1
|
Stuart CM, Varatharaj A, Zou Y, Darekar A, Domjan J, Gandini Wheeler-Kingshott CAM, Perry VH, Galea I. Systemic inflammation associates with and precedes cord atrophy in progressive multiple sclerosis. Brain Commun 2024; 6:fcae143. [PMID: 38712323 PMCID: PMC11073756 DOI: 10.1093/braincomms/fcae143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/05/2024] [Accepted: 04/17/2024] [Indexed: 05/08/2024] Open
Abstract
In preclinical models of multiple sclerosis, systemic inflammation has an impact on the compartmentalized inflammatory process within the central nervous system and results in axonal loss. It remains to be shown whether this is the case in humans, specifically whether systemic inflammation contributes to spinal cord or brain atrophy in multiple sclerosis. Hence, an observational longitudinal study was conducted to delineate the relationship between systemic inflammation and atrophy using magnetic resonance imaging: the SIMS (Systemic Inflammation in Multiple Sclerosis) study. Systemic inflammation and progression were assessed in people with progressive multiple sclerosis (n = 50) over two and a half years. Eligibility criteria included: (i) primary or secondary progressive multiple sclerosis; (ii) age ≤ 70; and (iii) Expanded Disability Status Scale ≤ 6.5. First morning urine was collected weekly to quantify systemic inflammation by measuring the urinary neopterin-to-creatinine ratio using a validated ultra-performance liquid chromatography mass spectrometry technique. The urinary neopterin-to-creatinine ratio temporal profile was characterized by short-term responses overlaid on a background level of inflammation, so these two distinct processes were considered as separate variables: background inflammation and inflammatory response. Participants underwent MRI at the start and end of the study, to measure cervical spinal cord and brain atrophy. Brain and cervical cord atrophy occurred on the study, but the most striking change was seen in the cervical spinal cord, in keeping with the corticospinal tract involvement that is typical of progressive disease. Systemic inflammation predicted cervical cord atrophy. An association with brain atrophy was not observed in this cohort. A time lag between systemic inflammation and cord atrophy was evident, suggesting but not proving causation. The association of the inflammatory response with cord atrophy depended on the level of background inflammation, in keeping with experimental data in preclinical models where the effects of a systemic inflammatory challenge on tissue injury depended on prior exposure to inflammation. A higher inflammatory response was associated with accelerated cord atrophy in the presence of background systemic inflammation below the median for the study population. Higher background inflammation, while associated with cervical cord atrophy itself, subdued the association of the inflammatory response with cord atrophy. Findings were robust to sensitivity analyses adjusting for potential confounders and excluding cases with new lesion formation. In conclusion, systemic inflammation associates with, and precedes, multiple sclerosis progression. Further work is needed to prove causation since targeting systemic inflammation may offer novel treatment strategies for slowing neurodegeneration in multiple sclerosis.
Collapse
Affiliation(s)
- Charlotte M Stuart
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Aravinthan Varatharaj
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Yukai Zou
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- Department of Medical Physics, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Angela Darekar
- Department of Medical Physics, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Janine Domjan
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Claudia A M Gandini Wheeler-Kingshott
- Department of Neuroinflammation, Faculty of Brain Sciences, NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Queen Square Institute of Neurology, University College London, London WC1B 5EH, UK
| | - V Hugh Perry
- School of Biological Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| |
Collapse
|
2
|
De Keersmaecker AV, Van Doninck E, Popescu V, Willem L, Cambron M, Laureys G, D’ Haeseleer M, Bjerke M, Roelant E, Lemmerling M, D’hooghe MB, Derdelinckx J, Reynders T, Willekens B. A metformin add-on clinical study in multiple sclerosis to evaluate brain remyelination and neurodegeneration (MACSiMiSE-BRAIN): study protocol for a multi-center randomized placebo controlled clinical trial. Front Immunol 2024; 15:1362629. [PMID: 38680485 PMCID: PMC11046490 DOI: 10.3389/fimmu.2024.1362629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/05/2024] [Indexed: 05/01/2024] Open
Abstract
Introduction Despite advances in immunomodulatory treatments of multiple sclerosis (MS), patients with non-active progressive multiple sclerosis (PMS) continue to face a significant unmet need. Demyelination, smoldering inflammation and neurodegeneration are important drivers of disability progression that are insufficiently targeted by current treatment approaches. Promising preclinical data support repurposing of metformin for treatment of PMS. The objective of this clinical trial is to evaluate whether metformin, as add-on treatment, is superior to placebo in delaying disease progression in patients with non-active PMS. Methods and analysis MACSiMiSE-BRAIN is a multi-center two-arm, 1:1 randomized, triple-blind, placebo-controlled clinical trial, conducted at five sites in Belgium. Enrollment of 120 patients with non-active PMS is planned. Each participant will undergo a screening visit with assessment of baseline magnetic resonance imaging (MRI), clinical tests, questionnaires, and a safety laboratory assessment. Following randomization, participants will be assigned to either the treatment (metformin) or placebo group. Subsequently, they will undergo a 96-week follow-up period. The primary outcome is change in walking speed, as measured by the Timed 25-Foot Walk Test, from baseline to 96 weeks. Secondary outcome measures include change in neurological disability (Expanded Disability Status Score), information processing speed (Symbol Digit Modalities Test) and hand function (9-Hole Peg test). Annual brain MRI will be performed to assess evolution in brain volumetry and diffusion metrics. As patients may not progress in all domains, a composite outcome, the Overall Disability Response Score will be additionally evaluated as an exploratory outcome. Other exploratory outcomes will consist of paramagnetic rim lesions, the 2-minute walking test and health economic analyses as well as both patient- and caregiver-reported outcomes like the EQ-5D-5L, the Multiple Sclerosis Impact Scale and the Caregiver Strain Index. Ethics and dissemination Clinical trial authorization from regulatory agencies [Ethical Committee and Federal Agency for Medicines and Health Products (FAMHP)] was obtained after submission to the centralized European Clinical Trial Information System. The results of this clinical trial will be disseminated at scientific conferences, in peer-reviewed publications, to patient associations and the general public. Trial registration ClinicalTrials.gov Identifier: NCT05893225, EUCT number: 2023-503190-38-00.
Collapse
Affiliation(s)
- Anna-Victoria De Keersmaecker
- Department of Neurology, Antwerp University Hospital, Edegem, Belgium
- Translational Neurosciences Research Group, Faculty of Medicine and Health Sciences, University of Antwerp, Edegem, Belgium
| | - Eline Van Doninck
- Department of Family Medicine and Population Health, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
- Center of Health Economic Research and Modelling Infectious Diseases, University of Antwerp, Wilrijk, Belgium
| | - Veronica Popescu
- Immunology and Infection, University of Hasselt, Diepenbeek, Belgium
- Biomedical Research Institute, University of Hasselt, Diepenbeek, Belgium
- Department of Neurology, Noorderhart Maria Hospital, Pelt, Belgium
- University Multiple Sclerosis Centre, University of Hasselt, Hasselt, Belgium
| | - Lander Willem
- Department of Family Medicine and Population Health, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
- Center of Health Economic Research and Modelling Infectious Diseases, University of Antwerp, Wilrijk, Belgium
| | - Melissa Cambron
- Faculty of Medicine and Health Sciences, University of Ghent, Ghent, Belgium
- Department of Neurology, Algemeen Ziekenhuis Sint Jan, Bruges, Belgium
| | - Guy Laureys
- Faculty of Medicine and Health Sciences, University of Ghent, Ghent, Belgium
- Department of Neurology, University Hospital Ghent, Ghent, Belgium
| | - Miguel D’ Haeseleer
- Department of Neurology, University Hospital Brussels, Brussels, Belgium
- Department of Neurology, National Multiple Sclerosis Center, Melsbroek, Belgium
- Department Neuroprotection and Neuromodulation, Center for Neurosciences, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Maria Bjerke
- Department Neuroprotection and Neuromodulation, Center for Neurosciences, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
- Neurochemistry Laboratory, Department of Clinical Biology, Brussels, University Hospital Brussels, Brussels, Belgium
- Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Ella Roelant
- Clinical Trial Center, Antwerp University Hospital, Edegem, Belgium
| | - Marc Lemmerling
- Department of Radiology, Antwerp University Hospital, Edegem, Wilrijk, Belgium
| | - Marie Beatrice D’hooghe
- Department of Neurology, University Hospital Brussels, Brussels, Belgium
- Department of Neurology, National Multiple Sclerosis Center, Melsbroek, Belgium
- Department Neuroprotection and Neuromodulation, Center for Neurosciences, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Judith Derdelinckx
- Department of Neurology, Antwerp University Hospital, Edegem, Belgium
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Tatjana Reynders
- Department of Neurology, Antwerp University Hospital, Edegem, Belgium
- Translational Neurosciences Research Group, Faculty of Medicine and Health Sciences, University of Antwerp, Edegem, Belgium
| | - Barbara Willekens
- Department of Neurology, Antwerp University Hospital, Edegem, Belgium
- Translational Neurosciences Research Group, Faculty of Medicine and Health Sciences, University of Antwerp, Edegem, Belgium
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
3
|
Nakamura K, Elliott C, Lee H, Narayanan S, Arnold DL. Brain volume increase after discontinuing natalizumab therapy: Evidence for reversible pseudoatrophy. Mult Scler Relat Disord 2024; 81:105123. [PMID: 37976981 DOI: 10.1016/j.msard.2023.105123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 09/02/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND The phenomenon of pseudoatropy after initiation of anti-inflammatory therapy is believed to be reversible, but a rebound in brain volume following cessation of highly-effective therapy has not been reported. OBJECTIVES To evaluate brain volume change in a treatment interruption study (RESTORE) in which relapsing-remitting multiple sclerosis (RRMS) patients were randomized to switch from natalizumab to placebo, from natalizumab to once-monthly intravenous methylprednisolone (IVMP), or to remain on natalizumab. METHODS T2 lesion volume (T2LV), baseline normalized brain volumes, and follow-up percent brain volume changes (PBVC) were calculated. Approximate T2 relaxation-time (pT2) was calculated within the brain mask and the T2 lesions to estimate changes in water content. Linear mixed effects models were used to detect differences in T2LV, pT2 in whole brain, pT2 in T2-weighted lesions, and PBVC among the placebo, natalizumab, and IVMP groups. We also estimated contributions of T2LV and pT2 (in whole brain and T2 lesions) to PBVC. RESULTS T2LV increased in the placebo group (by 0.66 ml/year, p<0.0001) and IVMP (+1.98 ml/year, p = 0.05) groups relative to the natalizumab group. The rates of PBVC were significantly different: -0.239%/year with continued natalizumab and +0.126 %/year after switch to placebo (p = 0.03), while the IVMP group showed brain volume loss (-0.74 %/ year, p = 0.08). pT2 was not statistically different between the groups (p ≥ 0.29) and did not have significant effects on PBVC (p ≥ 0.25). CONCLUSION The increase in the brain volume in patients witching from natalizumab to placebo is consistent with reversal of so-called pseudoatrophy after starting natalizumab.
Collapse
Affiliation(s)
- Kunio Nakamura
- McConnell Brain Imaging Centre, Montreal Neurological Institute Hospital, and Department of Neurology and Neurosurgery, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, ND20, Cleveland, Ohio 44195, USA.
| | - Colm Elliott
- Centre for Intelligent Machines, McGill University, 3480 Rue University, Montréal, QC H3A 2A7, Canada. NeuroRx Research, 3575 Park Avenue, Suite #5322, Montreal, Quebec H2 × 4B3, Canada; NeuroRx Research, 3575 Park Avenue, Suite #5322, Montreal, Quebec H2 × 4B3, Canada
| | - Hyunwoo Lee
- McConnell Brain Imaging Centre, Montreal Neurological Institute Hospital, and Department of Neurology and Neurosurgery, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; Division of Neurology, Department of Medicine, University of British Columbia S154-2211 Wesbrook Mall, Vancouver, BC V6T2B5, Canada
| | - Sridar Narayanan
- McConnell Brain Imaging Centre, Montreal Neurological Institute Hospital, and Department of Neurology and Neurosurgery, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; NeuroRx Research, 3575 Park Avenue, Suite #5322, Montreal, Quebec H2 × 4B3, Canada
| | - Douglas L Arnold
- McConnell Brain Imaging Centre, Montreal Neurological Institute Hospital, and Department of Neurology and Neurosurgery, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; NeuroRx Research, 3575 Park Avenue, Suite #5322, Montreal, Quebec H2 × 4B3, Canada
| |
Collapse
|
4
|
Morgan KE, White IR, Frost C. How important is the linearity assumption in a sample size calculation for a randomised controlled trial where treatment is anticipated to affect a rate of change? BMC Med Res Methodol 2023; 23:274. [PMID: 37990159 PMCID: PMC10664473 DOI: 10.1186/s12874-023-02093-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 11/03/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND For certain conditions, treatments aim to lessen deterioration over time. A trial outcome could be change in a continuous measure, analysed using a random slopes model with a different slope in each treatment group. A sample size for a trial with a particular schedule of visits (e.g. annually for three years) can be obtained using a two-stage process. First, relevant (co-) variances are estimated from a pre-existing dataset e.g. an observational study conducted in a similar setting. Second, standard formulae are used to calculate sample size. However, the random slopes model assumes linear trajectories with any difference in group means increasing proportionally to follow-up time. The impact of these assumptions failing is unclear. METHODS We used simulation to assess the impact of a non-linear trajectory and/or non-proportional treatment effect on the proposed trial's power. We used four trajectories, both linear and non-linear, and simulated observational studies to calculate sample sizes. Trials of this size were then simulated, with treatment effects proportional or non-proportional to time. RESULTS For a proportional treatment effect and a trial visit schedule matching the observational study, powers are close to nominal even for non-linear trajectories. However, if the schedule does not match the observational study, powers can be above or below nominal levels, with the extent of this depending on parameters such as the residual error variance. For a non-proportional treatment effect, using a random slopes model can lead to powers far from nominal levels. CONCLUSIONS If trajectories are suspected to be non-linear, observational data used to inform power calculations should have the same visit schedule as the proposed trial where possible. Additionally, if the treatment effect is expected to be non-proportional, the random slopes model should not be used. A model allowing trajectories to vary freely over time could be used instead, either as a second line analysis method (bearing in mind that power will be lost) or when powering the trial.
Collapse
Affiliation(s)
- Katy E Morgan
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK.
| | - Ian R White
- MRC Clinical Trials Unit at UCL, University College London, London, UK
| | - Chris Frost
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
5
|
Singh A, Arora S, Chavan M, Shahbaz S, Jabeen H. An Overview of the Neurotrophic and Neuroprotective Properties of the Psychoactive Drug Lithium as an Autophagy Modulator in Neurodegenerative Conditions. Cureus 2023; 15:e44051. [PMID: 37746513 PMCID: PMC10517711 DOI: 10.7759/cureus.44051] [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] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
For both short-term and long-term treatment of bipolar disorder, lithium is a prototypical mood stabilizer. Lithium's neuroprotective properties were revealed by cumulative translational research, which opened the door to reforming the chemical as a treatment for neurodegenerative illnesses. The control of homeostatic systems such as oxidative stress, autophagy, apoptosis, mitochondrial function, and inflammation underlies lithium's neuroprotective characteristics. The fact that lithium inhibits the enzymes inositol monophosphatase (IMPase) and glycogen synthase kinase (GSK)-3 may be the cause of the various intracellular reactions. In this article, we review lithium's neurobiological properties, as demonstrated by its neurotrophic and neuroprotective capabilities, as well as translational studies in cells in culture and in animal models of Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Prion disease, amyotrophic lateral sclerosis (ALS), ischemic stroke, and neuronal ceroid lipofuscinosis (NCL), discussing the justification for the drug's use in the treatment of these neurodegenerative disorders.
