SNPMiner Trials by Shray Alag


SNPMiner SNPMiner Trials (Home Page)


Report for Mutation T25W

Developed by Shray Alag, 2020.
SNP Clinical Trial Gene

There are 9 clinical trials

Clinical Trials


1 Project FARMS: Fall Risk Reduction in Multiple Sclerosis

Over half of persons with multiple sclerosis (MS) report falling over a 6-month period and a majority of those who fall require medical attention for injuries. Importantly, balance dysfunction, muscle weakness, and spasticity are modifiable risk factors for falls among community-dwelling older adults and likely persons with MS. Indeed, there is evidence that these physiological risk factors can be minimized with exercise training in persons with MS and this might translate into a decrease in fall risk as documented in community-dwelling older adults. The investigation will examine the effectiveness of a home-based exercise program that is designed to reduce fall risk by targeting specific fall risk factors including balance dysfunction and two of its latent causes, muscle weakness and spasticity in persons with multiple sclerosis. It is predicted that persons who receive home-based exercise program will have a reduction in fall risk.

NCT01837017 Multiple S Multiple Sclerosis Behavioral: Home-based Exercise
MeSH:Multiple Sclerosis Sclerosis

Mobility will be quantified with performance on timed 25 foot walk (T25W), timed up and go (TUG), Six spot step test, 6 minute walk and the MS walking scale-12.. Balance. --- T25W ---

Primary Outcomes

Description: Physiological fall risk will be determined by the physiological profile assessment which assesses physiological function related to fall risk by combining measures of vision, proprioception, lower-limb strength, postural sway, and cognitive function.

Measure: Physiological Fall risk

Time: 3 months

Secondary Outcomes

Description: Mobility will be quantified with performance on timed 25 foot walk (T25W), timed up and go (TUG), Six spot step test, 6 minute walk and the MS walking scale-12.

Measure: Mobility

Time: 3 months

Description: Balance will be quantified with the Berg Balance scale; self-report of balance impairment (ABC); and force platform metrics (sway range and velocity).

Measure: Balance

Time: 3 Months

Description: Spasticity will be assessed with the modified ashworth scale.

Measure: Spasticity

Time: 3 Months

2 Fall Risk Reduction in Multiple Sclerosis: Exercise Versus Behavior

Falls are a serious health concern for persons with multiple sclerosis (MS). Over 50% of persons with MS suffer a fall over a 6-month periodwith the majority of falls resulting in medical attention for injuries (i.e., lacerations, bone fractures, & head injuries). The effects of a fall are often compounded as it can lead to activity curtailment, physiological deconditioning, and institutionalization. Despite the importance of falls in persons with MS, the appropriate prevention strategies (i.e. rehabilitation approaches) are not clear. The purpose of this investigation is to determine whether exercise based or educational based interventions are more suited for fall prevention in older adults with MS.

NCT01956227 Multiple Sclerosis Adult Disease Behavioral: Home-based exercise Behavioral: Education Behavioral: Exercise plus Education
MeSH:Multiple Sclerosis Sclerosis

Walking speed was quantified with the Timed 25-Foot Walk (T25W), walking endurance was assessed with the 6-Minute Walk (6MW), and functional mobility was quantified with the Timed Up and Go (TUG) . --- T25W ---

Primary Outcomes

Measure: Fall incidence

Time: 3 months

Secondary Outcomes

Description: Physiological fall risk will be determined by the short form of the Physiological Profile Assessment (PPA)(Lord, 2003). The PPA is a standardized test battery which assesses vision (edge contrast sensitivity), lower limb proprioception, strength (knee extension), postural sway, and cognitive function (simple hand reaction time). The outcome of each test will be combined to generate an overall fall risk score

Measure: Physiological Fall Risk

Time: 3 Months

Description: Specific measures of walking speed, endurance, coordination and self-reported walking function scale will be employed to assess overall mobility of each person. Walking speed was quantified with the Timed 25-Foot Walk (T25W), walking endurance was assessed with the 6-Minute Walk (6MW), and functional mobility was quantified with the Timed Up and Go (TUG) . The Multiple Sclerosis Walking Scale-12 (MSWS-12) will be used as a self-reported measure of walking impairment.

Measure: Mobility

Time: 3 months

Description: To assess balance (e.g. postural control), we conducted a clinical assessment To measure balance the Berg Balance Scale (BBS) and self-reports of balance confidence will be used. The BBS is a clinical assessment of balance. Scores on the BBS range from 0-56 with higher scores indicating greater balance. The Activities-Specific Balance Confidence (ABC) scale was used as a measure of balance confidence.

Measure: Balance

Time: 3 Months

3 A Multicenter, Randomized, Double-Blind, Placebo-Controlled, Phase 3 Study to Evaluate the Efficacy and Safety of Satralizumab (SA237) as Monotherapy in Patients With Neuromyelitis Optica (NMO) and Neuromyelitis Optica Spectrum Disorder (NMOSD)

The objectives of this study are to evaluate the efficacy, safety, pharmacodynamic, pharmacokinetic and immunogenic profiles of satralizumab in participants with NMO and NMOSD.

