NE3107 Activity and Safety in Patients With Parkinson's Disease Using Levodopa

Sponsor
BioVie Inc. (Industry)
Overall Status
Recruiting
CT.gov ID
NCT05083260
Collaborator
(none)
40
15
2
18.9
2.7
0.1

Study Details

Study Description

Brief Summary

A 28-day phase 2a, double-blind, placebo-controlled (1:1), multi-center study of 20 mg NE3107, twice daily of safety, potential drug-drug interactions, and MDS-UPDRS defined activity in patients with Parkinson's disease . Study will enroll 40 patients that are currently taking immediate release levodopa/ carbidopa (IRLC) and have a practically defined early morning off-state for IRLC. Day one- baseline UPDRS and IRLC PK sampling; day 2- start NE3107 dosing, assess UPDRS during onset and NE3107 PK sampling, rescue meds as needed after 4 hours; day 3 and 14- NE3107 + IRLC UPDRS assessment and PK sampling; day 28- NE3107 + IRLC UPDRS assessments. Optional overnight stays in clinic prior to Day 1-3, 14, and 28.

Condition or Disease Intervention/Treatment Phase
Phase 1/Phase 2

Study Design

Study Type:
Interventional
Anticipated Enrollment :
40 participants
Allocation:
Randomized
Intervention Model:
Parallel Assignment
Masking:
Quadruple (Participant, Care Provider, Investigator, Outcomes Assessor)
Primary Purpose:
Treatment
Official Title:
A Double-Blind, Placebo-Controlled, Safety, Tolerability, and Pharmacokinetics Study in Parkinson's Disease (PD) Participants Treated With Carbidopa/Levodopa and NE3107
Actual Study Start Date :
Jan 4, 2021
Anticipated Primary Completion Date :
Aug 1, 2022
Anticipated Study Completion Date :
Aug 1, 2022

Arms and Interventions

Arm Intervention/Treatment
Experimental: NE3107

orally administered NE3107 20 mg twice daily (BID)

Drug: NE3107
NE3107 is an investigational orally bioavailable, blood-brain barrier permeable anti-inflammatory agent with a new mechanism of action targeting multiple mechanisms of pathology in Parkinson's disease

Placebo Comparator: placebo

orally administered placebo, twice daily

Drug: placebo
Hard gelatin capsule containing only common excipients for oral formulations

Outcome Measures

Primary Outcome Measures

  1. Change from baseline (day 1) in Motor disease society- Unified Parkinson's disease rating scale Part III total score for the patient in the "off-state" (without L-dopa for previous eight hours) [measured on day 1, 2, 3, 14, and 28]

    Unified Parkinson's disease rating scale Part III total score is the cumulative score of a 33 question assessment of disease impact on patient's ability to move, with each part scored on a scale of 0-4, with 0 being no effect of disease and 4 being severe disease. a total score of 0 indicates no disease and a total score of 132 indicates the most severe disease

  2. Change from baseline in the length of time during which L-dopa therapy is ineffective in reducing motor symptoms of disease (OFF time) [off time will be measured/recorded every day from day 2 to day 27]

    The total sum of time in which a patient self categorizes themselves as in the "off" state determined by diary entries. 0 hours is the minimum/best score, which indicates continuous L-dopa-like activity for the 24 hour period. 24 hours is the worst score and indicates that there was no L-dopa-like benefit at any time during the 24 hour period.

  3. Change from baseline in average Motor disease society- Unified Parkinson's disease rating scale Part III total score measured over the course of 8 hours after taking L-dopa and/or NE3107 [measured on day 1, 2, 3, 14, and 28]

    An average score will be calculated from assessments of the Unified Parkinson's disease rating scale Part III total score, which is the cumulative score of a 33 question assessment of disease impact on patient's ability to move, with each question scored on a scale of 0-4, with 0 being no effect of disease and 4 being severe disease. a total score of 0 indicates no disease and a total score of 132 indicates the most severe disease. Scores will be collected several times during the eight hours following L-dopa and/or NE3107 administration.

  4. Motor disease society- Unified Parkinson's disease rating scale Part I total score [measured on day 1, 2, 3, 14, and 28]

    the cumulative score of a 13 question assessment of disease impact on patient's cognitive impairment, with each question scored on a scale of 0-4, with 0 being no effect of disease and 4 being severe disease. a total score of 0 indicates no disease and a total score of 42 indicates the most severe disease. This test asks about the impact of disease on cognition, but does not directly measure cognitive capability.

  5. Motor disease society- Unified Parkinson's disease rating scale Part 2 total score [measured on day 1, 2, 3, 14, and 28]

    the cumulative score of a 13 question assessment of disease impact on patient's Motor Aspects of Experiences of Daily Living, with each question scored on a scale of 0-4, with 0 being no effect of disease and 4 being a severe effect. a total score of 0 indicates no disease and a total score of 42 indicates the most severe disease.