Collapse
Affiliation(s)
- Ajay Singh
- Internal Medicine, Sri Ram Murti Smarak Institute of Medical Sciences, Bareilly, IND
| | - Sanjiya Arora
- Health Department, Sub District Hospital (SDH) cum Civil Hospital, Fatehabad, Fatehabad, IND
| | - Manisha Chavan
- Internal Medicine, Kakatiya Medical College, Rangam Peta, Warangal, IND
| | - Samen Shahbaz
- Internal Medicine, Faisalabad Medical University, Faisalabad, PAK
| | - Hafsa Jabeen
- Internal Medicine, Dow University of Health Sciences, Nanakwara, PAK
| |
Collapse
|
6
|
N-Acetyl Cysteine as a Neuroprotective Agent in Progressive Multiple Sclerosis (NACPMS) trial: Study protocol for a randomized, double-blind, placebo-controlled add-on phase 2 trial. Contemp Clin Trials 2022; 122:106941. [PMID: 36182028 DOI: 10.1016/j.cct.2022.106941] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/16/2022] [Accepted: 09/25/2022] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Patients with progressive multiple sclerosis (PMS) experience relentless disability worsening. Current approved therapies have very modest effects on disability progression and purely focus on immunomodulation. While some inflammatory processes exist in non-active PMS, other biological processes such as neuronal injury from oxidative stress are likely more critical. N-acetyl cysteine (NAC) directly scavenges free radicals and restores neuronal glutathione, a major endogenous antioxidant. Our group has recently evaluated the safety of high dose NAC in a pilot trial in PMS with no tolerability concerns. We aim now to assess the safety, tolerability, and effect of NAC on progression of several MRI, clinical and biological markers in PMS patients. METHODS The NACPMS trial is a multi-site, randomized, double-blind, parallel-group, placebo-controlled add-on phase 2 trial. Ninety-eight PMS patients with EDSS 3.0-7.0 and aged 40-70 years will be randomized to NAC 1200 mg TID or matching placebo (1:1) as an add-on to the standard of care stratified by site and disease type during a 15-month intervention period. It is hypothesized that a reduction in oxidative stress injury will lessen brain atrophy estimated by MRI. The primary outcome analysis will compare the percent change over 12 months (Month 15 vs Month 3) between treatment and control arms using multivariable linear regression adjusted by age, sex, and disease duration. ETHICS This study was approved by the Institutional Review Board at the University of California, San Francisco (IRB21-34143), and an Investigational New Drug approval was obtained from the FDA (IND127184). TRIAL REGISTRATION NCT05122559.
Collapse
|
7
|
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: 0.5] [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
|
8
|
Rust R, Chien C, Scheel M, Brandt AU, Dörr J, Wuerfel J, Klumbies K, Zimmermann H, Lorenz M, Wernecke KD, Bellmann-Strobl J, Paul F. Epigallocatechin Gallate in Progressive MS: A Randomized, Placebo-Controlled Trial. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:e964. [PMID: 33622766 PMCID: PMC7954462 DOI: 10.1212/nxi.0000000000000964] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/17/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To examine whether treatment with epigallocatechin gallate (EGCG) influences progression of brain atrophy, reduces clinical and further radiologic disease activity markers, and is safe in patients with progressive multiple sclerosis (PMS). METHODS We enrolled 61 patients with primary or secondary PMS in a randomized double-blind, parallel-group, phase II trial on oral EGCG (up to 1,200 mg daily) or placebo for 36 months with an optional open-label EGCG treatment extension (OE) of 12-month duration. The primary end point was the rate of brain atrophy, quantified as brain parenchymal fraction (BPF). The secondary end points were radiologic and clinical disease parameters and safety assessments. RESULTS In our cohort, 30 patients were randomized to EGCG treatment and 31 to placebo. Thirty-eight patients (19 from each group) completed the study. The primary endpoint was not met, as in 36 months the rate of decrease in BPF was 0.0092 ± 0.0152 in the treatment group and -0.0078 ± 0.0159 in placebo-treated patients. None of the secondary MRI and clinical end points revealed group differences. Adverse events of EGCG were mostly mild and occurred with a similar incidence in the placebo group. One patient in the EGCG group had to stop treatment due to elevated aminotransferases (>3.5 times above normal limit). CONCLUSIONS In a phase II trial including patients with multiple sclerosis (MS) with progressive disease course, we were unable to demonstrate a treatment effect of EGCG on the primary and secondary radiologic and clinical disease parameters while confirming on overall beneficial safety profile. CLINICALTRIALGOV IDENTIFIER NCT00799890. CLASSIFICATION OF EVIDENCE This phase II trial provides Class II evidence that for patients with PMS, EGCG was safe, well tolerated, and did not significantly reduce the rate of brain atrophy.
Collapse
Affiliation(s)
- Rebekka Rust
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Claudia Chien
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Michael Scheel
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Alexander U Brandt
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Jan Dörr
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Jens Wuerfel
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Katharina Klumbies
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Hanna Zimmermann
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Mario Lorenz
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Klaus-Dieter Wernecke
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Judith Bellmann-Strobl
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Friedemann Paul
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland.
| |
Collapse
|
9
|
Bautin P, Cohen-Adad J. Minimum detectable spinal cord atrophy with automatic segmentation: Investigations using an open-access dataset of healthy participants. NEUROIMAGE: CLINICAL 2021; 32:102849. [PMID: 34624638 PMCID: PMC8503570 DOI: 10.1016/j.nicl.2021.102849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/07/2021] [Accepted: 09/28/2021] [Indexed: 11/20/2022] Open
Abstract
Evaluate the robustness of an automated analysis pipeline for detecting SC atrophy. Simulate spinal cord atrophy and scan-rescan variability. Fully automated analysis method available on an open access database. Evaluation of sample size and inter/intra-subject variability for T1w and T2w images.
Spinal cord atrophy is a well-known biomarker in multiple sclerosis (MS) and other diseases. It is measured by segmenting the spinal cord on an MRI image and computing the average cross-sectional area (CSA) over a few slices. Introduced about 25 years ago, this procedure is highly sensitive to the quality of the segmentation and is prone to rater-bias. Recently, fully-automated spinal cord segmentation methods, which remove the rater-bias and enable the automated analysis of large populations, have been introduced. A lingering question related to these automated methods is: How reliable are they at detecting atrophy? In this study, we evaluated the precision and accuracy of automated atrophy measurements by simulating scan-rescan experiments. Spinal cord MRI data from the open-access spine-generic project were used. The dataset aggregates 42 sites worldwide and consists of 260 healthy subjects and includes T1w and T2w contrasts. To simulate atrophy, each volume was globally rescaled at various scaling factors. Moreover, to simulate patient repositioning, random rigid transformations were applied. Using the DeepSeg algorithm from the Spinal Cord Toolbox, the spinal cord was segmented and vertebral levels were identified. Then, the average CSA between C3-C5 vertebral levels was computed for each Monte Carlo sample, allowing us to derive measures of atrophy, intra/inter-subject variability, and sample-size calculations. The minimum sample size required to detect an atrophy of 2% between unpaired study arms, commonly seen in MS studies, was 467 +/− 13.9 using T1w and 467 +/− 3.2 using T2w images. The minimum sample size to detect a longitudinal atrophy (between paired study arms) of 0.8% was 60 +/− 25.1 using T1w and 10 +/− 1.2 using T2w images. At the intra-subject level, the estimated CSA, observed in this study, showed good precision compared to other studies with COVs (across Monte Carlo transformations) of 0.8% for T1w and 0.6% for T2w images. While these sample sizes seem small, we would like to stress that these results correspond to a “best case” scenario, in that the dataset used here was of particularly good quality and the model for simulating atrophy does not encompass all the variability met in real-life datasets. The simulated atrophy and scan-rescan variability may over-simplify the biological reality. The proposed framework is open-source and available at https://csa-atrophy.readthedocs.io/.
Collapse
Affiliation(s)
- Paul Bautin
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada; Functional Neuroimaging Unit, CRIUGM, Université de Montréal, Montreal, QC, Canada; Mila - Quebec AI Institute, Montreal, QC, Canada.
| |
Collapse
|
10
|
Stuart CM, Varatharaj A, Domjan J, Philip S, Galea I. Physical activity monitoring to assess disability progression in multiple sclerosis. Mult Scler J Exp Transl Clin 2020; 6:2055217320975185. [PMID: 33343919 PMCID: PMC7727071 DOI: 10.1177/2055217320975185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/31/2020] [Indexed: 11/16/2022] Open
Abstract
Background Clinical outcome measurement in multiple sclerosis (MS) usually requires a physical visit. Remote activity monitoring (RAM) using wearable technology provides a rational alternative, especially desirable when distance is involved or in a pandemic setting. Objective To validate RAM in progressive MS using (1) traditional psychometric methods (2) brain atrophy. Methods 56 people with progressive MS participated in a longitudinal study over 2.5 years. An arm-worn RAM device measured activity over six days, every six months, and incorporated triaxial accelerometry and transcutaneous physiological variable measurement. Five RAM variables were assessed: physical activity duration, step count, active energy expenditure, metabolic equivalents and a composite RAM score incorporating all four variables. Other assessments every six months included EDSS, MSFC, MSIS-29, Chalder Fatigue Scale and Beck’s Depression Inventory. Annualized brain atrophy was measured using SIENA. Results RAM was tolerated well by people with MS; the device was worn 99.4% of the time. RAM had good convergent and divergent validity and was responsive, especially with respect to step count. Measurement of physical activity over one day was as responsive as six days. The composite RAM score positively correlated with brain volume loss. Conclusion Remote activity monitoring is a valid and acceptable outcome measure in MS.
Collapse
Affiliation(s)
- Charlotte M Stuart
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Aravinthan Varatharaj
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Janine Domjan
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Sheaba Philip
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | |
Collapse
|
11
|
Spinal cord atrophy in a primary progressive multiple sclerosis trial: Improved sample size using GBSI. NEUROIMAGE-CLINICAL 2020; 28:102418. [PMID: 32961403 PMCID: PMC7509079 DOI: 10.1016/j.nicl.2020.102418] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 08/29/2020] [Accepted: 09/03/2020] [Indexed: 01/18/2023]
Abstract
The GBSI provided clinically meaningful measurements of spinal cord atrophy, with low sample size. Deriving spinal cord atrophy from brain MRI using the GBSI is easier than spinal cord MRI. Spinal cord atrophy on GBSI could be used as a secondary outcome measure.
Background We aimed to evaluate the implications for clinical trial design of the generalised boundary-shift integral (GBSI) for spinal cord atrophy measurement. Methods We included 220 primary-progressive multiple sclerosis patients from a phase 2 clinical trial, with baseline and week-48 3DT1-weighted MRI of the brain and spinal cord (1 × 1 × 1 mm3), acquired separately. We obtained segmentation-based cross-sectional spinal cord area (CSA) at C1-2 (from both brain and spinal cord MRI) and C2-5 levels (from spinal cord MRI) using DeepSeg, and, then, we computed corresponding GBSI. Results Depending on the spinal cord segment, we included 67.4–98.1% patients for CSA measurements, and 66.9–84.2% for GBSI. Spinal cord atrophy measurements obtained with GBSI had lower measurement variability, than corresponding CSA. Looking at the image noise floor, the lowest median standard deviation of the MRI signal within the cerebrospinal fluid surrounding the spinal cord was found on brain MRI at the C1-2 level. Spinal cord atrophy derived from brain MRI was related to the corresponding measures from dedicated spinal cord MRI, more strongly for GBSI than CSA. Spinal cord atrophy measurements using GBSI, but not CSA, were associated with upper and lower limb motor progression. Discussion Notwithstanding the reduced measurement variability, the clinical correlates, and the possibility of using brain acquisitions, spinal cord atrophy using GBSI should remain a secondary outcome measure in MS studies, until further advancements increase the quality of acquisition and reliability of processing.
Collapse
|
12
|
Kapoor R, Smith KE, Allegretta M, Arnold DL, Carroll W, Comabella M, Furlan R, Harp C, Kuhle J, Leppert D, Plavina T, Sellebjerg F, Sincock C, Teunissen CE, Topalli I, von Raison F, Walker E, Fox RJ. Serum neurofilament light as a biomarker in progressive multiple sclerosis. Neurology 2020; 95:436-444. [PMID: 32675076 PMCID: PMC7538221 DOI: 10.1212/wnl.0000000000010346] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 06/26/2020] [Indexed: 01/06/2023] Open
Abstract
There is an unmet need in multiple sclerosis (MS) therapy for treatments to stop progressive disability. The development of treatments may be accelerated if novel biomarkers are developed to overcome the limitations of traditional imaging outcomes revealed in early phase trials. In January 2019, the International Progressive MS Alliance convened a standing expert panel to consider potential tissue fluid biomarkers in MS in general and in progressive MS specifically. The panel focused their attention on neurofilament light chain (NfL) in serum or plasma, examining data from both relapsing and progressive MS. Here, we report the initial conclusions of the panel and its recommendations for further research. Serum NfL (sNfL) is a plausible marker of neurodegeneration that can be measured accurately, sensitively, and reproducibly, but standard procedures for sample processing and analysis should be established. Findings from relapsing and progressive cohorts concur and indicate that sNfL concentrations correlate with imaging and disability measures, predict the future course of the disease, and can predict response to treatment. Importantly, disease activity from active inflammation (i.e., new T2 and gadolinium-enhancing lesions) is a large contributor to sNfL, so teasing apart disease activity from the disease progression that drives insidious disability progression in progressive MS will be challenging. More data are required on the effects of age and comorbidities, as well as the relative contributions of inflammatory activity and other disease processes. The International Progressive MS Alliance is well positioned to advance these initiatives by connecting and supporting relevant stakeholders in progressive MS.