NCT02073279 Neuromyelitis Optica (NMO) NMO Spectrum Disorder (NMOSD) Drug: Satralizumab Drug: Placebo
MeSH:Neuromyelitis Optica

A higher score reflects a better health state.. Change from Baseline Over Time in the Timed 25-Foot Walk (T25W). --- T25W ---

The T25W is the measurement to assess walking ability. --- T25W ---

Primary Outcomes

Measure: Time to First Protocol-Defined Relapse in the Double-Blind Period

Time: From the date of randomization until the first occurrence of a protocol-defined relapse throughout the double-blind period (up to approximately 50 months)

Secondary Outcomes

Description: The VAS for pain is a subjective measure and it consists of a 100 millimeter (mm) line with two end points representing 'no pain' to 'pain as bad as it could be'. Participants are asked to rate their pain by placing a mark on the line corresponding to their current level of pain. The distance along the line from the 'no pain' marker is then measured with a ruler giving a pain score out of 10.

Measure: Change from Baseline to Week 24 in the Visual Analogue Scale (VAS) Score for Pain

Time: Baseline, Week 24

Description: FACIT fatigue scale includes 13 statements, which measures fatigue/asthenia for participants with chronic, life-threatening illnesses. For each question, a participant rates his/her condition for the past week on a 5-point Likert scale ranging from 0 (not at all) to 4 (very much). A score is calculated by averaging the individual question scores, with lower scores indicative of less fatigue.

Measure: Change from Baseline to Week 24 in the Functional Assessment of Chronic Illness Therapy (FACIT) Fatigue Score

Time: Baseline, Week 24

Description: The SF-36 domain scores range from 0-100. Higher scores indicate better quality of life.

Measure: Change from Baseline Over Time in the Short Form Generic Health Survey (SF-36) Bodily Pain Domain Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The SF-36 domain scores range from 0-100. Higher scores indicate better quality of life.

Measure: Change from Baseline Over Time in the SF-36 General Health Domain Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The SF-36 domain scores range from 0-100. Higher scores indicate better quality of life.

Measure: Change from Baseline Over Time in the SF-36 Mental Health Domain Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The SF-36 domain scores range from 0-100. Higher scores indicate better quality of life.

Measure: Change from Baseline Over Time in the SF-36 Physical Functioning Domain Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The SF-36 domain scores range from 0-100. Higher scores indicate better quality of life.

Measure: Change from Baseline Over Time in the SF-36 Role-Emotional Domain Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The SF-36 domain scores range from 0-100. Higher scores indicate better quality of life.

Measure: Change from Baseline Over Time in the SF-36 Role-Physical Domain Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The SF-36 domain scores range from 0-100. Higher scores indicate better quality of life.

Measure: Change from Baseline Over Time in the SF-36 Social Role Functioning Domain Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The SF-36 domain scores range from 0-100. Higher scores indicate better quality of life.

Measure: Change from Baseline Over Time in the SF-36 Vitality Domain Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The SF-36 domain scores range from 0-100. Higher scores indicate better quality of life.

Measure: Change from Baseline Over Time in the SF-36 Mental Component Summary Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The SF-36 domain scores range from 0-100. Higher scores indicate better quality of life.

Measure: Change from Baseline Over Time in the SF-36 Physical Component Summary Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The EQ-5D is a participant-answered questionnaire measuring 5 dimensions of mobility, self-care, usual activities, pain/discomfort, and anxiety/depression with 3 possible response categories: 1) no problems; 2) some problems; 3) severe problems. The EQ-5D index score is scored on a scale of -0.2 to 1. A higher score reflects a better health state.

Measure: Change from Baseline Over Time in the EuroQoL-5 Dimensions (EQ-5D) Index Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The T25W is the measurement to assess walking ability. The time (in seconds) that it takes the participant to walk 25 feet is measured.

Measure: Change from Baseline Over Time in the Timed 25-Foot Walk (T25W)

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Measure: Percentage of Participants Who Are Relapse-Free Over Time

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Measure: Annualized Relapse Rate

Time: From Baseline until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The mRS is a 7-point disability scale that assesses the degree of disability in patients with neurological impairment. Possible scores range from 0 (no symptoms at all) up to 6 (death). Higher scores reflect increased disability.

Measure: Change from Baseline Over Time in Modified Rankin Scale (mRS) Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The ZBI is the measurement to assess caregiver burden. The 22 items ask for the strain caregivers perceive. Responses range from 0 (never) to 4 (nearly always). The overall ZBI score ranges from 0 to 88. The higher the total score, the heavier the perceived burden.

Measure: Change from Baseline Over Time in Zarit Burden Interview (ZBI) Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Description: The EDSS scale ranges from 0 to 10 in 0.5 unit increments that represent higher levels of disability.

