1
|
Reinhardt C, Angstwurm K, Freudenstein D, Lee DH, Wendl C, Linker RA. Real-world analysis of brain atrophy in multiple sclerosis patients with an artificial intelligence based software tool. Neurol Res Pract 2024; 6:40. [PMID: 39113151 PMCID: PMC11308334 DOI: 10.1186/s42466-024-00339-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Atrophy of white and grey matter volumes occurs early in the brains of people with multiple sclerosis (pwMS) and has great clinical relevance. In clinical trials, brain atrophy can be quantified by magnetic resonance imaging (MRI) with automated software tools. METHODS In this study, we analyze volumes of various brain regions with the software "md brain" based on routine MRI scans of 53 pwMS in a real-world setting. We compare brain volumes of pwMS with an EDSS ≥ 3.5 and a disease duration ≥ 10 years to the brain volumes of pwMS with an EDSS < 3.5 and a disease duration < 10 years as well as with or without immunotherapy. RESULTS pwMS with an EDSS ≥ 3.5 and a disease duration ≥ 10 years had significantly lower volumes of the total brain, the grey matter and of the frontal, temporal, parietal and occipital lobe regions as compared to pwMS with an EDSS < 3.5 and a disease duration < 10 years. Regional brain volumes were significantly lower in pwMS without immunotherapy. CONCLUSIONS The study showed that higher EDSS, longer disease duration and absence of immunotherapy was associated with lower volumes in a number of brain regions. Further real-world studies may include larger patient cohorts in longitudinal analyses.
Collapse
Affiliation(s)
- Caroline Reinhardt
- Department of Neurology, University of Regensburg, Universitätsstr. 84, 93053, Regensburg, Germany
| | - Klemens Angstwurm
- Department of Neurology, University of Regensburg, Universitätsstr. 84, 93053, Regensburg, Germany
| | - David Freudenstein
- Department of Neurology, University of Regensburg, Universitätsstr. 84, 93053, Regensburg, Germany
| | - De-Hyung Lee
- Department of Neurology, University of Regensburg, Universitätsstr. 84, 93053, Regensburg, Germany
| | - Christina Wendl
- Department of Neuroradiology, University of Regensburg, Regensburg, Germany
| | - Ralf A Linker
- Department of Neurology, University of Regensburg, Universitätsstr. 84, 93053, Regensburg, Germany.
| |
Collapse
|
2
|
Noteboom S, Strijbis EMM, Coerver EME, Colato E, van Kempen ZLE, Jasperse B, Vrenken H, Killestein J, Schoonheim MM, Steenwijk MD. Long-term neuroprotective effects of natalizumab and fingolimod in multiple sclerosis: Evidence from real-world clinical data. Mult Scler Relat Disord 2024; 87:105670. [PMID: 38772150 DOI: 10.1016/j.msard.2024.105670] [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: 11/29/2023] [Revised: 04/29/2024] [Accepted: 05/05/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND The long-term effect of high efficacy disease modifying therapy (DMT) on neurodegeneration in people with multiple sclerosis (pwMS) is largely unknown. The aim of this study was to evaluate the long-term effect of natalizumab (NTZ) or fingolimod (FTY) therapy on the evolution of brain atrophy compared to moderate efficacy DMT in a real-world clinical setting. METHODS A total of 438 pwMS with 2,439 MRI exams during treatment were analyzed: 252 pwMS treated with moderate efficacy DMT, 130 with NTZ and 56 with FTY. Evolution of brain atrophy was analyzed over an average follow-up of 6.6 years after treatment initiation. Brain segmentation was performed on clinical 3D-FLAIRs using SynthSeg and regional brain volume changes over time were compared between the treatment groups. RESULTS Total brain, white matter and deep gray matter atrophy rates did not differ between moderate efficacy DMTs, NTZ and FTY. Annualized ventricle growth rates were lower in pwMS treated with NTZ (1.1 %/year) compared with moderate efficacy DMT (2.4 %/year, p < 0.001) and similar to FTY (2.0 %/year, p = 0.051). Cortical atrophy rates were lower in NTZ (-0.08 %/year) compared with moderate efficacy DMT (-0.16 %/year, p = 0.048). CONCLUSION In a real-world clinical setting, pwMS treated with NTZ had slower ventricular expansion and cortical atrophy compared to those treated with moderate efficacy DMT.
Collapse
Affiliation(s)
- S Noteboom
- MS Center Amsterdam, Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, the Netherlands.
| | - E M M Strijbis
- MS Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - E M E Coerver
- MS Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - E Colato
- MS Center Amsterdam, Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing, University college London, UK
| | - Z L E van Kempen
- MS Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - B Jasperse
- MS Center Amsterdam, Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - H Vrenken
- MS Center Amsterdam, Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - J Killestein
- MS Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - M M Schoonheim
- MS Center Amsterdam, Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - M D Steenwijk
- MS Center Amsterdam, Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| |
Collapse
|
3
|
Hemond CC, Deshpande M, Berrios-Morales I, Zheng S, Meyer JS, Slavich GM, Cole SW. A single-arm, open-label pilot study of neuroimaging, behavioral, and peripheral inflammatory correlates of mindfulness-based stress reduction in multiple sclerosis. Sci Rep 2024; 14:14044. [PMID: 38890336 PMCID: PMC11189512 DOI: 10.1038/s41598-024-62960-w] [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: 02/14/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic neurological disease frequently associated with significant fatigue, anxiety, depression, and stress. These symptoms are difficult to treat, and prominently contribute to the decreases in quality of life observed with MS. The underlying mechanisms of these "silent" symptoms are not well understood and include not just the psychological responses to a chronic disease, but also biological contributions from bidirectional psycho-neuro-immune (dys)regulation of systemic inflammatory biology. To address these issues, we conducted a prospective, observational pilot study to investigate the psychological, biological, and neuroarchitecture changes associated with a mindfulness-based stress reduction (MBSR) program in MS. The overarching hypothesis was that MBSR modulates systemic and central nervous system inflammation via top-down neurocognitive control over forebrain limbic areas responsible for the neurobiological stress response. 23 patients were enrolled in MBSR and assessed pre/post-program with structural 3 T MRI, behavioral measures, hair cortisol, and blood measures of peripheral inflammation, as indexed by the Conserved Transcriptional Response to Adversity (CTRA) profile. MBSR was associated with improvements across a variety of behavioral outcomes, as well as on-study enlargement of the head of the right hippocampus. The CTRA analyses revealed that greater inflammatory gene expression was related to worse patient-reported anxiety, depression, stress, and loneliness, in addition to lower eudaimonic well-being. Hair cortisol did not significantly change from pre- to post-MBSR. These results support the use of MBSR in MS and elucidate inflammatory mechanisms related to key patient-reported outcomes in this population.
Collapse
Affiliation(s)
- Christopher C Hemond
- Department of Neurology, University of Massachusetts Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA.
| | - Mugdha Deshpande
- Department of Neurology, University of Massachusetts Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA
| | - Idanis Berrios-Morales
- Department of Neurology, University of Massachusetts Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA
| | - Shaokuan Zheng
- Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Jerrold S Meyer
- Department of Psychological & Brain Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, 90095, USA
| | - Steven W Cole
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, 90095, USA
| |
Collapse
|
4
|
Knudsen MH, Vestergaard MB, Lindberg U, Simonsen HJ, Frederiksen JL, Cramer SP, Larsson HBW. Age-related decline in cerebral oxygen consumption in multiple sclerosis. J Cereb Blood Flow Metab 2024; 44:1039-1052. [PMID: 38190981 PMCID: PMC11318400 DOI: 10.1177/0271678x231224502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024]
Abstract
Cerebral oxygen metabolism is altered in relapsing-remitting multiple sclerosis (RRMS), possibly a result of disease related cerebral atrophy with subsequent decreased oxygen demand. However, MS inflammation can also inhibit brain metabolism. Therefore, we measured cerebral blood flow (CBF) and cerebral metabolic rate of oxygen (CMRO2) using MRI phase contrast mapping and susceptibility-based oximetry in 44 patients with early RRMS and 36 healthy controls. Cerebral atrophy and white matter lesion load were assessed from high-resolution structural MRI. Expanded Disability Status Scale (EDSS) scores were collected from medical records. The CMRO2 was significantly lower in patients (-15%, p = 0.002) and decreased significantly with age in patients relative to the controls (-1.35 µmol/100 g/min/year, p = 0.036). The lower CMRO2 in RRMS was primarily driven by a higher venous oxygen saturation in the sagittal sinus (p = 0.007) and not a reduction in CBF (p = 0.69). There was no difference in cerebral atrophy between the groups, and no correlation between CMRO2 and MS lesion volume or EDSS score. Therefore, the progressive CMRO2 decline observed before the occurrence of significant cerebral atrophy and despite adequate CBF supports emerging evidence of dysfunctional cellular respiration as a potential pathogenic mechanism and therapeutic target in RRMS.
Collapse
Affiliation(s)
- Maria H Knudsen
- Functional Imaging Unit, Dept. of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Dept. of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen N, Denmark
| | - Mark B Vestergaard
- Functional Imaging Unit, Dept. of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Ulrich Lindberg
- Functional Imaging Unit, Dept. of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Helle J Simonsen
- Functional Imaging Unit, Dept. of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Jette L Frederiksen
- Dept. of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen N, Denmark
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Stig P Cramer
- Functional Imaging Unit, Dept. of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Henrik BW Larsson
- Functional Imaging Unit, Dept. of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Dept. of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
5
|
Nakamura K, Sun Z, Hara-Cleaver C, Bodhinathan K, Avila RL. Natalizumab reduces loss of gray matter and thalamic volume in patients with relapsing-remitting multiple sclerosis: A post hoc analysis from the randomized, placebo-controlled AFFIRM trial. Mult Scler 2024; 30:687-695. [PMID: 38469809 DOI: 10.1177/13524585241235055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
BACKGROUND Loss of brain gray matter fractional volume predicts multiple sclerosis (MS) progression and is associated with worsening physical and cognitive symptoms. Within deep gray matter, thalamic damage is evident in early stages of MS and correlates with physical and cognitive impairment. Natalizumab is a highly effective treatment that reduces disease progression and the number of inflammatory lesions in patients with relapsing-remitting MS (RRMS). OBJECTIVE To evaluate the effect of natalizumab on gray matter and thalamic atrophy. METHODS A combination of deep learning-based image segmentation and data augmentation was applied to MRI data from the AFFIRM trial. RESULTS This post hoc analysis identified a reduction of 64.3% (p = 0.0044) and 64.3% (p = 0.0030) in mean percentage gray matter volume loss from baseline at treatment years 1 and 2, respectively, in patients treated with natalizumab versus placebo. The reduction in thalamic fraction volume loss from baseline with natalizumab versus placebo was 57.0% at year 2 (p < 0.0001) and 41.2% at year 1 (p = 0.0147). Similar findings resulted from analyses of absolute gray matter and thalamic fraction volume loss. CONCLUSION These analyses represent the first placebo-controlled evidence supporting a role for natalizumab treatment in mitigating gray matter and thalamic fraction atrophy among patients with RRMS. CLINICALTRIALS.GOV IDENTIFIER NCT00027300URL: https://clinicaltrials.gov/ct2/show/NCT00027300.
Collapse
Affiliation(s)
- Kunio Nakamura
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH, USA
| | | | | | | | | |
Collapse
|
6
|
Ontaneda D, Chitnis T, Rammohan K, Obeidat AZ. Identification and management of subclinical disease activity in early multiple sclerosis: a review. J Neurol 2024; 271:1497-1514. [PMID: 37864717 PMCID: PMC10972995 DOI: 10.1007/s00415-023-12021-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/22/2023] [Accepted: 09/24/2023] [Indexed: 10/23/2023]
Abstract
IMPORTANCE Early treatment initiation in multiple sclerosis (MS) is crucial in preventing irreversible neurological damage and disability progression. The current assessment of disease activity relies on relapse rates and magnetic resonance imaging (MRI) lesion activity, but inclusion of other early, often "hidden," indicators of disease activity may describe a more comprehensive picture of MS. OBSERVATIONS Early indicators of MS disease activity other than relapses and MRI activity, such as cognitive impairment, brain atrophy, and fatigue, are not typically captured by routine disease monitoring. Furthermore, silent progression (neurological decline not clearly captured by standard methods) may occur undetected by relapse and MRI lesion activity monitoring. Consequently, patients considered to have no disease activity actually may have worsening disease, suggesting a need to revise MS management strategies with respect to timely initiation and escalation of disease-modifying therapy (DMT). Traditionally, first-line MS treatment starts with low- or moderate-efficacy therapies, before escalating to high-efficacy therapies (HETs) after evidence of breakthrough disease activity. However, multiple observational studies have shown that early initiation of HETs can prevent or reduce disability progression. Ongoing randomized clinical trials are comparing escalation and early HET approaches. CONCLUSIONS AND RELEVANCE There is an urgent need to reassess how MS disease activity and worsening are measured. A greater awareness of "hidden" indicators, potentially combined with biomarkers to reveal silent disease activity and neurodegeneration underlying MS, would provide a more complete picture of MS and allow for timely therapeutic intervention with HET or switching DMTs to address suboptimal treatment responses.
Collapse
Affiliation(s)
- Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Department of Neurology, Cleveland Clinic, Cleveland, OH, USA.
| | - Tanuja Chitnis
- Brigham Multiple Sclerosis Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kottil Rammohan
- Division of Multiple Sclerosis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ahmed Z Obeidat
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
7
|
Cortese R, Testa G, Assogna F, De Stefano N. Magnetic Resonance Imaging Evidence Supporting the Efficacy of Cladribine Tablets in the Treatment of Relapsing-Remitting Multiple Sclerosis. CNS Drugs 2024; 38:267-279. [PMID: 38489020 PMCID: PMC10980660 DOI: 10.1007/s40263-024-01074-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/15/2024] [Indexed: 03/17/2024]
Abstract
Numerous therapies are currently available to modify the disease course of multiple sclerosis (MS). Magnetic resonance imaging (MRI) plays a pivotal role in assessing treatment response by providing insights into disease activity and clinical progression. Integrating MRI findings with clinical and laboratory data enables a comprehensive assessment of the disease course. Among available MS treatments, cladribine is emerging as a promising option due to its role as a selective immune reconstitution therapy, with a notable impact on B cells and a lesser effect on T cells. This work emphasizes the assessment of MRI's contribution to MS treatment, particularly focusing on the influence of cladribine tablets on imaging outcomes, encompassing data from pivotal and real-world studies. The evidence highlights that cladribine, compared with placebo, not only exhibits a reduction in inflammatory imaging markers, such as T1-Gd+, T2 and combined unique active (CUA) lesions, but also mitigates the effect on brain volume loss, particularly within grey matter. Importantly, cladribine reveals early action by reducing CUA lesions within the first months of treatment, regardless of a patient's initial conditions. The selective mechanism of action, and sustained efficacy beyond year 2, combined with its early onset of action, collectively position cladribine tablets as a pivotal component in the therapeutic paradigm for MS. Overall, MRI, along with clinical measures, has played a substantial role in showcasing the effectiveness of cladribine in addressing both the inflammatory and neurodegenerative aspects of MS.