Collapse
Affiliation(s)
- Raju Kapoor
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Kathryn E Smith
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Mark Allegretta
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Douglas L Arnold
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - William Carroll
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Manuel Comabella
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Roberto Furlan
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Christopher Harp
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Jens Kuhle
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - David Leppert
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Tatiana Plavina
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Finn Sellebjerg
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Caroline Sincock
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Charlotte E Teunissen
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Ilir Topalli
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Florian von Raison
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Elizabeth Walker
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic
| | - Robert J Fox
- From the University College London (R.K.), United Kingdom; National Multiple Sclerosis Society (K.E.S., M.A.), New York; McGill University (D.L.A.), Montreal, Canada; Perron Institute (W.C.), Sir Charles Gairdner Hospital, Perth, Australia; University Hospital Vall d'Hebron (M.C.), Barcelona, Spain; San Raffaele Scientific Institute (R.F.), Milan, Italy; Genentech/Roche (C.H.), South San Francisco; University Hospital Basel (J.K., D.L.), Switzerland; Biogen (T.P.), Boston; Quanterix Corporation (T.P.), Billerica; Rigshospitalet (F.S.), University of Copenhagen, Denmark; Progressive Multiple Sclerosis Alliance (C.S.), Glasgow, United Kingdom; Amsterdam UMC (C.E.T.), the Netherlands; MedDay Pharma (I.T.), Paris, France; Novartis (F.v.R.), Basel, Switzerland; Elizabeth Walker Consulting (E.W.), Seattle; and Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic.
| |
Collapse
|
13
|
Kosa P, Wu T, Phillips J, Leinonen M, Masvekar R, Komori M, Wichman A, Sandford M, Bielekova B. Idebenone does not inhibit disability progression in primary progressive MS. Mult Scler Relat Disord 2020; 45:102434. [PMID: 32784117 PMCID: PMC9386688 DOI: 10.1016/j.msard.2020.102434] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/23/2020] [Accepted: 07/29/2020] [Indexed: 12/30/2022]
Abstract
Background: Multiple sclerosis (MS) is a chronic, immune-mediated neurodegenerative disorder of the central nervous system (CNS). While current MS therapies target the inflammatory processes, no treatment explicitly targets mitochondrial dysfunction and resulting axonal loss. Therefore, the aim of this study was to determine whether idebenone inhibits mitochondrial dysfunction and accumulation of disability in primary progressive MS (PPMS) and to enhance understanding of pathogenic mechanisms of PPMS progression using cerebrospinal fluid (CSF) biomarkers. Methods: The double-blind, placebo-controlled Phase I/II clinical trial of Idebenone in patients with Primary Progressive MS (IPPoMS; NCT00950248) was an adaptively designed, baseline-versus-treatment, placebo-controlled, CSF-biomarker-supported trial. Based on interim analysis of the 1-year pre-treatment data, change in the area under the curve of Combinatorial Weight-Adjusted Disability Score (CombiWISE) became the primary outcome, with >80% power to detect ≥40% efficacy with 28 patients/arm treated for 2 years in baseline versus treatment paradigm. Changes in traditional disability scales and in brain ventricular volume were secondary outcomes. Exploratory outcomes included CSF biomarkers of mitochondrial dysfunction (Growth/differentiation factor 15 [GDF15] and lactate), axonal damage (neurofilament light chain [NFL]), innate immunity (sCD14), blood brain barrier leakage (albumin quotient) and retinal nerve fiber layer thinning. Results: Idebenone was well tolerated but did not inhibit disability progression or CNS tissue destruction. Concentrations of GDF15, secreted predominantly by astrocytes and choroid plexus epithelium in vitro, increased after exposure to mitochondrial toxin rotenone, validating the ability of this biomarker to measure intrathecal mitochondrial damage. CSF GDF15 levels correlated strongly with age and MS patients had CSF levels of GDF15 significantly above age-adjusted healthy volunteers, with highest levels measured in PPMS. Idebenone did not change CSF GDF15 levels. Conclusion: Mitochondrial dysfunction exceeding normal aging reflected by age-adjusted CSF GDF15 is present in the majority of PPMS patients, but it is not inhibited by idebenone.
Collapse
Affiliation(s)
- Peter Kosa
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tianxia Wu
- Clinical trials Unit, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan Phillips
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mika Leinonen
- Santhera Pharmaceuticals (Switzerland) AG, Pratteln Switzerland
| | - Ruturaj Masvekar
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mika Komori
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alison Wichman
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mary Sandford
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bibiana Bielekova
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
14
|
Rinker JR, Meador WR, King P. Randomized feasibility trial to assess tolerance and clinical effects of lithium in progressive multiple sclerosis. Heliyon 2020; 6:e04528. [PMID: 32760832 PMCID: PMC7393418 DOI: 10.1016/j.heliyon.2020.e04528] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/14/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Disability accumulation in progressive multiple sclerosis (MS) results from inflammatory and neurodegenerative mechanisms. In animal models of MS, lithium acts to reduce inflammatory demyelination, and in models of neurodegenerative diseases, lithium also slows neuronal death. Prospective studies of lithium in MS patients have not been previously undertaken. OBJECTIVE To determine the tolerance and feasibility of using low-dose (150-300 mg/daily) lithium as a pharmaceutical intervention in a cohort of subjects with progressive MS, and to gauge preliminary effects of lithium on change in brain volume over time. METHODS Patients with primary or secondary progressive MS were recruited into a 2-year, single-blind crossover trial in which subjects were randomly assigned to take lithium in year 1 or 2. The primary outcomes of interest were tolerance of lithium and percentage brain volume change (PBVC) on vs. off lithium. Secondary outcomes included relapse rates, disability changes, and self-report scales assessing fatigue, mood, and quality of life (QOL). RESULTS Of 24 screened patients, 23 were randomized to take lithium during year 1 (n = 11) or 2 (n = 12). Two subjects discontinued the trial due to lithium side effects. Other reasons for discontinuation included personal reasons (n = 2), worsening MS (n = 1), and development of multiple myeloma (n = 1). For the 17 who completed the trial, change in PBVC on lithium (+0.107) did not significantly differ from the observation period (-0.355, p = 0.346). Disability measured by Expanded Disability Status Scale and MS Functional Composite did not differ by lithium treatment status. On patient reported measures of mental well-being, subjects reported fewer depressive symptoms on the Beck Depression Inventory (12.3 vs. 15.8, p = 0.016) and more favorably on the mental domains of the MSQOL inventory (56.7 vs. 52.4, p = 0.028). CONCLUSIONS Low-dose lithium is well tolerated in persons with MS. Taking lithium did not result in differences in PBVC, relapses, or disability, but conclusions were limited by study design and sample size. Despite concern for lithium-associated neurological side effects, subjects taking lithium did not report worsened fatigue or physical well-being. On measures of mood and mental health QOL, subjects scored more favorably while taking lithium. CLINICALTRIALSGOV IDENTIFIER NCT01259388.
Collapse
Affiliation(s)
- John R. Rinker
- Department of Neurology, University of Alabama at Birmingham, 1720 7 Avenue South, Birmingham, AL, 35294, USA
- Birmingham VA Medical Center, 700 19 Street South, Birmingham, AL, 35233, USA
| | - William R. Meador
- Department of Neurology, University of Alabama at Birmingham, 1720 7 Avenue South, Birmingham, AL, 35294, USA
| | - Peter King
- Birmingham VA Medical Center, 700 19 Street South, Birmingham, AL, 35233, USA
| |
Collapse
|
15
|
Ciampi E, Uribe-San-Martin R, Cárcamo C, Cruz JP, Reyes A, Reyes D, Pinto C, Vásquez M, Burgos RA, Hancke J. Efficacy of andrographolide in not active progressive multiple sclerosis: a prospective exploratory double-blind, parallel-group, randomized, placebo-controlled trial. BMC Neurol 2020; 20:173. [PMID: 32380977 PMCID: PMC7203851 DOI: 10.1186/s12883-020-01745-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/23/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic immune mediated disease and the progressive phase appears to have significant neurodegenerative mechanisms. The classification of the course of progressive MS (PMS) has been re-organized into categories of active vs. not active inflammatory disease and the presence vs. absence of gradual disease progression. Clinical trial experience to date in PMS with anti-inflammatory medications has shown limited effect. Andrographolide is a new class of anti-inflammatory agent, that has been proposed as a potential drug for autoimmune disorders, including MS. In the present trial, we perform an exploratory pilot study on the efficacy and safety of andrographolide (AP) compared to placebo in not active PMS. METHODS A pilot clinical trial using 140 mg oral AP or placebo twice daily for 24 months in patients with not active primary or secondary progressive MS was conducted. The primary efficacy endpoint was the mean percentage brain volume change (mPBVC). Secondary efficacy endpoints included 3-month confirmed disability progression (3-CDP) and mean EDSS change. RESULTS Forty-four patients were randomized: 23 were assigned to the AP group, and 21 were assigned to the placebo group. The median baseline EDSS of both groups was 6.0. Annualized mPBVC was - 0.679% for the AP group and - 1.069% for the placebo group (mean difference: -0.39; 95% CI [- 0.836-0.055], p = 0.08, relative reduction: 36.5%). In the AP group, 30% had 3-CDP compared to 41% in the placebo group (HR: 0.596; 95% CI [0.200-1.777], p = 0.06). The mean EDSS change was - 0.025 in the AP group and + 0.352 in the placebo group (mean difference: 0.63, p = 0.042). Adverse events related to AP were mild rash and dysgeusia. CONCLUSIONS AP was well tolerated and showed a potential effect in reducing brain atrophy and disability progression, that need to be further evaluated in a larger clinical trial. TRIAL REGISTRATION ClinicalTrials.gov NCT02273635 retrospectively registered on October 24th, 2014.
Collapse
Affiliation(s)
- Ethel Ciampi
- Neurology Department, Faculty of Medicine, Pontificia Universidad Católica de Chile, Diagonal Paraguay, 362, 5° floor, Santiago, Chile. .,Neurology, Hospital Dr. Sótero del Río, Av. Concha y Toro, 3459, Santiago, Chile.
| | - Reinaldo Uribe-San-Martin
- Neurology Department, Faculty of Medicine, Pontificia Universidad Católica de Chile, Diagonal Paraguay, 362, 5° floor, Santiago, Chile.,Neurology, Hospital Dr. Sótero del Río, Av. Concha y Toro, 3459, Santiago, Chile
| | - Claudia Cárcamo
- Neurology Department, Faculty of Medicine, Pontificia Universidad Católica de Chile, Diagonal Paraguay, 362, 5° floor, Santiago, Chile.
| | - Juan Pablo Cruz
- Radiology, Pontificia Universidad Católica de Chile, Diagonal Paraguay, 362, 5° floor, Santiago, Chile
| | - Ana Reyes
- Neurology Department, Faculty of Medicine, Pontificia Universidad Católica de Chile, Diagonal Paraguay, 362, 5° floor, Santiago, Chile
| | - Diego Reyes
- Neurology Department, Faculty of Medicine, Pontificia Universidad Católica de Chile, Diagonal Paraguay, 362, 5° floor, Santiago, Chile
| | - Carmen Pinto
- Neurology Department, Faculty of Medicine, Pontificia Universidad Católica de Chile, Diagonal Paraguay, 362, 5° floor, Santiago, Chile
| | - Macarena Vásquez
- Neurology Department, Faculty of Medicine, Pontificia Universidad Católica de Chile, Diagonal Paraguay, 362, 5° floor, Santiago, Chile
| | - Rafael A Burgos
- Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Independencia, 613, Valdivia, Chile
| | - Juan Hancke
- Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Independencia, 613, Valdivia, Chile
| |
Collapse
|
16
|
Efficacy of three neuroprotective drugs in secondary progressive multiple sclerosis (MS-SMART): a phase 2b, multiarm, double-blind, randomised placebo-controlled trial. Lancet Neurol 2020; 19:214-225. [PMID: 31981516 PMCID: PMC7029307 DOI: 10.1016/s1474-4422(19)30485-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/20/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
Background Neurodegeneration is the pathological substrate that causes major disability in secondary progressive multiple sclerosis. A synthesis of preclinical and clinical research identified three neuroprotective drugs acting on different axonal pathobiologies. We aimed to test the efficacy of these drugs in an efficient manner with respect to time, cost, and patient resource. Methods We did a phase 2b, multiarm, parallel group, double-blind, randomised placebo-controlled trial at 13 clinical neuroscience centres in the UK. We recruited patients (aged 25–65 years) with secondary progressive multiple sclerosis who were not on disease-modifying treatment and who had an Expanded Disability Status Scale (EDSS) score of 4·0–6·5. Participants were randomly assigned (1:1:1:1) at baseline, by a research nurse using a centralised web-based service, to receive twice-daily oral treatment of either amiloride 5 mg, fluoxetine 20 mg, riluzole 50 mg, or placebo for 96 weeks. The randomisation procedure included minimisation based on sex, age, EDSS score at randomisation, and trial site. Capsules were identical in appearance to achieve masking. Patients, investigators, and MRI readers were unaware of treatment allocation. The primary outcome measure was volumetric MRI percentage brain volume change (PBVC) from baseline to 96 weeks, analysed using multiple regression, adjusting for baseline normalised brain volume and minimisation criteria. The primary analysis was a complete-case analysis based on the intention-to-treat population (all patients with data at week 96). This trial is registered with ClinicalTrials.gov, NCT01910259. Findings Between Jan 29, 2015, and June 22, 2016, 445 patients were randomly allocated amiloride (n=111), fluoxetine (n=111), riluzole (n=111), or placebo (n=112). The primary analysis included 393 patients who were allocated amiloride (n=99), fluoxetine (n=96), riluzole (n=99), and placebo (n=99). No difference was noted between any active treatment and placebo in PBVC (amiloride vs placebo, 0·0% [95% CI −0·4 to 0·5; p=0·99]; fluoxetine vs placebo −0·1% [–0·5 to 0·3; p=0·86]; riluzole vs placebo −0·1% [–0·6 to 0·3; p=0·77]). No emergent safety issues were reported. The incidence of serious adverse events was low and similar across study groups (ten [9%] patients in the amiloride group, seven [6%] in the fluoxetine group, 12 [11%] in the riluzole group, and 13 [12%] in the placebo group). The most common serious adverse events were infections and infestations. Three patients died during the study, from causes judged unrelated to active treatment; one patient assigned amiloride died from metastatic lung cancer, one patient assigned riluzole died from ischaemic heart disease and coronary artery thrombosis, and one patient assigned fluoxetine had a sudden death (primary cause) with multiple sclerosis and obesity listed as secondary causes. Interpretation The absence of evidence for neuroprotection in this adequately powered trial indicates that exclusively targeting these aspects of axonal pathobiology in patients with secondary progressive multiple sclerosis is insufficient to mitigate neuroaxonal loss. These findings argue for investigation of different mechanistic targets and future consideration of combination treatment trials. This trial provides a template for future simultaneous testing of multiple disease-modifying medicines in neurological medicine. Funding Efficacy and Mechanism Evaluation (EME) Programme, an MRC and NIHR partnership, UK Multiple Sclerosis Society, and US National Multiple Sclerosis Society.
Collapse
|
17
|
Generalised boundary shift integral for longitudinal assessment of spinal cord atrophy. Neuroimage 2019; 209:116489. [PMID: 31877375 DOI: 10.1016/j.neuroimage.2019.116489] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/18/2019] [Accepted: 12/21/2019] [Indexed: 12/18/2022] Open
Abstract
Spinal cord atrophy measurements obtained from structural magnetic resonance imaging (MRI) are associated with disability in many neurological diseases and serve as in vivo biomarkers of neurodegeneration. Longitudinal spinal cord atrophy rate is commonly determined from the numerical difference between two volumes (based on 3D surface fitting) or two cross-sectional areas (CSA, based on 2D edge detection) obtained at different time-points. Being an indirect measure, atrophy rates are susceptible to variable segmentation errors at the edge of the spinal cord. To overcome those limitations, we developed a new registration-based pipeline that measures atrophy rates directly. We based our approach on the generalised boundary shift integral (GBSI) method, which registers 2 scans and uses a probabilistic XOR mask over the edge of the spinal cord, thereby measuring atrophy more accurately than segmentation-based techniques. Using a large cohort of longitudinal spinal cord images (610 subjects with multiple sclerosis from a multi-centre trial and 52 healthy controls), we demonstrated that GBSI is a sensitive, quantitative and objective measure of longitudinal spinal cord volume change. The GBSI pipeline is repeatable, reproducible, and provides more precise measurements of longitudinal spinal cord atrophy than segmentation-based methods in longitudinal spinal cord atrophy studies.