Measure: Change from Baseline Over Time in Expanded Disability Status Scale (EDSS) Score

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Measure: Change from Baseline Over Time in Visual Acuity (Snellen Chart)

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Measure: Change from Baseline Over Time in Low-Contrast Visual Acuity, as Assessed Using the Low-Contrast Sloan Letter Chart (LCSLC)

Time: Baseline and every 24 weeks thereafter until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Measure: Number of Participants with at Least One Adverse Event by Severity

Time: From Baseline until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Measure: Number of Participants with at Least One Serious Adverse Event by Severity

Time: From Baseline until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Measure: Number of Participants with Non-Serious Adverse Events of Special Interest by Severity

Time: From Baseline until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Measure: Number of Participants with Selected Adverse Events by Severity

Time: From Baseline until 12 weeks after last dose of satralizumab (up to approximately 7.25 years)

Measure: Number of Participants by Columbia-Suicide Severity Rating Scale (C-SSRS) Scores

Time: Baseline, Weeks 2, 4, 8, and every 4 weeks thereafter of double-blind period; every 4 weeks for first 48 weeks and every 12 weeks thereafter of open-label extension period (up to approximately 7.25 years)

Measure: Serum Satralizumab Concentration Over Time

Time: Baseline, Weeks 2, 4, 5, 6, 8, and every 4 weeks thereafter of double-blind period; every 4 weeks for first 48 weeks and every 24 weeks thereafter of open-label extension period (up to approximately 7.25 years)

Measure: Serum Interleukin-6 (IL-6) Concentration Over Time

Time: Baseline, Weeks 2, 4, 8, and every 4 weeks thereafter of double-blind period; every 4 weeks for first 48 weeks of open-label extension period (up to approximately 7.25 years)

Measure: Serum Soluble IL-6 Receptor (sIL-6R) Concentration Over Time

Time: Baseline, Weeks 2, 4, 8, and every 4 weeks thereafter of double-blind period; every 4 weeks for first 48 weeks of open-label extension period (up to approximately 7.25 years)

Measure: Serum High Sensitivity C-Reactive Protein (hsCRP) Concentration Over Time

Time: Baseline, Weeks 2, 4, 8, and every 4 weeks thereafter of double-blind period; every 4 weeks for first 48 weeks of open-label extension period (up to approximately 7.25 years)

Measure: Blood Anti-Aquaporin-4 (AQP4) Antibody Concentration Over Time

Time: Baseline, Weeks 2, 4, 8, 12, 24, 48, and every 24 weeks thereafter of double-blind period; every 24 weeks for first 48 weeks of open-label extension period (up to approximately 7.25 years)

Measure: Blood Plasmablast Concentration Over Time

Time: Baseline, Weeks 2, 4, 8, 12, 24, 48, and every 24 weeks thereafter of double-blind period (up to approximately 50 months)

Measure: Number of Participants with Anti-Drug Antibodies to Satralizumab

Time: Baseline and every 4 weeks thereafter of double-blind period; every 4 weeks for first 48 weeks and every 24 weeks thereafter of open-label extension period (up to approximately 7.25 years)

4 Open-label, Single-center, Single-arm Futility Trial Evaluating Oral Domperidone 10mg QID for Reducing Progression of Disability in Patients With Secondary Progressive Multiple Sclerosis (SPMS)

The purpose of this clinical trial is to determine if Domperidone in a dose of 40 mg daily can prevent worsening of walking ability in people secondary progressive MS. The number of participants in this study will be 62. A maximum of 75 people with secondary progressive MS will be included. Each patient will be followed for 12 months from inclusion. Domperidone is a medication which has been shown to increase levels of the hormone prolactin. The best understood function of prolactin is the stimulation of milk production in women after delivery. However, the increase in prolactin levels seen in patients treated with standard doses of Domperidone (in doses of up to 80mg per day) usually does not lead to clinical symptoms. Prolactin has been shown to improve myelin repair in mice. Domperidone therefore may also improve myelin repair in people with MS. Domperidone is currently approved in Canada to treat slow moving bowels and nausea, for instance in patients with Parkinson's Disease or Diabetes Mellitus, where too slowly moving bowels can cause constipation. Domperidone is available as a tablet that is usually taken four times per day. Doses up to 80mg per day may be used but we estimate that a dose of only 40mg daily will be needed to stimulate myelin repair. Domperidone is usually well tolerated.

NCT02308137 Multiple Sclerosis, Secondary Progressive Drug: Domperidone
MeSH:Neoplasm Metastasis Multiple Sclerosis Multiple Sclerosis, Chronic Progressive Sclerosis

Timed 25-Foot Walk (T25W). --- T25W ---

Primary Outcomes

Description: quantitative ambulation performance test

Measure: Timed 25-Foot Walk (T25W)

Time: up to 12 months

Secondary Outcomes

Description: brief, standardized, quantitative test of upper extremity

Measure: 9-Hole Peg Test

Time: administered at baseline, one month, 6 months, and 12 months

Description: measures cognitive processing speed and working memory

Measure: Symbol Digit Modalities Test

Time: administered at baseline, one month, 6 months, and 12 months

Description: EDSS is the standard measure of neurologic impairment that is used to describe disability in MS. The neurological assessment comprises seven functional systems.

