfL level differed across DMTs. Patients starting alemtuzumab displayed the highest reduction in pNfL concentration and lowest on-treatment pNfL concentrations, while those starting teriflunomide had the smallest decrease and highest on-treatment levels, but also starting from lower values. Both on-treatment pNfL and decrease in pNfL concentrations were highly dependent on baseline concentrations. CONCLUSION Choice of DMT in RRMS is significantly associated with degree of reduction in pNfL, which supports a role for pNfL as a drug response marker.
Collapse
Affiliation(s)
- Bénédicte Delcoigne
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden.
| | - Ali Manouchehrinia
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Christian Barro
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Pascal Benkert
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Zuzanna Michalak
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Ludwig Kappos
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - David Leppert
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Jon A Tsai
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Tatiana Plavina
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Bernd C Kieseier
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Jan Lycke
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Lars Alfredsson
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Ingrid Kockum
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Jens Kuhle
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Tomas Olsson
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Fredrik Piehl
- From the Department of Medicine Solna, Clinical Epidemiology Division (B.D.), The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience (A.M., I.K., T.O., F.P.), and Institute of Environmental Medicine (L.A.), Karolinska Institutet; Centre for Molecular Medicine (A.M., I.K., T.O., F.P.), Karolinska University Hospital, Stockholm, Sweden; Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research (C.B., Z.M., L.K., D.L., J.K.), and Clinical Trial Unit, Department of Clinical Research (P.B.), University Hospital Basel, University of Basel, Switzerland; Sanofi Genzyme (J.A.T.), Stockholm, Sweden; Biogen (T.P., B.C.K.), Cambridge, MA; Department of Neurology, Medical Faculty (B.C.K.), Heinrich-Heine University, Duesseldorf, Germany; and Institution of Neuroscience and Physiology (J.L.), Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
45
|
Cancer Risk for Fingolimod, Natalizumab, and Rituximab in Multiple Sclerosis Patients. Ann Neurol 2020; 87:688-699. [DOI: 10.1002/ana.25701] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 02/11/2020] [Accepted: 02/11/2020] [Indexed: 01/02/2023]
|
46
|
Luna G, Alping P, Burman J, Fink K, Fogdell-Hahn A, Gunnarsson M, Hillert J, Langer-Gould A, Lycke J, Nilsson P, Salzer J, Svenningsson A, Vrethem M, Olsson T, Piehl F, Frisell T. Infection Risks Among Patients With Multiple Sclerosis Treated With Fingolimod, Natalizumab, Rituximab, and Injectable Therapies. JAMA Neurol 2020; 77:184-191. [PMID: 31589278 PMCID: PMC6784753 DOI: 10.1001/jamaneurol.2019.3365] [Citation(s) in RCA: 328] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 08/16/2019] [Indexed: 12/25/2022]
Abstract
Importance Although highly effective disease-modifying therapies for multiple sclerosis (MS) have been associated with an increased risk of infections vs injectable therapies interferon beta and glatiramer acetate (GA), the magnitude of potential risk increase is not well established in real-world populations. Even less is known about infection risk associated with rituximab, which is extensively used off-label to treat MS in Sweden. Objective To examine the risk of serious infections associated with disease-modifying treatments for MS. Design, Setting, and Participants This nationwide register-based cohort study was conducted in Sweden from January 1, 2011, to December 31, 2017. National registers with prospective data collection from the public health care system were used. All Swedish patients with relapsing-remitting MS whose data were recorded in the Swedish MS register as initiating treatment with rituximab, natalizumab, fingolimod, or interferon beta and GA and an age-matched and sex-matched general population comparator cohort were included. Exposures Treatment with rituximab, natalizumab, fingolimod, and interferon beta and GA. Main Outcomes and Measures Serious infections were defined as all infections resulting in hospitalization. Additional outcomes included outpatient treatment with antibiotic or herpes antiviral medications. Adjusted hazard ratios (HRs) were estimated in Cox regressions. Results A total of 6421 patients (3260 taking rituximab, 1588 taking natalizumab, 1535 taking fingolimod, and 2217 taking interferon beta/GA) were included, plus a comparator cohort of 42 645 individuals. Among 6421 patients with 8600 treatment episodes, the mean (SD) age at treatment start ranged from 35.0 (10.1) years to 40.4 (10.6) years; 6186 patients were female. The crude rate of infections was higher in patients with MS taking interferon beta and GA than the general population (incidence rate, 8.9 [95% CI, 6.4-12.1] vs 5.2 [95% CI, 4.8-5.5] per 1000 person-years), and higher still in patients taking fingolimod (incidence rate, 14.3 [95% CI, 10.8-18.5] per 1000 person-years), natalizumab (incidence rate, 11.4 [95% CI, 8.3-15.3] per 1000 person-years), and rituximab (incidence rate, 19.7 [95% CI, 16.4-23.5] per 1000 person-years). After confounder adjustment, the rate remained significantly higher for rituximab (HR, 1.70 [95% CI, 1.11-2.61]) but not fingolimod (HR, 1.30 [95% CI, 0.84-2.03]) or natalizumab (HR, 1.12 [95% CI, 0.71-1.77]) compared with interferon beta and GA. In contrast, use of herpes antiviral drugs during rituximab treatment was similar to that of interferon beta and GA and lower than that of natalizumab (HR, 1.82 [1.34-2.46]) and fingolimod (HR, 1.71 [95% CI, 1.27-2.32]). Conclusions and Relevance Patients with MS are at a generally increased risk of infections, and this differs by treatment. The rate of infections was lowest with interferon beta and GA; among newer treatments, off-label use of rituximab was associated with the highest rate of serious infections. The different risk profiles should inform the risk-benefit assessments of these treatments.