Collapse
|
18
|
Sample Size for Oxidative Stress and Inflammation When Treating Multiple Sclerosis with Interferon-β1a and Coenzyme Q10. Brain Sci 2019; 9:brainsci9100259. [PMID: 31569668 PMCID: PMC6826871 DOI: 10.3390/brainsci9100259] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 11/16/2022] Open
Abstract
Studying multiple sclerosis (MS) and its treatments requires the use of biomarkers for underlying pathological mechanisms. We aim to estimate the required sample size for detecting variations of biomarkers of inflammation and oxidative stress. This is a post-hoc analysis on 60 relapsing-remitting MS patients treated with Interferon-β1a and Coenzyme Q10 for 3 months in an open-label crossover design over 6 months. At baseline and at the 3 and 6-month visits, we measured markers of scavenging activity, oxidative damage, and inflammation in the peripheral blood (180 measurements). Variations of laboratory measures (treatment effect) were estimated using mixed-effect linear regression models (including age, gender, disease duration, baseline expanded disability status scale (EDSS), and the duration of Interferon-β1a treatment as covariates; creatinine was also included for uric acid analyses), and were used for sample size calculations. Hypothesizing a clinical trial aiming to detect a 70% effect in 3 months (power = 80% alpha-error = 5%), the sample size per treatment arm would be 1 for interleukin (IL)-3 and IL-5, 4 for IL-7 and IL-2R, 6 for IL-13, 14 for IL-6, 22 for IL-8, 23 for IL-4, 25 for activation-normal T cell expressed and secreted (RANTES), 26 for tumor necrosis factor (TNF)-α, 27 for IL-1β, and 29 for uric acid. Peripheral biomarkers of oxidative stress and inflammation could be used in proof-of-concept studies to quickly screen the mechanisms of action of MS treatments.
Collapse
|
19
|
Moccia M, Prados F, Filippi M, Rocca MA, Valsasina P, Brownlee WJ, Zecca C, Gallo A, Rovira A, Gass A, Palace J, Lukas C, Vrenken H, Ourselin S, Gandini Wheeler‐Kingshott CAM, Ciccarelli O, Barkhof F. Longitudinal spinal cord atrophy in multiple sclerosis using the generalized boundary shift integral. Ann Neurol 2019; 86:704-713. [DOI: 10.1002/ana.25571] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/29/2019] [Accepted: 08/01/2019] [Indexed: 11/10/2022]
Affiliation(s)
- Marcello Moccia
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain SciencesUniversity College London London United Kingdom
- Multiple Sclerosis Clinical Care and Research Center, Department of NeurosciencesFederico II University Naples Italy
| | - Ferran Prados
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain SciencesUniversity College London London United Kingdom
- Centre for Medical Image Computing, Department of Medical Physics and BioengineeringUniversity College London London United Kingdom
- National Institute for Health ResearchUniversity College London Hospitals Biomedical Research Centre London United Kingdom
- Open University of Catalonia Barcelona Spain
| | - Massimo Filippi
- Division of Neuroscience, San Raffaele Scientific Institute, Vita‐Salute San Raffaele UniversityNeuroimaging Research Unit, Institute of Experimental Neurology Milan Italy
- Department of NeurologySan Raffaele Scientific Institute Milan Italy
| | - Maria A. Rocca
- Division of Neuroscience, San Raffaele Scientific Institute, Vita‐Salute San Raffaele UniversityNeuroimaging Research Unit, Institute of Experimental Neurology Milan Italy
- Department of NeurologySan Raffaele Scientific Institute Milan Italy
| | - Paola Valsasina
- Division of Neuroscience, San Raffaele Scientific Institute, Vita‐Salute San Raffaele UniversityNeuroimaging Research Unit, Institute of Experimental Neurology Milan Italy
| | - Wallace J. Brownlee
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain SciencesUniversity College London London United Kingdom
| | - Chiara Zecca
- Neurocenter of Southern SwitzerlandLugano Regional Hospital Lugano Switzerland
| | - Antonio Gallo
- 3T‐MRI Research Center, Department of Advanced Medical and Surgical SciencesUniversity of Campania Luigi Vanvitelli Naples Italy
| | - Alex Rovira
- Section of Neuroradiology, Department of RadiologyVall d'Hebron University Hospital, Autonomous University of Barcelona Barcelona Spain
| | - Achim Gass
- Department of NeurologyUniversitätsmedizin Mannheim, University of Heidelberg Mannheim Germany
| | - Jacqueline Palace
- Nuffield Department of Clinical NeurosciencesJohn Radcliffe Hospital Oxford United Kingdom
| | | | - Hugo Vrenken
- Department of Radiology and Nuclear MedicineVU University Medical Center Amsterdam the Netherlands
| | - Sebastien Ourselin
- Department of Imaging and Biomedical EngineeringKing's College London London United Kingdom
| | - Claudia A. M. Gandini Wheeler‐Kingshott
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain SciencesUniversity College London London United Kingdom
- Department of Brain and Behavioral SciencesUniversity of Pavia Pavia Italy
- Brain MRI 3T Research Center, Mondino FoundationScientific Institute for Research and Health Care Pavia Italy
| | - Olga Ciccarelli
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain SciencesUniversity College London London United Kingdom
- National Institute for Health ResearchUniversity College London Hospitals Biomedical Research Centre London United Kingdom
| | - Frederik Barkhof
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain SciencesUniversity College London London United Kingdom
- Centre for Medical Image Computing, Department of Medical Physics and BioengineeringUniversity College London London United Kingdom
- National Institute for Health ResearchUniversity College London Hospitals Biomedical Research Centre London United Kingdom
- Department of Radiology and Nuclear MedicineVU University Medical Center Amsterdam the Netherlands
| | | |
Collapse
|
20
|
Seif M, Gandini Wheeler-Kingshott CA, Cohen-Adad J, Flanders AE, Freund P. Guidelines for the conduct of clinical trials in spinal cord injury: Neuroimaging biomarkers. Spinal Cord 2019; 57:717-728. [PMID: 31267015 PMCID: PMC6760553 DOI: 10.1038/s41393-019-0309-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 12/16/2022]
Abstract
Traumatic spinal cord injury (SCI) leads to immediate neuronal and axonal damage at the focal injury site and triggers secondary pathologic series of events resulting in sensorimotor and autonomic dysfunction below the level of injury. Although there is no cure for SCI, neuroprotective and regenerative therapies show promising results at the preclinical stage. There is a pressing need to develop non-invasive outcome measures that can indicate whether a candidate therapeutic agent or a cocktail of therapeutic agents are positively altering the underlying disease processes. Recent conventional MRI studies have quantified spinal cord lesion characteristics and elucidated their relationship between severity of injury to clinical impairment and recovery. Next to the quantification of the primary cord damage, quantitative MRI measures of spinal cord (rostrocaudally to the lesion site) and brain integrity have demonstrated progressive and specific neurodegeneration of afferent and efferent neuronal pathways. MRI could therefore play a key role to ultimately uncover the relationship between clinical impairment/recovery and injury-induced neurodegenerative changes in the spinal cord and brain. Moreover, neuroimaging biomarkers hold promises to improve clinical trial design and efficiency through better patient stratification. The purpose of this narrative review is therefore to propose a guideline of clinically available MRI sequences and their derived neuroimaging biomarkers that have the potential to assess tissue damage at the macro- and microstructural level after SCI. In this piece, we make a recommendation for the use of key MRI sequences-both conventional and advanced-for clinical work-up and clinical trials.
Collapse
Affiliation(s)
- Maryam Seif
- Spinal Cord Injury Center, University Hospital Balgrist, University of Zurich, Zurich, Switzerland
| | - Claudia Am Gandini Wheeler-Kingshott
- Faculty of Brain Sciences, Queen Square MS Centre, UCL Queen Square Institute of Neurology, London, United Kingdom.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Brain MRI 3T Mondino Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
| | - Adam E Flanders
- Regional Spinal Cord Injury Center of the Delaware Valley, Department of Radiology, Division of Neuroradiology, Thomas Jefferson University, 1087 Main Building, 132 South 10th Street, Philadelphia, PA, 19107, USA
| | - Patrick Freund
- Spinal Cord Injury Center, University Hospital Balgrist, University of Zurich, Zurich, Switzerland. .,Faculty of Brain Sciences, Queen Square MS Centre, UCL Queen Square Institute of Neurology, London, United Kingdom. .,Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany. .,Department of Neurology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
21
|
Høgestøl EA, Kaufmann T, Nygaard GO, Beyer MK, Sowa P, Nordvik JE, Kolskår K, Richard G, Andreassen OA, Harbo HF, Westlye LT. Cross-Sectional and Longitudinal MRI Brain Scans Reveal Accelerated Brain Aging in Multiple Sclerosis. Front Neurol 2019; 10:450. [PMID: 31114541 PMCID: PMC6503038 DOI: 10.3389/fneur.2019.00450] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 04/12/2019] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disorder of the central nervous system. By combining longitudinal MRI-based brain morphometry and brain age estimation using machine learning, we tested the hypothesis that MS patients have higher brain age relative to chronological age than healthy controls (HC) and that longitudinal rate of brain aging in MS patients is associated with clinical course and severity. Seventy-six MS patients [71% females, mean age 34.8 years (range 21-49) at inclusion] were examined with brain MRI at three time points with a mean total follow up period of 4.4 years (±0.4 years). We used additional cross-sectional MRI data from 235 HC for case-control comparison. We applied a machine learning model trained on an independent set of 3,208 HC to estimate individual brain age and to calculate the difference between estimated and chronological age, termed brain age gap (BAG). We also assessed the longitudinal change rate in BAG in individuals with MS. MS patients showed significantly higher BAG (4.4 ± 6.6 years) compared to HC (Cohen's D = 0.69, p = 4.0 × 10-6). Longitudinal estimates of BAG in MS patients showed high reliability and suggested an accelerated rate of brain aging corresponding to an annual increase of 0.41 (SE = 0.15) years compared to chronological aging (p = 0.008). Multiple regression analyses revealed higher rate of brain aging in patients with more brain atrophy (Cohen's D = 0.86, p = 4.3 × 10-15) and increased white matter lesion load (WMLL) (Cohen's D = 0.55, p = 0.015). On average, patients with MS had significantly higher BAG compared to HC. Progressive brain aging in patients with MS was related to brain atrophy and increased WMLL. No significant clinical associations were found in our sample, future studies are warranted on this matter. Brain age estimation is a promising method for evaluation of subtle brain changes in MS, which is important for predicting clinical outcome and guide choice of intervention.
Collapse
Affiliation(s)
| | - Tobias Kaufmann
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gro O. Nygaard
- 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
| | - Piotr Sowa
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | | | - Knut Kolskår
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Sunnaas Rehabilitation Hospital HT, Nesodden, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Geneviève Richard
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Sunnaas Rehabilitation Hospital HT, Nesodden, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Ole A. Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hanne F. Harbo
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Lars T. Westlye
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
Moccia M, Ruggieri S, Ianniello A, Toosy A, Pozzilli C, Ciccarelli O. Advances in spinal cord imaging in multiple sclerosis. Ther Adv Neurol Disord 2019; 12:1756286419840593. [PMID: 31040881 PMCID: PMC6477770 DOI: 10.1177/1756286419840593] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/03/2019] [Indexed: 11/18/2022] Open
Abstract
The spinal cord is frequently affected in multiple sclerosis (MS), causing motor, sensory and autonomic dysfunction. A number of pathological abnormalities, including demyelination and neuroaxonal loss, occur in the MS spinal cord and are studied in vivo with magnetic resonance imaging (MRI). The aim of this review is to summarise and discuss recent advances in spinal cord MRI. Advances in conventional spinal cord MRI include improved identification of MS lesions, recommended spinal cord MRI protocols, enhanced recognition of MRI lesion characteristics that allow MS to be distinguished from other myelopathies, evidence for the role of spinal cord lesions in predicting prognosis and monitoring disease course, and novel post-processing methods to obtain lesion probability maps. The rate of spinal cord atrophy is greater than that of brain atrophy (-1.78% versus -0.5% per year), and reflects neuroaxonal loss in an eloquent site of the central nervous system, suggesting that it can become an important outcome measure in clinical trials, especially in progressive MS. Recent developments allow the calculation of spinal cord atrophy from brain volumetric scans and evaluation of its progression over time with registration-based techniques. Fully automated analysis methods, including segmentation of grey matter and intramedullary lesions, will facilitate the use of spinal cord atrophy in trial designs and observational studies. Advances in quantitative imaging techniques to evaluate neuroaxonal integrity, myelin content, metabolic changes, and functional connectivity, have provided new insights into the mechanisms of damage in MS. Future directions of research and the possible impact of 7T scanners on spinal cord imaging will be discussed.
Collapse
Affiliation(s)
- Marcello Moccia
- Queen Square MS Centre, NMR Research Unit, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
- Multiple Sclerosis Clinical Care and Research Centre, Department of Neurosciences, Federico II University of Naples, via Sergio Pansini, 5, Edificio 17 - piano terra, Napoli, 80131 Naples, Italy
| | - Serena Ruggieri
- Department of Human Neuroscience, Sapienza University of Rome, Italy
| | - Antonio Ianniello
- Department of Human Neuroscience, Sapienza University of Rome, Italy
| | - Ahmed Toosy
- Queen Square MS Centre, NMR Research Unit, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Carlo Pozzilli
- Department of Human Neuroscience, Sapienza University of Rome, Italy
| | - Olga Ciccarelli
- Queen Square MS Centre, NMR Research Unit, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
- National Institute for Health Research, University College London Hospitals Biomedical Research Centre, London, UK
| |
Collapse
|
23
|
Winges KM, Murchison CF, Bourdette DN, Spain RI. Longitudinal optical coherence tomography study of optic atrophy in secondary progressive multiple sclerosis: Results from a clinical trial cohort. Mult Scler 2019; 25:55-62. [PMID: 29111873 PMCID: PMC5930161 DOI: 10.1177/1352458517739136] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Limited prospective information exists regarding spectral-domain optical coherence tomography (SD-OCT) in secondary progressive multiple sclerosis (SPMS). OBJECTIVE Document cross-sectional and longitudinal retinal nerve fiber layer (RNFL) and macular ganglion cell plus inner plexiform layer (GCIPL) features of an SPMS clinical trial cohort. METHODS Prospective, observational study using a 2-year randomized placebo-controlled SPMS trial cohort with yearly SD-OCT testing. Post hoc analysis determined influences of optic neuritis (ON), disease duration, and baseline SD-OCT on annualized atrophy rates and on correlations between OCT and brain atrophy. RESULTS Mean RNFL and GCIPL values of patients ( n = 47, mean age = 59 years, mean disease duration = 30 years) were significantly lower among eyes with prior ON than those without (no history of ON (NON)). Annualized RNFL (-0.31 µm/year) and GCIPL (-0.29 µm/year) atrophy rates did not differ between ON and NON eyes. Baseline RNFL thickness >75 µm was associated with greater annualized RNFL atrophy (-0.85 µm/year). Neither RNFL nor GCIPL atrophy correlated with whole-brain atrophy. CONCLUSION This study suggests that eyes with and without ON history may be pooled for atrophy analysis in SPMS clinical trials using SD-OCT. Low baseline RNFL, small retinal atrophy rates, and lack of correlation with whole-brain atrophy in this population are important trial design considerations.