Measure: Functional Systems and Expanded Disability Status Scale (EDSS)

Time: administered at baseline, one month, 6 months, and 12 months

Description: structured, self-report questionnaire with 21 itmes concerning how fatigue impacts patient's life

Measure: Modified Fatigue Impact Scale (MFIS)

Time: administered at baseline, one month, 6 months, and 12 months

Description: 54-item multidimensional health-related quality of life measure that combines both generic and MS-specific items

Measure: Multiple Sclerosis Quality of Life Scale 54 item version

Time: administered at baseline, one month, 6 months, and 12 months

5 A Phase 1, Two-part, Open-label Dose-escalation and Double-blind, Placebo-controlled Dose-expansion Study With an Open-label Extension to Evaluate the Safety and Efficacy of ATA188 in Subjects With Progressive Multiple Sclerosis

The purpose of this study is to evaluate the safety and tolerability of ATA188 as a monotherapy in Parts 1 and 2, to determine the recommended Part 2 dose (RP2D) of ATA188 as monotherapy in Part 1, and to evaluate the effect of ATA188 treatment on biological markers of disease activity in cerebral spinal fluid in Part 2 in participants with progressive forms of multiple sclerosis (MS) (primary progressive multiple sclerosis [PPMS] and secondary progressive multiple sclerosis [SPMS]).

NCT03283826 Primary Progressive Multiple Sclerosis Secondary Progressive Multiple Sclerosis Biological: ATA188 Drug: Placebo
MeSH:Multiple Sclerosis Multiple Sclerosis, Chronic Progressive Sclerosis

Part 2: Change from baseline in clinical disability as assessed by the EDSS score and/or Timed 25 foot Walk (T25W) and/or 9-hole Peg Test (9HPT). --- T25W ---

Primary Outcomes

Description: Safety and tolerability

Measure: Part 1 and Part 2: Incidence of adverse events

Time: At 12 months after the first dose of study drug

Measure: Part 1: Incidence of clinically significant changes in laboratory tests, electrocardiograms (ECGs), and vital signs

Time: At 12 months after the first dose of study drug

Description: Dose assessment

Measure: Part 1: Recommended Part 2 dose of ATA188 monotherapy

Time: Day 1 to Day 35 of Cycle 1 for each participant in dose escalation part (approximately 1 year)

Description: Antibody assessment and quantification

Measure: Part 2: Change from baseline in immunoglobulin G (IgG) index, including quantification of IgG production

Time: At 12 months after the first dose of study drug

Secondary Outcomes

Description: Changes in disability score

Measure: Part 1: Change from baseline in expanded disability status scale (EDSS) score

Time: At 12 months after the first dose of study drug

Description: Changes in disability score

Measure: Part 2: Change from baseline in clinical disability as assessed by the EDSS score and/or Timed 25 foot Walk (T25W) and/or 9-hole Peg Test (9HPT)

Time: At 12 months after the first dose of study drug

Description: Change in MRI activity

Measure: Part 2: Change from baseline in cervical spinal cord volume and whole brain volume on MRI scans

Time: At 12 months after the first dose of study drug

Description: Change in MRI activity

Measure: Part 2: Change from baseline in the number of Gadolinium-enhancing and new or enlarging T2 lesions on brain MRI scans

Time: At 12 months after the first dose of study drug

6 Effect of Alemtuzumab on Microglial Activation Assessed Using Novel [F-18]-Based Positron Emission Tomography (PET) Ligand in Multiple Sclerosis

Specific Aims The specific aims of the study are: - Primary Objective: To assess the effect of alemtuzumab on microglial activation in MS patients. The hypothesis is that alemtuzumab reduces microglial activation in MS, which may mediate its effect on reducing conversion of RRMS patients to SPMS, and its effects on cognition, including cognitive fatigue. - Secondary Objective: To determine the time course of effect of alemtuzumab on microglial activation. The hypothesis is that alemtuzumab reduces microglial activation at 6 months after initiation of treatment and this effect persists and is accentuated at 18 years, i.e. after administration of the second course

NCT03983252 Multiple Sclerosis Drug: [F-18]PBR06
MeSH:Multiple Sclerosis Sclerosis

Non Imaging/Clinical Data The following non-imaging/clinical data will be obtained: Expanded Disability Status Scale (EDSS) Timed 25-feet walk (T25W) MS Functional Composite (MSFC) Minimal Assessment of Cognitive Function Scale in MS (MACFIMS) battery Symbol digit modalities test (SDMT) MSQOL-54 scale (QOL) Modified fatigue Impact Scale (MFIS) Pittsburgh Sleep Quality Index (PSQI) Beck's Depression Inventory (BDI) Center for Epidemiological Studies-Depression Scale (CES-D) Hospital Anxiety and Depression Scale (HADS) --- T25W ---

