search
Back to results

Mexiletine in Sporadic Amyotrophic Lateral Sclerosis (Mexiletine-2)

Primary Purpose

Sporadic Amyotrophic Lateral Sclerosis

Status
Completed
Phase
Phase 2
Locations
United States
Study Type
Interventional
Intervention
Mexiletine
Placebo
Sponsored by
University of Washington
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Sporadic Amyotrophic Lateral Sclerosis focused on measuring SALS, Mexiletine, TMS, NCS

Eligibility Criteria

18 Years - undefined (Adult, Older Adult)All SexesDoes not accept healthy volunteers

Inclusion Criteria:

  1. Sporadic ALS diagnosed as possible, laboratory-supported probable, probable, or definite ALS as defined by revised El Escorial criteria.
  2. Age 18 years or older.
  3. Symptom onset of weakness or spasticity due to ALS ≤ 60 months prior to Screening Visit.
  4. Slow vital capacity (SVC) measure ≥50% of predicted for gender, height, and age at the screening visit.
  5. Must be able to swallow capsules throughout the course of the study, according to Site Investigator judgment.
  6. Capable of providing informed consent and following trial procedures.
  7. For TMS: a resting motor threshold defined as 50% of pulses eliciting a motor evoked potential (MEP) of amplitude ≥ 50 µV.
  8. For TTNCS: median Compound Muscle Action Potential (CMAP) ≥ 1.5 mV.
  9. Subjects must not have taken riluzole for at least 30 days or be on a stable dose of riluzole for at least 30 days prior to the Screening Visit and continue on the stable dose throughout the course of the study (riluzole-naïve subjects are permitted in the study).
  10. Subjects must not have taken medication for muscle cramping such as cyclobenzaprine, baclofen, carisoprodol, or methocarbamol, for at least 30 days prior to screening or be on a stable dose for at least 60 days prior to screening.
  11. Geographic accessibility to the site.
  12. Women must not become pregnant for the duration of the study and must be willing to use two contraceptive therapies and have a negative pregnancy test throughout the course of the study.
  13. Use of medications known to affect the neurophysiology measures in the study must be scheduled, not as needed (pro re nata, PRN). A subject must have been on a fixed dose for 30 days prior to the Screening Visit, and there must be no reason to believe that a subsequent change would be necessary during the course of the study. These medications include: benzodiazepines, muscle relaxants, tricyclic antidepressants, selective serotonin reuptake inhibitors, non-selective serotonin reuptake inhibitors, hypnotics (including anti-histamines) and anti-cholinergics.

Exclusion Criteria:

  1. Invasive ventilator dependence, such as tracheostomy.
  2. Creatinine level greater than 1.5 mg/dL at screening.
  3. Serum Glutamic-Oxaloacetic (SGOT/AST) / Serum Glutamic-Pyruvic (SGPT/ALT) greater than 3 times the upper limit of normal at screening.
  4. History of known sensitivity or intolerability to mexiletine or lidocaine.
  5. Any history of either substance abuse within the past year, unstable psychiatric disease, cognitive impairment, or dementia.
  6. Clinically significant conduction abnormalities on electrocardiogram or a known history of cardiac arrhythmia.
  7. Known history of epilepsy.
  8. Known history of congestive heart failure (CHF) or history of myocardial infarction within the past 24 months.
  9. Use of mexiletine for 30 days prior to Screening Visit.
  10. Exposure to any other experimental agent (off-label use or investigational) including high dose creatine (>10 grams a day) within 30 days prior to Screening Visit.
  11. Metal in the head and neck region, cardiac pacemaker or brain stimulator, cochlear implants, implanted infusion device or personal history of epilepsy.
  12. Use of amiodarone, flecainide, duloxetine, tizanidine, or clozapine.
  13. Pregnant women or women currently breastfeeding.
  14. Placement of Diaphragm Pacing System (DPS) device < 60 days prior to Screening Visit.
  15. Planned DPS device implantation during study participation

Sites / Locations

  • Barrow Neurological Institute
  • University of California, Irvine
  • Augusta University
  • Beth Israel Deaconess Medical Center
  • University of Michigan
  • Columbia Universtiy Medical Center
  • Pennsylvania State Hershey Medical Center
  • University of Pittsburgh
  • Medical University of South Carolina
  • University of Washington

Arms of the Study

Arm 1

Arm 2

Arm 3

Arm Type

Active Comparator

Active Comparator

Placebo Comparator

Arm Label

Mexiletine, 300 milligrams

Mexiletine, 600 milligrams

Placebo

Arm Description

Mexiletine, 300 milligrams by mouth per day for 4 weeks.