Collapse
Affiliation(s)
- Kimberly M. Winges
- Department of Ophthalmology, VA Portland Health Care System, Portland, OR, USA
- Department of Neurology, Oregon Health & Science University; Portland, OR, USA
- Departments of Casey Eye Institute, Oregon Health & Science University; Portland, OR, USA
| | | | - Dennis N. Bourdette
- Department of Neurology, Oregon Health & Science University; Portland, OR, USA
| | - Rebecca I. Spain
- Department of Neurology, VA Portland Health Care System, Portland, OR, USA
- Department of Neurology, Oregon Health & Science University; Portland, OR, USA
| |
Collapse
|
24
|
Petracca M, Margoni M, Bommarito G, Inglese M. Monitoring Progressive Multiple Sclerosis with Novel Imaging Techniques. Neurol Ther 2018; 7:265-285. [PMID: 29956263 PMCID: PMC6283788 DOI: 10.1007/s40120-018-0103-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Indexed: 02/04/2023] Open
Abstract
Imaging markers for monitoring disease progression in progressive multiple sclerosis (PMS) are scarce, thereby limiting the possibility to monitor disease evolution and to test effective treatments in clinical trials. Advanced imaging techniques that have the advantage of metrics with increased sensitivity to short-term tissue changes and increased specificity to the structural abnormalities characteristic of PMS have recently been applied in clinical trials of PMS. In this review, we (1) provide an overview of the pathological features of PMS, (2) summarize the findings of research and clinical trials conducted in PMS which have applied conventional and advanced magnetic resonance imaging techniques and (3) discuss recent advancements and future perspectives in monitoring PMS with imaging techniques.
Collapse
Affiliation(s)
- Maria Petracca
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Monica Margoni
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Multiple Sclerosis Centre, Department of Neurosciences DNS, University Hospital, University of Padua, Padua, Italy
| | - Giulia Bommarito
- Department of Neuroscience, Rehabilitation, Genetics and Maternal and Perinatal Sciences, University of Genoa, Genoa, Italy
| | - Matilde Inglese
- Department of Neuroscience, Rehabilitation, Genetics and Maternal and Perinatal Sciences, University of Genoa, Genoa, Italy.
- Departments of Neurology, Radiology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
25
|
Nakamura K, Eskildsen SF, Narayanan S, Arnold DL, Collins DL. Improving the SIENA performance using BEaST brain extraction. PLoS One 2018; 13:e0196945. [PMID: 30235215 PMCID: PMC6147402 DOI: 10.1371/journal.pone.0196945] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/23/2018] [Indexed: 01/30/2023] Open
Abstract
We present an improved image analysis pipeline to detect the percent brain volume change (PBVC) using SIENA (Structural Image Evaluation, using Normalization, of Atrophy) in populations with Alzheimer’s dementia. Our proposed approach uses the improved brain extraction mask from BEaST (Brain Extraction based on nonlocal Segmentation Technique) instead of the conventional BET (Brain Extraction Tool) for SIENA. We compared four varying options of BET as well as BEaST and applied these five methods to analyze scan-rescan MRIs in ADNI from 332 subjects, longitudinal ADNI MRIs from the same 332 subjects, their repeat scans over time, and OASIS longitudinal MRIs from 123 subjects. The results showed that BEaST brain masks were consistent in scan-rescan reproducibility. The cross-sectional scan-rescan error in the absolute percent brain volume difference measured by SIENA was smallest (p≤0.0187) with the proposed BEaST-SIENA. We evaluated the statistical power in terms of effect size, and the best performance was achieved with BEaST-SIENA (1.2789 for ADNI and 1.095 for OASIS). The absolute difference in PBVC between scan-dataset (volume change from baseline to year-1) and rescan-dataset (volume change from baseline repeat scan to year-1 repeat scan) was also the smallest with BEaST-SIENA compared to the BET-based SIENA and had the highest correlation when compared to the BET-based SIENA variants. In conclusion, our study shows that BEaST was robust in terms of reproducibility and consistency and that SIENA’s reproducibility and statistical power are improved in multiple datasets when used in combination with BEaST.
Collapse
Affiliation(s)
- Kunio Nakamura
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- * E-mail:
| | - Simon F. Eskildsen
- Centre of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - Sridar Narayanan
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- NeuroRx Research, Montreal, Quebec, Canada
| | - Douglas L. Arnold
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- NeuroRx Research, Montreal, Quebec, Canada
| | - D. Louis Collins
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
26
|
Krotenkova IA, Bryukhov VV, Krotenkova MV, Zakharova MN, Askarova LS. [Brain atrophy and perfusion changes in patients with remitting and secondary progressive multiple sclerosis]. Zh Nevrol Psikhiatr Im S S Korsakova 2018; 118:47-54. [PMID: 30160668 DOI: 10.17116/jnevro201811808247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
AIM To study the relationship of brain atrophy and changes in perfusion with an increase in the level of disability in patients with multiple sclerosis (MS). MATERIAL AND METHODS Twenty patients with remitting MS, 20 patients with secondary progressive multiple sclerosis (SPMS) and 20 healthy people were studied. The level of neurological deficit was assessed with EDSS and cognitive status with PASAT. MRI of the brain (standard impulse sequences and 3D-T1-MPR for voxel MRI-morphometry) and perfusion computed tomography with the assessment of visually intact white matter (VIWM) and thalamus were performed. RESULTS Compared to the control group, patients with MS had a significant atrophy of subcortical gray matter. Patients with SPMS in addition had an atrophy of some cortical areas which was correlated with EDSS scores (p<0.05). The correlation between cognitive impairment and the volume of the left inferior parietal lobule (r=0.677; p=0.011) and worsening of perfusion of VIWM of frontal and parietal lobes, thalamus on both sides was observed in patients with SPMS compared to those with remitting MS. That was correlated with cognitive performance assessed by PASAT. CONCLUSION Patterns of atrophy distribution in different types of MS were determined. The level of disability is correlated with the severity of brain atrophy. Hypoperfusion of VIWM that was correlated with cognitive impairment was found in patients with SPMS.
Collapse
|
27
|
Connick P, De Angelis F, Parker RA, Plantone D, Doshi A, John N, Stutters J, MacManus D, Prados Carrasco F, Barkhof F, Ourselin S, Braisher M, Ross M, Cranswick G, Pavitt SH, Giovannoni G, Gandini Wheeler-Kingshott CA, Hawkins C, Sharrack B, Bastow R, Weir CJ, Stallard N, Chandran S, Chataway J. Multiple Sclerosis-Secondary Progressive Multi-Arm Randomisation Trial (MS-SMART): a multiarm phase IIb randomised, double-blind, placebo-controlled clinical trial comparing the efficacy of three neuroprotective drugs in secondary progressive multiple sclerosis. BMJ Open 2018; 8:e021944. [PMID: 30166303 PMCID: PMC6119433 DOI: 10.1136/bmjopen-2018-021944] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION The major unmet need in multiple sclerosis (MS) is for neuroprotective therapies that can slow (or ideally stop) the rate of disease progression. The UK MS Society Clinical Trials Network (CTN) was initiated in 2007 with the purpose of developing a national, efficient, multiarm trial of repurposed drugs. Key underpinning work was commissioned by the CTN to inform the design, outcome selection and drug choice including animal models and a systematic review. This identified seven leading oral agents for repurposing as neuroprotective therapies in secondary progressive MS (SPMS). The purpose of the Multiple Sclerosis-Secondary Progressive Multi-Arm Randomisation Trial (MS-SMART) will be to evaluate the neuroprotective efficacy of three of these drugs, selected with distinct mechanistic actions and previous evidence of likely efficacy, against a common placebo arm. The interventions chosen were: amiloride (acid-sensing ion channel antagonist); fluoxetine (selective serotonin reuptake inhibitor) and riluzole (glutamate antagonist). METHODS AND ANALYSIS Patients with progressing SPMS will be randomised 1:1:1:1 to amiloride, fluoxetine, riluzole or matched placebo and followed for 96 weeks. The primary outcome will be the percentage brain volume change (PBVC) between baseline and 96 weeks, derived from structural MR brain imaging data using the Structural Image Evaluation, using Normalisation, of Atrophy method. With a sample size of 90 per arm, this will give 90% power to detect a 40% reduction in PBVC in any active arm compared with placebo and 80% power to detect a 35% reduction (analysing by analysis of covariance and with adjustment for multiple comparisons of three 1.67% two-sided tests), giving a 5% overall two-sided significance level. MS-SMART is not powered to detect differences between the three active treatment arms. Allowing for a 20% dropout rate, 110 patients per arm will be randomised. The study will take place at Neuroscience centres in England and Scotland. ETHICS AND DISSEMINATION MS-SMART was approved by the Scotland A Research Ethics Committee on 13 January 2013 (REC reference: 13/SS/0007). Results of the study will be submitted for publication in a peer-reviewed journal. TRIAL REGISTRATION NUMBERS NCT01910259; 2012-005394-31; ISRCTN28440672.
Collapse
Affiliation(s)
- Peter Connick
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Floriana De Angelis
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - Richard A Parker
- Edinburgh Clinical Trials Unit, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Domenico Plantone
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - Anisha Doshi
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - Nevin John
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - Jonathan Stutters
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - David MacManus
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - Ferran Prados Carrasco
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
- Department of Medical Physics and Biomedical Engineering, Translational Imaging Group (TIG), Centre for Medical Image Computing (CMIC), UCL, London, UK
| | - Frederik Barkhof
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
- Department of Radiology and Nuclear Medicine, VU University Medical Centre, Amsterdam, The Netherlands
| | - Sebastien Ourselin
- Department of Medical Physics and Biomedical Engineering, Translational Imaging Group (TIG), Centre for Medical Image Computing (CMIC), UCL, London, UK
| | - Marie Braisher
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - Moira Ross
- Edinburgh Clinical Trials Unit, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Gina Cranswick
- Edinburgh Clinical Trials Unit, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Sue H Pavitt
- Dental Translational and Clinical Research Unit (part of the NIHR Leeds CRF), University of Leeds, Leeds, UK
| | - Gavin Giovannoni
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Claudia Angela Gandini Wheeler-Kingshott
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
- Brain MRI 3T Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Clive Hawkins
- Keele Medical School and Institute for Science and Technology in Medicine, Keele University, Keele, UK
| | - Basil Sharrack
- Department of Neuroscience, Royal Hallamshire Hospital, Sheffield, UK
| | | | - Christopher J Weir
- Edinburgh Clinical Trials Unit, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Nigel Stallard
- Statistics and Epidemiology, Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | | | - Jeremy Chataway
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| |
Collapse
|
28
|
Sinnecker T, Granziera C, Wuerfel J, Schlaeger R. Future Brain and Spinal Cord Volumetric Imaging in the Clinic for Monitoring Treatment Response in MS. Curr Treat Options Neurol 2018; 20:17. [PMID: 29679165 DOI: 10.1007/s11940-018-0504-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW Volumetric analysis of brain imaging has emerged as a standard approach used in clinical research, e.g., in the field of multiple sclerosis (MS), but its application in individual disease course monitoring is still hampered by biological and technical limitations. This review summarizes novel developments in volumetric imaging on the road towards clinical application to eventually monitor treatment response in patients with MS. RECENT FINDINGS In addition to the assessment of whole-brain volume changes, recent work was focused on the volumetry of specific compartments and substructures of the central nervous system (CNS) in MS. This included volumetric imaging of the deep brain structures and of the spinal cord white and gray matter. Volume changes of the latter indeed independently correlate with clinical outcome measures especially in progressive MS. Ultrahigh field MRI and quantitative MRI added to this trend by providing a better visualization of small compartments on highly resolving MR images as well as microstructural information. New developments in volumetric imaging have the potential to improve sensitivity as well as specificity in detecting and hence monitoring disease-related CNS volume changes in MS.
Collapse
Affiliation(s)
- Tim Sinnecker
- Neurologic Clinic and Policlinic, Departments of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Petersgraben 4, 4031, Basel, Switzerland
- Translational Imaging in Neurology (ThINK) Basel, Department of Medicine and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Medical Image Analysis Center Basel AG, Basel, Switzerland
- NeuroCure Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Cristina Granziera
- Neurologic Clinic and Policlinic, Departments of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Petersgraben 4, 4031, Basel, Switzerland
- Translational Imaging in Neurology (ThINK) Basel, Department of Medicine and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Jens Wuerfel
- Medical Image Analysis Center Basel AG, Basel, Switzerland
- NeuroCure Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Ultrahigh Field Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Regina Schlaeger
- Neurologic Clinic and Policlinic, Departments of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Petersgraben 4, 4031, Basel, Switzerland.
- Translational Imaging in Neurology (ThINK) Basel, Department of Medicine and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland.
| |
Collapse
|
29
|
Azevedo CJ, Cen SY, Khadka S, Liu S, Kornak J, Shi Y, Zheng L, Hauser SL, Pelletier D. Thalamic atrophy in multiple sclerosis: A magnetic resonance imaging marker of neurodegeneration throughout disease. Ann Neurol 2018; 83:223-234. [PMID: 29328531 DOI: 10.1002/ana.25150] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 11/17/2017] [Accepted: 11/26/2017] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Thalamic volume is a candidate magnetic resonance imaging (MRI)-based marker associated with neurodegeneration to hasten development of neuroprotective treatments. Our objective is to describe the longitudinal evolution of thalamic atrophy in MS and normal aging, and to estimate sample sizes for study design. METHODS Six hundred one subjects (2,632 MRI scans) were analyzed. Five hundred twenty subjects with relapse-onset MS (clinically isolated syndrome, n = 90; relapsing-remitting MS, n = 392; secondary progressive MS, n = 38) underwent annual standardized 3T MRI scans for an average of 4.1 years, including a 1mm3 3-dimensional T1-weighted sequence (3DT1; 2,485 MRI scans). Eighty-one healthy controls (HC) were scanned longitudinally on the same scanner using the same protocol (147 MRI scans). 3DT1s were processed using FreeSurfer's longitudinal pipeline after lesion inpainting. Rates of normalized thalamic volume loss in MS and HC were compared in linear mixed effects models. Simulation-based sample size calculations were performed incorporating the rate of atrophy in HC. RESULTS Thalamic volume declined significantly faster in MS subjects compared to HC, with an estimated decline of -0.71% per year (95% confidence interval [CI] = -0.77% to -0.64%) in MS subjects and -0.28% per year (95% CI = -0.58% to 0.02%) in HC (p for difference = 0.007). The rate of decline was consistent throughout the MS disease duration and across MS clinical subtypes. Eighty or 100 subjects per arm (α = 0.1 or 0.05, respectively) would be needed to detect the maximal effect size with 80% power in a 24-month study. INTERPRETATION Thalamic atrophy occurs early and consistently throughout MS. Preliminary sample size calculations appear feasible, adding to its appeal as an MRI marker associated with neurodegeneration. Ann Neurol 2018;83:223-234.
Collapse
Affiliation(s)
| | - Steven Y Cen
- Department of Neurology, University of Southern California, Los Angeles, CA
| | | | - Shuang Liu
- Department of Neurology, Yale University, New Haven, CT
| | - John Kornak
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA
| | - Yonggang Shi
- Department of Neurology, University of Southern California, Los Angeles, CA
| | - Ling Zheng
- Department of Neurology, University of Southern California, Los Angeles, CA
| | - Stephen L Hauser
- Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Daniel Pelletier
- Department of Neurology, University of Southern California, Los Angeles, CA
| |
Collapse
|
30
|
Affiliation(s)
- Daniel S Reich
- From the Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda (D.S.R.), and the Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore (P.A.C.) - both in Maryland; and the Department of Neurology, Mayo Clinic, Rochester, MN (C.F.L.)
| | - Claudia F Lucchinetti
- From the Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda (D.S.R.), and the Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore (P.A.C.) - both in Maryland; and the Department of Neurology, Mayo Clinic, Rochester, MN (C.F.L.)
| | - Peter A Calabresi
- From the Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda (D.S.R.), and the Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore (P.A.C.) - both in Maryland; and the Department of Neurology, Mayo Clinic, Rochester, MN (C.F.L.)
| |
Collapse
|
31
|
Abstract
Understanding the clinico-radiological paradox is important in the search for more sensitive and specific surrogates of relapses and disability progression (such that they can be used to inform treatment choices in individual people with multiple sclerosis) and to gain a better understanding of the pathophysiological basis of disability in multiple sclerosis (to identify and assess key therapeutic targets). In this brief review, we will consider themes and issues underlying the clinico-radiological paradox and recent advances in its resolution.