Primary Outcomes

Description: PET outcome measure change at 18 months from baseline

Measure: PET Uptake/Standardized uptake value ratio (SUVR) change

Time: baseline and 18 months

Secondary Outcomes

Description: PET outcome measure change at 6 months from baseline

Measure: PET Uptake/Standardized uptake value ratio (SUVR) change

Time: baseline and 6 months

Description: MRI outcome measure change at 18 months from baseline

Measure: T2/FLAIR lesion load change

Time: baseline and 18 months

Description: MRI outcome measure change at 18 months from baseline

Measure: Whole brain/deep gray matter atrophy change

Time: baseline and 18 months

Description: Clinical outcome measure change at 18 months from baseline; Scale Range: 0-10; Higher values represent worse outcomes

Measure: Expanded Disability Status Scale (EDSS) change

Time: baseline and 18 months

Description: Clinical outcome measure change at 18 months from baseline

Measure: Timed 25-foot walk (T25FW) change

Time: baseline and 18 months

Description: Clinical outcome measure change at 18 months from baseline; Physical subscale range: 0-36; Cognitive subscale range: 0-40; Psychosocial subscale range: 0-8; Total MFIS Score scale range (Subscales Summed): 0-84; Higher values represent worse outcomes

Measure: Modified Fatigue Impact Scale (MIFS) change

Time: baseline and 18 months

Description: Clinical outcome measure change at 18 months from baseline

Measure: Minimal Assessment of Cognitive Function in MS (MACFIMS) change

Time: baseline and 18 months

7 A Multicenter, Longitudinal, Open-Label, Single-Arm Study Describing Cognitive Processing Speed Changes in Relapsing Multiple Sclerosis Subjects Treated With Ozanimod (RPC-1063)

This is a multicenter, longitudinal, single-arm, open-label study to describe the change from baseline in cognitive processing speed, measured by the SDMT, in subjects with RMS treated with ozanimod HCl 1 mg at 3 years. All subjects will receive orally administered ozanimod HCl 1 mg. The primary efficacy endpoint is the proportion of subjects with a clinically meaningful increase in raw score of ≥ 4 points or 10% from baseline (improved). The treatment period is 36 months. For subjects who discontinue the study, there will be a 30-day (± 15 days) and a 90-day (± 10 days) Safety Follow-up Visit. There is no planned protocol extension following the end of the study. Approximately 250 subjects with RMS will be recruited for this study. Subjects with RMS will be enrolled in this study if they have received ≤1 DMT, have an EDSS ≤ 3.5, and have been diagnosed with RMS within 5 years of study entry. The Investigator will be responsible for the overall conduct of the study at the site, confirmation of subject eligibility, routine study subject clinical management including for MS relapses, and management of AEs.

NCT04140305 Multiple Sclerosis Drug: RPC-1063
MeSH:Multiple Sclerosis Sclerosis

Timed 25-foot Walk (T25W). --- T25W ---

Disability progression assessed by 20% worsening from baseline over 3 years on T25W. --- T25W ---

Primary Outcomes

Description: Symbol Digit Modalities Test

Measure: Proportion of subjects with an increase in raw score of ≥ 4 points or 10% from baseline (improved)

Time: Up to approximately 3 years

Secondary Outcomes

Description: Symbol Digit Modalities Test

Measure: Proportion of subjects with a decrease in raw score of ≥ 4 points or 10% from baseline (worsened)

Time: Up to approximately 3 years

Description: Symbol Digit Modalities Test

Measure: Proportion of subjects with a raw score change from baseline who do not meet the improved or worsened definition (stable)

Time: Up to approximately 3 years

Description: Symbol Digit Modalities Test

Measure: Proportion of subjects with an increase in raw score of ≥ 3 points from baseline

Time: Up to approximately 3 years

Description: Symbol Digit Modalities Test

Measure: Proportion of subjects with a decrease in raw score of ≥ 3 points from baseline

Time: Up to approximately 3 years

Description: The SDMT is a measure of cognitive processing speed

Measure: Change from baseline in Symbol Digit Modalities Test (SMDT)

Time: Up to approximately 3 years

Description: Magnetic resonance imaging (MRI) brain volume

Measure: Percent change from baseline in thalamic, cortical grey matter, whole brain, lateral ventricular, and MOV volumes

Time: Up to approximately 3 years

Description: Magnetic Resonance Imaging

Measure: Proportion of subjects free of gadolinium enhancing (GdE) lesions over 3 years

Time: Up to approximately 3 years

Description: Magnetic Resonance Imaging

Measure: GdE lesion volume over 3 years

Time: Up to approximately 3 years

Description: Magnetic Resonance Imaging

Measure: Number of unique new or enlarging hyperintense T2-weighted lesions and their volume from baseline to Year 3