Collapse
Affiliation(s)
- Rosa Cortese
- Department of Medicine, Surgery and Neuroscience, University of Siena, Viale Bracci 2, 53100, Siena, Italy
| | - Giovanna Testa
- Merck Serono S.p.A. Italy, An Affiliate of Merck KGaA, Rome, Italy
| | | | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Viale Bracci 2, 53100, Siena, Italy.
| |
Collapse
|
8
|
Chertcoff A, Schneider R, Azevedo CJ, Sicotte N, Oh J. Recent Advances in Diagnostic, Prognostic, and Disease-Monitoring Biomarkers in Multiple Sclerosis. Neurol Clin 2024; 42:15-38. [PMID: 37980112 DOI: 10.1016/j.ncl.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2023]
Abstract
Multiple sclerosis (MS) is a highly heterogeneous disease. Currently, a combination of clinical features, MRI, and cerebrospinal fluid markers are used in clinical practice for diagnosis and treatment decisions. In recent years, there has been considerable effort to develop novel biomarkers that better reflect the pathologic substrates of the disease to aid in diagnosis and early prognosis, evaluation of ongoing inflammatory activity, detection and monitoring of disease progression, prediction of treatment response, and monitoring of disease-modifying treatment safety. In this review, the authors provide an overview of promising recent developments in diagnostic, prognostic, and disease-monitoring/treatment-response biomarkers in MS.
Collapse
Affiliation(s)
- Anibal Chertcoff
- Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, 30 Bond Street, PGT 17-742, Toronto, Ontario M5B 1W8, Canada
| | - Raphael Schneider
- Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, 30 Bond Street, PGT 17-742, Toronto, Ontario M5B 1W8, Canada
| | - Christina J Azevedo
- Department of Neurology, Keck School of Medicine, University of Southern California, HCT 1520 San Pablo Street, Health Sciences Campus, Los Angeles, CA 90033, USA
| | - Nancy Sicotte
- Department of Neurology, Cedars-Sinai Medical Center, 127 S San Vicente Boulevard, 6th floor, Suite A6600, Los Angeles, CA 90048, USA
| | - Jiwon Oh
- Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, 30 Bond Street, PGT 17-742, Toronto, Ontario M5B 1W8, Canada; Department of Neurology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
9
|
Lin T, Chien C, Kuchling J, Asseyer S, Motamedi S, Bellmann‐Strobl J, Schmitz‐Hübsch T, Ruprecht K, Brandt AU, Zimmermann HG, Paul F. Interactions of optic radiation lesions with retinal and brain atrophy in early multiple sclerosis. Ann Clin Transl Neurol 2024; 11:45-56. [PMID: 37903651 PMCID: PMC10791029 DOI: 10.1002/acn3.51931] [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: 07/06/2023] [Revised: 09/27/2023] [Accepted: 10/15/2023] [Indexed: 11/01/2023] Open
Abstract
OBJECTIVE Retrograde trans-synaptic neuroaxonal degeneration is considered a key pathological factor of subclinical retinal neuroaxonal damage in multiple sclerosis (MS). We aim to evaluate the longitudinal association of optic radiation (OR) lesion activity with retinal neuroaxonal damage and its role in correlations between retinal and brain atrophy in people with clinically isolated syndrome and early MS (pweMS). METHODS Eighty-five pweMS were retrospectively screened from a prospective cohort (Berlin CIS cohort). Participants underwent 3T magnetic resonance imaging (MRI) for OR lesion volume and brain atrophy measurements and optical coherence tomography (OCT) for retinal layer thickness measurements. All pweMS were followed with serial OCT and MRI over a median follow-up of 2.9 (interquartile range: 2.6-3.4) years. Eyes with a history of optic neuritis prior to study enrollment were excluded. Linear mixed models were used to analyze the association of retinal layer thinning with changes in OR lesion volume and brain atrophy. RESULTS Macular ganglion cell-inner plexiform layer (GCIPL) thinning was more pronounced in pweMS with OR lesion volume increase during follow-up compared to those without (Difference: -0.82 μm [95% CI:-1.49 to -0.15], p = 0.018). Furthermore, GCIPL thinning correlated with both OR lesion volume increase (β [95% CI] = -0.27 [-0.50 to -0.03], p = 0.028) and brain atrophy (β [95% CI] = 0.47 [0.25 to 0.70], p < 0.001). Correlations of GCIPL changes with brain atrophy did not differ between pweMS with or without OR lesion increase (η p 2 = 5.92e-7 , p = 0.762). INTERPRETATION Faster GCIPL thinning rate is associated with increased OR lesion load. Our results support the value of GCIPL as a sensitive biomarker reflecting both posterior visual pathway pathology and global brain neurodegeneration.
Collapse
Affiliation(s)
- Ting‐Yi Lin
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin BerlinBerlinGermany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Max‐Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | - Claudia Chien
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin BerlinBerlinGermany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Max‐Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Department of Psychiatry and PsychotherapyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu BerlinBerlinGermany
| | - Joseph Kuchling
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin BerlinBerlinGermany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Max‐Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Department of NeurologyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Susanna Asseyer
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin BerlinBerlinGermany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Max‐Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Neuroscience Clinical Research CenterCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Seyedamirhosein Motamedi
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin BerlinBerlinGermany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Max‐Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Neuroscience Clinical Research CenterCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Judith Bellmann‐Strobl
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin BerlinBerlinGermany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Max‐Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Neuroscience Clinical Research CenterCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Tanja Schmitz‐Hübsch
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin BerlinBerlinGermany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Max‐Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Neuroscience Clinical Research CenterCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Klemens Ruprecht
- Department of NeurologyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Alexander U. Brandt
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin BerlinBerlinGermany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Max‐Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | - Hanna G. Zimmermann
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin BerlinBerlinGermany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Max‐Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Neuroscience Clinical Research CenterCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Einstein Center Digital FutureBerlinGermany
| | - Friedemann Paul
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin BerlinBerlinGermany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Max‐Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Department of NeurologyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Neuroscience Clinical Research CenterCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| |
Collapse
|
10
|
Mattiesing RM, Kramer E, Strijbis EMM, Brouwer I, van Schijndel RA, Gentile G, Battaglini M, De Stefano N, Uitdehaag BMJ, Barkhof F, Vrenken H, Schoonheim MM. Disease progression in the first 5 years of treatment in multiple sclerosis: Predictive value of early brain and lesion volume changes. Mult Scler 2024; 30:44-54. [PMID: 38018502 PMCID: PMC10782656 DOI: 10.1177/13524585231212879] [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: 06/28/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Whether the degree of inflammation (and its resolution) and neurodegeneration after treatment initiation predicts disease progression in multiple sclerosis (MS) remains unclear. OBJECTIVES To assess the predictive value of magnetic resonance imaging (MRI)-derived brain and lesion volume (LV) changes in years 1 and 2 of treatment for disease progression. METHODS Patients receiving early interferon beta-1a treatment in REFLEX/REFLEXION (N = 262) were included. Predictive regression models included new/enlarging LV (positive activity), disappearing/shrinking LV (negative activity), and global/central atrophy during years 1 and 2. RESULTS Faster global atrophy and/or pseudo-atrophy and positive lesion activity in years 1 and 2 related to an increased probability and faster conversion to clinically definite multiple sclerosis (CDMS). Negative lesion activity in year 1 and slower central atrophy in year 2 were predictive of confirmed disability progression (9-Hole Peg Test). Positive lesion activity in year 2 was predictive of faster global atrophy, while positive lesion activity in years 1 and 2 was predictive of faster central atrophy. CONCLUSIONS A higher degree of global atrophy and/or pseudo-atrophy in year 1 was predictive of CDMS. Positive lesion activity in any year was related to CDMS and neurodegeneration. Disability was related to negative lesion activity in year 1 and slower central atrophy in year 2.
Collapse
Affiliation(s)
- Rozemarijn M Mattiesing
- MS Center Amsterdam, Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Eline Kramer
- MS Center Amsterdam, Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Eva MM Strijbis
- MS Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Iman Brouwer
- MS Center Amsterdam, Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Ronald A van Schijndel
- MS Center Amsterdam, Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Giordano Gentile
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy/SIENA Imaging SRL, Siena, Italy
| | - Marco Battaglini
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy/SIENA Imaging SRL, Siena, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Bernard MJ Uitdehaag
- MS Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Frederik Barkhof
- MS Center Amsterdam, Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, UK
| | - Hugo Vrenken
- MS Center Amsterdam, Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Menno M Schoonheim
- MS Center Amsterdam, Department of Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Newsome SD, Binns C, Kaunzner UW, Morgan S, Halper J. No Evidence of Disease Activity (NEDA) as a Clinical Assessment Tool for Multiple Sclerosis: Clinician and Patient Perspectives [Narrative Review]. Neurol Ther 2023; 12:1909-1935. [PMID: 37819598 PMCID: PMC10630288 DOI: 10.1007/s40120-023-00549-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023] Open
Abstract
The emergence of high-efficacy therapies for multiple sclerosis (MS), which target inflammation more effectively than traditional disease-modifying therapies, has led to a shift in MS management towards achieving the outcome assessment known as no evidence of disease activity (NEDA). The most common NEDA definition, termed NEDA-3, is a composite of three related measures of disease activity: no clinical relapses, no disability progression, and no radiological activity. NEDA has been frequently used as a composite endpoint in clinical trials, but there is growing interest in its use as an assessment tool to help patients and healthcare professionals navigate treatment decisions in the clinic. Raising awareness about NEDA may therefore help patients and clinicians make more informed decisions around MS management and improve overall MS care. This review aims to explore the potential utility of NEDA as a clinical decision-making tool and treatment target by summarizing the literature on its current use in the context of the expanding treatment landscape. We identify current challenges to the use of NEDA in clinical practice and detail the proposed amendments, such as the inclusion of alternative outcomes and biomarkers, to broaden the clinical information captured by NEDA. These themes are further illustrated with the real-life perspectives and experiences of our two patient authors with MS. This review is intended to be an educational resource to support discussions between clinicians and patients on this evolving approach to MS-specialized care.
Collapse
Affiliation(s)
- Scott D Newsome
- Johns Hopkins University School of Medicine, 600 North Wolfe Street, Pathology 627, Baltimore, MD, 21287, USA.
| | - Cherie Binns
- Multiple Sclerosis Foundation, 6520 N Andrews Avenue, Fort Lauderdale, FL, 33309, USA
| | | | - Seth Morgan
- National Multiple Sclerosis Society, 1 M Street SE, Suite 510, Washington, DC, 20003, USA
| | - June Halper
- Consortium of Multiple Sclerosis Centers, 3 University Plaza Drive Suite A, Hackensack, NJ, 07601, USA
| |
Collapse
|
12
|
Maxwell DL, Orian JM. Cerebellar pathology in multiple sclerosis and experimental autoimmune encephalomyelitis: current status and future directions. J Cent Nerv Syst Dis 2023; 15:11795735231211508. [PMID: 37942276 PMCID: PMC10629308 DOI: 10.1177/11795735231211508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 10/15/2023] [Indexed: 11/10/2023] Open
Abstract
Recent decades have witnessed significant progress in understanding mechanisms driving neurodegeneration and disease progression in multiple sclerosis (MS), but with a focus on the cerebrum. In contrast, there have been limited studies of cerebellar disease, despite the common occurrence of cerebellar symptoms in this disorder. These rare studies, however, highlight the early cerebellar involvement in disease development and an association between the early occurrence of cerebellar lesions and risk of worse prognosis. In parallel developments, it has become evident that far from being a region specialized in movement control, the cerebellum plays a crucial role in cognitive function, via circuitry connecting the cerebellum to association areas of the cerebrum. This complexity, coupled with challenges in imaging of the cerebellum have been major obstacles in the appreciation of the spatio-temporal evolution of cerebellar damage in MS and correlation with disability and progression. MS studies based on animal models have relied on an induced neuroinflammatory disease known as experimental autoimmune encephalomyelitis (EAE), in rodents and non-human primates (NHP). EAE has played a critical role in elucidating mechanisms underpinning tissue damage and been validated for the generation of proof-of-concept for cerebellar pathological processes relevant to MS. Additionally, rodent and NHP studies have formed the cornerstone of current knowledge of functional anatomy and cognitive processes. Here, we propose that improved insight into consequences of cerebellar damage in MS at the functional, cellular and molecular levels would be gained by more extensive characterization of EAE cerebellar pathology combined with the power of experimental paradigms in the field of cognition. Such combinatorial approaches would lead to improved potential for the development of MS sensitive markers and evaluation of candidate therapeutics.
Collapse
Affiliation(s)
- Dain L. Maxwell
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Jacqueline M. Orian
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
13
|
Nold AK, Wittayer M, Weber CE, Platten M, Gass A, Eisele P. Short-term brain atrophy evolution after initiation of immunotherapy in a real-world multiple sclerosis cohort. J Neuroimaging 2023; 33:904-908. [PMID: 37491626 DOI: 10.1111/jon.13146] [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: 07/11/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND AND PURPOSE In multiple sclerosis (MS), brain atrophy measurements have emerged as an important biomarker reflecting neurodegeneration and disability progression. However, due to several potential confounders, investigation of brain atrophy in clinical routine and even in controlled clinical studies can be challenging. The aim of this study was to investigate the short-term dynamics of brain atrophy development after initiation of disease-modifying therapy (DMT) in a "real-world setting." METHODS In this retrospective study, we included MS patients starting DMT (natalizumab, fingolimod, dimethyl fumarate, or interferon-ß1a) or without DMT, availability of a baseline MRI, and two annual follow-up scans on the same MRI system. Two-timepoint percentage brain volume changes (PBVCs) were calculated. RESULTS Fifty-five MS patients (12 patients starting DMT with natalizumab, 7 fingolimod, 14 dimethyl fumarate, 11 interferon-ß1a, and 11 patients without DMT) were included. We found the highest PBVCs in the first 12 months after initiation of natalizumab treatment. Furthermore, the PBVCs in our study were very much comparable to the results observed by other groups, as well as for fingolimod, dimethyl fumarate, and interferon-ß1a. CONCLUSION We found PBVCs that are comparable to the results of previous studies, suggesting that brain atrophy, assessed on 3D MRI data sets acquired on the same 3T MRI, provides a robust MS biomarker.
Collapse
Affiliation(s)
- Ann-Kathrin Nold
- Department of Neurology, Medical Faculty Mannheim and Mannheim Center of Translational Neurosciences (MCTN), Heidelberg University, Mannheim, Germany
| | - Matthias Wittayer
- Department of Neurology, Medical Faculty Mannheim and Mannheim Center of Translational Neurosciences (MCTN), Heidelberg University, Mannheim, Germany
| | - Claudia E Weber
- Department of Neurology, Medical Faculty Mannheim and Mannheim Center of Translational Neurosciences (MCTN), Heidelberg University, Mannheim, Germany
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim and Mannheim Center of Translational Neurosciences (MCTN), Heidelberg University, Mannheim, Germany
| | - Achim Gass
- Department of Neurology, Medical Faculty Mannheim and Mannheim Center of Translational Neurosciences (MCTN), Heidelberg University, Mannheim, Germany
| | - Philipp Eisele
- Department of Neurology, Medical Faculty Mannheim and Mannheim Center of Translational Neurosciences (MCTN), Heidelberg University, Mannheim, Germany
| |
Collapse
|
14
|
Cen S, Gebregziabher M, Moazami S, Azevedo CJ, Pelletier D. Toward precision medicine using a "digital twin" approach: modeling the onset of disease-specific brain atrophy in individuals with multiple sclerosis. Sci Rep 2023; 13:16279. [PMID: 37770560 PMCID: PMC10539386 DOI: 10.1038/s41598-023-43618-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/26/2023] [Indexed: 09/30/2023] Open
Abstract
Digital Twin (DT) is a novel concept that may bring a paradigm shift for precision medicine. In this study we demonstrate a DT application for estimating the age of onset of disease-specific brain atrophy in individuals with multiple sclerosis (MS) using brain MRI. We first augmented longitudinal data from a well-fitted spline model derived from a large cross-sectional normal aging data. Then we compared different mixed spline models through both simulated and real-life data and identified the mixed spline model with the best fit. Using the appropriate covariate structure selected from 52 different candidate structures, we augmented the thalamic atrophy trajectory over the lifespan for each individual MS patient and a corresponding hypothetical twin with normal aging. Theoretically, the age at which the brain atrophy trajectory of an MS patient deviates from the trajectory of their hypothetical healthy twin can be considered as the onset of progressive brain tissue loss. With a tenfold cross validation procedure through 1000 bootstrapping samples, we found the onset age of progressive brain tissue loss was, on average, 5-6 years prior to clinical symptom onset. Our novel approach also discovered two clear patterns of patient clusters: earlier onset versus simultaneous onset of brain atrophy.