Collapse
Affiliation(s)
- Declan Chard
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH), Biomedical Research Centre, London, UK.,NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - S Anand Trip
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH), Biomedical Research Centre, London, UK.,NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| |
Collapse
|
32
|
Sormani MP, Pardini M. Assessing Repair in Multiple Sclerosis: Outcomes for Phase II Clinical Trials. Neurotherapeutics 2017; 14:924-933. [PMID: 28695472 PMCID: PMC5722763 DOI: 10.1007/s13311-017-0558-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multiple Sclerosis (MS) pathology is complex and includes inflammatory processes, neurodegeneration, and demyelination. While multiple drugs have been developed to tackle MS-related inflammation, to date there is scant evidence regarding which therapeutic approach, if any, could be used to reverse demyelination, foster tissue repair, and thus positively impact on chronic disability. Here, we reviewed the current structural and functional markers (magnetic resonance imaging, positron emission tomography, optical coherence tomography, and visual evoked potentials) which could be used in phase II clinical trials of new compounds aimed to foster tissue repair in MS. Magnetic transfer ratio recovery in newly formed lesions currently represents the most widely used biomarker of tissue repair in MS, even if other markers, such as optical coherence tomography and positron emission tomography hold great promise to complement magnetic transfer ratio in tissue repair clinical trials. Future studies are needed to better characterize the different possible biomarkers to study tissue repair in MS, especially regarding their pathological specificity, sensitivity to change, and their relationship with disease activity.
Collapse
Affiliation(s)
- Maria Pia Sormani
- Biostatistics Unit, Department of Health Sciences, University of Genoa, Genoa, Italy.
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal and Child Health, University of Genoa, Genoa, Italy
- Policlinic San Martino-IST, Genoa, Italy
| |
Collapse
|
33
|
Moccia M, de Stefano N, Barkhof F. Imaging outcome measures for progressive multiple sclerosis trials. Mult Scler 2017; 23:1614-1626. [PMID: 29041865 PMCID: PMC5650056 DOI: 10.1177/1352458517729456] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 07/28/2017] [Indexed: 11/16/2022]
Abstract
Imaging markers that are reliable, reproducible and sensitive to neurodegenerative changes in progressive multiple sclerosis (MS) can enhance the development of new medications with a neuroprotective mode-of-action. Accordingly, in recent years, a considerable number of imaging biomarkers have been included in phase 2 and 3 clinical trials in primary and secondary progressive MS. Brain lesion count and volume are markers of inflammation and demyelination and are important outcomes even in progressive MS trials. Brain and, more recently, spinal cord atrophy are gaining relevance, considering their strong association with disability accrual; ongoing improvements in analysis methods will enhance their applicability in clinical trials, especially for cord atrophy. Advanced magnetic resonance imaging (MRI) techniques (e.g. magnetization transfer ratio (MTR), diffusion tensor imaging (DTI), spectroscopy) have been included in few trials so far and hold promise for the future, as they can reflect specific pathological changes targeted by neuroprotective treatments. Positron emission tomography (PET) and optical coherence tomography have yet to be included. Applications, limitations and future perspectives of these techniques in clinical trials in progressive MS are discussed, with emphasis on measurement sensitivity, reliability and sample size calculation.
Collapse
Affiliation(s)
- Marcello Moccia
- NMR Research Unit, Queen Square MS Centre, UCL Institute of Neurology, University College London, London, UK; Multiple Sclerosis Clinical Care and Research Centre, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Nicola de Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Frederik Barkhof
- NMR Research Unit, Queen Square MS Centre, UCL Institute of Neurology, University College London, London, UK; Translational Imaging Group, UCL Institute of Healthcare Engineering, University College London, London, UK; Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
34
|
Spain R, Powers K, Murchison C, Heriza E, Winges K, Yadav V, Cameron M, Kim E, Horak F, Simon J, Bourdette D. Lipoic acid in secondary progressive MS: A randomized controlled pilot trial. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2017; 4:e374. [PMID: 28680916 PMCID: PMC5489387 DOI: 10.1212/nxi.0000000000000374] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/15/2017] [Indexed: 01/22/2023]
Abstract
Objective: To determine whether lipoic acid (LA), an endogenously produced antioxidant, slowed the whole-brain atrophy rate and was safe in secondary progressive MS (SPMS). Methods: Patients with SPMS aged 40–70 years enrolled in a single center, 2-year, double-blind, randomized trial of daily oral 1,200 mg LA vs placebo. Primary outcome was change in annualized percent change brain volume (PCBV). Secondary outcomes were changes in rates of atrophy of segmented brain, spinal cord, and retinal substructures, disability, quality of life, and safety. Intention-to-treat analysis used linear mixed models. Results: Participation occurred between May 2, 2011, and August 14, 2015. Study arms of LA (n = 27) and placebo (n = 24) were matched with mean age of 58.5 (SD 5.9) years, 61% women, mean disease duration of 29.6 (SD 9.5) years, and median Expanded Disability Status Score of 6.0 (interquartile range 1.75). After 2 years, the annualized PCBV was significantly less in the LA arm compared with placebo (−0.21 [standard error of the coefficient estimate (SEE) 0.14] vs −0.65 [SEE 0.10], 95% confidence interval [CI] 0.157–0.727, p = 0.002). Improved Timed 25-Foot Walk was almost but not significantly better in the LA than in the control group (−0.535 [SEE 0.358] vs 0.137 [SEE 0.247], 95% CI −1.37 to 0.03, p = 0.06). Significantly more gastrointestinal upset and fewer falls occurred in LA patients. Unexpected renal failure (n = 1) and glomerulonephritis (n = 1) occurred in the LA cohort. Compliance, measured by pill counts, was 87%. Conclusions: LA demonstrated a 68% reduction in annualized PCBV and suggested a clinical benefit in SPMS while maintaining favorable safety, tolerability, and compliance over 2 years. ClinicalTrials.gov identifier: NCT01188811. Classification of evidence: This study provides Class I evidence that for patients with SPMS, LA reduces the rate of brain atrophy.
Collapse
Affiliation(s)
- Rebecca Spain
- Neurology Division (R.S., V.Y., M.C., E.K., D.B.), Research Service (K.P., E.H.), and Department of Ophthalmology (K.W.), Veterans Affairs Portland Health Care System, OR; and Department of Neurology (R.S., C.M., K.W., V.Y., E.K., F.H., J.S., D.B.), Advanced Imaging Research Center (K.P.), and Casey Eye Institute (K.W.), Oregon Health & Science University, Portland
| | - Katherine Powers
- Neurology Division (R.S., V.Y., M.C., E.K., D.B.), Research Service (K.P., E.H.), and Department of Ophthalmology (K.W.), Veterans Affairs Portland Health Care System, OR; and Department of Neurology (R.S., C.M., K.W., V.Y., E.K., F.H., J.S., D.B.), Advanced Imaging Research Center (K.P.), and Casey Eye Institute (K.W.), Oregon Health & Science University, Portland
| | - Charles Murchison
- Neurology Division (R.S., V.Y., M.C., E.K., D.B.), Research Service (K.P., E.H.), and Department of Ophthalmology (K.W.), Veterans Affairs Portland Health Care System, OR; and Department of Neurology (R.S., C.M., K.W., V.Y., E.K., F.H., J.S., D.B.), Advanced Imaging Research Center (K.P.), and Casey Eye Institute (K.W.), Oregon Health & Science University, Portland
| | - Elizabeth Heriza
- Neurology Division (R.S., V.Y., M.C., E.K., D.B.), Research Service (K.P., E.H.), and Department of Ophthalmology (K.W.), Veterans Affairs Portland Health Care System, OR; and Department of Neurology (R.S., C.M., K.W., V.Y., E.K., F.H., J.S., D.B.), Advanced Imaging Research Center (K.P.), and Casey Eye Institute (K.W.), Oregon Health & Science University, Portland
| | - Kimberly Winges
- Neurology Division (R.S., V.Y., M.C., E.K., D.B.), Research Service (K.P., E.H.), and Department of Ophthalmology (K.W.), Veterans Affairs Portland Health Care System, OR; and Department of Neurology (R.S., C.M., K.W., V.Y., E.K., F.H., J.S., D.B.), Advanced Imaging Research Center (K.P.), and Casey Eye Institute (K.W.), Oregon Health & Science University, Portland
| | - Vijayshree Yadav
- Neurology Division (R.S., V.Y., M.C., E.K., D.B.), Research Service (K.P., E.H.), and Department of Ophthalmology (K.W.), Veterans Affairs Portland Health Care System, OR; and Department of Neurology (R.S., C.M., K.W., V.Y., E.K., F.H., J.S., D.B.), Advanced Imaging Research Center (K.P.), and Casey Eye Institute (K.W.), Oregon Health & Science University, Portland
| | - Michelle Cameron
- Neurology Division (R.S., V.Y., M.C., E.K., D.B.), Research Service (K.P., E.H.), and Department of Ophthalmology (K.W.), Veterans Affairs Portland Health Care System, OR; and Department of Neurology (R.S., C.M., K.W., V.Y., E.K., F.H., J.S., D.B.), Advanced Imaging Research Center (K.P.), and Casey Eye Institute (K.W.), Oregon Health & Science University, Portland
| | - Ed Kim
- Neurology Division (R.S., V.Y., M.C., E.K., D.B.), Research Service (K.P., E.H.), and Department of Ophthalmology (K.W.), Veterans Affairs Portland Health Care System, OR; and Department of Neurology (R.S., C.M., K.W., V.Y., E.K., F.H., J.S., D.B.), Advanced Imaging Research Center (K.P.), and Casey Eye Institute (K.W.), Oregon Health & Science University, Portland
| | - Fay Horak
- Neurology Division (R.S., V.Y., M.C., E.K., D.B.), Research Service (K.P., E.H.), and Department of Ophthalmology (K.W.), Veterans Affairs Portland Health Care System, OR; and Department of Neurology (R.S., C.M., K.W., V.Y., E.K., F.H., J.S., D.B.), Advanced Imaging Research Center (K.P.), and Casey Eye Institute (K.W.), Oregon Health & Science University, Portland
| | - Jack Simon
- Neurology Division (R.S., V.Y., M.C., E.K., D.B.), Research Service (K.P., E.H.), and Department of Ophthalmology (K.W.), Veterans Affairs Portland Health Care System, OR; and Department of Neurology (R.S., C.M., K.W., V.Y., E.K., F.H., J.S., D.B.), Advanced Imaging Research Center (K.P.), and Casey Eye Institute (K.W.), Oregon Health & Science University, Portland
| | - Dennis Bourdette
- Neurology Division (R.S., V.Y., M.C., E.K., D.B.), Research Service (K.P., E.H.), and Department of Ophthalmology (K.W.), Veterans Affairs Portland Health Care System, OR; and Department of Neurology (R.S., C.M., K.W., V.Y., E.K., F.H., J.S., D.B.), Advanced Imaging Research Center (K.P.), and Casey Eye Institute (K.W.), Oregon Health & Science University, Portland
| |
Collapse
|
35
|
Ontaneda D, Thompson AJ, Fox RJ, Cohen JA. Progressive multiple sclerosis: prospects for disease therapy, repair, and restoration of function. Lancet 2017; 389:1357-1366. [PMID: 27889191 DOI: 10.1016/s0140-6736(16)31320-4] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/11/2016] [Accepted: 08/02/2016] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis is a major cause of neurological disability, which accrues predominantly during progressive forms of the disease. Although development of multifocal inflammatory lesions is the underlying pathological process in relapsing-remitting multiple sclerosis, the gradual accumulation of disability that characterises progressive multiple sclerosis seems to result more from diffuse immune mechanisms and neurodegeneration. As a result, the 14 anti-inflammatory drugs that have regulatory approval for treatment of relapsing-remitting multiple sclerosis have little or no efficacy in progressive multiple sclerosis without inflammatory lesion activity. Effective therapies for progressive multiple sclerosis that prevent worsening, reverse damage, and restore function are a major unmet need. In this Series paper we summarise the current status of therapy for progressive multiple sclerosis and outline prospects for the future.
Collapse
Affiliation(s)
- Daniel Ontaneda
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alan J Thompson
- Department of Brain Repair and Rehabilitation, University College London, Institute of Neurology, Faculty of Brain Sciences, London, UK
| | - Robert J Fox
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jeffrey A Cohen
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
36
|
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system. Magnetic resonance imaging (MRI) is sensitive to lesion formation both in the brain and spinal cord. Imaging plays a prominent role in the diagnosis and monitoring of MS. Over a dozen anti-inflammatory therapies are approved for MS and the development of many of these medications was made possible through the use of contrast-enhancing lesions on MRI as a phase II outcome. A similar phase II outcome method for the neurodegeneration that underlies progressive courses of the disease is still unavailable. Although magnetic resonance is an invaluable tool for the diagnosis and monitoring of treatment effects in MS, several imaging barriers still exist. In general, MRI is less sensitive to gray matter lesions, lacks pathological specificity, and does not provide quantitative data easily. Several advanced imaging methods including diffusion tensor imaging, magnetization transfer, functional MRI, myelin water fraction imaging, ultra-high field MRI, positron emission tomography, and optical coherence tomography of the retina study promising ways of overcoming the difficulties in MS imaging.
Collapse
Affiliation(s)
- Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.
| | - Robert J Fox
- Mellen Center for Multiple Sclerosis, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
37
|
Kosa P, Ghazali D, Tanigawa M, Barbour C, Cortese I, Kelley W, Snyder B, Ohayon J, Fenton K, Lehky T, Wu T, Greenwood M, Nair G, Bielekova B. Development of a Sensitive Outcome for Economical Drug Screening for Progressive Multiple Sclerosis Treatment. Front Neurol 2016; 7:131. [PMID: 27574516 PMCID: PMC4983704 DOI: 10.3389/fneur.2016.00131] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/29/2016] [Indexed: 11/28/2022] Open
Abstract
Therapeutic advance in progressive multiple sclerosis (MS) has been very slow. Based on the transformative role magnetic resonance imaging (MRI) contrast-enhancing lesions had on drug development for relapsing-remitting MS, we consider the lack of sensitive outcomes to be the greatest barrier for developing new treatments for progressive MS. The purpose of this study was to compare 58 prospectively acquired candidate outcomes in the real-world situation of progressive MS trials to select and validate the best-performing outcome. The 1-year pre-treatment period of adaptively designed IPPoMS (ClinicalTrials.gov #NCT00950248) and RIVITaLISe (ClinicalTrials.gov #NCT01212094) Phase II trials served to determine the primary outcome for the subsequent blinded treatment phase by comparing 8 clinical, 1 electrophysiological, 1 optical coherence tomography, 7 MRI volumetric, 9 quantitative T1 MRI, and 32 diffusion tensor imaging MRI outcomes. Fifteen outcomes demonstrated significant progression over 1 year (Δ) in the predetermined analysis and seven out of these were validated in two independent cohorts. Validated MRI outcomes had limited correlations with clinical scales, relatively poor signal-to-noise ratios (SNR) and recorded overlapping values between healthy subjects and MS patients with moderate-severe disability. Clinical measures correlated better, even though each reflects a somewhat different disability domain. Therefore, using machine-learning techniques, we developed a combinatorial weight-adjusted disability score (CombiWISE) that integrates four clinical scales: expanded disability status scale (EDSS), Scripps neurological rating scale, 25 foot walk and 9 hole peg test. CombiWISE outperformed all clinical scales (Δ = 9.10%; p = 0.0003) and all MRI outcomes. CombiWISE recorded no overlapping values between healthy subjects and disabled MS patients, had high SNR, and predicted changes in EDSS in a longitudinal assessment of 98 progressive MS patients and in a cross-sectional cohort of 303 untreated subjects. One point change in EDSS corresponds on average to 7.50 point change in CombiWISE with a standard error of 0.10. The novel validated clinical outcome, CombiWISE, outperforms the current broadly utilized MRI brain atrophy outcome and more than doubles sensitivity in detecting clinical deterioration in progressive MS in comparison to the scale traditionally used for regulatory approval, EDSS.