Time: Up to approximately 3 years

Description: Magnetic Resonance Imaging

Measure: Number of unique new or enlarging hypointense T1 weighted lesions and their volume from baseline to Year 3

Time: Up to approximately 3 years

Description: Change is TSQM score over 3 years

Measure: Treatment Satisfaction Questionnaire for Medication (TSQM v1.4)

Time: Up to approximately 3 years

Description: Change in WPAI score over 3 years

Measure: Work Productivity and Activity Impairment-Multiple Sclerosis (WPAI-MS)

Time: Up to approximately 3 years

Description: The Fatigue Severity Scale (FSS) questionnaire contains nine statements that attempt to explore severity of fatigue symptoms.

Measure: Fatigue Severity Scale (FSS)

Time: Up to approximately 3 years

Description: The MSQOL-54 is a multidimensional health-related QOL measure that combines both generic and MS-specific items into a single instrument

Measure: Multiple Sclerosis Quality of Life-54 (MSQOL-54)

Time: Up to approximately 3 years

Description: The HADS was developed to identify anxiety disorders and depression among subjects in nonpsychiatric hospital clinics

Measure: Hospital Anxiety and Depression Scale (HADS)

Time: Up to approximately 3 years

Description: Change in relapse rate over 3 years

Measure: Annualized relapse rate (ARR)

Time: Up to approximately 3 years

Description: Disability progression assessed by 20% worsening from baseline over 3 years on T25W

Measure: Timed 25-foot Walk (T25W)

Time: Up to approximately 3 years

Description: Change from baseline in the time in seconds needed to complete test activity

Measure: Nine-hole Peg Test (9-HPT)

Time: Up to approximately 3 years

Description: Change from baseline in EDSS score (0-10) yearly and at 3 years

Measure: Expanded Disability Status Scale (EDSS)

Time: Up to approximately 3 years

Description: An AE is any noxious, unintended, or untoward medical occurrence that may appear or worsen in a subject during the course of a study. It may be a new intercurrent illness, a worsening concomitant illness, an injury, or any concomitant impairment of the subject's health, including laboratory test values, regardless of etiology. Any worsening (ie, any clinically significant adverse change in the frequency or intensity of a pre-existing condition) should be considered an AE.

Measure: Adverse Events (AEs)

Time: Up to approximately 3 years

8 Role of Microglial Activation and Norepinephrine Transporter Abnormalities in Pathogenesis of MS-related Fatigue

The overarching aim is to assess the role of microglial activation and norepinephrine transporter binding in pathogenesis of MS-related fatigue, using novel Positron Emission Tomography (PET) radiotracers, [F-18]PBR06 and [C-11]MRB. Specific Aims: Specific Aim 1: To determine the relationship of cerebral microglial activation, as assessed by [F-18]PBR06 PET, with MS-related fatigue. Specific Aim 2: To determine the relationship of norepinephrine transporter (NET) binding, as assessed by [C-11]MRB PET, with MS-related fatigue. Specific Aim 3: To determine the relationship of microglial activation and NET binding, with grey matter pathology (lesion load and brain atrophy) assessed using 7T MRI, and evaluate their independent contribution in development of MS-related fatigue.

NCT04144257 Multiple Sclerosis Drug: [F-18]PBR06 Drug: [C-11]Methylreboxetine
MeSH:Multiple Sclerosis Fatigue
HPO:Fatigue

Clinical Data The following non-imaging, clinical data will be obtained: Modified fatigue Impact Scale (MFIS) Fatigue Severity Status Scale (FSSS) Expanded Disability Status Scale (EDSS) Timed 25-feet walk (T25W) MS Functional Composite (MSFC) Symbol digit modalities test (SDMT) MSQOL-54 scale (QOL) Pittsburgh Sleep Quality Index (PSQI) Beck's Depression Inventory (BDI) Center for Epidemiological Studies-Depression Scale (CES-D) Hospital Anxiety and Depression Scale (HADS) --- T25W ---

Primary Outcomes

Description: PET outcome measure

Measure: Standardized Uptake Value (SUV)/Standardized Uptake Value Ratio (SUVR)

Time: Baseline

Secondary Outcomes

Description: Clinical outcome measure; Physical subscale range: 0-36; Cognitive subscale range: 0-40; Psychosocial subscale range: 0-8; Total MFIS Score scale range (Subscales Summed): 0-84; Higher values represent worse outcomes.