Collapse
Affiliation(s)
- Steven Cen
- Department of Radiology/Neurology, University of Southern California, Los Angeles, USA.
| | - Mulugeta Gebregziabher
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, USA
| | - Saeed Moazami
- Department of Aerospace and Mechanical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, USA
| | - Christina J Azevedo
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Daniel Pelletier
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| |
Collapse
|
15
|
Donatelli G, Cecchi P, Migaleddu G, Cencini M, Frumento P, D'Amelio C, Peretti L, Buonincontri G, Pasquali L, Tosetti M, Cosottini M, Costagli M. Quantitative T1 mapping detects blood-brain barrier breakdown in apparently non-enhancing multiple sclerosis lesions. Neuroimage Clin 2023; 40:103509. [PMID: 37717382 PMCID: PMC10514220 DOI: 10.1016/j.nicl.2023.103509] [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: 06/21/2023] [Revised: 08/09/2023] [Accepted: 09/10/2023] [Indexed: 09/19/2023]
Abstract
OBJECTIVES The disruption of the blood-brain barrier (BBB) is a key and early feature in the pathogenesis of demyelinating multiple sclerosis (MS) lesions and has been neuropathologically demonstrated in both active and chronic plaques. The local overt BBB disruption in acute demyelinating lesions is captured as signal hyperintensity in post-contrast T1-weighted images because of the contrast-related shortening of the T1 relaxation time. On the contrary, the subtle BBB disruption in chronic lesions is not visible at conventional radiological evaluation but it might be of clinical relevance. Indeed, persistent, subtle BBB leakage might be linked to low-grade inflammation and plaque evolution. Here we hypothesised that 3D Quantitative Transient-state Imaging (QTI) was able to reveal and measure T1 shortening (ΔT1) reflecting small amounts of contrast media leakage in apparently non-enhancing lesions (ANELs). MATERIALS AND METHODS Thirty-four patients with relapsing remitting MS were included in the study. All patients underwent a 3 T MRI exam of the brain including conventional sequences and QTI acquisitions (1.1 mm isotropic voxel) performed both before and after contrast media administration. For each patient, a ΔT1 map was obtained via voxel-wise subtraction of pre- and post- contrast QTI-derived T1 maps. ΔT1 values measured in ANELs were compared with those recorded in enhancing lesions and in the normal appearing white matter. A reference distribution of ΔT1 in the white matter was obtained from datasets acquired in 10 non-MS patients with unrevealing MR imaging. RESULTS Mean ΔT1 in ANELs (57.45 ± 48.27 ms) was significantly lower than in enhancing lesions (297.71 ± 177.52 ms; p < 0. 0001) and higher than in the normal appearing white matter (36.57 ± 10.53 ms; p < 0.005). Fifty-two percent of ANELs exhibited ΔT1 higher than those observed in the white matter of non-MS patients. CONCLUSIONS QTI-derived quantitative ΔT1 mapping enabled to measure contrast-related T1 shortening in ANELs. ANELs exhibiting ΔT1 values that deviate from the reference distribution in non-MS patients may indicate persistent, subtle, BBB disruption. Access to this information may be proved useful to better characterise pathology and objectively monitor disease activity and response to therapy.
Collapse
Affiliation(s)
- Graziella Donatelli
- Neuroradiology Unit, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; Imago7 Research Foundation, Pisa, Italy
| | - Paolo Cecchi
- Neuroradiology Unit, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; Imago7 Research Foundation, Pisa, Italy
| | | | - Matteo Cencini
- National Institute for Nuclear Physics (INFN), Pisa Division, Pisa, Italy
| | - Paolo Frumento
- Department of Political Sciences, University of Pisa, Pisa, Italy
| | - Claudio D'Amelio
- Neuroradiology Unit, Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Luca Peretti
- Imago7 Research Foundation, Pisa, Italy; Laboratory of Medical Physics and Magnetic Resonance, IRCCS Stella Maris, Pisa, Italy
| | - Guido Buonincontri
- Laboratory of Medical Physics and Magnetic Resonance, IRCCS Stella Maris, Pisa, Italy
| | - Livia Pasquali
- Neurology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Michela Tosetti
- Laboratory of Medical Physics and Magnetic Resonance, IRCCS Stella Maris, Pisa, Italy
| | - Mirco Cosottini
- Neuroradiology Unit, Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.
| | - Mauro Costagli
- Laboratory of Medical Physics and Magnetic Resonance, IRCCS Stella Maris, Pisa, Italy; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Sciences (DINOGMI), University of Genoa, Genoa, Italy
| |
Collapse
|
16
|
Conway Kleven BD, Chien LC, Labus B, Cross CL, Ritter A, Randall R, Montes A, Bernick C. Longitudinal Changes in Regional Brain Volumes and Cognition of Professional Fighters With Traumatic Encephalopathy Syndrome. Neurology 2023; 101:e1118-e1126. [PMID: 37380429 PMCID: PMC10513890 DOI: 10.1212/wnl.0000000000207594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/12/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Due to current limitations in diagnosing chronic traumatic encephalopathy (CTE) clinically, traumatic encephalopathy syndrome (TES) has been proposed as the clinical presentation of suspected CTE. This study aimed to determine whether there was an association between a clinical diagnosis of TES and subsequent temporal decline in cognitive or MRI volumetric measures. METHODS This was a secondary analysis of the Professional Athletes Brain Health Study (PABHS), inclusive of active and retired professional fighters older than 34 years. All athletes were adjudicated as TES positive (TES+) or TES negative (TES-) based on the 2021 clinical criteria. General linear mixed models were used to compare MRI regional brain volumes and cognitive performance between groups. RESULTS A total of 130 fighters met inclusion criteria for consensus conference. Of them, 52 fighters (40%) were adjudicated as TES+. Athletes with a TES+ diagnosis were older and had significantly lower education. Statistically significant interactions and between-group total mean differences were found in all MRI volumetric measurements among the TES+ group compared with those among the TES- group. The rate of volumetric change indicated a significantly greater increase for lateral (estimate = 5,196.65; 95% CI = 2642.65, 7750.66) and inferior lateral ventricles (estimate = 354.28; 95% CI = 159.90, 548.66) and a decrease for the hippocampus (estimate = -385.04, 95% CI = -580.47, -189.62), subcortical gray matter (estimate = -4,641.08; 95% CI = -6783.98, -2498.18), total gray matter (estimate = -26492.00; 95% CI = -50402.00, -2582.32), and posterior corpus callosum (estimate = -147.98; 95% CI = -222.33, -73.62). Likewise, the rate of cognitive decline was significantly greater for reaction time (estimate = 56.31; 95% CI = 26.17, 86.45) and other standardized cognitive scores in the TES+ group. DISCUSSION The 2021 TES criteria clearly distinguishes group differences in the longitudinal presentation of volumetric loss in select brain regions and cognitive decline among professional fighters 35 years and older. This study suggests that a TES diagnosis may be useful in professional sports beyond football, such as boxing and mixed martial arts. These findings further suggest that the application of TES criteria may be valuable clinically in predicting cognitive decline.
Collapse
Affiliation(s)
- Brooke D Conway Kleven
- From the School of Public Health (B.D.C.K., L.-C.C., B.L., C.L.C., A.M.), University of Nevada, Las Vegas; and Cleveland Clinic Lou Ruvo Center for Brain Health (A.R., R.R., A.M., C.B.), Las Vegas, NV.
| | - Lung-Chang Chien
- From the School of Public Health (B.D.C.K., L.-C.C., B.L., C.L.C., A.M.), University of Nevada, Las Vegas; and Cleveland Clinic Lou Ruvo Center for Brain Health (A.R., R.R., A.M., C.B.), Las Vegas, NV
| | - Brian Labus
- From the School of Public Health (B.D.C.K., L.-C.C., B.L., C.L.C., A.M.), University of Nevada, Las Vegas; and Cleveland Clinic Lou Ruvo Center for Brain Health (A.R., R.R., A.M., C.B.), Las Vegas, NV
| | - Chad L Cross
- From the School of Public Health (B.D.C.K., L.-C.C., B.L., C.L.C., A.M.), University of Nevada, Las Vegas; and Cleveland Clinic Lou Ruvo Center for Brain Health (A.R., R.R., A.M., C.B.), Las Vegas, NV
| | - Aaron Ritter
- From the School of Public Health (B.D.C.K., L.-C.C., B.L., C.L.C., A.M.), University of Nevada, Las Vegas; and Cleveland Clinic Lou Ruvo Center for Brain Health (A.R., R.R., A.M., C.B.), Las Vegas, NV
| | - Rebekah Randall
- From the School of Public Health (B.D.C.K., L.-C.C., B.L., C.L.C., A.M.), University of Nevada, Las Vegas; and Cleveland Clinic Lou Ruvo Center for Brain Health (A.R., R.R., A.M., C.B.), Las Vegas, NV
| | - Arturo Montes
- From the School of Public Health (B.D.C.K., L.-C.C., B.L., C.L.C., A.M.), University of Nevada, Las Vegas; and Cleveland Clinic Lou Ruvo Center for Brain Health (A.R., R.R., A.M., C.B.), Las Vegas, NV
| | - Charles Bernick
- From the School of Public Health (B.D.C.K., L.-C.C., B.L., C.L.C., A.M.), University of Nevada, Las Vegas; and Cleveland Clinic Lou Ruvo Center for Brain Health (A.R., R.R., A.M., C.B.), Las Vegas, NV
| |
Collapse
|
17
|
Pogoda-Wesołowska A, Dziedzic A, Maciak K, Stȩpień A, Dziaduch M, Saluk J. Neurodegeneration and its potential markers in the diagnosing of secondary progressive multiple sclerosis. A review. Front Mol Neurosci 2023; 16:1210091. [PMID: 37781097 PMCID: PMC10535108 DOI: 10.3389/fnmol.2023.1210091] [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: 04/24/2023] [Accepted: 08/25/2023] [Indexed: 10/03/2023] Open
Abstract
Approximately 70% of relapsing-remitting multiple sclerosis (RRMS) patients will develop secondary progressive multiple sclerosis (SPMS) within 10-15 years. This progression is characterized by a gradual decline in neurological functionality and increasing limitations of daily activities. Growing evidence suggests that both inflammation and neurodegeneration are associated with various pathological processes throughout the development of MS; therefore, to delay disease progression, it is critical to initiate disease-modifying therapy as soon as it is diagnosed. Currently, a diagnosis of SPMS requires a retrospective assessment of physical disability exacerbation, usually over the previous 6-12 months, which results in a delay of up to 3 years. Hence, there is a need to identify reliable and objective biomarkers for predicting and defining SPMS conversion. This review presents current knowledge of such biomarkers in the context of neurodegeneration associated with MS, and SPMS conversion.
Collapse
Affiliation(s)
| | - Angela Dziedzic
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Karina Maciak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Adam Stȩpień
- Clinic of Neurology, Military Institute of Medicine–National Research Institute, Warsaw, Poland
| | - Marta Dziaduch
- Medical Radiology Department of Military Institute of Medicine – National Research Institute, Warsaw, Poland
| | - Joanna Saluk
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| |
Collapse
|
18
|
Loomis SJ, Sadhu N, Fisher E, Gafson AR, Huang Y, Yang C, Hughes EE, Marshall E, Herman A, John S, Runz H, Jia X, Bhangale T, Bronson PG. Genome-wide study of longitudinal brain imaging measures of multiple sclerosis progression across six clinical trials. Sci Rep 2023; 13:14313. [PMID: 37652990 PMCID: PMC10471679 DOI: 10.1038/s41598-023-41099-0] [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: 03/07/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023] Open
Abstract
While the genetics of MS risk susceptibility are well-described, and recent progress has been made on the genetics of disease severity, the genetics of disease progression remain elusive. We therefore investigated the genetic determinants of MS progression on longitudinal brain MRI: change in brain volume (BV) and change in T2 lesion volume (T2LV), reflecting progressive tissue loss and increasing disease burden, respectively. We performed genome-wide association studies of change in BV (N = 3401) and change in T2LV (N = 3513) across six randomized clinical trials from Biogen and Roche/Genentech: ADVANCE, ASCEND, DECIDE, OPERA I & II, and ORATORIO. Analyses were adjusted for randomized treatment arm, age, sex, and ancestry. Results were pooled in a meta-analysis, and were evaluated for enrichment of MS risk variants. Variant colocalization and cell-specific expression analyses were performed using published cohorts. The strongest peaks were in PTPRD (rs77321193-C/A, p = 3.9 × 10-7) for BV change, and NEDD4L (rs11398377-GC/G, p = 9.3 × 10-8) for T2LV change. Evidence of colocalization was observed for NEDD4L, and both genes showed increased expression in neuronal and/or glial populations. No association between MS risk variants and MRI outcomes was observed. In this unique, precompetitive industry partnership, we report putative regions of interest in the neurodevelopmental gene PTPRD, and the ubiquitin ligase gene NEDD4L. These findings are distinct from known MS risk genetics, indicating an added role for genetic progression analyses and informing drug discovery.
Collapse
|
19
|
Chylińska M, Komendziński J, Wyszomirski A, Karaszewski B. Brain Atrophy as an Outcome of Disease-Modifying Therapy for Remitting-Relapsing Multiple Sclerosis. Mult Scler Int 2023; 2023:4130557. [PMID: 37693228 PMCID: PMC10484652 DOI: 10.1155/2023/4130557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/21/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction Currently, clinical trials of DMTs strive to determine their effect on neuroinflammation and neurodegeneration. We aimed to determine the impact of currently used DMTs on brain atrophy and disability in RRMS. The main goal of this review is to evaluate the neuroprotective potential of MS therapy and assess its impact on disability. Methods We performed a systematic analysis of clinical trials that used brain atrophy as an outcome or performed post hoc analysis of volumetric MRI parameters to assess the neuroprotective potential of applied therapies. Trials between 2008 and 2019 that included published results of brain parenchymal fraction (BPF) change and brain volume loss (BVL) in the period from baseline to week 96 or longer were considered. Results Twelve from 146 clinical trials met the inclusion criteria and were incorporated into the analysis. DMTs that presented a large reduction in BVL also exhibited robust effects on clinical disability worsening, e.g., alemtuzumab with a 42% risk reduction in 6-month confirmed disability accumulation (p = 0.0084), ocrelizumab with a 40% risk reduction in 6-month confirmed disability progression (p = 0.003), and other DMTs (cladribine and teriflunomide) with moderate influence on brain atrophy were also associated with a marked impact on disability worsening. Dimethyl fumarate (DEFINE) and fingolimod (FREEDOMS I) initially exhibited significant effect on BVL; however, this effect was not confirmed in further clinical trials: CONFIRM and FREEDOMS II, respectively. Peg-IFN-β1a shows a modest effect on BVL and disability worsening. Conclusion Our results show that BVL in one of the components of clinical disability worsening, together with other variables (lesion volume and annualized relapse rate). Standardization of atrophy measurement technique as well as harmonization of disability worsening and progression criteria in further clinical trials are of utmost importance as they enable a reliable comparison of neuroprotective potential of DMTs.
Collapse
Affiliation(s)
| | - Jakub Komendziński
- Department of Adult Neurology, Gdańsk Medical University, Gdańsk, Poland
| | - Adam Wyszomirski
- Department of Adult Neurology, Gdańsk Medical University, Gdańsk, Poland
| | | |
Collapse
|
20
|
McGuire J, Muto C, Marcello C. Multiple sclerosis: Implications for the primary care NP. Nurse Pract 2023; 48:38-47. [PMID: 37487047 DOI: 10.1097/01.npr.0000000000000083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
ABSTRACT Multiple sclerosis is a demyelinating disease of the central nervous system. It contributes to a variety of symptoms affecting different areas of the body. The primary care NP must be familiar with the disease, therapies, and social impact to provide proper care to affected patients.