Collapse
Affiliation(s)
- Peter Kosa
- Neuroimmunological Diseases Unit, National Institute of Neurological Diseases and Stroke, National Institute of Health , Bethesda, MD , USA
| | - Danish Ghazali
- Neuroimmunological Diseases Unit, National Institute of Neurological Diseases and Stroke, National Institute of Health , Bethesda, MD , USA
| | - Makoto Tanigawa
- Neuroimmunological Diseases Unit, National Institute of Neurological Diseases and Stroke, National Institute of Health , Bethesda, MD , USA
| | - Chris Barbour
- Neuroimmunological Diseases Unit, National Institute of Neurological Diseases and Stroke, National Institute of Health, Bethesda, MD, USA; Department of Mathematical Sciences, Montana State University, Bozeman, MT, USA
| | - Irene Cortese
- Neuroimmunology Clinic, National Institute of Neurological Diseases and Stroke, National Institute of Health , Bethesda, MD , USA
| | - William Kelley
- Neuroimmunological Diseases Unit, National Institute of Neurological Diseases and Stroke, National Institute of Health , Bethesda, MD , USA
| | - Blake Snyder
- Neuroimmunological Diseases Unit, National Institute of Neurological Diseases and Stroke, National Institute of Health , Bethesda, MD , USA
| | - Joan Ohayon
- Neuroimmunology Clinic, National Institute of Neurological Diseases and Stroke, National Institute of Health , Bethesda, MD , USA
| | - Kaylan Fenton
- Neuroimmunology Clinic, National Institute of Neurological Diseases and Stroke, National Institute of Health , Bethesda, MD , USA
| | - Tanya Lehky
- EMG Section, National Institute of Neurological Diseases and Stroke, National Institute of Health , Bethesda, MD , USA
| | - Tianxia Wu
- Clinical Trials Unit, National Institute of Neurological Diseases and Stroke, National Institute of Health , Bethesda, MD , USA
| | - Mark Greenwood
- Department of Mathematical Sciences, Montana State University , Bozeman, MT , USA
| | - Govind Nair
- Neuroimmunology Clinic, National Institute of Neurological Diseases and Stroke, National Institute of Health , Bethesda, MD , USA
| | - Bibiana Bielekova
- Neuroimmunological Diseases Unit, National Institute of Neurological Diseases and Stroke, National Institute of Health , Bethesda, MD , USA
| |
Collapse
|
38
|
Fox RJ, Coffey CS, Cudkowicz ME, Gleason T, Goodman A, Klawiter EC, Matsuda K, McGovern M, Conwit R, Naismith R, Ashokkumar A, Bermel R, Ecklund D, Koepp M, Long J, Natarajan S, Ramachandran S, Skaramagas T, Thornell B, Yankey J, Agius M, Bashir K, Cohen B, Coyle P, Delgado S, Dewitt D, Flores A, Giesser B, Goldman M, Jubelt B, Lava N, Lynch S, Miravalle A, Moses H, Ontaneda D, Perumal J, Racke M, Repovic P, Riley C, Severson C, Shinnar S, Suski V, Weinstock-Gutman B, Yadav V, Zabeti A. Design, rationale, and baseline characteristics of the randomized double-blind phase II clinical trial of ibudilast in progressive multiple sclerosis. Contemp Clin Trials 2016; 50:166-77. [PMID: 27521810 DOI: 10.1016/j.cct.2016.08.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/02/2016] [Accepted: 08/09/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Primary and secondary progressive multiple sclerosis (MS), collectively called progressive multiple sclerosis (PMS), is characterized by gradual progression of disability. The current anti-inflammatory treatments for MS have little or no efficacy in PMS in the absence of obvious active inflammation. Optimal biomarkers for phase II PMS trials is unknown. Ibudilast is an inhibitor of macrophage migration inhibitor factor and phosphodiesterases-4 and -10 and exhibits possible neuroprotective properties. The goals of SPRINT-MS study are to evaluate the safety and efficacy of ibudilast in PMS and to directly compare several imaging metrics for utility in PMS trials. METHODS SPRINT-MS is a randomized, placebo-controlled, phase II trial of ibudilast in patients with PMS. Eligible subjects were randomized 1:1 to receive either ibudilast (100mg/day) or placebo for 96weeks. Imaging is conducted every 24weeks for whole brain atrophy, magnetization transfer ratio, diffusion tensor imaging, cortical brain atrophy, and retinal nerve fiber layer thickness. Clinical outcomes include neurologic disability and patient reported quality of life. Safety assessments include laboratory testing, electrocardiography, and suicidality screening. RESULTS A total of 331 subjects were enrolled, of which 255 were randomized onto active study treatment. Randomized subjects were 53.7% female and mean age 55.7 (SD 7.3) years. The last subject is projected to complete the study in May 2017. CONCLUSION SPRINT-MS is designed to evaluate the safety and efficacy of ibudilast as a treatment for PMS while simultaneously validating five different imaging biomarkers as outcome metrics for use in future phase II proof-of-concept PMS trials.
Collapse
Affiliation(s)
- Robert J Fox
- Cleveland Clinic, Neurological Institute, Cleveland, OH, United States.
| | - Christopher S Coffey
- Data Coordinating Center, NeuroNEXT, University of Iowa, Iowa City, IA, United States
| | - Merit E Cudkowicz
- Clinical Coordinating Center, NeuroNEXT, Harvard Partners, Boston, MA, United States
| | | | - Andrew Goodman
- University of Rochester Medical Center, Rochester, NY, United States
| | | | | | - Michelle McGovern
- Clinical Coordinating Center, NeuroNEXT, Harvard Partners, Boston, MA, United States
| | - Robin Conwit
- National Institutes of Neurological Disease and Stroke, Bethesda, MD, United States
| | - Robert Naismith
- Washington University School of Medicine, St. Louis, MO, United States
| | - Akshata Ashokkumar
- Data Coordinating Center, NeuroNEXT, University of Iowa, Iowa City, IA, United States
| | - Robert Bermel
- Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
| | - Dixie Ecklund
- Data Coordinating Center, NeuroNEXT, University of Iowa, Iowa City, IA, United States
| | - Maxine Koepp
- Data Coordinating Center, NeuroNEXT, University of Iowa, Iowa City, IA, United States
| | - Jeffrey Long
- Data Coordinating Center, NeuroNEXT, University of Iowa, Iowa City, IA, United States
| | - Sneha Natarajan
- Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
| | | | - Thomai Skaramagas
- Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
| | - Brenda Thornell
- Clinical Coordinating Center, NeuroNEXT, Harvard Partners, Boston, MA, United States
| | - Jon Yankey
- Data Coordinating Center, NeuroNEXT, University of Iowa, Iowa City, IA, United States
| | - Mark Agius
- University of California at Davis, Sacramento, CA; currently at Barrows Neurological Institute, Phoenix, AZ, United States
| | - Khurram Bashir
- University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bruce Cohen
- Northwestern University, Chicago, IL, United States
| | - Patricia Coyle
- State University of New York, Stony Brook, NY, United States
| | - Silvia Delgado
- University of Miami School of Medicine, Miami, FL, United States
| | - Dana Dewitt
- University of Utah, Salt Lake City, UT, United States
| | - Angela Flores
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Barbara Giesser
- University of California at Los Angeles, Los Angeles, CA, United States
| | - Myla Goldman
- University of Virginia at Charlottesville, Charlottesville, VA, United States
| | - Burk Jubelt
- State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Neil Lava
- Emory University, Atlanta, GA, United States
| | - Sharon Lynch
- University of Kansas Medical Center, Kansas City, KS, United States
| | | | - Harold Moses
- Vanderbilt University, Nashville, TN, United States
| | - Daniel Ontaneda
- Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
| | - Jai Perumal
- Weill Cornell Medical College, New York, NY, United States
| | - Michael Racke
- The Ohio State University, Columbus, OH, United States
| | - Pavle Repovic
- Swedish Medical Center at Seattle, Seattle, WA, United States
| | - Claire Riley
- Columbia University Medical Center, New York, NY, United States
| | | | | | - Valerie Suski
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | | | - Vijayshree Yadav
- Oregon Health and Science University, Portland, OR, United States
| | - Aram Zabeti
- University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
39
|
Cover KS, van Schijndel RA, Versteeg A, Leung KK, Mulder ER, Jong RA, Visser PJ, Redolfi A, Revillard J, Grenier B, Manset D, Damangir S, Bosco P, Vrenken H, van Dijk BW, Frisoni GB, Barkhof F. Reproducibility of hippocampal atrophy rates measured with manual, FreeSurfer, AdaBoost, FSL/FIRST and the MAPS-HBSI methods in Alzheimer's disease. Psychiatry Res Neuroimaging 2016; 252:26-35. [PMID: 27179313 DOI: 10.1016/j.pscychresns.2016.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 02/16/2016] [Accepted: 04/08/2016] [Indexed: 11/23/2022]
Abstract
The purpose of this study is to assess the reproducibility of hippocampal atrophy rate measurements of commonly used fully-automated algorithms in Alzheimer disease (AD). The reproducibility of hippocampal atrophy rate for FSL/FIRST, AdaBoost, FreeSurfer, MAPS independently and MAPS combined with the boundary shift integral (MAPS-HBSI) were calculated. Back-to-back (BTB) 3D T1-weighted MPRAGE MRI from the Alzheimer's Disease Neuroimaging Initiative (ADNI1) study at baseline and year one were used. Analysis on 3 groups of subjects was performed - 562 subjects at 1.5T, a 75 subject group that also had manual segmentation and 111 subjects at 3T. A simple and novel statistical test based on the binomial distribution was used that handled outlying data points robustly. Median hippocampal atrophy rates were -1.1%/year for healthy controls, -3.0%/year for mildly cognitively impaired and -5.1%/year for AD subjects. The best reproducibility was observed for MAPS-HBSI (1.3%), while the other methods tested had reproducibilities at least 50% higher at 1.5T and 3T which was statistically significant. For a clinical trial, MAPS-HBSI should require less than half the subjects of the other methods tested. All methods had good accuracy versus manual segmentation. The MAPS-HBSI method has substantially better reproducibility than the other methods considered.
Collapse
Affiliation(s)
- Keith S Cover
- VU University Medical Center, Amsterdam, Netherlands.
| | | | | | | | - Emma R Mulder
- VU University Medical Center, Amsterdam, Netherlands
| | - Remko A Jong
- VU University Medical Center, Amsterdam, Netherlands
| | | | | | | | | | | | | | - Paolo Bosco
- IRCCS San Giovanni di Dio Fatebenefratelli, Italy
| | - Hugo Vrenken
- VU University Medical Center, Amsterdam, Netherlands
| | | | - Giovanni B Frisoni
- IRCCS San Giovanni di Dio Fatebenefratelli, Italy; University Hospitals and University of Geneva, Switzerland
| | | |
Collapse
|
40
|
Chard D. Brain atrophy measurements should be used to guide therapy monitoring in MS - Commentary. Mult Scler 2016; 22:1526-1527. [PMID: 27335097 DOI: 10.1177/1352458516656061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Declan Chard
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Institute of Neurology, London, UK/National Institute for Health Research (NIHR) Biomedical Research Centre, University College London Hospitals (UCLH), London, UK
| |
Collapse
|
41
|
Zivadinov R, Hojnacki D, Bergsland N, Kennedy C, Hagemeier J, Melia R, Ramasamy DP, Durfee J, Carl E, Dwyer MG, Weinstock-Guttman B. Effect of natalizumab on brain atrophy and disability progression in multiple sclerosis patients over 5 years. Eur J Neurol 2016; 23:1101-9. [DOI: 10.1111/ene.12992] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/02/2016] [Indexed: 01/21/2023]
Affiliation(s)
- R. Zivadinov
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
- MR Imaging Clinical Translational Research Center; School of Medicine and Biomedical Sciences; University at Buffalo; State University of New York; Buffalo NY USA
| | - D. Hojnacki
- Jacobs MS Center; Department of Neurology; University at Buffalo; State University of New York; Buffalo NY USA
| | - N. Bergsland
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
- Magnetic Resonance Laboratory; IRCCS Don Gnocchi Foundation; Milan Italy
| | - C. Kennedy
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - J. Hagemeier
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - R. Melia
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - D. P. Ramasamy
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - J. Durfee
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - E. Carl
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - M. G. Dwyer
- Department of Neurology; Buffalo Neuroimaging Analysis Center; University at Buffalo; State University of New York; Buffalo NY USA
| | - B. Weinstock-Guttman
- Jacobs MS Center; Department of Neurology; University at Buffalo; State University of New York; Buffalo NY USA
| |
Collapse
|
42
|
Brown JWL, Chard DT. The role of MRI in the evaluation of secondary progressive multiple sclerosis. Expert Rev Neurother 2016; 16:157-71. [PMID: 26692498 DOI: 10.1586/14737175.2016.1134323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Magnetic resonance imaging already has an established role in the diagnosis of multiple sclerosis, but it also has the potential to provide prognostic information, and to monitor [corrected] disease progression in clinical trials and practice. Magnetic resonance imaging measures are increasingly being used as the primary outcome in early phase clinical trials of immunomodulatory therapies (for example brain white matter lesion counts or volumes, and gadolinium contrast enhancing lesions) and putatively neuroprotective agents (for example measures of whole brain atrophy), and trials of agents that promote remyelination are also likely to follow suit. In this review we consider the use of magnetic resonance imaging measures as predictors and markers of disease progression in multiple sclerosis, and explore possible future directions in this rapidly developing field.