Mexiletine, 600 milligrams by mouth per day for 4 weeks.

Placebo, by mouth per day for 4 weeks.

Outcomes

Primary Outcome Measures

Change in Resting Motor Threshold
The resting motor threshold (RMT) assessed from single pulse transcranial magnetic stimulation (TMS) measurements made before treatment, after 4 weeks of treatment, and then again after a 4 week washout, was used as the primary pharmacodynamic marker of cortical hyperexcitability. RMT is the stimulus intensity required to produce and maintain a 0.2 mV peak-to-peak motor evoked potential of the abductor pollicis brevis muscle by TMS. A smaller RMT is thought to suggest greater neuronal excitability.

Secondary Outcome Measures

Effect on Short-interval Intracortical Inhibition
Short-interval intracortical inhibition (SICI) is a measure of neuronal excitability measured by dual pulse TMS with a conditioned (80% of RMT) and test pulses (120% of RMT) to generate a stable MEP amplitude of 0.2 mV, averaged over interstimulus intervals of 1-7 ms. It is thought to reflect refractory cortical axons and subsequent resynchronization of cortico-cortical and corticomotoneuronal volleys or activation of non-GABAergic cortical inhibitory circuits (initial phase) and synaptic neurotransmission through GABAA receptors (second phase). The value for SICI thought to be maximally sensitive for detecting in changes in ALS subjects compared to controls is is derived by measuring the motor evoked potential amplitude (MEP) at an interstimulus interval of 3 ms (ISI 3 ms) and normalizing to the MEP amplitude at 120% of the resting motor threshold (MEP 120% RMT). A reduction in SICI reflecting greater excitability would generate a larger ratio of MEP ISI 3 ms/MEP 120% RMT.
Change in Motor Evoked Potential Amplitude
The motor evoked potential (MEP) amplitude is taken from single pulse transcranial magnetic stimulation (TMS) and reflects the density of corticomotoneuronal projections onto motor neurons and is affected by cortical hyperexcitability early in ALS where it is thought to be larger than age-matched controls and axonal degeneration later in the disease when it decreases in amplitude. The MEP is most reliable in assessing cortical motor neuronal preservation and excitability when normalized to the peak compound nerve action potential (CMAP) amplitude which reflects the integrity of peripheral motor nerve axons. It is also normalized here to 120% of the RMT to derive a ratio of MEP at 120% RMT/peak CMAP.
Effect on Cortical Silent Period
The cortical silent period (CSP) is recorded with single pulse TMS as a duration from the onset of the MEP response to resumption of voluntary electromyography activity with the patient performing a voluntary contraction, set to 30% of maximal voluntary contraction. A shorter CSP compared to controls would reflect greater excitability.
Effect on Strength Duration Time Constant
The strength duration time constant (SDTC) is used in threshold tracking nerve axonal excitability studies and is interpreted as a measure of axonal excitability that is dependent upon the biophysical properties of the axonal membrane at the node of Ranvier, especially persistent sodium current. It is derived from the relationship between stimulus duration and intensity. A higher SDTC would reflect greater excitability of motor nerve axons.
Effect on Depolarizing Threshold Electrotonus (90-100 ms)
Depolarizing threshold electrotonus (90-100 ms) (TEd 90-100 ms) is used in threshold tracking nerve axonal excitability studies in which long-lasting subthreshold depolarizing currents are generated, measured at 90-100 ms following the stimulus. This measure is associated with a decrease in the membrane excitability threshold due to opening of potassium channels on the axonal membrane. Intrinsic changes in axonal excitability properties, such as thought to occur in ALS, could possibly alter this measure, presumably by decreasing TEd 90-100 ms more substantially than normal.
Effect on Hyperpolarizing Threshold Electrotonus (90-100 ms)
Hyperpolarizing threshold electrotonus (90-100 ms) (TEh 90-100 ms) is used in threshold tracking nerve axonal excitability studies in which long-lasting subthreshold hyperpolarizing currents are generated, measured at 90-100 ms following the stimulus. This measure is associated with an increase in the membrane excitability threshold due to closure of potassium channels causing increased resistance of the internodal axonal membrane. Intrinsic changes in axonal excitability properties, such as thought to occur in ALS, could possibly alter this measure.
Effect on Superexcitability
Superexcitability is a component of recovery cycle analysis assessing motor axonal excitability, employing threshold tracking nerve conduction study. It is a depolarizing afterpotential measured following a single supramaximal stimulus followed by a second smaller stimulus of variable intensity and reflects passive depolarization of the internodal axon.
Effect on Subexcitability
Subexcitability is a component of recovery cycle analysis assessing motor axonal excitability, employing threshold tracking nerve conduction study. It is a late hyperpolarizing after potential measured following a single supramaximal stimulus followed by a second smaller stimulus of variable intensity and is related to the very slow turn-off of slow potassium channels.
Effect on Frequency of Muscle Cramps
Will be assessed using a daily muscle cramps diary tabulated weekly beginning at Baseline.
Effect on Frequency of Fasciculations (Muscle Twitching)
Will be assessed using a daily fasciculations diary tabulated as a percentage of days from weeks 3-4.