Collapse
|
21
|
Zhan G, Wang D, Cabezas M, Bai L, Kyle K, Ouyang W, Barnett M, Wang C. Learning from pseudo-labels: deep networks improve consistency in longitudinal brain volume estimation. Front Neurosci 2023; 17:1196087. [PMID: 37483345 PMCID: PMC10358358 DOI: 10.3389/fnins.2023.1196087] [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: 03/29/2023] [Accepted: 06/16/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction Brain atrophy is a critical biomarker of disease progression and treatment response in neurodegenerative diseases such as multiple sclerosis (MS). Confounding factors such as inconsistent imaging acquisitions hamper the accurate measurement of brain atrophy in the clinic. This study aims to develop and validate a robust deep learning model to overcome these challenges; and to evaluate its impact on the measurement of disease progression. Methods Voxel-wise pseudo-atrophy labels were generated using SIENA, a widely adopted tool for the measurement of brain atrophy in MS. Deformation maps were produced for 195 pairs of longitudinal 3D T1 scans from patients with MS. A 3D U-Net, namely DeepBVC, was specifically developed overcome common variances in resolution, signal-to-noise ratio and contrast ratio between baseline and follow up scans. The performance of DeepBVC was compared against SIENA using McLaren test-retest dataset and 233 in-house MS subjects with MRI from multiple time points. Clinical evaluation included disability assessment with the Expanded Disability Status Scale (EDSS) and traditional imaging metrics such as lesion burden. Results For 3 subjects in test-retest experiments, the median percent brain volume change (PBVC) for DeepBVC and SIENA was 0.105 vs. 0.198% (subject 1), 0.061 vs. 0.084% (subject 2), 0.104 vs. 0.408% (subject 3). For testing consistency across multiple time points in individual MS subjects, the mean (± standard deviation) PBVC difference of DeepBVC and SIENA were 0.028% (± 0.145%) and 0.031% (±0.154%), respectively. The linear correlation with baseline T2 lesion volume were r = -0.288 (p < 0.05) and r = -0.249 (p < 0.05) for DeepBVC and SIENA, respectively. There was no significant correlation of disability progression with PBVC as estimated by either method (p = 0.86, p = 0.84). Discussion DeepBVC is a deep learning powered brain volume change estimation method for assessing brain atrophy used T1-weighted images. Compared to SIENA, DeepBVC demonstrates superior performance in reproducibility and in the context of common clinical scan variances such as imaging contrast, voxel resolution, random bias field, and signal-to-noise ratio. Enhanced measurement robustness, automation, and processing speed of DeepBVC indicate its potential for utilisation in both research and clinical environments for monitoring disease progression and, potentially, evaluating treatment effectiveness.
Collapse
Affiliation(s)
- Geng Zhan
- Brain and Mind Center, The University of Sydney, Sydney, NSW, Australia
- Sydney Neuroimaging Analysis Center, Sydney, NSW, Australia
| | - Dongang Wang
- Brain and Mind Center, The University of Sydney, Sydney, NSW, Australia
- Sydney Neuroimaging Analysis Center, Sydney, NSW, Australia
| | - Mariano Cabezas
- Brain and Mind Center, The University of Sydney, Sydney, NSW, Australia
| | - Lei Bai
- Shanghai AI Laboratory, Shanghai, China
| | - Kain Kyle
- Brain and Mind Center, The University of Sydney, Sydney, NSW, Australia
- Sydney Neuroimaging Analysis Center, Sydney, NSW, Australia
| | | | - Michael Barnett
- Brain and Mind Center, The University of Sydney, Sydney, NSW, Australia
- Sydney Neuroimaging Analysis Center, Sydney, NSW, Australia
| | - Chenyu Wang
- Brain and Mind Center, The University of Sydney, Sydney, NSW, Australia
- Sydney Neuroimaging Analysis Center, Sydney, NSW, Australia
| |
Collapse
|
22
|
Lorefice L, Mellino P, Fenu G, Cocco E. How to measure the treatment response in progressive multiple sclerosis: Current perspectives and limitations in clinical settings'. Mult Scler Relat Disord 2023; 76:104826. [PMID: 37327601 DOI: 10.1016/j.msard.2023.104826] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/04/2023] [Accepted: 06/09/2023] [Indexed: 06/18/2023]
Abstract
New treatment options are available for active progressive multiple sclerosis (MS), including primary and secondary progressive forms. Several pieces of evidence have recently suggested a "window of beneficial treatment opportunities," principally in the early stages of progression. However, for progressive MS, which is characterised by an inevitable tendency to get worse, it is crucial to redefine the "response to treatment" beyond the concept of "no evidence of disease activity" (NEDA-3), which was initially conceived to evaluate disease outcomes in relapsing-remitting form, albeit it is currently applied to all MS cases in clinical practice. This review examines the current perspectives and limitations in assessing the effectiveness of DMTs and disease outcomes in progressive MS, the current criteria applied in defining the response to DMTs, and the strengths and limitations of clinical scales and tools for evaluating MS evolution and patient perception. Additionally, the impact of age and comorbidities on the assessment of MS outcomes was examined.
Collapse
Affiliation(s)
- L Lorefice
- Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Address: via Is Guadazzonis 2, Cagliari 09126, Italy.
| | - P Mellino
- Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Address: via Is Guadazzonis 2, Cagliari 09126, Italy
| | - G Fenu
- Department of Neurosciences, ARNAS Brotzu, Cagliari, Italy
| | - E Cocco
- Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Address: via Is Guadazzonis 2, Cagliari 09126, Italy
| |
Collapse
|
23
|
Matthews PM, Gupta D, Mittal D, Bai W, Scalfari A, Pollock KG, Sharma V, Hill N. The association between brain volume loss and disability in multiple sclerosis: A systematic review. Mult Scler Relat Disord 2023; 74:104714. [PMID: 37068369 DOI: 10.1016/j.msard.2023.104714] [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: 12/19/2022] [Revised: 03/23/2023] [Accepted: 04/08/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic, inflammatory, demyelinating, degenerative disease of the central nervous system that affects approximately 2.8 million people worldwide. Compelling evidence from observational studies and clinical trials indicates a strong association between brain volume loss (BVL) and the accumulation of disability in MS. However, the considerable heterogeneity in study designs and methods of assessment of BVL invites questions concerning the generalizability of the reported findings. Therefore, we conducted this systematic review to characterize the relationship between BVL and physical disability in patients with MS. METHODS A systematic literature search of MEDLINE and EMBASE databases was performed supplemented by gray literature searches. The following study designs were included: prospective/retrospective cohort, cross-sectional and case-control. Only English language articles published from 2010 onwards were eligible for final inclusion. There were no restrictions on MS subtype, age, or ethnicity. Of the 1620 citations retrieved by the structured searches, 50 publications met our screening criteria and were included in the final data set. RESULTS Across all BVL measures, there was considerable heterogeneity in studies regarding the underlying study population, the definitions of BVL and image analysis methodologies, the physical disability measure used, the measures of association reported and whether the analysis conducted was univariable or multivariable. A total of 36 primary studies providing data on the association between whole BVL and physical disability in MS collectively suggest that whole brain atrophy is associated with greater physical disability progression in MS patients. Similarly, a total of 15 primary studies providing data on the association between ventricular atrophy and physical disability in MS suggest that ventricular atrophy is associated with greater physical disability progression in MS patients. Along similar lines, the existing evidence based on a total of 13 primary studies suggests that gray matter atrophy is associated with greater physical disability progression in MS patients. Four primary studies suggest that corpus callosum atrophy is associated with greater physical disability progression in MS patients. The majority of the existing evidence (6 primary studies) suggests no association between white matter atrophy and physical disability in MS. It is difficult to assign a relationship between basal ganglia volume loss and physical disability as well as medulla oblongata width and physical disability in MS due to very limited data. CONCLUSION The evidence gathered from this systematic review, although very heterogeneous, suggests that whole brain atrophy is associated with greater physical disability progression in MS patients. Our review can help define future imaging biomarkers for physical disability progression and treatment monitoring in MS.
Collapse
Affiliation(s)
- Paul M Matthews
- Department of Brain Sciences and UK Dementia Research Institute at Imperial College London, Burlington Danes Building, Hammersmith Hospital, DuCane Road, London, UK.
| | - Digant Gupta
- Bridge Medical Consulting Limited, 2 Marsault Court, 11 Kew Foot Road, Richmond, London, TW9 2SS, UK
| | - Deepali Mittal
- Bridge Medical Consulting Limited, 2 Marsault Court, 11 Kew Foot Road, Richmond, London, TW9 2SS, UK
| | - Wenjia Bai
- Department of Brain Sciences and UK Dementia Research Institute at Imperial College London, Burlington Danes Building, Hammersmith Hospital, DuCane Road, London, UK; Department of Computing, Imperial College London, William Penny Building, South Kensington Campus, London, UK
| | - Antonio Scalfari
- Imperial College Healthcare Trust, Centre of Neuroscience, Department of Medicine, Charing Cross Hospital, Fulham Palace Rd, London W6 8RF, UK
| | - Kevin G Pollock
- Bristol-Myers Squibb, Uxbridge Business Park, Sanderson Road, Uxbridge, UB8 1DH, UK
| | - Vishal Sharma
- Bristol-Myers Squibb, Uxbridge Business Park, Sanderson Road, Uxbridge, UB8 1DH, UK
| | - Nathan Hill
- Bristol-Myers Squibb, Uxbridge Business Park, Sanderson Road, Uxbridge, UB8 1DH, UK
| |
Collapse
|
24
|
Gentile G, Mattiesing RM, Brouwer I, van Schijndel RA, Uitdehaag BMJ, Twisk JWR, Kappos L, Freedman MS, Comi G, Jack D, Barkhof F, De Stefano N, Vrenken H, Battaglini M. The spatio-temporal relationship between concurrent lesion and brain atrophy changes in early multiple sclerosis: A post-hoc analysis of the REFLEXION study. Neuroimage Clin 2023; 38:103397. [PMID: 37086648 PMCID: PMC10300577 DOI: 10.1016/j.nicl.2023.103397] [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: 12/22/2022] [Revised: 03/30/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND White matter (WM) lesions and brain atrophy are present early in multiple sclerosis (MS). However, their spatio-temporal relationship remains unclear. METHODS Yearly magnetic resonance images were analysed in 387 patients with a first clinical demyelinating event (FCDE) from the 5-year REFLEXION study. Patients received early (from baseline; N = 258; ET) or delayed treatment (from month-24; N = 129; DT) with subcutaneous interferon beta-1a. FSL-SIENA/VIENA were used to provide yearly percentage volume change of brain (PBVC) and ventricles (PVVC). Yearly total lesion volume change (TLVC) was determined by a semi-automated method. Using linear mixed models and voxel-wise analyses, we firstly investigated the overall relationship between TLVC and PBVC and between TLVC and PVVC in the same follow-up period. Analyses were then separately performed for: the untreated period of DT patients (first two years), the first year of treatment (year 1 for ET and year 3 for DT), and a period where patients had received at least 1 year of treatment (stable treatment; ET: years 2, 3, 4, and 5; DT: years 4 and 5). RESULTS Whole brain: across the whole study period, lower TLVC was related to faster atrophy (PBVC: B = 0.046, SE = 0.013, p < 0.001; PVVC: B = -0.466, SE = 0.118, p < 0.001). Within the untreated period of DT patients, lower TLVC was related to faster atrophy (PBVC: B = 0.072, SE = 0.029, p = 0.013; PVVC: B = -0.917, SE = 0.306, p = 0.003). A similar relationship was found within the first year of treatment of ET patients (PBVC: B = 0.081, SE = 0.027, p = 0.003; PVVC: B = -1.08, SE = 0.284, p < 0.001), consistent with resolving oedema and pseudo-atrophy. Voxel-wise: overall, higher TLVC was related to faster ventricular enlargement. Lower TLVC was related to faster widespread atrophy in year 1 in both ET (first year of treatment) and DT (untreated) patients. In the second untreated year of DT patients and within the stable treatment period of ET patients (year 4), faster periventricular and occipital lobe atrophy was associated with higher TLVC. CONCLUSIONS WM lesion changes and atrophy occurred simultaneously in early MS. Spatio-temporal correspondence of these two processes involved mostly the periventricular area. Within the first year of the study, in both treatment groups, faster atrophy was linked to lower lesion volume changes, consistent with higher shrinking and disappearing lesion activity. This might reflect the pseudo-atrophy phenomenon that is probably related to the therapy driven (only in ET patients, as they received treatment from baseline) and "natural" (both ET and DT patients entered the study after a FCDE) resolution of oedema. In an untreated period and later on during stable treatment, (real) atrophy was related to higher lesion volume changes, consistent with increased new and enlarging lesion activity.
Collapse
Affiliation(s)
- Giordano Gentile
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy; Siena Imaging SRL, 53100 Siena, Italy.
| | - Rozemarijn M Mattiesing
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, De Boelelaan 1118, 1081 HZ Amsterdam, the Netherlands
| | - Iman Brouwer
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, De Boelelaan 1118, 1081 HZ Amsterdam, the Netherlands
| | - Ronald A van Schijndel
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, De Boelelaan 1118, 1081 HZ Amsterdam, the Netherlands
| | - Bernard M J Uitdehaag
- MS Center Amsterdam, Neurology, Amsterdam Neuroscience, Amsterdam UMC location VUmc, De Boelelaan 1118, 1081 HZ Amsterdam, the Netherlands
| | - Jos W R Twisk
- Epidemiology and Data Science, Amsterdam UMC location VUmc, De Boelelaan 1118, 1081 HZ Amsterdam, the Netherlands
| | - Ludwig Kappos
- Research Center for Clinical Neuroimmunology, and Neuroscience Basel (RC2NB), University Hospital Basel, CH-4031 Basel, Switzerland; Neurology Departments of Head, Spine and Neuromedicine, Biomedical Engineering and Clinical Research, University of Basel, Basel, Switzerland
| | - Mark S Freedman
- Department of Medicine, University of Ottawa, Ottawa ON, K1N 6N5, Ontario, Canada; Ottawa Hospital Research Institute, Ottawa ON, K1H 8L6, Ontario, Canada
| | - Giancarlo Comi
- Università Vita Salute San Raffaele, Casa di Cura del Policlinico, 20132 Milan, Italy
| | - Dominic Jack
- Merck Serono Ltd, Feltham, TW14 8HD, UK, an affiliate of Merck KGaA
| | - Frederik Barkhof
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, De Boelelaan 1118, 1081 HZ Amsterdam, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering, London, WC1E 6BT, UK
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Hugo Vrenken
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, De Boelelaan 1118, 1081 HZ Amsterdam, the Netherlands
| | - Marco Battaglini
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy; Siena Imaging SRL, 53100 Siena, Italy
| |
Collapse
|
25
|
Bourre B, Casez O, Ciron J, Gueguen A, Kwiatkowski A, Moisset X, Montcuquet A, Ayrignac X. Paradigm shifts in multiple sclerosis management: Implications for daily clinical practice. Rev Neurol (Paris) 2023; 179:256-264. [PMID: 36621364 DOI: 10.1016/j.neurol.2022.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 01/09/2023]
Abstract
Multiple sclerosis (MS) is the most common chronic inflammatory neurological disease. The emergence of disease-modifying therapies (DMTs) has greatly improved disease activity control and progression of disability in MS patients. DMTs differ in their mode of action, route of administration, efficacy, and safety profiles, offering multiple options for clinicians. Personalized medicine aims at tailoring the therapeutic strategy to patients' characteristics and disease activity but also patients' needs and preferences. New therapeutic options have already changed treatment paradigms for patients with active relapsing MS (RMS). The traditional approach consists in initiating treatment with moderate-efficacy DMTs and subsequently, escalating to higher-efficacy DMTs when there is evidence of clinical and/or radiological breakthrough activity. Recent real-world studies suggest that initiation of high-efficacy DMTs from disease onset can improve long-term outcomes for RMS patients. In this article, we review different treatment strategies and discuss challenges associated with personalized therapy.