Collapse
Affiliation(s)
- Gustavo Luna
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Peter Alping
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Joachim Burman
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Katharina Fink
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin Gunnarsson
- Center for Health and Medical Psychology, Örebro University, Örebro, Sweden
| | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Annette Langer-Gould
- Clinical and Translational Neuroscience, Southern California Permanente Medical Group, Kaiser Permanente, Pasadena
| | - Jan Lycke
- Department of Clinical Neuroscience and Rehabilitation, University of Gothenburg, Gothenburg, Sweden
| | - Petra Nilsson
- Department of Clinical Sciences, Division of Neurology, Lund University, Lund, Sweden
| | - Jonatan Salzer
- Department of Pharmacology and Clinical Neuroscience, Umea University, Umea, Sweden
| | - Anders Svenningsson
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Vrethem
- Department of Clinical and Experimental Medicine, Linkoping University, Linkoping, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Academic Specialist Center, Stockholm Health Services, Stockholm, Sweden
| | - Thomas Frisell
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
47
|
Murley C, Friberg E, Hillert J, Alexanderson K, Yang F. Validation of multiple sclerosis diagnoses in the Swedish National Patient Register. Eur J Epidemiol 2019; 34:1161-1169. [PMID: 31493189 PMCID: PMC7010617 DOI: 10.1007/s10654-019-00558-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/29/2019] [Indexed: 11/28/2022]
Abstract
Population-based registers are widely used in epidemiological studies. We aimed to estimate the validity of multiple sclerosis (MS) diagnoses registered in the Swedish National Patient Register (NPR) by two sequential register-based case-definition algorithms. Prevalent MS patients aged 16–64 years were identified from the in- and specialised out-patient NPR in 2001–2013, using International Classification of Diseases code G35. These identified MS diagnoses were validated through two sequential register-based case-definition algorithms, as the ‘gold-standard’ reference, by linking individual-level data longitudinally to other nationwide registers. The primary algorithm first sought to corroborate the MS diagnoses with MS-specific information in other nationwide registers. The exploratory secondary algorithm identified individuals with MS-related information in other registers and those who were unable to be followed sufficiently. Through multi-register linkage, we estimated the number of confirmed and uncertain individuals with an MS diagnosis recorded in the NPR. A total of 19,781 individuals (mean age at first visit 45.2 years; 69.5% women) had at least one MS diagnosis recorded in the NPR during 2001–2013. Using the two case-definition algorithms, 92.5% (n = 18,291) of the MS diagnoses recorded in the NPR were confirmed, while 7.5% (n = 1490) remained uncertain. Our findings indicate that a very high percentage of patients coded with an MS diagnosis in the Swedish NPR actually have MS, and supports the use of the NPR as a viable source to identify individuals with an MS diagnosis for population-based research. This exploratory methods paper suggests an alternative novel method to verify individuals’ diagnoses in register-based settings.
Collapse
Affiliation(s)
- Chantelle Murley
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Emilie Friberg
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Jan Hillert
- Division of Neurology, Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Kristina Alexanderson
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Fei Yang
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| |
Collapse
|
48
|
Granqvist M, Burman J, Gunnarsson M, Lycke J, Nilsson P, Olsson T, Sundström P, Svenningsson A, Vrethem M, Frisell T, Piehl F. Comparative effectiveness of dimethyl fumarate as the initial and secondary treatment for MS. Mult Scler 2019; 26:1532-1539. [DOI: 10.1177/1352458519866600] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Population-based real-world evidence studies of the effectiveness and tolerability of dimethyl fumarate in relation to common treatment alternatives are still limited. Objective: To evaluate the clinical effectiveness and tolerability of dimethyl fumarate (DMF) as the initial and secondary treatment for relapsing-remitting multiple sclerosis (RRMS) patients compared with common treatment alternatives in Sweden. Methods: We conducted a nationwide retrospective observational cohort study of all RRMS patients identified through the Swedish MS registry initiating DMF ( n = 641) or interferons/glatiramer acetate (IFN/GA; n = 555) as the initial therapy, or DMF ( n = 703) or fingolimod (FGL; n = 194) after switch from IFN/GA between 1 January 2014 and 31 December 2016. Results: The discontinuation rate was lower with DMF as the initial treatment than IFN/GA (adjusted hazard rate (HR): 0.46, 95% confidence interval (CI): 0.37–0.58, p < 0.001), but higher than FGL as the secondary treatment (HR: 1.51, CI: 1.08–2.09, p < 0.05). Annualized relapse rate (ARR) was lower with DMF compared to IFN/GA (0.04, CI: 0.03–0.06 vs 0.10, CI: 0.07–0.13; p < 0.05), but not FGL (0.03, CI: 0.02–0.05 vs 0.02, CI: 0.01–0.04; p = 0.41). Finally, time to first relapse (TTFR) was longer with DMF as the initial, but not secondary, therapy ( p < 0.05 and p = 0.20, respectively). Conclusion: Our findings indicate that DMF performs better than IFN/GA as the initial treatment for RRMS. Compared to FGL, DMF displayed a lower tolerability, but largely similar effectiveness outcomes.
Collapse
Affiliation(s)
- Mathias Granqvist
- Department of Clinical Neuroscience, Center for Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
| | - Joachim Burman
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | | | - Jan Lycke
- Department of Neuroscience and Physiology, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Petra Nilsson
- Neurology Clinic, Skåne University Hospital, Lund, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Center for Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
| | - Peter Sundström
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Anders Svenningsson
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Vrethem
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Thomas Frisell
- Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Center for Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|