Full Information

First Posted
May 10, 2016
Last Updated
November 17, 2019
Sponsor
University of Washington
Collaborators
Massachusetts General Hospital
search

1. Study Identification

Unique Protocol Identification Number
NCT02781454
Brief Title
Mexiletine in Sporadic Amyotrophic Lateral Sclerosis
Acronym
Mexiletine-2
Official Title
Effect of Mexiletine on Cortical Hyperexcitability in Sporadic Amyotrophic Lateral Sclerosis (SALS)
Study Type
Interventional

2. Study Status

Record Verification Date
November 2019
Overall Recruitment Status
Completed
Study Start Date
October 2016 (Actual)
Primary Completion Date
September 30, 2018 (Actual)
Study Completion Date
September 30, 2018 (Actual)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Principal Investigator
Name of the Sponsor
University of Washington
Collaborators
Massachusetts General Hospital

4. Oversight

Data Monitoring Committee
No

5. Study Description

Brief Summary
The purpose of this research study is to find out whether the drug mexiletine will be effective in lowering motor neuron electrical activity in the brains and nerves in the arms of people with ALS. The investigators will also determine if there are any signs that the drug may slow down the progression of ALS and reduce muscle cramps and muscle twitching. This will be determined through transcranial magnetic stimulation (TMS) and threshold tracking nerve conduction studies (TTNCS). In this trial, the participants will be taking either 300mg/day of mexiletine, 600mg/day of mexiletine, or placebo (non-active study drug).
Detailed Description
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder affecting primarily motor neurons, for which treatment designed to slow or arrest progression remains lacking. Mexiletine is a use-dependent sodium channel blocker that has been FDA-approved for decades for the treatment of cardiac arrhythmias and more recently to treat neuropathic pain in diabetic polyneuropathy. Mexiletine has been shown also to be protective of neurons following spinal cord, head injury, and cerebral ischemia, largely by blocking excitotoxicity. Based on previous studies, mexiletine appears to penetrate into the central nervous system at concentrations sufficient to confer significant protection. Recent unpublished studies in the laboratory of Dr. Robert Brown at the University of Massachusetts have also demonstrated that mexiletine ingestion in mice genetically engineered to express high levels of mutant cytosolic copper-zinc superoxide dismutase-1 (SOD1) transgene prolongs survival in these animals. As mexiletine already has FDA-approval as an anti-arrhythmic agent, much is known about the pharmacology and safety of this drug in non-ALS patients. We anticipate that by excluding subjects with a known history of cardiac disease and with the known neuroprotectant properties of this medication, mexiletine is a good choice for further study in an ALS clinical trial.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Sporadic Amyotrophic Lateral Sclerosis
Keywords
SALS, Mexiletine, TMS, NCS

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 2
Interventional Study Model
Parallel Assignment
Masking
ParticipantCare ProviderInvestigatorOutcomes Assessor
Allocation
Randomized
Enrollment
20 (Actual)