Collapse
Affiliation(s)
- B Bourre
- Rouen University Hospital, Rouen, France.
| | - O Casez
- Pathologies Inflammatoires du Système Nerveux, Neurologie, Department of Neurology, CRC-SEP, CHU of Grenoble-Alpes and T-RAIG (Translational Research in Autoimmunity and Inflammation Group), University of Grenoble-Alpes, Rouen, France
| | - J Ciron
- Toulouse University Hospital, Toulouse, France
| | - A Gueguen
- Department of Neurology, Rothschild Foundation, Paris, France
| | - A Kwiatkowski
- Department of Neurology, Lille Catholic University, Lille Catholic Hospitals, Lille, France
| | - X Moisset
- Inserm, NEURODOL, CHU of Clermont-Ferrand, University of Clermont Auvergne, Clermont-Ferrand, France
| | - A Montcuquet
- Department of Neurology, CHU of Limoges, Limoges, France
| | - X Ayrignac
- Inserm, INM, Department of Neurology, MS Center and National Reference Center of Adult Leukodystrophies, University of Montpellier, Montpellier University Hospital, Montpellier, France
| |
Collapse
|
26
|
Skorve E, Lundervold AJ, Torkildsen Ø, Riemer F, Grüner R, Myhr KM. Brief international cognitive assessment for MS (BICAMS) and global brain volumes in early stages of MS - A longitudinal correlation study. Mult Scler Relat Disord 2023; 69:104398. [PMID: 36462469 DOI: 10.1016/j.msard.2022.104398] [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: 12/27/2021] [Revised: 08/04/2022] [Accepted: 11/03/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Cognitive impairment is common in patients with multiple sclerosis, even in the early stages of the disease. The Brief International Cognitive Assessment for multiple sclerosis (BICAMS) is a short screening tool developed to assess cognitive function in everyday clinical practice. OBJECTIVE To investigate associations between volumetric brain measures derived from a magnetic resonance imaging (MRI) examination and performance on BICAMS subtests in early stages of multiple sclerosis (MS). METHODS BICAMS was used to assess cognitive function in 49 MS patients at baseline and after one and two years. The patients were separated into two groups (with or without cognitive impairment) based on their performances on BICAMSs subtests. MRI data were analysed by a software tool (MSMetrix), yielding normalized measures of global brain volumes and lesion volumes. Associations between cognitive tests and brain MRI measures were analysed by running correlation analyses, and differences between subgroups and changes over time with independent and paired samples tests, respectively. RESULTS The strongest baseline correlations were found between the BICAMS subtests and normalized whole brain volume (NBV) and grey matter volume (NGV); processing speed r = 0.54/r = 0.48, verbal memory r = 0.49/ r = 0.42, visual memory r = 0.48 /r = 0.39. Only the verbal memory test had significant correlations with T2 and T1 lesion volumes (LV) at both time points; T2LV r = 0.39, T1LV r = 0.38. There were significant loss of grey matter and white matter volume overall (NGV p<0.001, NWV p = 0.003), as well as an increase in T1LV (p = 0.013). The longitudinally defined confirmed cognitively impaired (CCI) and preserved (CCP) patients showed significant group differences on all MRI volume measures at both time points, except for NWV. Only the CCI subgroup showed significant white matter atrophy (p = 0.006) and increase in T2LV (p = 0.029). CONCLUSIONS The present study found strong correlations between whole brain and grey matter volumes and performance on the BICAMS subtests as well as significant changes in global volumes from baseline to follow-up with clear differences between patients defined as cognitively impaired and preserved at both baseline and follow-up.
Collapse
Affiliation(s)
- Ellen Skorve
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Astri J Lundervold
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Øivind Torkildsen
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Frank Riemer
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Renate Grüner
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway; Department of Physics and Technology, University of Bergen, N-5007 Bergen, Norway
| | - Kjell-Morten Myhr
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
27
|
Mendelsohn Z, Pemberton HG, Gray J, Goodkin O, Carrasco FP, Scheel M, Nawabi J, Barkhof F. Commercial volumetric MRI reporting tools in multiple sclerosis: a systematic review of the evidence. Neuroradiology 2023; 65:5-24. [PMID: 36331588 PMCID: PMC9816195 DOI: 10.1007/s00234-022-03074-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE MRI is integral to the diagnosis of multiple sclerosis (MS) and is important for clinical prognostication. Quantitative volumetric reporting tools (QReports) can improve the accuracy and objectivity of MRI-based assessments. Several QReports are commercially available; however, validation can be difficult to establish and does not currently follow a common pathway. To aid evidence-based clinical decision-making, we performed a systematic review of commercial QReports for use in MS including technical details and published reports of validation and in-use evaluation. METHODS We categorized studies into three types of testing: technical validation, for example, comparison to manual segmentation, clinical validation by clinicians or interpretation of results alongside clinician-rated variables, and in-use evaluation, such as health economic assessment. RESULTS We identified 10 companies, which provide MS lesion and brain segmentation and volume quantification, and 38 relevant publications. Tools received regulatory approval between 2006 and 2020, contextualize results to normative reference populations, ranging from 620 to 8000 subjects, and require T1- and T2-FLAIR-weighted input sequences for longitudinal assessment of whole-brain volume and lesions. In MS, six QReports provided evidence of technical validation, four companies have conducted clinical validation by correlating results with clinical variables, only one has tested their QReport by clinician end-users, and one has performed a simulated in-use socioeconomic evaluation. CONCLUSION We conclude that there is limited evidence in the literature regarding clinical validation and in-use evaluation of commercial MS QReports with a particular lack of clinician end-user testing. Our systematic review provides clinicians and institutions with the available evidence when considering adopting a quantitative reporting tool for MS.
Collapse
Affiliation(s)
- Zoe Mendelsohn
- grid.83440.3b0000000121901201Neuroradiological Academic Unit, UCL Queen Square Institute of Neurology, University College London, London, UK ,grid.83440.3b0000000121901201Department of Medical Physics and Bioengineering, Centre for Medical Image Computing (CMIC), University College London, London, UK ,grid.83440.3b0000000121901201Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, University College London, London, UK ,grid.6363.00000 0001 2218 4662Department of Neuroradiology, Charité School of Medicine and University Hospital Berlin, Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Radiology, Charité School of Medicine and University Hospital Berlin, Berlin, Germany
| | - Hugh G. Pemberton
- grid.83440.3b0000000121901201Neuroradiological Academic Unit, UCL Queen Square Institute of Neurology, University College London, London, UK ,grid.83440.3b0000000121901201Department of Medical Physics and Bioengineering, Centre for Medical Image Computing (CMIC), University College London, London, UK ,grid.420685.d0000 0001 1940 6527GE Healthcare, Amersham, UK
| | - James Gray
- grid.416626.10000 0004 0391 2793Stepping Hill Hospital, NHS Foundation Trust, Stockport, UK
| | - Olivia Goodkin
- grid.83440.3b0000000121901201Neuroradiological Academic Unit, UCL Queen Square Institute of Neurology, University College London, London, UK ,grid.83440.3b0000000121901201Department of Medical Physics and Bioengineering, Centre for Medical Image Computing (CMIC), University College London, London, UK ,grid.83440.3b0000000121901201Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, University College London, London, UK
| | - Ferran Prados Carrasco
- grid.83440.3b0000000121901201Department of Medical Physics and Bioengineering, Centre for Medical Image Computing (CMIC), University College London, London, UK ,grid.83440.3b0000000121901201Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, University College London, London, UK ,grid.36083.3e0000 0001 2171 6620E-Health Centre, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Michael Scheel
- grid.6363.00000 0001 2218 4662Department of Neuroradiology, Charité School of Medicine and University Hospital Berlin, Berlin, Germany
| | - Jawed Nawabi
- grid.6363.00000 0001 2218 4662Department of Radiology, Charité School of Medicine and University Hospital Berlin, Berlin, Germany ,grid.484013.a0000 0004 6879 971XBerlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Digital Clinician Scientist Program, Berlin, Germany
| | - Frederik Barkhof
- grid.83440.3b0000000121901201Neuroradiological Academic Unit, UCL Queen Square Institute of Neurology, University College London, London, UK ,grid.83440.3b0000000121901201Department of Medical Physics and Bioengineering, Centre for Medical Image Computing (CMIC), University College London, London, UK ,grid.83440.3b0000000121901201Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, University College London, London, UK ,grid.12380.380000 0004 1754 9227Radiology & Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
28
|
El Garhy NM, El Toukhy MM, Fatouh MM. MR volumetry in detection of brain atrophic changes in MS patients and its implication on disease prognosis: retrospective study. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2022. [DOI: 10.1186/s43055-022-00726-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Multiple sclerosis is a chronic demyelinating disease of the central nervous system. It may lead to disability and cognitive impairment. Our study aimed at evaluation of the role of MR volumetry technique in detection of brain atrophic changes in patients with multiple sclerosis and its impact on disease prognosis.
Results
This study was carried out on thirty healthy control with mean age 26.23 years and thirty patients with remitting relapsing multiple sclerosis, with a mean age of 28.18 years. Patients with multiple sclerosis were distributed across six subgroups based on the z-score cut-off of − 1.96 for regional and whole brain atrophy. We found that 2 patients (6.6%) showed no thalamic or brain atrophy, 28 patients (93.3%) showed whole brain atrophy only and 10 patients (33.3%) showed both, thalamic and BP atrophy. No patients showed only thalamic atrohy, 4 patients showed whole brain atrophy with other structure atrophy rather than thalamus (13.3%), 10 patients with whole brain and more than one structure atrophy (33.3%). Relation between subgroups and degree of increase in the Expanded Disability Status Scale (EDSS) as well as presence of cognitive decline were assessed. No significant relation were found between RRMS patients subgroups with whole brain atrophy, subgroup with isolated thalamic atrophy or subgroup with multiple structure atrophy and increase of EDSS or cognitive decline.
Conclusion
We found that MRI volumetry is a very useful technique in the assessment of the atrophic changes that occur as a consequence of multiple sclerosis affecting the whole brain, deep grey matter as well as corpus callosum. Although our study did not prove significant relation between presence of brain atrophic changes and disability or cognitive impairment, presence of atrophy warrants careful clinical evaluation of those patients to detect any possible further progression of disability or cognitive decline.
Collapse
|
29
|
The spatio-temporal relationship between white matter lesion volume changes and brain atrophy in clinically isolated syndrome and early multiple sclerosis. Neuroimage Clin 2022; 36:103220. [PMID: 36274376 PMCID: PMC9668617 DOI: 10.1016/j.nicl.2022.103220] [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: 05/27/2022] [Revised: 09/15/2022] [Accepted: 10/01/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND White matter lesions and brain atrophy are both present early in multiple sclerosis. However, the spatio-temporal relationship between atrophy and lesion processes remains unclear. METHODS Yearly magnetic resonance images were analyzed in 392 patients with clinically isolated syndrome from the 5-year REFLEX/REFLEXION studies. Patients received early treatment (from baseline; N = 262) or delayed treatment (from month-24; N = 130) with subcutaneous interferon beta-1a. Global and central atrophy were assessed using FSL-SIENA to provide yearly percentage volume change of brain and ventricles, respectively. Yearly total lesion volume change was calculated by subtracting the sum of the negative lesion volume change (disappearing + shrinking) from the positive lesion volume change (new + enlarging) for each yearly interval, as determined by an in-house developed semi-automated method. Using linear mixed models, during the period where patients had received ≥1 year of treatment, we investigated whether total lesion volume change was associated with percentage brain volume change or percentage ventricular volume change in the next year, and vice versa. RESULTS Higher total lesion volume change was related to significantly faster global atrophy (percentage brain volume change) in the next year (B = - 0.113, SE = 0.022, p < 0.001). In patients receiving early treatment only, total lesion volume change was also associated with percentage ventricular volume change in the next year (B = 1.348, SE = 0.181, p < 0.001). Voxel-wise analyses showed that in patients receiving early treatment, higher total lesion volume change in years 2, 3, and 4 was related to faster atrophy in the next year, and in year 4 this relationship was stronger in patients receiving delayed treatment. Interestingly, faster atrophy was related to higher total lesion volume change in the next year (percentage brain volume change: B = - 0.136, SE = 0.062, p = 0.028; percentage ventricular volume change: B = 0.028, SE = 0.008, p < 0.001). CONCLUSIONS Higher lesion volume changes were associated with faster atrophy in the next year. Interestingly, there was also an association between faster atrophy and higher lesion volume changes in the next year.
Collapse
|
30
|
Nooraei A, Khazaeel K, Darvishi M, Ghotbeddin Z, Basir Z. Dimorphic evaluation of hippocampal changes in rat model of demyelination: A comparative functional, morphometric, and histological study. Brain Behav 2022; 12:e32723. [PMID: 35861689 PMCID: PMC9392515 DOI: 10.1002/brb3.2723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 07/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is the most common autoimmune disease. Progressive depletion of the brain and spinal cord tissue appears at the onset of the disease. Several studies have shown the increased size of the ventricles of the brain and decreases in the area of the corpus callosum and the width of the brain. Other important symptoms of this disease are cognitive, learning, and memory disorders. AIM The aim of this study was to compare morphometric, histological, and functional changes in the demyelination model in both sexes. MATERIALS AND METHODS In this experimental study, male and female Wistar rats were studied in four experimental groups. Demyelination was induced by the injection of ethidium bromide in the ventricular region. The chronic effect of demyelination on spatial memory, movement, and coordination was investigated using the Morris Water Maze (MWM), and clinical and balance beam tests, respectively. Myelin degradation, cell death and neurogenesis were estimated using Luxol Fast Blue staining and immunohistochemistry (Caspase-3 and Nestin markers). In addition, morphometric findings were recorded for the brain and hippocampus (weight, volume, length, width). RESULT Demyelination increased the time and distance index and decreased the residence time in the target quarter in the water maze test (p < .001). It also increases the neuromuscular and modified neurological severity score (p < .01). Demyelination increases caspase-3 (p < .05) expression and decreases Nestin expression (p < .001), which are directly related to the extent of damage. CONCLUSION This study showed an interaction between hippocampal structural and functional networks in explaining spatial learning and memory in the early stages of MS.