Collapse
Affiliation(s)
- J William L Brown
- a Department of Clinical Neurosciences , University of Cambridge , Cambridge , UK.,b NMR Research Unit, Queen Square Multiple Sclerosis Centre, Institute of Neurology , University College London (UCL) , London , UK
| | - Declan T Chard
- b NMR Research Unit, Queen Square Multiple Sclerosis Centre, Institute of Neurology , University College London (UCL) , London , UK.,c Biomedical Research Centre, National Institute for Health Research (NIHR) , University College London Hospitals (UCLH) , London , UK
| |
Collapse
|
43
|
Popescu V, Schoonheim MM, Versteeg A, Chaturvedi N, Jonker M, Xavier de Menezes R, Gallindo Garre F, Uitdehaag BMJ, Barkhof F, Vrenken H. Grey Matter Atrophy in Multiple Sclerosis: Clinical Interpretation Depends on Choice of Analysis Method. PLoS One 2016; 11:e0143942. [PMID: 26745873 PMCID: PMC4706325 DOI: 10.1371/journal.pone.0143942] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/11/2015] [Indexed: 11/27/2022] Open
Abstract
Background Studies disagree on the location of grey matter (GM) atrophy in the multiple sclerosis (MS) brain. Aim To examine the consistency between FSL, FreeSurfer, SPM for GM atrophy measurement (for volumes, patient/control discrimination, and correlations with cognition). Materials and Methods 127 MS patients and 50 controls were included and cortical and deep grey matter (DGM) volumetrics were performed. Consistency of volumes was assessed with Intraclass Correlation Coefficient/ICC. Consistency of patients/controls discrimination was assessed with Cohen’s d, t-tests, MANOVA and a penalized double-loop logistic classifier. Consistency of association with cognition was assessed with Pearson correlation coefficient and ANOVA. Voxel-based morphometry (SPM-VBM and FSL-VBM) and vertex-wise FreeSurfer were used for group-level comparisons. Results The highest volumetry ICC were between SPM and FreeSurfer for cortical regions, and the lowest between SPM and FreeSurfer for DGM. The caudate nucleus and temporal lobes had high consistency between all software, while amygdala had lowest volumetric consistency. Consistency of patients/controls discrimination was largest in the DGM for all software, especially for thalamus and pallidum. The penalized double-loop logistic classifier most often selected the thalamus, pallidum and amygdala for all software. FSL yielded the largest number of significant correlations. DGM yielded stronger correlations with cognition than cortical volumes. Bilateral putamen and left insula volumes correlated with cognition using all methods. Conclusion GM volumes from FreeSurfer, FSL and SPM are different, especially for cortical regions. While group-level separation between MS and controls is comparable, correlations between regional GM volumes and clinical/cognitive variables in MS should be cautiously interpreted.
Collapse
Affiliation(s)
- Veronica Popescu
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam (NCA), VU University Medical Center, Amsterdam, The Netherlands
| | - Menno M. Schoonheim
- Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam (NCA), VU University Medical Center, Amsterdam, The Netherlands
| | - Adriaan Versteeg
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam (NCA), VU University Medical Center, Amsterdam, The Netherlands
| | - Nimisha Chaturvedi
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Marianne Jonker
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Renee Xavier de Menezes
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Francisca Gallindo Garre
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Bernard M. J. Uitdehaag
- Department of Neurology, Neuroscience Campus Amsterdam (NCA), VU University Medical Center, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam (NCA), VU University Medical Center, Amsterdam, The Netherlands
| | - Hugo Vrenken
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam (NCA), VU University Medical Center, Amsterdam, The Netherlands
- Department of Physics and Medical Technology, Neuroscience Campus Amsterdam (NCA), VU University Medical Center, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
44
|
Benjamin P, Zeestraten E, Lambert C, Chis Ster I, Williams OA, Lawrence AJ, Patel B, MacKinnon AD, Barrick TR, Markus HS. Progression of MRI markers in cerebral small vessel disease: Sample size considerations for clinical trials. J Cereb Blood Flow Metab 2016; 36:228-40. [PMID: 26036939 PMCID: PMC4758545 DOI: 10.1038/jcbfm.2015.113] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/18/2015] [Accepted: 04/30/2015] [Indexed: 11/09/2022]
Abstract
Detecting treatment efficacy using cognitive change in trials of cerebral small vessel disease (SVD) has been challenging, making the use of surrogate markers such as magnetic resonance imaging (MRI) attractive. We determined the sensitivity of MRI to change in SVD and used this information to calculate sample size estimates for a clinical trial. Data from the prospective SCANS (St George’s Cognition and Neuroimaging in Stroke) study of patients with symptomatic lacunar stroke and confluent leukoaraiosis was used (n = 121). Ninety-nine subjects returned at one or more time points. Multimodal MRI and neuropsychologic testing was performed annually over 3 years. We evaluated the change in brain volume, T2 white matter hyperintensity (WMH) volume, lacunes, and white matter damage on diffusion tensor imaging (DTI). Over 3 years, change was detectable in all MRI markers but not in cognitive measures. WMH volume and DTI parameters were most sensitive to change and therefore had the smallest sample size estimates. MRI markers, particularly WMH volume and DTI parameters, are more sensitive to SVD progression over short time periods than cognition. These markers could significantly reduce the size of trials to screen treatments for efficacy in SVD, although further validation from longitudinal and intervention studies is required.
Collapse
Affiliation(s)
- Philip Benjamin
- Neurosciences Research Centre, Cardiovascular and Cell Sciences Research Institute, St George’s University of London, London, UK
| | - Eva Zeestraten
- Neurosciences Research Centre, Cardiovascular and Cell Sciences Research Institute, St George’s University of London, London, UK
| | - Christian Lambert
- Neurosciences Research Centre, Cardiovascular and Cell Sciences Research Institute, St George’s University of London, London, UK
| | - Irina Chis Ster
- Institute of Infection and Immunity, St George's University of London, London, UK
| | - Owen A Williams
- Neurosciences Research Centre, Cardiovascular and Cell Sciences Research Institute, St George’s University of London, London, UK
| | | | - Bhavini Patel
- Neurosciences Research Centre, Cardiovascular and Cell Sciences Research Institute, St George’s University of London, London, UK
| | - Andrew D MacKinnon
- Atkinson Morley Regional Neuroscience Centre, St George’s NHS Healthcare Trust, London, UK
| | - Thomas R Barrick
- Neurosciences Research Centre, Cardiovascular and Cell Sciences Research Institute, St George’s University of London, London, UK
| | - Hugh S Markus
- Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
45
|
Reich DS, White R, Cortese IC, Vuolo L, Shea CD, Collins TL, Petkau J. Sample-size calculations for short-term proof-of-concept studies of tissue protection and repair in multiple sclerosis lesions via conventional clinical imaging. Mult Scler 2015; 21:1693-704. [PMID: 25662351 DOI: 10.1177/1352458515569098] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 12/16/2014] [Indexed: 01/15/2023]
Abstract
BACKGROUND New multiple sclerosis (MS) lesion activity on magnetic resonance imaging (MRI) can test immunomodulatory therapies in proof-of-concept trials. Comparably powerful endpoints to assess tissue protection or repair are lacking. OBJECTIVE The objective of this paper is to report sample-size calculations for assessment of new lesion recovery. METHODS In two sets of six active MS cases, new lesions were observed by monthly MRI for approximately 12 months. Averages and quartiles of normalized (proton density/T1/T2 weighted) and quantitative (T1/T2 and mean diffusivity maps for dataset 1, T2 and magnetization transfer ratio maps for dataset 2) measures were used to compare the lesion area before lesion appearance to afterward. A linear mixed-effects model incorporating lesion- and participant-specific random effects estimated average levels and variance components for sample-size calculations. RESULTS In both datasets, greatest statistical sensitivity was observed for the 25th percentile of normalized proton density-weighted signal. At 3T, using new lesions ⩾15 mm(3), as few as nine participants/arm may be required for a six-month placebo-controlled add-on trial postulating a therapeutic effect size of 20% and statistical power of 90%. CONCLUSION Lesion recovery is a powerful outcome measure for proof-of-concept clinical trials of tissue protection and repair in MS. The trial design requires active cases and is therefore best implemented near disease onset.
Collapse
Affiliation(s)
- Daniel S Reich
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, USA
| | - Richard White
- Department of Statistics, University of British Columbia, Canada
| | - Irene Cm Cortese
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, USA
| | - Luisa Vuolo
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, USA
| | - Colin D Shea
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, USA
| | | | - John Petkau
- Department of Statistics, University of British Columbia, Canada
| |
Collapse
|
46
|
Ontaneda D, Fox RJ, Chataway J. Clinical trials in progressive multiple sclerosis: lessons learned and future perspectives. Lancet Neurol 2015; 14:208-23. [PMID: 25772899 PMCID: PMC4361791 DOI: 10.1016/s1474-4422(14)70264-9] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Progressive multiple sclerosis is characterised clinically by the gradual accrual of disability independent of relapses and can occur with disease onset (primary progressive) or can be preceded by a relapsing disease course (secondary progressive). An effective disease-modifying treatment for progressive multiple sclerosis has not yet been identified, and so far the results of clinical trials have generally been disappointing. Ongoing advances in the knowledge of pathogenesis, in the identification of novel targets for neuroprotection, and in improved outcome measures could lead to effective treatments for progressive multiple sclerosis. In this Series paper, we summarise the lessons learned from completed clinical trials and perspectives from trials in progress in progressive multiple sclerosis. We review promising clinical, imaging, and biological markers, along with novel designs, for clinical trials. The use of more refined outcomes and truly neuroprotective drugs, coupled with more efficient trial design, has the capacity to deliver a new era of therapeutic discovery in this challenging area.
Collapse
Affiliation(s)
- Daniel Ontaneda
- Mellen Center for Multiple Sclerosis Treatment and Research, Cleveland Clinic, Cleveland, OH, USA.
| | - Robert J Fox
- Mellen Center for Multiple Sclerosis Treatment and Research, Cleveland Clinic, Cleveland, OH, USA
| | - Jeremy Chataway
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Institute of Neurology, University College London, London, UK; National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
47
|
Lavery AM, Verhey LH, Waldman AT. Outcome measures in relapsing-remitting multiple sclerosis: capturing disability and disease progression in clinical trials. Mult Scler Int 2014; 2014:262350. [PMID: 24883205 PMCID: PMC4026972 DOI: 10.1155/2014/262350] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/25/2014] [Indexed: 11/24/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disease that manifests as acute relapses and progressive disability. As a primary endpoint for clinical trials in MS, disability is difficult to both characterize and measure. Furthermore, the recovery from relapses and the rate of disability vary considerably among patients. Given these challenges, investigators have developed and studied the performance of various outcome measures and surrogate endpoints in MS clinical trials. This review defines the outcome measures and surrogate endpoints used to date in MS clinical trials and presents challenges in the design of both adult and pediatric trials.
Collapse
Affiliation(s)
- Amy M. Lavery
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Leonard H. Verhey
- The Pediatric Demyelinating Disease Program, Program in Neuroscience & Mental Health, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada M5G 1X8
| | - Amy T. Waldman
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Departments of Neurology and Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
48
|
Simon JH. MRI outcomes in the diagnosis and disease course of multiple sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2014; 122:405-25. [PMID: 24507528 DOI: 10.1016/b978-0-444-52001-2.00017-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Despite major advances in MRI, including practical implementations of multiple quantitative MRI methods, the conventional measures of focal, macroscopic disease remain the core MRI outcome measures in clinical trials. MRI enhancing lesion counts are used to assess inflammation, and new T2-lesions provide an index of (interval) activity between scans. These simple MRI measures also have immediate significance for early diagnosis as components of the 2010 revised dissemination in space and time criteria, and they provide a mechanism to monitor the subclinical disease in patients, including after treatment is initiated. The focal macroscopic injury, which includes demyelination and axonal damage, is at least partially linked to the diffuse injury through pathophysiologic mechanisms, such as secondary degeneration, but the diffuse diseases is largely independent. Quantitative measures of the more widespread pathology of the normal appearing white and gray matter currently remain applicable to populations of patients rather than individuals. Gray matter pathology, including focal lesions of the cortical gray matter and diffuse changes in the deep and cortical gray has emerged as both early and clinically relevant, as has atrophy. Major technical improvements in MRI hardware and pulse sequence design allow more specific and potentially more sensitive treatment metrics required for targeting outcomes most relevant to neuronal degeneration, remyelination and repair.
Collapse
Affiliation(s)
- Jack H Simon
- Oregon Health and Sciences University and Portland VA Medical Center, Portland, OR, USA.
| |
Collapse
|
49
|
Paulsen JS, Smith MM, Long JD. Cognitive decline in prodromal Huntington Disease: implications for clinical trials. J Neurol Neurosurg Psychiatry 2013; 84:1233-9. [PMID: 23911948 PMCID: PMC3795884 DOI: 10.1136/jnnp-2013-305114] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND OBJECTIVE Controversy exists regarding the feasibility of preventive clinical trials in prodromal Huntington disease (HD). A primary limitation is a lack of outcome measures for persons with the gene mutation who have not yet been diagnosed with HD. Many longitudinal studies of cognitive decline in prodromal HD have not stratified samples based on disease progression, thereby obscuring differences between symptomatic and nonsymptomatic individuals. METHODS Prodromal participants from PREDICT-HD were stratified by disease progression into one of three groups: those having a High, Medium, or Low probability of motor manifestation within the next 5 years. Data from a total of N=1299 participants with up to 5950 data points were subjected to linear mixed effects regression on 29 longitudinal cognitive variables, controlling for age, education, depression, and gender. RESULTS Performance of the three prodromal HD groups was characterised by insidious and significant cognitive decline over time. Twenty-one variables from 19 distinct cognitive tasks revealed evidence of a disease progression gradient, meaning that the rate of deterioration varied as a function of progression level, with faster deterioration associated with greater disease progression. Nineteen measures showed significant longitudinal change in the High group, nine showed significant change in the Medium group and four showed significant cognitive decline in the Low group. CONCLUSIONS Results indicate that clinical trials may be conducted in prodromal HD using the outcome measures and methods specified. The findings may help inform interventions in HD as well as other neurodegenerative disorders.
Collapse
Affiliation(s)
- Jane S Paulsen
- Department of Neurology, Carver College of Medicine, Univeristy of Iowa, , Iowa City, Iowa, USA
| | | | | |
Collapse
|
50
|
Long JD, Paulsen JS, Marder K, Zhang Y, Kim JI, Mills JA. Tracking motor impairments in the progression of Huntington's disease. Mov Disord 2013; 29:311-9. [PMID: 24150908 DOI: 10.1002/mds.25657] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 07/23/2013] [Accepted: 07/31/2013] [Indexed: 11/05/2022] Open
Abstract
The Unified Huntington's Disease Rating Scale is used to characterize motor impairments and establish motor diagnosis. Little is known about the timing of diagnostic confidence level categories and the trajectory of motor impairments during the prodromal phase. Goals of this study were to estimate the timing of categories, model the prodromal trajectory of motor impairments, estimate the rate of motor impairment change by category, and provide required sample size estimates for a test of efficacy in clinical trials. In total, 1010 gene-expanded participants from the Neurobiological Predictors of Huntington's Disease (PREDICT-HD) trial were analyzed. Accelerated failure time models were used to predict the timing of categories. Linear mixed effects regression was used to model the longitudinal motor trajectories. Age and length of gene expansion were incorporated into all models. The timing of categories varied significantly by gene expansion, with faster progression associated with greater expansion. For the median expansion, the third diagnostic confidence level category was estimated to have a first occurrence 1.5 years before diagnosis, and the second and first categories were estimated to occur 6.75 years and 19.75 years before diagnosis, respectively. Motor impairments displayed a nonlinear prodromal course. The motor impairment rate of change increased as the diagnostic confidence level increased, with added acceleration for higher progression scores. Motor items can detect changes in motor impairments before diagnosis. Given a sufficiently high progression score, there is evidence that the diagnostic confidence level can be used for prodromal staging. Implications for Huntington's disease research and the planning of clinical trials of efficacy are discussed.
Collapse
Affiliation(s)
- Jeffery D Long
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA; Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | | | | | | | | | | | | |
Collapse
|