8. Arms, Groups, and Interventions

Arm Title
Mexiletine, 300 milligrams
Arm Type
Active Comparator
Arm Description
Mexiletine, 300 milligrams by mouth per day for 4 weeks.
Arm Title
Mexiletine, 600 milligrams
Arm Type
Active Comparator
Arm Description
Mexiletine, 600 milligrams by mouth per day for 4 weeks.
Arm Title
Placebo
Arm Type
Placebo Comparator
Arm Description
Placebo, by mouth per day for 4 weeks.
Intervention Type
Drug
Intervention Name(s)
Mexiletine
Other Intervention Name(s)
Mexitil
Intervention Type
Drug
Intervention Name(s)
Placebo
Primary Outcome Measure Information:
Title
Change in Resting Motor Threshold
Description
The resting motor threshold (RMT) assessed from single pulse transcranial magnetic stimulation (TMS) measurements made before treatment, after 4 weeks of treatment, and then again after a 4 week washout, was used as the primary pharmacodynamic marker of cortical hyperexcitability. RMT is the stimulus intensity required to produce and maintain a 0.2 mV peak-to-peak motor evoked potential of the abductor pollicis brevis muscle by TMS. A smaller RMT is thought to suggest greater neuronal excitability.
Time Frame
Accessed at Screening, Baseline, Week 4, and Week 8; change from Baseline to Week 4 and from Week 4 to Week 8 reported
Secondary Outcome Measure Information:
Title
Effect on Short-interval Intracortical Inhibition
Description
Short-interval intracortical inhibition (SICI) is a measure of neuronal excitability measured by dual pulse TMS with a conditioned (80% of RMT) and test pulses (120% of RMT) to generate a stable MEP amplitude of 0.2 mV, averaged over interstimulus intervals of 1-7 ms. It is thought to reflect refractory cortical axons and subsequent resynchronization of cortico-cortical and corticomotoneuronal volleys or activation of non-GABAergic cortical inhibitory circuits (initial phase) and synaptic neurotransmission through GABAA receptors (second phase). The value for SICI thought to be maximally sensitive for detecting in changes in ALS subjects compared to controls is is derived by measuring the motor evoked potential amplitude (MEP) at an interstimulus interval of 3 ms (ISI 3 ms) and normalizing to the MEP amplitude at 120% of the resting motor threshold (MEP 120% RMT). A reduction in SICI reflecting greater excitability would generate a larger ratio of MEP ISI 3 ms/MEP 120% RMT.
Time Frame
Accessed at Screening, Baseline, Week 4, and Week 8; change from Baseline to Week 4 reported
Title
Change in Motor Evoked Potential Amplitude
Description
The motor evoked potential (MEP) amplitude is taken from single pulse transcranial magnetic stimulation (TMS) and reflects the density of corticomotoneuronal projections onto motor neurons and is affected by cortical hyperexcitability early in ALS where it is thought to be larger than age-matched controls and axonal degeneration later in the disease when it decreases in amplitude. The MEP is most reliable in assessing cortical motor neuronal preservation and excitability when normalized to the peak compound nerve action potential (CMAP) amplitude which reflects the integrity of peripheral motor nerve axons. It is also normalized here to 120% of the RMT to derive a ratio of MEP at 120% RMT/peak CMAP.
Time Frame
Accessed at Screening, Baseline, Week 4, and Week 8; change from Baseline to Week 4 reported
Title
Effect on Cortical Silent Period
Description
The cortical silent period (CSP) is recorded with single pulse TMS as a duration from the onset of the MEP response to resumption of voluntary electromyography activity with the patient performing a voluntary contraction, set to 30% of maximal voluntary contraction. A shorter CSP compared to controls would reflect greater excitability.
Time Frame
Accessed at Screening, Baseline, Week 4, and Week 8; change from Baseline to Week 4 reported
Title
Effect on Strength Duration Time Constant
Description
The strength duration time constant (SDTC) is used in threshold tracking nerve axonal excitability studies and is interpreted as a measure of axonal excitability that is dependent upon the biophysical properties of the axonal membrane at the node of Ranvier, especially persistent sodium current. It is derived from the relationship between stimulus duration and intensity. A higher SDTC would reflect greater excitability of motor nerve axons.