Collapse
Affiliation(s)
- Aref Nooraei
- Faculty of Veterinary Medicine, Department of Basic Sciences, Shahid Chamran University of Ahvaz, Iran
| | - Kaveh Khazaeel
- Faculty of Veterinary Medicine, Department of Basic Sciences, Shahid Chamran University of Ahvaz, Iran.,Stem Cell and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Marzieh Darvishi
- Faculty of Medicine, Department of Anatomy, Ilam University of Medical Sciences, Ilam, Iran.,Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Zohreh Ghotbeddin
- Faculty of Veterinary Medicine, Department of Basic Sciences, Shahid Chamran University of Ahvaz, Iran.,Stem Cell and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Zahra Basir
- Faculty of Veterinary Medicine, Department of Basic Sciences, Shahid Chamran University of Ahvaz, Iran
| |
Collapse
|
31
|
Niiranen M, Koikkalainen J, Lötjönen J, Selander T, Cajanus A, Hartikainen P, Simula S, Vanninen R, Remes AM. Grey matter atrophy in patients with benign multiple sclerosis. Brain Behav 2022; 12:e2679. [PMID: 35765699 PMCID: PMC9304852 DOI: 10.1002/brb3.2679] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/22/2022] [Accepted: 06/03/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Brain atrophy appears during the progression of multiple sclerosis (MS) and is associated with the disability caused by the disease. METHODS We investigated global and regional grey matter (GM) and white matter (WM) volumes, WM lesion load, and corpus callosum index (CCI), in benign relapsing-remitting MS (BRRMS, n = 35) with and without any treatment and compared those to aggressive relapsing-remitting MS (ARRMS, n = 46). Structures were analyzed by using an automated MRI quantification tool (cNeuro®). RESULTS The total brain and cerebral WM volumes were larger in BRRMS than in ARRMS (p = .014, p = .017 respectively). In BRRMS, total brain volumes, regional GM volumes, and CCI were found similar whether or not disease-modifying treatment (DMT) was used. The total (p = .033), as well as subcortical (p = .046) and deep WM (p = .041) lesion load volumes were larger in BRRMS patients without DMT. Cortical GM volumes did not differ between BRRMS and ARRMS, but the volumes of total brain tissue (p = .014) and thalami (p = .003) were larger in patients with BRRMS compared to ARRMS. A positive correlation was found between CCI and whole-brain volume in both BRRMS (r = .73, p < .001) and ARRMS (r = .80, p < .01). CONCLUSIONS Thalamic volume is the most prominent measure to differentiate BRRMS and ARRMS. Validation of automated quantification of CCI provides an additional applicable MRI biomarker to detect brain atrophy in MS.
Collapse
Affiliation(s)
- Marja Niiranen
- Neuro Center, Neurology, Kuopio University Hospital, Kuopio, Finland
| | | | | | - Tuomas Selander
- Science Service Center, Kuopio University Hospital, Kuopio, Finland
| | - Antti Cajanus
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Päivi Hartikainen
- Neuro Center, Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Sakari Simula
- Department of Neurology, Mikkeli Central Hospital, Mikkeli, Finland
| | - Ritva Vanninen
- Institute of Clinical Medicine - Radiology, University of Eastern Finland, Kuopio, Finland.,Department of Radiology, Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Anne M Remes
- Unit of Clinical Neuroscience, Neurology, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
32
|
Zelilidou SP, Tripoliti EE, Vlachos KI, Konitsiotis S, Fotiadis DI. Segmentation and volume quantification of MR Images for the detection and monitoring multiple sclerosis progression. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:4745-4748. [PMID: 36085727 DOI: 10.1109/embc48229.2022.9871533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Multiple Sclerosis (MS) lesions detection and disease's progression monitoring at the same time, play an important role. The purpose of this research is to demonstrate a method for detecting MS plaques and volume estimation from MR Images for monitoring the progression of the disease and the brain atrophy caused. In the proposed research, a clustering-based method is utilized in order to delineate MS plaques in brain, based on anatomical information, brain geometry and lesion features. In addition to volumetric information concerning lesions and whole brain volume, volume quantification is employed to estimate MS atrophy by measuring Brain Parenchymal Fraction (BPF). In the present study, Fluid Attenuated Inversion Recovery (FLAIR) images were utilized for the detection of MS lesions and BPF evaluation, while Tl-weighted MR Images utilized in volume estimation. 30 MS patients were included in a dataset consisted of 3D FLAIR and T1-weighted MR images in order to evaluate the proposed technique. MRI scans performed in two different clinical visits, a baseline and a visit after 6 months. The results extracted in segmentation of MS lesions in terms of sensitivity is 73.80 %. The BPF at baseline estimated to 0.82 ± 0.01, and at 1stfollow up, 0.83 ± 0.01. Finally, the brain volume loss between baseline and after 6 months is 0.4%.
Collapse
|
33
|
Bhargava P, Hartung HP, Calabresi PA. Contribution of B cells to cortical damage in multiple sclerosis. Brain 2022; 145:3363-3373. [PMID: 35775595 DOI: 10.1093/brain/awac233] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/06/2022] [Accepted: 06/17/2022] [Indexed: 11/14/2022] Open
Abstract
Multiple sclerosis is associated with lesions not just in the white matter, but also involving the cortex. Cortical involvement has been linked to greater disease severity and hence understanding the factor underlying cortical pathology could help identify new therapeutic strategies for multiple sclerosis. The critical role of B cells in multiple sclerosis has been clarified by multiple pivotal trials of B cell depletion in people with multiple sclerosis. The presence of B cell rich areas of meningeal inflammation in multiple sclerosis has been identified at all stages of multiple sclerosis. Leptomeningeal inflammation is associated with greater extent of cortical demyelination and neuronal loss and with greater disease severity. Recent studies have identified several potential mechanisms by which B cells may mediate cortical injury including antibody production, extracellular vesicles containing neurotoxic substances and production of pro-inflammatory cytokines. Additionally, B cells may indirectly mediate cortical damage through effects on T cells, macrophages or microglia. Several animal models replicate the meningeal inflammation and cortical injury noted in people with multiple sclerosis. Studies in these models have identified BTK inhibition and type II anti-CD20 antibodies as potential agents that can impact meningeal inflammation. Trials of anti-CD20 monoclonal antibodies in people with multiple sclerosis have unsuccessfully attempted to eliminate B cells in the leptomeninges. New strategies to target B cells in multiple sclerosis include BTK inhibition and cell-based therapies aimed at B cells infected with Epstein Barr virus. Future studies will clarify the mechanisms by which B cells mediate cortical injury and treatment strategies that can target B cells in the leptomeninges and CNS parenchyma.
Collapse
Affiliation(s)
- Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hans Peter Hartung
- Department of Neurology, Heinrich-Heine University, Dusseldorf, Germany.,Brain and Mind Center, University of Sydney, Sydney, Australia.,Department of Neurology, Medical University of Vienna, Vienna, Austria.,Department of Neurology, Palacky University Olomouc, Olomouc, Czech Republic
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
34
|
Vestergaard MB, Frederiksen JL, Larsson HBW, Cramer SP. Cerebrovascular Reactivity and Neurovascular Coupling in Multiple Sclerosis-A Systematic Review. Front Neurol 2022; 13:912828. [PMID: 35720104 PMCID: PMC9198441 DOI: 10.3389/fneur.2022.912828] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/04/2022] [Indexed: 11/25/2022] Open
Abstract
The inflammatory processes observed in the central nervous system in multiple sclerosis (MS) could damage the endothelium of the cerebral vessels and lead to a dysfunctional regulation of vessel tonus and recruitment, potentially impairing cerebrovascular reactivity (CVR) and neurovascular coupling (NVC). Impaired CVR or NVC correlates with declining brain health and potentially plays a causal role in the development of neurodegenerative disease. Therefore, we examined studies on CVR or NVC in MS patients to evaluate the evidence for impaired cerebrovascular function as a contributing disease mechanism in MS. Twenty-three studies were included (12 examined CVR and 11 examined NVC). Six studies found no difference in CVR response between MS patients and healthy controls. Five studies observed reduced CVR in patients. This discrepancy can be because CVR is mainly affected after a long disease duration and therefore is not observed in all patients. All studies used CO2 as a vasodilating stimulus. The studies on NVC demonstrated diverse results; hence a conclusion that describes all the published observations is difficult to find. Future studies using quantitative techniques and larger study samples are needed to elucidate the discrepancies in the reported results.
Collapse
Affiliation(s)
- Mark B Vestergaard
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
| | - Jette L Frederiksen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Henrik B W Larsson
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark.,Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
| | - Stig P Cramer
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
| |
Collapse
|
35
|
Brain Structural and Functional Alterations in Multiple Sclerosis-Related Fatigue: A Systematic Review. Neurol Int 2022; 14:506-535. [PMID: 35736623 PMCID: PMC9228847 DOI: 10.3390/neurolint14020042] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 01/27/2023] Open
Abstract
Fatigue is one of the most disabling symptoms of multiple sclerosis (MS); it influences patients’ quality of life. The etiology of fatigue is complex, and its pathogenesis is still unclear and debated. The objective of this review was to describe potential brain structural and functional dysfunctions underlying fatigue symptoms in patients with MS. To reach this purpose, a systematic review was conducted of published studies comparing functional brain activation and structural brain in MS patients with and without fatigue. Electronic databases were searched until 24 February 2021. The structural and functional outcomes were extracted from eligible studies and tabulated. Fifty studies were included: 32 reported structural brain differences between patients with and without fatigue; 14 studies described functional alterations in patients with fatigue compared to patients without it; and four studies showed structural and functional brain alterations in patients. The results revealed structural and functional abnormalities that could correlate to the symptom of fatigue in patients with MS. Several studies reported the differences between patients with fatigue and patients without fatigue in terms of conventional magnetic resonance imaging (MRI) outcomes and brain atrophy, specifically in the thalamus. Functional studies showed abnormal activation in the thalamus and in some regions of the sensorimotor network in patients with fatigue compared to patients without it. Patients with fatigue present more structural and functional alterations compared to patients without fatigue. Specifically, abnormal activation and atrophy of the thalamus and some regions of the sensorimotor network seem linked to fatigue.
Collapse
|
36
|
Brune S, Høgestøl EA, de Rodez Benavent SA, Berg-Hansen P, Beyer MK, Leikfoss IS, Bos SD, Sowa P, Brunborg C, Andorra M, Pulido Valdeolivas I, Asseyer S, Brandt A, Chien C, Scheel M, Blennow K, Zetterberg H, Kerlero de Rosbo N, Paul F, Uccelli A, Villoslada P, Berge T, Harbo HF. Serum neurofilament light chain concentration predicts disease worsening in multiple sclerosis. Mult Scler 2022; 28:1859-1870. [PMID: 35658739 PMCID: PMC9493412 DOI: 10.1177/13524585221097296] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Serum neurofilament light (sNfL) chain is a promising biomarker reflecting
neuro-axonal injury in multiple sclerosis (MS). However, the ability of sNfL
to predict outcomes in real-world MS cohorts requires further
validation. Objective: The aim of the study is to investigate the associations of sNfL
concentration, magnetic resonance imaging (MRI) and retinal optical
coherence tomography (OCT) markers with disease worsening in a longitudinal
European multicentre MS cohort. Methods: MS patients (n = 309) were prospectively enrolled at four
centres and re-examined after 2 years (n = 226). NfL
concentration was measured by single molecule array assay in serum. The
patients’ phenotypes were thoroughly characterized with clinical
examination, retinal OCT and MRI brain scans. The primary outcome was
disease worsening at median 2-year follow-up. Results: Patients with high sNfL concentrations (⩾8 pg/mL) at baseline had increased
risk of disease worsening at median 2-year follow-up (odds ratio (95%
confidence interval) = 2.8 (1.5–5.3), p = 0.001). We found
no significant associations of MRI or OCT measures at baseline with risk of
disease worsening. Conclusion: Serum NfL concentration was the only factor associated with disease
worsening, indicating that sNfL is a useful biomarker in MS that might be
relevant in a clinical setting.
Collapse
Affiliation(s)
- Synne Brune
- Institute of clinical Medicine, University of Oslo, Oslo, Norway/Department of Neurology, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Einar A Høgestøl
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway/Department of Neurology, Oslo University Hospital, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | | | - Pål Berg-Hansen
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Mona K Beyer
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway/Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Ingvild Sørum Leikfoss
- Department of Neurology, Oslo University Hospital, Oslo, Norway/Department of Research, Innovation and Education, Oslo University Hospital, Oslo, Norway
| | - Steffan D Bos
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway/Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Piotr Sowa
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Cathrine Brunborg
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Magi Andorra
- Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain
| | | | - Susanna Asseyer
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Alexander Brandt
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitaetsmedizin Berlin, Berlin, Germany/NeuroCure Clinical Research Center, Charité-Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Claudia Chien
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitaetsmedizin Berlin, Berlin, Germany/NeuroCure Clinical Research Center, Charité-Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Michael Scheel
- NeuroCure Clinical Research Center, Charité-Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany/Department of Neuroradiology, Charité-Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden/Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden/Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK/UK Dementia Research Institute at UCL, London, UK/Hong Kong Center for Neurodegenerative Diseases, Shatin, Hong Kong, China
| | - Nicole Kerlero de Rosbo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitaetsmedizin Berlin, Berlin, Germany/NeuroCure Clinical Research Center, Charité-Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Antonio Uccelli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy/Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy/IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Pablo Villoslada
- Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain
| | - Tone Berge
- Department of Research, Innovation and Education, Oslo University Hospital, Oslo, Norway/Department of Mechanical, Electronic and Chemical Engineering, Oslo Metropolitan University, Oslo, Norway
| | - Hanne F Harbo
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway/Department of Neurology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
37
|
Bells S, Longoni G, Berenbaum T, de Medeiros CB, Narayanan S, Banwell BL, Arnold DL, Mabbott DJ, Ann Yeh E. Patterns of white and gray structural abnormality associated with paediatric demyelinating disorders. Neuroimage Clin 2022; 34:103001. [PMID: 35381508 PMCID: PMC8980471 DOI: 10.1016/j.nicl.2022.103001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 11/26/2022]
Abstract
A multi-modal approach was used to evaluate the visual pathway from anterior (retina) to posterior (visual cortex) in both paediatric MOGAD and MS patients. MS patients exhibited more widespread white matter abnormalities; MOGAD patients exhibited white matter changes primarily within the optic radiation. The pattern of cortical thinning differed in MS and MOGAD patients. Reduced RNFLT was associated with lower axonal density in MOGAD and tortuosity in MS.
The impact of multiple sclerosis (MS) and myelin oligodendrocyte glycoprotein (MOG) - associated disorders (MOGAD) on brain structure in youth remains poorly understood. Reductions in cortical mantle thickness on structural MRI and abnormal diffusion-based white matter metrics (e.g., diffusion tensor parameters) have been well documented in MS but not in MOGAD. Characterizing structural abnormalities found in children with these disorders can help clarify the differences and similarities in their impact on neuroanatomy. Importantly, while MS and MOGAD affect the entire CNS, the visual pathway is of particular interest in both groups, as most patients have evidence for clinical or subclinical involvement of the anterior visual pathway. Thus, the visual pathway is of key interest in analyses of structural abnormalities in these disorders and may distinguish MOGAD from MS patients. In this study we collected MRI data on 18 MS patients, 14 MOGAD patients and 26 age- and sex-matched typically developing children (TDC). Full-brain group differences in fixel diffusion measures (fibre-bundle populations) and cortical thickness measures were tested using age and sex as covariates. Visual pathway analysis was performed by extracting mean diffusion measures within lesion free optic radiations, cortical thickness within the visual cortex, and retinal nerve fibre layer (RNFL) and ganglion cell layer thickness measures from optical coherence tomography (OCT). Fixel based analysis (FBA) revealed MS patients have widespread abnormal white matter within the corticospinal tract, inferior longitudinal fasciculus, and optic radiations, while within MOGAD patients, non-lesional impact on white matter was found primarily in the right optic radiation. Cortical thickness measures were reduced predominately in the temporal and parietal lobes in MS patients and in frontal, cingulate and visual cortices in MOGAD patients. Additionally, our findings of associations between reduced RNFLT and axonal density in MOGAD and TORT in MS patients in the optic radiations imply widespread axonal and myelin damage in the visual pathway, respectively. Overall, our approach of combining FBA, cortical thickness and OCT measures has helped evaluate similarities and differences in brain structure in MS and MOGAD patients in comparison to TDC.