  6. Change from baseline in the length of time during which L-dopa-like effects are felt by the patient [baseline and day 1, 2, 3, 14, and 28]

    The total sum of time in which a patient self categorizes themselves as in the "on" state, with or without dyskinesia determined by diary entries. 0 hours is the minimum/worst score, which indicates no L-dopa-like activity for the 24 hour period. 24 hours is the best score and indicates that there was no L-dopa-like benefit at any time during the 24 hour period.

  7. change from baseline in L-dopa induced dyskinesia measured with the abnormal involuntary movement scale (AIMS) [AIMS will be measured during the 8 hour period of observation on Day 1, 2, 3, 14, and 28]

    AIMS is a 12-item clinician-rated scale to assess severity of dyskinesias (orofacial movements and extremity and truncal movements) in patients taking L-dopa. Questions assess the overall severity, incapacitation, and the patient's level of awareness of the movements, and distress associated with them. Questions are scored from 0 (no symptom) to 4 (severe). the minimum total score of 0 indicates no dyskinesia and the maximum total score of 48 indicates severe dyskinesia.

  8. change from baseline in time to onset of L-dopa-like activity [measured on Day 1, 2, 3, 14, and 28]

    Length of time from L-dopa administration to beginning of L-dopa activity producing "on-state". Patient self assessment of the time L-dopa-like activity begins after L-dopa administration. A decrease in the time to onset may shorten the time in the off-state and be considered a patient benefit. An increase in time to onset may lengthen time in the off-state and be considered a general worsening of response to treatment.

  9. Change from baseline in Non-Motor Symptom Assessment Scale for Parkinson Disease (NMSS) [Measured on Day 1, 2, 3, 14, and 28]

    The Non-Motor Symptoms Scale (NMSS) is a 30-item rater-based scale to assess a wide range of non-motor symptoms in patients with Parkinson's disease (PD). The NMSS measures the severity and frequency of non-motor symptoms across nine dimensions. The scale can be used for patients at all stages of PD. For each question, scores of severity (0 to 3, with 0 being none and 3 being major source of distress) and frequency (1 to 4, 1= rarely and 4 = very frequent) are recorded and the product of the two calculated. The sum of the products yields the overall score. The minimum total score of 0 indicates no non-motor symptoms and the maximum score of 360 indicates severe disease.

  10. change in area under the levodopa plasma concentration vs. time curve when administered alone compared to being co-administered with NE3107 [Blood samples will be collected on Day 1, 2, 3, and 14]

    the plasma concentration vs time curve for L-dopa will be calculated from the L-dopa concentration in plasma samples collected prior to dosing and at 0.5, 1, 2, 3, 4, and 8 hours after L-dopa administration on days on which NE3107 is not administered (Day 1) and three days on which NE3107 is co-administered (Day 2, 3, and 14). Changes in L-dopa area under the curve could be associated with changes in activity against motor symptoms of disease.

  11. Change in the maximum plasma concentration (Cmax) of levodopa when administered alone compared to being co-administered with NE3107. [Blood samples will be collected on Day 1, 2, 3, and 14]

    the maximum plasma concentration for L-dopa will be determined in plasma samples collected prior to dosing and at 0.5, 1, 2, 3, 4, and 8 hours after L-dopa administration on days on which NE3107 is not administered (Day 1) and three days on which NE3107 is co-administered (Day 2, 3, and 14). Changes in L-dopa Cmax could be associated with changes in activity against motor symptoms of disease.

  12. The area under the NE3107 plasma concentration vs. time curve when administered alone compared to being co-administered with [Blood samples will be collected on Day 2, 3, and 14]

    the plasma concentration vs time curve for NE3107 will be calculated from the NE3107 concentration in plasma samples collected prior to dosing and at 0.5, 1, 2, 3, 4, and 8 hours

  13. Change in the maximum plasma concentration (Cmax) of NE3107 when administered alone compared to being co-administered with L-dopa [Blood samples will be collected on Day 2, 3, and 14]

    the maximum plasma concentration for NE3107 will be determined in plasma samples collected prior to dosing and at 0.5, 1, 2, 3, 4, and 8 hours after NE3107 administration

Eligibility Criteria

Criteria

Ages Eligible for Study:
30 Years to 80 Years
Sexes Eligible for Study:
All
Accepts Healthy Volunteers:
No
Inclusion Criteria:
  1. Men or women at least 30 and no more 80 years of age

  2. Diagnosis of PD consistent with UK Brain Bank Criteria or MDS Research Criteria for the Diagnosis of PD, with bradykinesia and a clear motor response to levodopa

  3. Stable doses of all PD medications for at least 4 weeks prior to Screening

  4. Carbidopa/levodopa dose of at least 300 mg daily, distributed over a minimum of 3 dosing intervals during waking hours

  5. Participants must have history of motor fluctuations with reliable early-morning OFF episodes and a history of a good response to levodopa, in the judgement of the investigator

  6. If of reproductive potential, willing and able to use a highly effective form of birth control during the study and for 30 days following last dose of study drug. Examples of highly effective forms of birth control are:

  7. Surgical sterility (via vasectomy, hysterectomy, or bilateral tubal ligation) or postmenopausal status in females

  8. Sexual partner who is sterile or of the same sex

  9. Double-barrier method (any combination of physical and chemical methods)

  10. Intrauterine device in females not containing hormones.

  11. Able and willing to comply with study drug administration, scheduled visits, treatment plan, laboratory tests, and other study-related procedures to complete the study

  12. Investigational Review Board/Ethics Committee-approved consent form signed and date by the participant

  13. Assessed as an appropriate and suitable candidate by the Enrollment Authorization Committee (EAC)

Exclusion Criteria:
  1. Diagnosis of secondary or atypical parkinsonism

  2. Severe or disabling fluctuations or dyskinesias that would, in the opinion of the investigator, interfere with completion of the study

  3. Clinically significant cognitive impairment

  4. Clinically significant hallucinations or delusions

  5. Clinically significant orthostatic hypotension

  6. Currently active major depression as determined by BDI-II score of >19

  7. Previous surgical procedure for PD (Duopa, DBS, etc.)

  8. History of small bowel or gastric surgery

  9. History of clinically significant GI abnormality (inflammatory bowel disease, significant motility disorder or emesis of any cause, etc.)

  10. Use of long-acting levodopa formulations (Sinemet CR, ER, Rytary, etc.)

  11. Routine use of proton pump inhibitors or H2 blockers

  12. Routine use of medications that may influence gastric motility (opiates, TCA antidepressants, anticholinergics, etc.)

  13. Other clinically significant medical, surgical, psychiatric, or laboratory abnormality that, in the judgment of the investigator, is likely to interfere with study compliance or assessment of safety or efficacy

  14. Alanine aminotransferase (ALT) or aspartate aminotransferase (AST) levels greater than 2.5 times the upper limit of normal (ULN)

  15. Significant renal impairment as determined by creatinine clearance (CrCL) less than or equal to 50 mL/min

  16. Participant has an ECG or clinical evidence of potentially unstable heart disease, including the following:

  17. QTcF > 470 msec females; > 450 msec males

  18. Complete right or left bundle branch block

  19. Ischemia or myocardial infarct within 1 year prior to the Screening Visit

  20. Clinically significant atrial or ventricular dysrhythmia; the heart must be in predominantly normal sinus rhythm

  21. Second- or third-degree AV block

  22. Heart failure of NYHA classification III or greater

  23. Serious cardiomyopathy or cardiac structural abnormality

  24. Symptomatic coronary artery or ischemic cardiac disease

  25. Any other cardiac condition that the Investigator feels may predispose the participant to ischemia or arrhythmia.

  26. Current (or within past 12 months) diagnosis or history of substance abuse, including alcohol (excluding nicotine or caffeine) by Diagnostic and Statistical Manual of Mental Disorders 5 criteria, or positive urine drug screen for tetrahydrocannabinol or any drugs that may affect participant safety or interfere with efficacy assessments

  27. Medical or recreational use of marijuana or CBD within 3 months of the Screening Visit

  28. Active suicidal ideation within 1 year prior to Screening Visit as determined by a positive response to Question 4 or 5 on the C-SSRS

  29. Currently lactating or pregnant, or planning to become pregnant during the study

  30. Current participation in another investigational clinical study and/or receipt of any investigational drug within 90 days prior to screening

  31. Prior randomization into this study

  32. Diabetes requiring insulin treatment

Contacts and Locations

Locations

Site City State Country Postal Code
1 Rocky Mountain Movement Disorders Center Englewood Colorado United States 80113
2 Parkinson's Disease & Movement Disorders Center Of Boca Raton Boca Raton Florida United States 33486
3 Velocity Hallandale Beach Florida United States 33009
4 Charter Research Lady Lake Florida United States 32159
5 First Excellent Research Group Miami Florida United States 33172
6 First Excellent Research Miami Florida United States 33172
7 EZY Medical Research Miami Florida United States 33175
8 Charter Research Winter Park Florida United States 32792
9 Quest Research Institute Farmington Hills Michigan United States 48334
10 Duke University Durham North Carolina United States 27705
11 M3 Wake Raleigh North Carolina United States 27612
12 Wake Research Raleigh North Carolina United States 27612
13 University of Toledo Toledo Ohio United States 43614
14 Texas Institute for Neurological Disorders Sherman Texas United States 75092
15 Inland Northwest Research Spokane Washington United States 99202

Sponsors and Collaborators

  • BioVie Inc.

Investigators

None specified.

Study Documents (Full-Text)

None provided.

More Information

Publications

None provided.
Responsible Party:
BioVie Inc.
ClinicalTrials.gov Identifier:
NCT05083260
Other Study ID Numbers:
  • NM201
First Posted:
Oct 19, 2021
Last Update Posted:
Aug 2, 2022
Last Verified:
Jul 1, 2022
Studies a U.S. FDA-regulated Drug Product:
Yes
Studies a U.S. FDA-regulated Device Product:
No
Keywords provided by BioVie Inc.
Additional relevant MeSH terms:

Study Results

No Results Posted as of Aug 2, 2022