Time Frame
Accessed at Screening, Baseline, Week 4, and Week 8; change from Baseline to Week 4 reported
Title
Effect on Depolarizing Threshold Electrotonus (90-100 ms)
Description
Depolarizing threshold electrotonus (90-100 ms) (TEd 90-100 ms) is used in threshold tracking nerve axonal excitability studies in which long-lasting subthreshold depolarizing currents are generated, measured at 90-100 ms following the stimulus. This measure is associated with a decrease in the membrane excitability threshold due to opening of potassium channels on the axonal membrane. Intrinsic changes in axonal excitability properties, such as thought to occur in ALS, could possibly alter this measure, presumably by decreasing TEd 90-100 ms more substantially than normal.
Time Frame
Accessed at Screening, Baseline, Week 4, and Week 8; change from Baseline to Week 4 reported
Title
Effect on Hyperpolarizing Threshold Electrotonus (90-100 ms)
Description
Hyperpolarizing threshold electrotonus (90-100 ms) (TEh 90-100 ms) is used in threshold tracking nerve axonal excitability studies in which long-lasting subthreshold hyperpolarizing currents are generated, measured at 90-100 ms following the stimulus. This measure is associated with an increase in the membrane excitability threshold due to closure of potassium channels causing increased resistance of the internodal axonal membrane. Intrinsic changes in axonal excitability properties, such as thought to occur in ALS, could possibly alter this measure.
Time Frame
Accessed at Screening, Baseline, Week 4, and Week 8; change from Baseline to Week 4 reported
Title
Effect on Superexcitability
Description
Superexcitability is a component of recovery cycle analysis assessing motor axonal excitability, employing threshold tracking nerve conduction study. It is a depolarizing afterpotential measured following a single supramaximal stimulus followed by a second smaller stimulus of variable intensity and reflects passive depolarization of the internodal axon.
Time Frame
Accessed at Screening, Baseline, Week 4, and Week 8; change from Baseline to Week 4 reported
Title
Effect on Subexcitability
Description
Subexcitability is a component of recovery cycle analysis assessing motor axonal excitability, employing threshold tracking nerve conduction study. It is a late hyperpolarizing after potential measured following a single supramaximal stimulus followed by a second smaller stimulus of variable intensity and is related to the very slow turn-off of slow potassium channels.
Time Frame
Accessed at Screening, Baseline, Week 4, and Week 8; change from Baseline to Week 4 reported
Title
Effect on Frequency of Muscle Cramps
Description
Will be assessed using a daily muscle cramps diary tabulated weekly beginning at Baseline.
Time Frame
Accessed at Screening, Baseline, Week 4, and Week 8; comparisons of treatments at Weeks 3-4 reported
Title
Effect on Frequency of Fasciculations (Muscle Twitching)
Description
Will be assessed using a daily fasciculations diary tabulated as a percentage of days from weeks 3-4.
Time Frame
Accessed at Screening, Baseline, Week 4, and Week 8; comparisons of treatments at Weeks 3-4 reported
Other Pre-specified Outcome Measures:
Title
Change in the Amyotrophic Lateral Sclerosis Functional Rating Scale - Revised
Description
The Amyotrophic Lateral Sclerosis Functional Rating Scale - Revised (ALSFRS-R) is an instrument for evaluating the functional status of patients with ALS that includes functions related to speech, swallowing, salivation, fine motor control, gross motor function, and respiration. The score is the sum of 12 items (range 0 to 48) with higher scores reflecting better function.
Time Frame
Accessed at Screening, Baseline, Week 4, and Week 8; change from Baseline to Week 4 reported
Title
Change in Slow Vital Capacity
Description
Measure of decline in respiratory muscle strength
Time Frame
Accessed at Screening, Baseline, Week 4, and Week 8; change from Baseline to Week 4 reported