Collapse
Affiliation(s)
- Sonya Bells
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children, Toronto, Canada; Pediatric Neurology, Spectrum Health Helen Devos Children's Hospital, Grand Rapids, USA; Department of Pediatrics and Human Development, Michigan State University, East Lansing, USA
| | - Giulia Longoni
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children, Toronto, Canada; Department of Neurology, Hospital for Sick Children, Toronto, Canada; Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Tara Berenbaum
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Cynthia B de Medeiros
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Sridar Narayanan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Brenda L Banwell
- Division of Child Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, USA
| | - Douglas L Arnold
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Donald J Mabbott
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children, Toronto, Canada; Department of Psychology, University of Toronto, Toronto, Canada
| | - E Ann Yeh
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children, Toronto, Canada; Department of Neurology, Hospital for Sick Children, Toronto, Canada; Department of Paediatrics, University of Toronto, Toronto, Canada.
| |
Collapse
|
38
|
Bose G, Healy BC, Lokhande HA, Sotiropoulos MG, Polgar‐Turcsanyi M, Anderson M, Glanz BI, Guttman CRG, Bakshi R, Weiner HL, Chitnis T. Early predictors of clinical and MRI outcomes using LASSO in multiple sclerosis. Ann Neurol 2022; 92:87-96. [DOI: 10.1002/ana.26370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/28/2022] [Accepted: 04/10/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Gauruv Bose
- Harvard Medical School Boston MA US
- Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital Boston MA US
| | - Brian C. Healy
- Harvard Medical School Boston MA US
- Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital Boston MA US
| | - Hrishikesh A. Lokhande
- Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital Boston MA US
| | - Marinos G. Sotiropoulos
- Harvard Medical School Boston MA US
- Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital Boston MA US
| | - Mariann Polgar‐Turcsanyi
- Harvard Medical School Boston MA US
- Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital Boston MA US
| | - Mark Anderson
- Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital Boston MA US
| | - Bonnie I. Glanz
- Harvard Medical School Boston MA US
- Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital Boston MA US
| | - Charles R. G. Guttman
- Harvard Medical School Boston MA US
- Center for Neurological Imaging, Department of Radiology, Brigham and Women’s Hospital Boston MA US
| | - Rohit Bakshi
- Harvard Medical School Boston MA US
- Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital Boston MA US
| | - Howard L. Weiner
- Harvard Medical School Boston MA US
- Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital Boston MA US
| | - Tanuja Chitnis
- Harvard Medical School Boston MA US
- Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital Boston MA US
| |
Collapse
|
39
|
Predictive MRI Biomarkers in MS—A Critical Review. Medicina (B Aires) 2022; 58:medicina58030377. [PMID: 35334554 PMCID: PMC8949449 DOI: 10.3390/medicina58030377] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 02/12/2022] [Accepted: 02/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Objectives: In this critical review, we explore the potential use of MRI measurements as prognostic biomarkers in multiple sclerosis (MS) patients, for both conventional measurements and more novel techniques such as magnetization transfer, diffusion tensor, and proton spectroscopy MRI. Materials and Methods: All authors individually and comprehensively reviewed each of the aspects listed below in PubMed, Medline, and Google Scholar. Results: There are numerous MRI metrics that have been proven by clinical studies to hold important prognostic value for MS patients, most of which can be readily obtained from standard 1.5T MRI scans. Conclusions: While some of these parameters have passed the test of time and seem to be associated with a reliable predictive power, some are still better interpreted with caution. We hope this will serve as a reminder of how vast a resource we have on our hands in this versatile tool—it is up to us to make use of it.
Collapse
|
40
|
No Changes in Functional Connectivity After Dimethyl Fumarate Treatment in Multiple Sclerosis. Neurol Ther 2022; 11:471-479. [PMID: 35119678 PMCID: PMC8857342 DOI: 10.1007/s40120-022-00328-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/19/2022] [Indexed: 11/06/2022] Open
Abstract
Introduction Despite the increased availability of disease-modifying therapies (DMTs) for treating relapsing-remitting multiple sclerosis (RR-MS), only a few studies have evaluated DMT-associated brain functional changes. Methods We investigated whether significant resting-state functional connectivity (FC) changes occurred in RR-MS patients after 6 and 12 months of dimethyl fumarate (DMF) treatment using both a seed-based and data-driven approach. Results Thirty patients were followed up after 6 months of therapy, and 27 of them reached a 12-month follow-up. Three patients at baseline and only one after 12 months showed gadolinium-enhancing lesions. We did not find any significant FC changes after therapy at either time point. After 12 months of DMF, we observed relatively modest brain volume loss and a significant improvement in Paced Auditory Serial Addition Test 3 s and 25-Foot Walk Test scores. Conclusion The absence of FC changes could be due to the low degree of baseline inflammation in our patients, though we cannot exclude that more time may be required to observe such changes. No FC changes may reflect a beneficial effect of DMF therapy, as supported by conventional MRI findings and clinical improvement.
Collapse
|
41
|
Ajitomi S, Fujimori J, Nakashima I. Usefulness of two-dimensional measurements for the evaluation of brain volume and disability in multiple sclerosis. Mult Scler J Exp Transl Clin 2022; 8:20552173211070749. [PMID: 35024162 PMCID: PMC8743968 DOI: 10.1177/20552173211070749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/15/2021] [Indexed: 11/29/2022] Open
Abstract
Background Two-dimensional (2D) measures have been proposed as potential proxies for whole-brain volume in multiple sclerosis (MS). Objective To verify whether 2D measurements by routine MRI are useful in predicting brain volume or disability in MS. Methods In this cross-sectional analysis, eighty-five consecutive Japanese MS patients—relapsing-remitting MS (81%) and progressive MS (19%)—underwent 1.5 Tesla T1-weighted 3D MRI examinations to measure whole-brain and grey matter volume. 2D measurements, namely, third ventricle width, lateral ventricle width (LVW), brain width, bicaudate ratio, and corpus callosum index (CCI), were obtained from each scan. Correlations between 2D measurements and 3D measurements, the Expanded Disability Status Scale (EDSS), or processing speed were analysed. Results The third and lateral ventricle widths were well-correlated with the whole-brain volume (p < 0.0001), grey matter volume (p < 0.0001), and EDSS scores (p = 0.0001, p = .0004, respectively).The least squares regression model revealed that 78% of the variation in whole-brain volume could be explained using five explanatory variables, namely, LVW, CCI, age, sex, and disease duration. By contrast, the partial correlation coefficient excluding the effect of age showed that the CCI was significantly correlated with the EDSS and processing speed (p < 0.0001). Conclusion Ventricle width correlated well with brain volumes, while the CCI correlated well with age-independent (i.e. disease-induced) disability.
Collapse
Affiliation(s)
- Satori Ajitomi
- School of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Juichi Fujimori
- Division of Neurology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Ichiro Nakashima
- Division of Neurology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
42
|
Miscioscia A, Puthenparampil M, Miante S, Pengo M, Rinaldi F, Perini P, Gallo P. Retinal inner nuclear layer thinning is decreased and associates with the clinical outcome in ocrelizumab-treated primary progressive multiple sclerosis. J Neurol 2022; 269:5436-5442. [PMID: 35648233 PMCID: PMC9467948 DOI: 10.1007/s00415-022-11183-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Ocrelizumab was found to decrease brain atrophy rate in primary progressive multiple sclerosis (PPMS), but no data are currently available on the effect of ocrelizumab on retinal layer thicknesses in the PPMS population. OBJECTIVE To assess retinal layer changes in ocrelizumab-treated PPMS and test their possible application as biomarkers of therapy response. METHODS 36 PPMS patients, treated with ocrelizumab for at least 6 months, and 39 sex- and age-matched healthy controls (HC) were included in a blind, longitudinal study. Spectrum-domain optical coherence tomography (SD-OCT) was performed at study entry (T0) and after 6 (T6) and 12 months (T12). At month 24 (T24), patients were divided into responders (no evidence of 1-year confirmed disability progression, 1y-CDP) and non-responders (evidence of 1y-CDP). RESULTS At T24, 23/36 (64%) patients were considered responders and 13/36 (36%) non-responders. At T0, peripapillary retinal nerve fiber layer (pRNFL) thickness, macular ganglion cell-inner plexiform layer (GCIPL) and inner retinal layer (IRL) volume were significantly lower in PPMS compared to HC (p = 0.001 for all comparisons). At T6 and T12, non-responders significantly differed in the inner nuclear layer (INL) thinning rate compared to responders (p = 0.005 at both time-points). CONCLUSIONS Ocrelizumab significantly slows down INL thinning rate in PPMS responders. The longitudinal analysis of retina layer changes by means of OCT may be a promising prognostic test, and merits further investigations.
Collapse
Affiliation(s)
- Alessandro Miscioscia
- Department of Neuroscience DNS, School of Medicine, University of Padua, Via Giustiniani, 5, 35128 Padua, Veneto Region Italy ,Multiple Sclerosis Centre, University Hospital of Padua, Padua, Veneto Region Italy
| | - Marco Puthenparampil
- Department of Neuroscience DNS, School of Medicine, University of Padua, Via Giustiniani, 5, 35128 Padua, Veneto Region Italy ,Multiple Sclerosis Centre, University Hospital of Padua, Padua, Veneto Region Italy
| | - Silvia Miante
- Department of Neuroscience DNS, School of Medicine, University of Padua, Via Giustiniani, 5, 35128 Padua, Veneto Region Italy ,Multiple Sclerosis Centre, University Hospital of Padua, Padua, Veneto Region Italy ,Present Address: Neurology Unit, Ospedale dell’Angelo, Mestre, Italy
| | - Marta Pengo
- Department of Neuroscience DNS, School of Medicine, University of Padua, Via Giustiniani, 5, 35128 Padua, Veneto Region Italy ,Multiple Sclerosis Centre, University Hospital of Padua, Padua, Veneto Region Italy ,Present Address: Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesca Rinaldi
- Multiple Sclerosis Centre, University Hospital of Padua, Padua, Veneto Region Italy
| | - Paola Perini
- Multiple Sclerosis Centre, University Hospital of Padua, Padua, Veneto Region Italy
| | - Paolo Gallo
- Department of Neuroscience DNS, School of Medicine, University of Padua, Via Giustiniani, 5, 35128 Padua, Veneto Region Italy ,Multiple Sclerosis Centre, University Hospital of Padua, Padua, Veneto Region Italy
| |
Collapse
|
43
|
Wang C, Barton J, Kyle K, Ly L, Barnett Y, Hartung HP, Reddel SW, Beadnall H, Taha M, Klistorner A, Barnett MH. Multiple sclerosis: structural and functional integrity of the visual system following alemtuzumab therapy. J Neurol Neurosurg Psychiatry 2021; 92:1319-1324. [PMID: 34187865 DOI: 10.1136/jnnp-2021-326164] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/02/2021] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To investigate potential neuroprotective and pro-remyelinating effects of alemtuzumab in multiple sclerosis (MS), using the visual pathway as a model. METHODS We monitored clinical, multifocal visual evoked potential (mfVEP) and MRI outcomes in 30 patients commencing alemtuzumab for relapsing MS, and a reference group of 20 healthy controls (HCs), over 24 months. Change in mfVEP latency was the primary endpoint; change in optic radiation (OR) lesion diffusion metrics and Mars letter contrast sensitivity over the course of the study were secondary endpoints. RESULTS In patients, we observed a mean shortening of mfVEP latency of 1.21 ms over the course of the study (95% CI 0.21 to 2.21, p=0.013), not altered by correction for age, gender, disease duration or change in OR T2 lesion volume. Mean mfVEP latency in the HC group increased over the course of the study by 0.72 ms (not significant). Analysis of chronic OR T2 lesions (patients) showed an increase in normalised fractional anisotropy and axial diffusivity between baseline and 24 months (both p<0.01). Mean Mars letter contrast sensitivity was improved at 24 months vs baseline (p<0.001), and driven by an early improvement, in both patients and HC. CONCLUSION We found evidence of partial lesion remyelination after alemtuzumab therapy, indicating either natural restoration in the context of a 'permissive' local milieu; or potentially an independent, pro-reparative mechanism of action. The visual system presents a unique opportunity to study function-structure specific effects of therapy and inform the design of future phase 2 MS remyelination trials.
Collapse
Affiliation(s)
- Chenyu Wang
- Sydney Neuroimaging Analysis Centre, Camperdown, New South Wales, Australia.,Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Joshua Barton
- Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Kain Kyle
- Sydney Neuroimaging Analysis Centre, Camperdown, New South Wales, Australia.,Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Linda Ly
- Sydney Neuroimaging Analysis Centre, Camperdown, New South Wales, Australia
| | - Yael Barnett
- Sydney Neuroimaging Analysis Centre, Camperdown, New South Wales, Australia.,Radiology Department, St Vincent's Hospital Sydney, Darlinghurst, New South Wales, Australia
| | - Hans-Peter Hartung
- Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia.,Clinic for Neurology, Heinrich Heine University Düsseldorf, Dusseldorf, Germany
| | - Stephen W Reddel
- Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Heidi Beadnall
- Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia.,Neurology Department, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Marinda Taha
- Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Alexander Klistorner
- Sydney Neuroimaging Analysis Centre, Camperdown, New South Wales, Australia.,Save Sight Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Michael Harry Barnett
- Sydney Neuroimaging Analysis Centre, Camperdown, New South Wales, Australia .,Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia.,Neurology Department, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| |
Collapse
|
44
|
Cerebellar Contributions to Motor Impairments in People with Multiple Sclerosis. THE CEREBELLUM 2021; 21:1052-1060. [PMID: 34657272 DOI: 10.1007/s12311-021-01336-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/10/2021] [Indexed: 12/25/2022]
Abstract
Although Charcot characterized classic cerebellar symptoms in people with multiple sclerosis (PwMS) in 1877, the impact of cerebellar dysfunction on MS symptoms has predominately been evaluated in the last two decades. Recent studies have clearly demonstrated the association between cerebellar pathology, including atrophy and reduced fractional anisotropy in the peduncles, and motor impairments, such as reduced gait velocity and time to complete walking tasks. However, future studies using novel imaging techniques are needed to elucidate all potential pathophysiology that is associated with disability in PwMS. Additionally, future studies are required to determine the most effective treatments for motor impairments in PwMS, including the specific type and duration of exercise interventions, and potential means to amplify their effects, such as transcranial direct current stimulation (tDCS). This mini-review critically discusses the distinct role of cerebellar dysfunction in motor impairments in PwMS, potential treatments, and directions for future studies.
Collapse
|
45
|
Uher T, Havrdova EK, Benkert P, Bergsland N, Krasensky J, Srpova B, Dwyer M, Tyblova M, Meier S, Vaneckova M, Horakova D, Zivadinov R, Leppert D, Kalincik T, Kuhle J. Measurement of neurofilaments improves stratification of future disease activity in early multiple sclerosis. Mult Scler 2021; 27:2001-2013. [PMID: 34612753 DOI: 10.1177/13524585211047977] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND The added value of neurofilament light chain levels in serum (sNfL) to the concept of no evidence of disease activity-3 (NEDA-3) has not yet been investigated in detail. OBJECTIVE To assess whether combination of sNfL with NEDA-3 status improves identification of patients at higher risk of disease activity during the following year. METHODS We analyzed 369 blood samples from 155 early relapsing-remitting MS patients on interferon beta-1a. We compared disease activity, including the rate of brain volume loss in subgroups defined by NEDA-3 status and high or low sNfL (> 90th or < 90th percentile). RESULTS In patients with disease activity (EDA-3), those with higher sNFL had higher odds of EDA-3 in the following year than those with low sNFL (86.5% vs 57.9%; OR = 4.25, 95% CI: [2.02, 8.95]; p = 0.0001) and greater whole brain volume loss during the following year (β = -0.36%; 95% CI = [-0.60, -0.13]; p = 0.002). Accordingly, NEDA-3 patients with high sNfL showed numerically higher disease activity (EDA-3) in the following year compared with those with low sNfL (57.1% vs 31.1%). CONCLUSION sNfL improves the ability to identify patients at higher risk of future disease activity, beyond their NEDA-3 status. Measurement of sNfL may assist clinicians in decision-making by providing more sensitive prognostic information.