Measure: Modified Fatigue Impact Scale (MFIS)

Time: Baseline

Description: PET outcome measure

Measure: Binding Potential (BPnd)

Time: Baseline

Description: MRI outcome measure

Measure: MRI grey matter lesional load/brain atrophy

Time: Baseline

Description: PET outcome measure

Measure: Tissue Volume of distribution (Vt)/Distribution Volume Ratios (DVR)

Time: Baseline

Description: MRI outcome measure

Measure: MRI global/regional volumetrics

Time: Baseline

9 Open-label, Observational, Prospective, 9-month Study to Assess the Efficacy of Ofatumumab on Microglia in Patients With Relapsing Forms of Multiple Sclerosis

We aim to assess the effect of Ofatumumab on microglial activation using [F-18]PBR06 PET in MS patients in relation to changes in serum markers, MRI abnormalities and clinical impairment longitudinally over 9 months. Specific Aims: Specific Aim 1: To determine the effect of Ofatumumab on microglial activation in MS over 9 months. Specific Aim 2: To determine the time course of effect of Ofatumumab on microglial activation and its relationship with peripheral B-cell depletion, serum neurofilament light (sNfL) chain and glial-fibrillary acid protein (GFAP) levels and other serum biomarkers Specific Aim 3: To determine the relationship of PET changes following Ofatumumab initiation with 3T MRI changes and clinical parameters.

NCT04510220 Relapsing Multiple Sclerosis Drug: Ofatumumab Drug: [F-18]PBR06
MeSH:Multiple Sclerosis Sclerosis

Clinical Data: The following non-imaging, clinical data will be obtained: Expanded Disability Status Scale (EDSS), Timed 25-feet walk (T25W), 9-Hole Peg Test (9HPT), Four component MS Functional Composite (MSFC-4), Symbol-Digit Modality test (SDMT), cognitive and symptom questionnaires, vision testing, Levels of serum biomarkers. --- T25W ---

Primary Outcomes

Description: The primary endpoint of the study will be the change in regional microglial activity in the brain using standardized uptake value ratio (SUVR) measurements at early and late time-points (5, 28, 90 and 273 days) as compared to baseline.

Measure: Effect of Ofatumumab on microglial activity

Time: Baseline to 9 months

Secondary Outcomes

Description: Association of change in regional microglial activity in the brain using standardized uptake value ratio (SUVR) measurements with changes in the peripheral CD19 counts at 5, 28, 90 and 273 days as compared to baseline. We will calculate the change from day 0 to each of the other time points for each of the measures, and we will estimate the correlation between the changes using Pearson's correlation coefficient.

Measure: Relationship between changes in Microglial activity and CD19 counts

Time: Baseline to 9 months

Description: Association of change in regional microglial activity in the brain using standardized uptake value ratio (SUVR) measurements with changes in the serum glial fibrillary acid protein (GFAP) level measurements at 5, 28, 90 and 273 days as compared to baseline. We will calculate the change from day 0 to each of the other time points for each of the measures, and we will estimate the correlation between the changes using Pearson's correlation coefficient.

Measure: Relationship between changes in Microglial activity and glial fibrillary acid protein (GFAP)

Time: Baseline to 9 months

Description: Association of change in regional microglial activity in the brain using standardized uptake value ratio (SUVR) measurements with changes in the serum neurofilament light chain levels at 5, 28, 90 and 273 days as compared to baseline. We will calculate the change from day 0 to each of the other time points for each of the measures, and we will estimate the correlation between the changes using Pearson's correlation coefficient.

Measure: Relationship between changes in Microglial activity and neurofilament light chain

Time: Baseline to 9 months

Description: Association of change in regional microglial activity in the brain using standardized uptake value ratio (SUVR) measurements with changes in MRI -based brain volume (in ml) measurements at 5, 28, 90 and 273 days as compared to baseline. We will calculate the change from day 0 to each of the other time points for each of the measures, and we will estimate the correlation between the changes using Pearson's correlation coefficient.

Measure: Relationship between changes in Microglial activity and MRI-based brain atrophy changes

Time: Baseline to 9 months

Description: Association of changes in regional microglial activity in the brain using standardized uptake value ratio (SUVR) measurements with changes in disease severity (measured using expanded disability status scale or EDSS) at 5, 28, 90 and 273 days as compared to baseline. We will calculate the change from day 0 to each of the other time points for each of the measures, and we will estimate the correlation between the changes using Pearson's correlation coefficient.

Measure: Relationship between changes in Microglial activity and physical disability

Time: Baseline to 9 months

Description: Association of changes in regional microglial activity in the brain using standardized uptake value ratio (SUVR) measurements with changes in severity of cognitive impairment (measured using symbol digit modality test or SDMT) at 5, 28, 90 and 273 days as compared to baseline. We will calculate the change from day 0 to each of the other time points for each of the measures, and we will estimate the correlation between the changes using Pearson's correlation coefficient.