10. Eligibility

Sex
All
Minimum Age & Unit of Time
18 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
Inclusion Criteria: Sporadic ALS diagnosed as possible, laboratory-supported probable, probable, or definite ALS as defined by revised El Escorial criteria. Age 18 years or older. Symptom onset of weakness or spasticity due to ALS ≤ 60 months prior to Screening Visit. Slow vital capacity (SVC) measure ≥50% of predicted for gender, height, and age at the screening visit. Must be able to swallow capsules throughout the course of the study, according to Site Investigator judgment. Capable of providing informed consent and following trial procedures. For TMS: a resting motor threshold defined as 50% of pulses eliciting a motor evoked potential (MEP) of amplitude ≥ 50 µV. For TTNCS: median Compound Muscle Action Potential (CMAP) ≥ 1.5 mV. Subjects must not have taken riluzole for at least 30 days or be on a stable dose of riluzole for at least 30 days prior to the Screening Visit and continue on the stable dose throughout the course of the study (riluzole-naïve subjects are permitted in the study). Subjects must not have taken medication for muscle cramping such as cyclobenzaprine, baclofen, carisoprodol, or methocarbamol, for at least 30 days prior to screening or be on a stable dose for at least 60 days prior to screening. Geographic accessibility to the site. Women must not become pregnant for the duration of the study and must be willing to use two contraceptive therapies and have a negative pregnancy test throughout the course of the study. Use of medications known to affect the neurophysiology measures in the study must be scheduled, not as needed (pro re nata, PRN). A subject must have been on a fixed dose for 30 days prior to the Screening Visit, and there must be no reason to believe that a subsequent change would be necessary during the course of the study. These medications include: benzodiazepines, muscle relaxants, tricyclic antidepressants, selective serotonin reuptake inhibitors, non-selective serotonin reuptake inhibitors, hypnotics (including anti-histamines) and anti-cholinergics. Exclusion Criteria: Invasive ventilator dependence, such as tracheostomy. Creatinine level greater than 1.5 mg/dL at screening. Serum Glutamic-Oxaloacetic (SGOT/AST) / Serum Glutamic-Pyruvic (SGPT/ALT) greater than 3 times the upper limit of normal at screening. History of known sensitivity or intolerability to mexiletine or lidocaine. Any history of either substance abuse within the past year, unstable psychiatric disease, cognitive impairment, or dementia. Clinically significant conduction abnormalities on electrocardiogram or a known history of cardiac arrhythmia. Known history of epilepsy. Known history of congestive heart failure (CHF) or history of myocardial infarction within the past 24 months. Use of mexiletine for 30 days prior to Screening Visit. Exposure to any other experimental agent (off-label use or investigational) including high dose creatine (>10 grams a day) within 30 days prior to Screening Visit. Metal in the head and neck region, cardiac pacemaker or brain stimulator, cochlear implants, implanted infusion device or personal history of epilepsy. Use of amiodarone, flecainide, duloxetine, tizanidine, or clozapine. Pregnant women or women currently breastfeeding. Placement of Diaphragm Pacing System (DPS) device < 60 days prior to Screening Visit. Planned DPS device implantation during study participation
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Michael Weiss, MD
Organizational Affiliation
University of Washington
Official's Role
Principal Investigator
Facility Information:
Facility Name
Barrow Neurological Institute
City
Phoenix
State/Province
Arizona
ZIP/Postal Code
85013
Country
United States
Facility Name
University of California, Irvine
City
Orange
State/Province
California
ZIP/Postal Code
92868
Country
United States
Facility Name
Augusta University
City
Augusta
State/Province
Georgia
ZIP/Postal Code
30912
Country
United States
Facility Name
Beth Israel Deaconess Medical Center
City
Boston
State/Province
Massachusetts
ZIP/Postal Code
02215
Country
United States
Facility Name
University of Michigan
City
Ann Arbor
State/Province
Michigan
ZIP/Postal Code
48109
Country
United States
Facility Name
Columbia Universtiy Medical Center
City
New York
State/Province
New York
ZIP/Postal Code
10032
Country
United States
Facility Name
Pennsylvania State Hershey Medical Center
City
Hershey
State/Province
Pennsylvania
ZIP/Postal Code
17033
Country
United States
Facility Name
University of Pittsburgh
City
Pittsburgh
State/Province
Pennsylvania
ZIP/Postal Code
15213
Country
United States
Facility Name
Medical University of South Carolina
City
Charleston
State/Province
South Carolina
ZIP/Postal Code
29425
Country
United States
Facility Name
University of Washington
City
Seattle
State/Province
Washington
ZIP/Postal Code
98195
Country
United States

12. IPD Sharing Statement

Plan to Share IPD
No
Links:
URL
http://www.alsconsortium.org/
Description
Northeast ALS Consortium Website

Learn more about this trial

Mexiletine in Sporadic Amyotrophic Lateral Sclerosis

We'll reach out to this number within 24 hrs