Collapse
Affiliation(s)
- Tomas Uher
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Katerinska 30, 120 00 Prague, Czech Republic.,Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic/CORe, Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
| | - Eva Kubala Havrdova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Pascal Benkert
- Clinical Trial Unit, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA/IRCCS, Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Jan Krasensky
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Barbora Srpova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Michael Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Michaela Tyblova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Stephanie Meier
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Manuela Vaneckova
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Dana Horakova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA/Center for Biomedical Imaging, Clinical and Translational Science Institute, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - David Leppert
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Tomas Kalincik
- CORe, Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia; Melbourne MS Centre, Department of Neurology, The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
46
|
Krajnc N, Bsteh G, Berger T. Clinical and Paraclinical Biomarkers and the Hitches to Assess Conversion to Secondary Progressive Multiple Sclerosis: A Systematic Review. Front Neurol 2021; 12:666868. [PMID: 34512500 PMCID: PMC8427301 DOI: 10.3389/fneur.2021.666868] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/06/2021] [Indexed: 12/11/2022] Open
Abstract
Conversion to secondary progressive (SP) course is the decisive factor for long-term prognosis in relapsing multiple sclerosis (MS), generally considered the clinical equivalent of progressive MS-associated neuroaxonal degeneration. Evidence is accumulating that both inflammation and neurodegeneration are present along a continuum of pathologic processes in all phases of MS. While inflammation is the prominent feature in early stages, its quality changes and relative importance to disease course decreases while neurodegenerative processes prevail with ongoing disease. Consequently, anti-inflammatory disease-modifying therapies successfully used in relapsing MS are ineffective in SPMS, whereas specific treatment for the latter is increasingly a focus of MS research. Therefore, the prevention, but also the (anticipatory) diagnosis of SPMS, is of crucial importance. The problem is that currently SPMS diagnosis is exclusively based on retrospectively assessing the increase of overt physical disability usually over the past 6–12 months. This inevitably results in a delay of diagnosis of up to 3 years resulting in periods of uncertainty and, thus, making early therapy adaptation to prevent SPMS conversion impossible. Hence, there is an urgent need for reliable and objective biomarkers to prospectively predict and define SPMS conversion. Here, we review current evidence on clinical parameters, magnetic resonance imaging and optical coherence tomography measures, and serum and cerebrospinal fluid biomarkers in the context of MS-associated neurodegeneration and SPMS conversion. Ultimately, we discuss the necessity of multimodal approaches in order to approach objective definition and prediction of conversion to SPMS.
Collapse
Affiliation(s)
- Nik Krajnc
- Department of Neurology, Medical University of Vienna, Vienna, Austria.,Department of Neurology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Gabriel Bsteh
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
47
|
Measuring Treatment Response in Progressive Multiple Sclerosis-Considerations for Adapting to an Era of Multiple Treatment Options. Biomolecules 2021; 11:biom11091342. [PMID: 34572555 PMCID: PMC8470215 DOI: 10.3390/biom11091342] [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] [Received: 08/03/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 12/15/2022] Open
Abstract
Disability in multiple sclerosis accrues predominantly in the progressive forms of the disease. While disease-modifying treatment of relapsing MS has drastically evolved over the last quarter-century, the development of efficient drugs for preventing or at least delaying disability in progressive MS has proven more challenging. In that way, many drugs (especially disease-modifying treatments) have been researched in the aspect of delaying disability progression in patients with a progressive course of the disease. While there are some disease-modifying treatments approved for progressive multiple sclerosis, their effect is moderate and limited mostly to patients with clinical and/or radiological signs of disease activity. Several phase III trials have used different primary outcomes with different time frames to define disease progression and to evaluate the efficacy of a disease-modifying treatment. The lack of sufficiently sensitive outcome measures could be a possible explanation for the negative clinical trials in progressive multiple sclerosis. On the other hand, even with a potential outcome measure that would be sensitive enough to determine disease progression and, thus, the efficacy or failure of a disease-modifying treatment, the question of clinical relevance remains unanswered. In this systematic review, we analyzed outcome measures and definitions of disease progression in phase III clinical trials in primary and secondary progressive multiple sclerosis. We discuss advantages and disadvantages of clinical and paraclinical outcome measures aiming for practical ways of combining them to detect disability progression more sensitively both in future clinical trials and current clinical routine.
Collapse
|
48
|
Barnett M, Bergsland N, Weinstock-Guttman B, Butzkueven H, Kalincik T, Desmond P, Gaillard F, van Pesch V, Ozakbas S, Rojas JI, Boz C, Altintas A, Wang C, Dwyer MG, Yang S, Jakimovski D, Kyle K, Ramasamy DP, Zivadinov R. Brain atrophy and lesion burden are associated with disability progression in a multiple sclerosis real-world dataset using only T2-FLAIR: The NeuroSTREAM MSBase study. NEUROIMAGE-CLINICAL 2021; 32:102802. [PMID: 34469848 PMCID: PMC8408519 DOI: 10.1016/j.nicl.2021.102802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/28/2021] [Accepted: 08/18/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Methodological challenges limit the use of brain atrophy and lesion burden measures in the follow-up of multiple sclerosis (MS) patients on clinical routine datasets. OBJECTIVE To determine the feasibility of T2-FLAIR-only measures of lateral ventricular volume (LVV) and salient central lesion volume (SCLV), as markers of disability progression (DP) in MS. METHODS A total of 3,228 MS patients from 9 MSBase centers in 5 countries were enrolled. Of those, 2,875 (218 with clinically isolated syndrome, 2,231 with relapsing-remitting and 426 with progressive disease subtype) fulfilled inclusion and exclusion criteria. Patients were scanned on either 1.5 T or 3 T MRI scanners, and 5,750 brain scans were collected at index and on average after 42.3 months at post-index. Demographic and clinical data were collected from the MSBase registry. LVV and SCLV were measured on clinical routine T2-FLAIR images. RESULTS Longitudinal LVV and SCLV analyses were successful in 96% of the scans. 57% of patients had scanner-related changes over the follow-up. After correcting for age, sex, disease duration, disability, disease-modifying therapy and LVV at index, and follow-up time, MS patients with DP (n = 671) had significantly greater absolute LVV change compared to stable (n = 1,501) or disability improved (DI, n = 248) MS patients (2.0 mL vs. 1.4 mL vs. 1.1 mL, respectively, ANCOVA p < 0.001, post-hoc pair-wise DP vs. Stable p = 0.003; and DP vs. DI, p = 0.002). Similar ANCOVA model was also significant for SCLV (p = 0.03). CONCLUSIONS LVV-based atrophy and SCLV-based lesion outcomes are feasible on clinically acquired T2-FLAIR scans in a multicenter fashion and are associated with DP over mid-term.
Collapse
Affiliation(s)
- Michael Barnett
- Sydney Neuroimaging Analysis Centre, Camperdown, Sydney, Australia; Brain and Mind Centre, University of Sydney, Sydney, Australia.
| | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, USA; IRCCS, Fondazione Don Carlo Gnocchi ONLUS, Italy
| | - Bianca Weinstock-Guttman
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, USA
| | | | - Tomas Kalincik
- CORe, Department of Medicine, The University of Melbourne, Melbourne, Australia; MS Centre, The Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
| | - Patricia Desmond
- Department of Radiology, Royal Melbourne Hospital, The University of Melbourne, Melbourne, Australia
| | - Frank Gaillard
- Department of Radiology, Royal Melbourne Hospital, The University of Melbourne, Melbourne, Australia
| | | | | | | | - Cavit Boz
- KTU Medical Faculty Farabi Hospital, Trabzon, Turkey
| | - Ayse Altintas
- Koç University School of Medicine, Koç University Research Center for Translational Medicine (KUTTAM), İstanbul, Turkey
| | - Chenyu Wang
- Sydney Neuroimaging Analysis Centre, Camperdown, Sydney, Australia; Brain and Mind Centre, University of Sydney, Sydney, Australia
| | - Michael G Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, USA; Center for Biomedical Imaging, Clinical Translational Science Institute, USA; University at Buffalo, NY, USA
| | - Suzie Yang
- Sydney Neuroimaging Analysis Centre, Camperdown, Sydney, Australia
| | - Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, USA
| | - Kain Kyle
- Sydney Neuroimaging Analysis Centre, Camperdown, Sydney, Australia; Brain and Mind Centre, University of Sydney, Sydney, Australia
| | - Deepa P Ramasamy
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, USA; Center for Biomedical Imaging, Clinical Translational Science Institute, USA; University at Buffalo, NY, USA
| |
Collapse
|
49
|
Alvarez E, Nair KV, Hoyt BD, Seale RA, Sillau S, Miravalle A, Engebretson E, Schurr B, Corboy JR, Vollmer TL, Honce JM. Brain atrophy rates in patients with multiple sclerosis on long term natalizumab resembles healthy controls. Mult Scler Relat Disord 2021; 55:103170. [PMID: 34364034 DOI: 10.1016/j.msard.2021.103170] [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: 04/13/2021] [Revised: 06/28/2021] [Accepted: 07/22/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND Clinically stable multiple sclerosis (MS) patients often have negligible inflammatory MRI changes. Brain atrophy may provide insight into subclinical disease progression. The objective was to compare brain atrophy rates in stable patients on long term natalizumab treatment vs. age and gender matched healthy non-MS controls (HC) prospectively over two-years examining brain volume, cognition, and patient reported outcomes (PROs). METHODS MS patients treated with natalizumab for a minimum of 2 years, age 18-60 were recruited and compared with age- and gender-matched healthy controls (HC). Both groups were followed prospectively to obtain two years of consecutive magnetic resonance imaging, clinical and PRO data. Baseline normalized brain volume (NBV), yearly T2 lesion volume (T2LV), and percent brain volume change (PBVC) were measured using SIENAX, JIM 6.0, and SIENA respectively. Neuropsychological tests from the MACFIMS battery were selected to optimize assessments for impairments in the domains of information processing speed and memory. Patient reported outcomes (PROs) for domains of physical, mental and social quality of life were evaluated using the NeuroQol short forms. RESULTS Forty-eight natalizumab and 62 HC completed all study visits. At baseline, unadjusted mean NBV (natalizumab=1508.80cm (Popescu et al., 2013) vs. HC=1539.23cm (Popescu et al., 2013); p=0.033) and median baseline T2LV (natalizumab=1724.62mm (Popescu et al., 2013) vs. HC=44.20mm (Popescu et al., 2013); p=<0.0001) were different. The mean PBVC at year 2, adjusted for gender and baseline age was -0.57% (CI: 0.7620, -0.3716) for natalizumab and -0.50% (-0.7208, -0.2831) for HC, but the difference between groups was not statistically significant (0.073%; p=0.62). Over the 2-year period, HC demonstrated mild improvements in some cognitive tests vs. natalizumab subjects. However, PROs were similar between the two groups. CONCLUSION Stable MS patients on natalizumab have similar brain volume loss as people who do not have MS, suggesting normalization of brain atrophy.
Collapse
Affiliation(s)
- Enrique Alvarez
- Department of Neurology, Rocky Mountain Multiple Sclerosis Center at the University of Colorado, 1635 Aurora Court, Aurora, CO 80045 USA
| | - Kavita V Nair
- Department of Neurology, Rocky Mountain Multiple Sclerosis Center at the University of Colorado, 1635 Aurora Court, Aurora, CO 80045 USA; Department of Clinical Pharmacy, University of Colorado, 12850 East Montview Boulevard, Aurora, CO 80045 USA
| | - Brian D Hoyt
- Department of Neurosurgery, University of Colorado, 12631 East 17th Avenue, Aurora, CO 80045 USA
| | - Rebecca A Seale
- Department of Neurology, Rocky Mountain Multiple Sclerosis Center at the University of Colorado, 1635 Aurora Court, Aurora, CO 80045 USA
| | - Stefan Sillau
- Department of Neurology, Rocky Mountain Multiple Sclerosis Center at the University of Colorado, 1635 Aurora Court, Aurora, CO 80045 USA
| | - Augusto Miravalle
- Advanced Neurology, 2121 E Harmony Rd Ste #180, Fort Collins, CO 80528 USA
| | - Eric Engebretson
- Department of Neurology, Rocky Mountain Multiple Sclerosis Center at the University of Colorado, 1635 Aurora Court, Aurora, CO 80045 USA
| | - Brittany Schurr
- Department of Neurology, Rocky Mountain Multiple Sclerosis Center at the University of Colorado, 1635 Aurora Court, Aurora, CO 80045 USA
| | - John R Corboy
- Department of Neurology, Rocky Mountain Multiple Sclerosis Center at the University of Colorado, 1635 Aurora Court, Aurora, CO 80045 USA
| | - Timothy L Vollmer
- Department of Neurology, Rocky Mountain Multiple Sclerosis Center at the University of Colorado, 1635 Aurora Court, Aurora, CO 80045 USA
| | - Justin M Honce
- Department of Radiology, University of Colorado Hospital, 12401 East 17th Avenue, Aurora, CO 80045 USA.
| |
Collapse
|
50
|
Giedraitiene N, Drukteiniene E, Kizlaitiene R, Cimbalas A, Asoklis R, Kaubrys G. Cognitive Decline in Multiple Sclerosis Is Related to the Progression of Retinal Atrophy and Presence of Oligoclonal Bands: A 5-Year Follow-Up Study. Front Neurol 2021; 12:678735. [PMID: 34326806 PMCID: PMC8315759 DOI: 10.3389/fneur.2021.678735] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/14/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Brain atrophy, which is associated with cognitive impairment and retinal nerve fiber layer (RNFL) atrophy, is the main biomarker of neurodegeneration in multiple sclerosis (MS). However, data on the relationship between inflammatory markers, such as oligoclonal bands (OCBs) in the cerebrospinal fluid (CSF), and cognition, RNFL atrophy, and brain atrophy are scarce. The aim of this study was to assess the influence of RNFL thickness, brain atrophy markers, intrathecal OCBs, and the immunoglobulin G (IgG) index on cognitive decline over a 5-year period in patients with MS. Methods: This prospective, single-center, observational cohort study included 49 patients with relapsing MS followed up over 5 years. At baseline, the patients underwent brain magnetic resonance imaging (MRI). Cognitive evaluation was performed using the Brief International Cognitive Assessment for MS (BICAMS), and RNFL thickness was assessed using optical coherence tomography (OCT). OCBs and IgG levels in the CSF were evaluated at baseline. The BICAMS, OCT, and MRI findings were re-evaluated after 5 years. Results: A significant reduction in information processing speed, visual learning, temporal RNFL thickness, the Huckman index, and third ventricle mean diameter was found in all 49 patients with relapsing MS over the observation period (p < 0.05). Of the patients, 63.3% had positive OCBs and 59.2% had elevated IgG indices. The atrophy of the temporal segment and papillomacular bundle and the presence of OCBs were significantly related to a decline in information processing speed in these patients (p < 0.05). However, brain atrophy markers were not found to be significant on the general linear models. Conclusions: RNFL atrophy and the presence of OCBs were related to cognitive decline in patients with MS over a 5-year follow-up period, thereby suggesting their utility as potential biomarkers of cognitive decline in MS.
Collapse
Affiliation(s)
- Natasa Giedraitiene
- Center of Neurology, Clinic of Neurology and Neurosurgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Egle Drukteiniene
- Center of Eye Diseases, Clinic of Ear, Nose, Throat, and Eye Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Rasa Kizlaitiene
- Center of Neurology, Clinic of Neurology and Neurosurgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Andrius Cimbalas
- Center of Eye Diseases, Clinic of Ear, Nose, Throat, and Eye Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Rimvydas Asoklis
- Center of Eye Diseases, Clinic of Ear, Nose, Throat, and Eye Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Gintaras Kaubrys
- Center of Neurology, Clinic of Neurology and Neurosurgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|