Measure: Relationship between changes in Microglial activity and cognitive disability

Time: Baseline to 9 months


HPO Nodes


HP:0012378: Fatigue
Genes 398
PFN1 SOX3 ATRX VHL LBR BIRC3 BLNK GNAS CD79A SH2B3 STAT5B EPCAM NF1 MEFV IL12A-AS1 CTLA4 MLX PON1 PNPLA8 RPL5 RUNX1 KCNQ1 RRM2B GPR35 ELANE PPARGC1A IGH GLA DBH CCR1 MLH3 XPC MSH2 CCND1 KLRC4 ATP7A KCNQ3 PSTPIP1 PALLD HLA-B TCF4 CDKN2A RUNX1 BCOR ALB AGK GBA NEK1 CFI IRF2BP2 ALAS2 MRAP OPTN MYD88 CAV3 NKX2-1 BCL2 CCDC78 PALB2 C9ORF72 UNC13A DNAJC6 SDHA SMAD4 TBX19 C4A ATM PDE8B RPS28 NUMA1 SCN8A MMACHC NFKB2 PRKAR1A MST1 HBA1 RPL11 SOX3 SLC25A11 SCNN1A ERBB4 UBQLN2 SLC18A3 PODXL HFE SQSTM1 ARMC5 HLA-DPB1 PRRT2 MET NPM1 SLC26A4 NAGS FAS CCNF TSHR STAR SDHD IL12A SPIB PREPL RET POU1F1 WIPF1 DLST LHX4 TSC1 HNF1A TET2 DCTN1 STEAP3 FOXE1 TREM2 NABP1 FGFR1 PYGM HESX1 WAS SLC5A5 INSR NEFH KCNN4 IL23R SOD1 HESX1 ERAP1 RPS27 RPS7 FOXP1 STAT6 CALR CFH DMPK DUOX2 BRCA1 VCP TARDBP GCK DYSF TBL1XR1 CCND1 MSH6 PMS1 COL1A2 ACADM BCR SDHB IGLL1 MEN1 EPOR PRTN3 UNC93B1 MDH2 DNMT3A HLA-B BRCA2 JAK2 HMGCL DDB2 RPS17 CHMP2B CFAP410 IL12RB1 PAX8 LHX4 CPT1A ERCC2 RPL35A MMEL1 RPS20 PRKACA COL5A2 RET RPL31 PYGL RPL15 IRF5 NKX2-5 HLA-DRB1 EPHA4 ABL1 ERCC4 RPL27 DNM1L SLC25A4 SMAD3 POU1F1 HAVCR2 OPA1 PLEC SLC26A4 JAK2 KCNE1 CBL SDHB KIT ATP13A2 RPS24 PGM1 DAO GATA1 NAB2 SDHB SDHAF2 VHL DMD PDGFRA PMS2 ALB ERCC5 SLC11A1 PON2 CPT2 IGHM RPS26 PROP1 PML PIGT AP2S1 SCNN1B HNRNPA1 SLC18A2 PROP1 TSHR POMGNT1 COQ2 RPL26 RPS29 SDHC MALT1 SDHD HESX1 FUS SDHC FH SCN2A GLE1 C1QBP GPR101 LHX3 MATR3 RPS10 CPT2 PON3 CHRND BCL10 BCL6 ZBTB16 AIP GLT8D1 TAZ TGFBR2 KRAS SERPINA6 HLA-B TWNK IKZF1 UBAC2 ABCC2 RARA MORC2 MYH7 ANXA11 POLG EPAS1 CDH23 SLC2A10 PTPN22 TFR2 SEMA4A POLG2 GCH1 PROKR2 KCNQ2 TLR3 TAF15 TRAF3 FIP1L1 ASXL1 CDC73 SOX2 TNPO3 ATXN2 TXNRD2 TRNK HLA-DPA1 LRRC8A MPL TSC2 DUOXA2 TLR4 CTNNB1 PIK3CA NLRP3 FAN1 PIGA IGH TWNK KIT TICAM1 PROP1 RPS15A NR3C1 TPO XPA PRKAR1A BMPR1A ERCC3 TET2 KIF23 TBK1 TSHB MC2R MEN1 ATP13A2 TK2 CDC73 IL10 IL12B TET2 RPL18 STAT4 MMADHC POU2AF1 FIG4 STAT3 ARMC5 VPS13A MPL PRPH TSR2 NNT HNF4A TP53 CDH23 COL5A1 BTNL2 GLI2 PTPN22 PAX8 FOXA2 CHCHD10 PTPN3 TMEM127 CD79B GATA2 IL12A MLH1 TBK1 ADA2 SRSF2 SLC12A3 IGH KRAS ARNT2 IYD TG SLC3A1 PHKG2 SMAD3 PDE11A COL1A1 ANG VAPB SLC40A1 NLRP3 OTX2 NR3C1 RPL35 HELLPAR MAX KIF1B FGF23 TCF3 PIEZO1 PIK3R1 AIP PHKA2 SDHA OTX2 HLA-DRB1 SLC25A26 TNXB CDH23 RPS19 CD46 BTK SLC4A1 SYNJ1 FTL TNFSF15 SCNN1G VHL USP8 TET2 TET2 JAK2 HBA2
Protein Mutations 3
T25W V158M V18M
SNP 0